Skip to main content

Full text of "Vascular-Surgery-Books_1aim.net"

See other formats


ascular 
Embolotherapy 



MEDICAL RADIOLOGY 
Diagnostic Imaging 



Editors: 

A. L. Baert, Leuven 

K. Sartor, Heidelberg 



3HRMAP 



J. Golzarian • S. Sun • M. J. Sharafuddin (Eds.) 

Vascular 
Embolotherapy 

A Comprehensive Approach 

Volume 2 

Oncology, Trauma, Gene Therapy, 

Vascular Malformations, and Neck 



With Contributions by 

K. Ahrar ■ H. Alvarez ■ J. C. Chaloupka ■ M. D.Darcy ■ J. Dubois ■ J. R. Duncan ■ D. Elias ■ L. Garel 
J.-F. Geschwind ■ C. B. Glaiberman ■ C. Georgiades ■ J. Golzarian ■ M. Hayakawa ■ S. Heye 
S.-W. Hsu ■ M. S. Johnson ■ J. R. Kachura ■ N. M. Khilnani ■ P. Klurfen ■ P. Lasjaunias ■ S. K. Lee 
E. Liapi ■ W. S. Lesley ■ D. C. Madoff ■ G. Maleux ■ F. Marshalleck ■ R. J. Min ■ A. C. Roberts 
A. J. Roche ■ G. Rodesch ■ R. Salem ■ M. Sharafuddin ■ G. P. Siskin ■ S. Sun ■ K. T. Tan ■ M. Thijs 
K. G. Thurston ■ R. Verma ■ L. Wibbenmeyer ■ J. J. Wong 

Foreword by 

A.L.Baert 



i 368 Separate Illustn 



4y Spring 



er 



Jafar Golzarian, MD 

Professor of Radiology, Department of Radio 

University of Iowa Hospitals and Clinics 

Carver College of Medicine 

200 Hawkins Drive 

Iowa City, IA 52242 

USA 

Shiliang Sun, MD 

Associate Professor of Radiology 

University of Iowa Hospitals and Clinics 

Carver College of Medicine 

200 Hawkins Drive 

Iowa City, IA 52242 

USA 

M. J. Sharafuddin, MD 

Departments of Radiology and Surgery 

University of Iowa Hospitals and Clinics 

Carver College of Medicine 

200 Hawkins Drive 

Iowa City, IA 52242 

USA 



Medical Radiology ■ Diagnostic Imaging and Radiation Oncology 

Series Editors: A. L. Baert ■ L. W. Brady ■ H.-P. Heilmann ■ M. Molls ■ K. Sartor 

Continuation of Handbuch der medizinischen Radiologic 

Iv.iitVi.lC'pi-ii],! ol Mediatl Kadiology 



Library of Congress Control Number: 2005923494 

ISBN 3-540-21491-7 Springer Berlin Heidelberg New York 
ISBN 978-3-540-21491-5 Springer Berlin Heidelberg New York 



caily the rights oi inniri.UK.n. fiprinting, jeuse A uu<t 

provisions of the German Coryrighi Law of September ? 
be obtained from Springer-Verlag. Violations are liable f 


ofthii 
jrpros 


: public 


LLim:, broadcasting, reproduction i 

Trent version, and permission for 
under the German Copyright Lav, 


E; 


nderthe 
st always 


Springer \i p.;:; of Springer v-ience+riuiiinjis Msdis 












htlp//www.springer.com 
■ Spnng;r-\;iiag Bef:.n Hedelberg 2006 












Printed in Germany 












The use of general descriptive names, trademarks, etc. in 


fllis pi 


iblicatic 


n does not imply, even in the abse 


ice of j 


i specific 


statement, that such names aie eiempt from the relevant 


protect 


ivelaws 


and regulations and therefore free 


for gen 


eraluse. 


ProdiiL!liahility:The publishers cannot guarantee theact 
in this book. In every case the user must check such infi 


uracyi 


nby^cc 


formation about dosage and applic 


atlonc 


stained 


Medical Editor: Dr. Ute Heilmann, Heidelberg 












Desk Editor: Ursula N. Davis, Heidelberg 












Production Editor: Kurt Teichmann, Mauer 












Cover-Design and Typesetting: Verlagsservice Tekhman 


n.Mau 


* 









io my parents 

a wellspring of love and support without limit. 

I owe you everything. 

io my wonderful wife, Elham 
and my children Sina and Sadra 

Dr. Golzarian 

To my wife, Shuzhen, and daughter, Yue 
for their selfless support 



lo my wife Lucy, and children Jacob and Evan 
Dr. Sharafuddin 



To all o 



Foreword 



Percutaneous image-guided treatment is now well recognized as an effective minimally 
invasive treatment modality in modern medicine. Its field of application is growing 
every year due to the availability of more and more sophisticated materials, tools and 
devices, but also because of the technical progress in reduction of the dose of ionizing 
irradiation incurred by both patient and radiologist during fluoroscopy. 

Vascular embolotherapy is now one of the main forms of endovascular percutaneous 
treatment of diseases of the vascular system. 

The editors of the two volumes of "Vascular Embolotherapy: a Comprehensive 
Approach", J. Golzarian, S. Sun and M.J. Sharafuddin, leading experts in the field, were 
successful in obtaining the collaboration of many other internationally renowned inter- 
ventional radiologists. I am particularly indebted to Professor Golzarian for his origi- 
nal concept for these books and his relentless efforts to complete the project in good 

I would like to congratulate the editors and authors on producing these well-written, 
superbly illustrated and exhaustive volumes covering all aspects of vascular embolo- 
therapy. The readers will find comprehensive up-to-date information as a source of 
knowledge and as a guideline for their daily clinical work. 

These two outstanding books will certainly meet with high interest from interven- 
tional radiologists and vascular surgeons. They - and therefore their patients - will 
greatly benefit from its contents. Also referring physicians may find these books very 
useful to learn more about the indications, possibilities and limitations of modern vas- 
cular embolotherapy 

I am confident that these two volumes will encounter the same success with readers 
as the previous books in this series. 

Leuven Albert L. Baert 



Preface 



Therapeutic embolization has now become a major part of modern interventional prac- 
tice, and its applications have becomean integral component of the modern multimodal- 
ity management paradigms in trauma, gastrointestinal hemorrhage and oncology, and 
the endovascular therapy of vascular malformations and aneurysms. The past decade 
has also marked the emergence of several new indications for therapeutic embolization, 
such as uterine fibroid embolization, and the widespread acceptance of embolization 
therapy as an effective non-operative management modality for major hepatic, splenic 
and renal injuries that once posed tremendous challenge to the trauma surgeon. Emboli- 
zation therapy has also become an integral facet of the modern oncology center, offering 
solid-organ chemoembolization, preoperative devascularization, hepatic growth stimu- 
lation prior to resection, and direct gene therapy delivery. 

Despite this remarkable growth, there are currently few references available to sum- 
marize this major field in vascular interventional therapy. The purpose of our two- 
volume book was to organize and present the current state of the art of embolotherapy 
in a comprehensive yet manageable manner. Our goal was to provide a user-friendly, 
well- illustrated, and easy- to -browse resource to enable both experts and novices in 
this field to quickly derive high-yield clinically relevant information when needed. In 
addition to standard applications of embolotherapy, we have also included a number 
of closely relakvi applications that have become intimately associated with the field of 
therapeutic embolization, such as stent- graft placement and radiofrequency ablation. 
The two volumes constitute the combined experience of many of the leading experts in 
the field and have been generously supplemented with helpful tables, illustrations and 
detailed imaging material. We have also striven to include insightful discussions and a 
"cookbook" segment in each topic to provide a quick outline of procedural preparation 
and technique. We have included a chapter on monitoring and resuscitation of the hem- 
orrhaging patient that should be a "must-read" for the interventionist who is not well 
versed in surgical critical care. Readers will also find important coverage of pathophysi- 
ology and of diagnostic clinical as well as imaging workup. 

We hope this reference will meet the needs of physicians providing therapeutic embo- 
lization, whether they are trainees, recent graduates or even well-established interven- 
tionists who wish to refresh their memory or learn the opinion of some of the field's 
renowned experts before embarking on a difficult case or trying a new technique or 

Iowa City Jafar Golzarian 

Shiliang Sun 
Melhem I.Sharafuddin 



Contents 



Vascular Malformation 

1 Percutaneous Management of Hemangiomas and Vascular Malfor mat ic 
Francis Marshalleck and Matthew S. foi 

2 Predominantly Venous Malformation 
Josee Dubois 



Trauma and Iatrogenic Lesions 33 

3 Recognition and Treatment of Medical Emergencies in the Trauma Patient 

Lucy Wibbenmeyer and Melhem J. Sharafuddin 35 



Visceral and Abdominal Solid Organ Trauma 
Gary Siskin and [afar Golzarian 



Embolization and Pelvic Trauma 
Jeffrey J. Wong and Anne C. Roberts . . 



Postcatheterization Femoral Artery Injuries 
Geert Maleux, Sam Heye, and Maria Thijs . , 



Iatrogenic Lesions 
Michael Darcy. . . 



Visceral Aneurysm 

8 Embolization of Visceral Arterial Aneurysms 

Craig B. Glaiberman and D.Michael D. Darcy . 



Venous Ablation 117 

9 Endovenous Thermal Ablation of Incompetent Truncal Veins in Patients with 

Superficial Venous Insulrieienv 

Neil M. Khilnani and Robert J. Min 119 



Embolo therapy Applications in Oncology . 



10 Chemo-Embolization for Liver 

Christos Georgiades and Jean Francois Gesc 



XII Contents 

11 Radioactive Microspheres for the Treatment of HCC 

Christos Georgiades, Riad Salem, and Jean-Francois Geschwind 141 

12 Yttrium-90 Radioembolization for the Treatment of Liver Metastases 

Riad Salem, Kenneth G.Thurston, and [ean-Francois Geschwind 149 

13 Portal Vein Embolization 

Alain J. Roche and Dominique Elias 163 

14 Embolotherapy for Neuroendocrine Tumor Hepatic Metastases 

Kong Teng Tan and John R.Kachura 177 

1 5 Bone Metastases from Renal Cell Carcinoma: Preoperative Embolization 
Shiliang Sun 189 

16 Embolotherapy for Organ Ablation 

David C. Madoff, Rajiv Verm a, and Kamran Ahrar 201 

17 Research and Future Directions in Oncology Embolotherapy 

Eleni Liapi and Jean-Francois H. Geschwind 221 



External Carotid 233 

18 Technical and Anatomical Considerations of the External Carotid System 

Paula Klurfan and Seon Kyu Lee 235 

19 Endovascular Management for Head and Neck Tumors 

Paula Klurfan and Seon-Kyu Lee 247 

20 Embolization oi Hpiiaxis 

Georges Rodesch, Hortensi a Alvarez, and Pierre Lasjaunias 257 

21 Diagnosis and EndoYasail.ir Surgical Management of Carotid 
Blowout Syndrome 

John C.Chaloupka, Walter S. Lesley, Minako Hayakawa, 

and Shih-Wei Hsu 271 



Gene Therapy and Pediatri 



22 Embolotlu-rapv Applications in Gene Therapy 
James R.Duncan 



23 Embolotherapy in Pediatrics 

Josee Dubois and Laurent Garel . 



Subject Index 321 

List of Contributors 331 

Contents and List of Contributors of Volume 1 335 



Vascular Malformation 



Percutaneous Management of Hemangiomas 
and Vascular Malformations 



Classification 3 

Hemangioma 4 

Clinical Presentation 4 

I i iagr.ostic Imaging 4 
i Treatment 5 

Kaposiform Hemangioendothelioma 5 

Hep.iiic Hemai:[ :•<:*. iKhehoma 7 

Vascular Malfo-maluins .1 

Arteriovenous Malformations S 

Clinical Presentation S 

! ^agnostic Imaging 9 
■ Treatment 10 

Arteriovenous Fistulae 11 

Treatment 12 

venous Malformations 12 

Lymphatic Malformations 12 

Clinical Presentation 12 

I 'iagnostic Imaging 13 
i Treatment 13 

Capillary Malformations 16 

Complications of Embolization 

ja.7. Sclerotherapy 16 

Preprocedural Preparation 16' 

Post-Procedural Care and Fo!low-Up 16 

Conclusion 17 

References J S 






Classification 



Vascular birthmarks have intrigued physi 
centuries. Many attempts have been madt 
sify vascular birthmarks, resulting in much con- 
fusion. Historically, various classifications have 
been developed, each with its own shortcomings. 
Initially, classifications were largely descriptive [1], 



F. Marsh alleck, MD 

Assistant Professor or' K.; ■.:.,:■ I ogy, Indiana University School of 
Medicine, Indiana University Hospital, Room 0279, 550 North 
University Boulevard, Indianapolis, IN 26202, USA 
M. Johnson, MD 

Associate Professor of Radiology, Director, Section of Inter- 
ventional Radiology. Indiana University Hospital, UH0279, 
Department of Radiology, 550 "o'niversily Boulevard, India- 
napolis, IN 46202-5253, USA 



Although the descriptive classification allowed dif- 
ferentiation between benign and more serious forms 
of vascular malformations, because many different 
malformations can have similar external appear- 
ances, it was limited in its value in differentiating 
between them. The histopathological classification 
[1] represented an improvement in the attempt to 
classify vascular malformations. Its broad use of 
the word "hemangioma" and lack of clinical cor- 
relation limited its usefulness because hemangio- 
mas and vascular maltorin.it ions Jitter in pathology 
and are treated differently. The embryological clas- 
sification [1] was based on the theory that vascular 
malformations were due to improper development 
of various cellular lines (arteries, veins, capillaries, 
and lymphatics). Although the premise was sound, 
the embryological classification was not clinically 
useful to direct treatment. To date, the most perti- 
nent classification of vascular birthmarks has been 
published by John Mulliken and Julie Glowaki 
[1-3]. This biological classification separates vas- 
cular birthmarks into hemangiomas (vascular 
tumors) and vascular malformations (malformed 
vessels) (Table 1.1). Hemangiomas are characterized 
by a proliferating phase and subsequent involution 
phase, distinguishing them from vascular malfor- 
mations which do not spontaneously involute. Vas- 
cular malformations may be high-flow lesions (e.g. 
arteriovenous malformations, arteriovenous fistu- 
lae) or low-flow lesions (e.g. venous malformations, 
capillary malformations, lymphatic malformations, 
combined or mixed lesions). Vascular malforma- 
tions are best managed by a vascular anomalies 
team in a facility equipped and experienced in the 
management of vascular anomalies. Such avascular 

tionalist, dermatologist, plastic surgeon, orthope- 
dic surgeon and/or neurosurgeon, pediatrician, and 
physiotherapist. The percutaneous management of 
these lesions including clinical diagnosis, radiologi- 
cal diagnosis, percutaneous treatment (emboliza- 
tion, sclerotherapy) and post-procedure care will be 



F. Marshalleck and M. Johnson 



HcnitiiigiC'triiii 






PiV'lirer.'.ting 






\::v- ■|i.:ii:'.;_' 






Vascular malfc 






High-flow 








Arteric 


venous malformations 




Arteric 


venous fistulae 


Low-flow 








Venout 


malformations 




Lymph 


atic malformations 




Capilla 


ry malformations 




Mixed 


m alter mitt ions 



1.1.1 

Hemangioma 

A hemangioma is a benign vascular endothelial 
cell neoplasm characterized by a period of intense 
cellular and endothelial proliferation resulting in 
the formation of a cellular mass. During the pro- 
liferation phase, there is formation of new feeding 
and draining vessels similar to that of a high-flow 
vascular malformation. Proliferation is followed by 
involution and finally regression. This distinguishes 
a hemangioma from a vascular malformation. 



:al Presentatioi 






Unlike vascular maltoniialions, hemangi' 
commonly present at birth but u su ally be. 
during the first month of life. They are m 
in Caucasians, females, and premature infants and 
have a predilection for the head and neck. Hemangio- 
mas are the most common tumor of infancy with a 
reported incidence of 10%-12% [4, 5]. 

A hemangioma^ location determines its presen- 
tation. When it is superficial, it typically presents 
as a small red macule or patch which proliferates at 
a rapid rate during the first 6-12 months of life. A 
superficial lesion may produce a mass (a "strawberry" 
lesion) which can grow so large as to become disfig- 
uring. The strawberry appearance is produced by 
the presence of multiple reddened superficial vessels 
which result in an irregular raised "pebbly" surface 
([4]Fig. 1.1). When the hemangioma is deeper in loca- 
tion, the overlying skin may in fact be normal in color 
or may show bluish discoloration. The mass is usually 
warm and may be pulsatile during the proliferative 
phase. After the first 12 months of life, the majority 
of hemangiomas undergo an involution phase which 




Fig 1.1. Typical "str.;\vbe:ry" heauagiciv.Li. |[m 
through the courtesy of Dr. Phillip John, MD ] 



can last more than 5 years. Complete resolution of 
hemangiomas occurs in greater than 50% of children 
by age 5 years and in over 70% by the age of 7 years 
[I]. As the hemangioma involutes, it softens, shrinks, 
loses its red color and becomes dull grey due to its 
replacement with fibrofatty tissue. Depending on the 
original size of the hemangioma, the overlying skin 
may become loose with a "crepe paper" -like appear- 
ance. Occasionally, scars ortelangiectasias are seen at 
the site of an involuted hemangioma [■■!]. 

Complications of hemangiomas usually occur 
during the first o months of lite. The most common 
complication is ulceration, which occurs in up to 
10% of patients, especially when the lips or genital 
areas are involved [1, 4]. Occasionally, there may be 
associated bleeding, which is usually not significant. 
Hemangiomas may also result in congestive cardiac 
failure (e.g. hepatic hemangioendotheliomas) or 
platelet consumption (Kasabach-Merritt phenom- 
enon). Both entities will be discussed later in this 
chapter. When diffuse, hemangiomas may compro- 
mise the airway, obstruct vision, or impair hearing 
[1]. Associated osseous deformities are uncommon 
[1]. Rarely, hemangiomas may be associated with 
other anomalies, such as posterior fossa malforma- 
tions, right aortic arch, coarctation of the aorta, gen- 
itourinary anomalies, and spinal dysraphism [6]. 



1.1.1.2 

Diagnostic Imaging 



>, when superficial, are easily diag- 
lly as previously discussed. Appro- 
priate treatment of a symptomatic hemangioma, 



Hemangi 
nosed cl. 



Cerent.! neon:. Minaprine nl of Hem.iisgiomjs .m.". Vascular M..i liV-iiii:":! .-.:■: 



however, requires delineation of its extent. Diag- 
nostic imaging is also useful when the diagnosis 
is in doubt. On CT and MR imaging, hemangio- 
mas are well-circumscribed lobulated masses that 
demonstrate intense parenchymal enhancement fol- 
lowing the administration of intravenous contrast 
(Fig. 1.2a,b). During the proliferating phase, dilated 
vessels representing feeding arteries and draining 
veins are seen. MR is the optimal modality for the 
diagnosis and evaluation of hemangiomas [5]. The 
vessels are seen as flow voids on Tl- and T2 (spin 
echo) -weigh ted MR images. A proliferating hem- 
angioma is hypointense to muscle on Tl-weighted 
images and hyperintense on T2-weighted images. 
During involution, there may be a preponderance 
of fat (high signal on Tl-weighted images} with 
lack of flow voids. If a lesion lacks the classic clini- 
cal and imaging findings already discussed for a 
hemangioma, then a biopsy should be performed to 
exclude other potentially more serious tumors such 
as rhabdomyosarcoma, infantile fibrosarcoma, or 
neurofibroma. 



1.1.1.3 
Treatment 

About 75% of hemangiomas will regress on their own 
without treatment [1, 4]. Multiple factors will deter- 
tnine whether ,i hemangioma requites treatment, 
including the child's age and emotional needs, the 
location of the lesion, and symptomatology. When 
hemangiomas are small or are already decreasing in 
size before the child enters school, observation and 
reassurance are all that is needed. When treatment 
is deemed necessary, systemic corticosteroids have 
been the therapeutic mainstay, with a nearly 90% 
response [8]. Side effects of systemic steroids include 
gastrointestinal symptoms, weight gain, hyperten- 
sion, immunosuppression, and growth retardation 
[7-9]. Intralesional corticosteroids have been used 
to treat rapidly growing hemangiomas with the dose 
limited by the size of the hemangioma [7-9]. When 
steroids fail to cause adequate response, alpha inter- 
feron, chemotherapeutic agents, and radiotherapy 
have also been used [10-12]. The use of a- interferon 
is now limited to refractory cases due to its effects on 
the central nervous system such as spastic diplegia 
[13]. Laser therapy has been used to treat areas of 
ulceration, bleeding, telangiectasias, and skin dis- 
coloration [14]. 

Surgical removal becomes warranted in cases of 
ocular hemangioma unresponsive to medical ther- 




Fig 1.2a,b. Proliferating hemangioma of the ringer demon- 
strating: a low siga.il on Tl -iveielileJ image and b intense 
parenchymal enhancement with gadolinium 



apy and for airway compromise. Cosmetic needs may 
dictate surgical removal depending on the parents' 
and patient's wishes especially for head and neck 
hemangiomas. After involution, surgical resection 
may be required to remove excess skin and fibro- 
fatty tissue [4]. In the minority of cases in which a 
hemangioma fails to involute (non involuting hem- 
angioma) despite medical management, surgical 
resection of the lesion, if possible, is indicated [15]. 
Percutaneous embolization prior to surgical resec- 
tion has also been successful [16]. 

Rarely, arterial embolization is required to treat 
life-threatening hemorrhage, high-output cardiac 
failure, or platelet consumption (Kasabach-Merrift 
phenomenon) ([17], Fig. 1.3a-c). 



1.1.2 

Kaposiform Hemangioendothelioma 

Kaposiform hemangioendothelioma is an infil- 
trative variant of pediatric hemangioma. It com- 
monly affects the trunk and extremities, produc- 
ing an edematous mass of variable size with purple 
skin discoloration (Fig.l.4a,b). It proliferates and 
involutes like a typical hemangioma but persists, 
infiltrates, and consumes platelets (Kasabach-Mer- 
ritt phenomenon) resulting in hemorrhage [18, 19]. 
Rarely, it may resemble a classic hemangioma [20]. 
Platelets decrease to low levels (< 5000) despite 
repeated transfusions. Management involves a 
multidiscipli nan approach [21]. Chemotherapy, ste- 



F. M..irsh..illeckand M.John; 



bach-Merritt ] 
parenchymal ■ 
c Angiographi 




Tia of the trunk resuming .11 K.-Sji- 

n. b CT miner ■Jemonr.iTitii'.ii 

it with contrast aJnumstrjiiun. 

j demons! fa ;ing multiple enlarged 

emboli zed with PV'A p.'.rticles prior 




Fig 1.4. a Kaposi form HeiiiiinHioeiiJoLieliomj p resen I mi: 
findings in the sjiiis pniieiil deT.cnsrr.'.tmg .1 diffuse piirendr 
Dr. Phillip John, MD) 



!- : ei"ciit.':[iC'jLi; : Maaagerr.ent of He: 



■s an;: Vascular Malformatio: 



roids, ct-interteron, and radiation have all been tried 
[22, 23]. Surgical resection can be curative [24] but, 
in many cases, may not be possible due to the risk of 
hemorrhage. ] ntervenlional management comprises 
treatment of the associated platelet consumption by 
endovascular embolization using a microcatheter 
technique. PVA (polyvinyl alcohol) particles and/or 
absolute alcohol are typically used [25-27]. These 
cases are usually difficult to treat and time-con- 
suming due to the presence of multiple feeders. The 
long-term effects of endovascular embolization have 
not yet been established. 



1.1.3 

Hepatic Hemangioendothelioma 

Hepatic hemangioendothelioma (multiple hepatic 
hemangiomas, congenital hepatic hemangioma) of 
the newborn is characterized by a liver mass, an audi- 
ble bruit, and congestive heart failure with or without 

cutaneous heman^iomata ([]], l-'ii:. 1.5a- cj. The hish- 
output cardiac state could be fatal. Hirst-line manage- 
ment is medical with the use of steroids, interferon, 



or chemotherapy. Endovascular embolization can be 
performed as a temporizing measure to liver trans- 
plantation if surgical treatment (hepatic resection, 
hepatic artery ligation) is not possible and if medi- 
cal therapy fails [28], The vascular anatomy may be 
complex with multiple collaterals and various shunts 
(arteriovenous, arterioportal, portovenous) resulting 
in the high-output state [29, 30]. In order to treat the 
severe AV shunting in the liver, endovascular embo- 
lization can be performed. Selective hepatic arterial 
embolization has been used to treat arteriovenous 
shunts. Coils [31, 32], detachable balloons [47], and 
PVA particles [33] have been used. Coils and detach- 
able balloons result in permanent occlusion and their 
use depends on personal preference. The hepatic 
artery canbe accessed via I lie femoral artery, a central 
vein, or femoral vein by '.vay ot a patent foramen ovale 
[32] or via a patent ductus arteriosus in neonates [34], 
In cases of portovenous shunts, the portal vein can 
be accessed using a transjugular approach, transhe- 
patic approach or via the umbilical vein in neonates 
[34] to allow for coil embolization. In cases where 
portovenous shunts are dominant, hepatic arterial 
embolization alone may not prove to be of benefit 




Fig 1.5, ii C".' so:;:; image in a newb'.'ra . : . eni-.jj";SL:":i: ij:g !ir.\ 
.y.bCT arterial phase cenr 'ii : a:a:iiig :::ul;iple hypo J, 
fs Kidiiii the liver, c CT port.-! venous phase 
lultiple masses with nodular enhancement 



h M.ir>h.iU.:k -mi V. !■ 



and may result in hepatic necrosis [30, 35]. It is there- 
fore imperative to perform an angiographic study to 
include the portal venous circulation and potential 
collateral vessels to allow for careful planning before 
embolization is performed [29]. 



Vascular Malformations 

Vascular malformations, unlike hemangiomas, 
are not neoplasms, but instead represent errors 
of vascular morphogenesis resulting in abnormal 
blood vessels and lymphatics [!]. They are classified 
into high-flow and low-flow lesions based on their 
hemodynamic properties. High-flow lesions include 
arteriovenous malformations (AVMs) and arterio- 
venous fistulae (AVFs). Low-flow lesions include 
venous malformations, lymphatic malformations, 
capillary malformations, and combined malforma- 
tions. Vascular malformations tend to be present at 
birth and grow commensurate with the growth of 
the child. The majority ot vascular malformations 
can be diagnosed with history and physical exami- 
nation and confirmed by diagnostic imaging. 



1.2.1 

Arteriovenous Malformations 

Arteriovenous malformations (AVMs) consist of 
multiple small abnormal connections (nidi) con- 
necting large arterial feeding arteries to large drain- 
ing veins without an intervening capillary bed. 



Clinical Presentation 

AVMs affect males and females equally. Their growth 
is known to be stimulated from hormonal changes 
during puberty, hormonal therapy, and pregnancy 
[1]. AVMs are classified into four clinical stages 
according to the International Society for the Study 
of Vascular Anomalies (ISSVA) Schobinger clas- 
sification [36]. Stage 1 represents a dormant AVM 
which present like a capillary skin stain or a small 
pulsatile skin mass. In stage 2, an AVM is larger and 
presents as a warm, tender, red, pulsatile mass with 
visible large draining veins and an audible bruit. In 
stage 3, the AVM is complicated by ulceration, bleed- 
ing, and associated destructive osseous changes. In 



stage 4 (2.5% of cases), the AVM results in conges- 
tive cardiac failure due to increased arteriovenous 
shunting. AVMs may affect the head and neck, 
extremities, and viscera (e.g. lungs, liver, kidneys, 
spleen, and pancreas). AVMs maybe focal, but more 
frequently are diffuse and cross tissue planes. AVMs 
become symptomatic when they bleed, ulcerate, or 
exhibit mass effect on nearby structures. 

In the extremities, AVMs typically present as a 
soft tissue mass with hyperthermia, redness, ten- 
derness, and swelling (Fig. l.oa). The draining veins 
are usually visibly distended and associated with a 
palpable thrill and an audible bruit. There is usu- 
ally associated tissue ischemia and edema which can 
lead to ulceration. It has been postulated that the 
skin necrosis is due in part to arteriovenous shunt- 
ing but also due to associated venous hypertension 
and mass effect [1]. Ulceration can ultimately lead to 
life-threatening hemorrhage and can also be compli- 
cated by infection. Spontaneous bleeding is uncom- 
mon in the absence of ulceration or trauma. When 
intramuscular, AVMs may produce significant pain 
[1]. Pelvic AVMs are rare and typically present with 
pelvic pain, pedal edema, menoi rliagia, hemorrhage 
(antepartum, postpartum) or a pulsatile mass on 
pelvic exam. In males, dysuria, frequency, impo- 
tence, tenesmus, and hematuria can occur [37-39]. 
AVMs may produce lytic osseous lesions or result in 
limb overgrowth. Congestive heart failure can result 
if the AVM is large or if it occurs in infancy. 

When affecting the brain, an AVM can present 
with hemorrhage, stroke, seizures, or focal neuro- 
logical deficits. Spinal AVMs present with hemor- 
rhage or a myeloradiculopathy. Dental AVMs can 
present with life-threatening hemorrhage after tooth 
extraction, eruption, or infection [1]. 

AVMs of the abdominal viscera are uncommon, 
but when they do occur, they have an increased prob- 
ability of bleeding due to the proximity to mucosa. 
True AVMs of the liver in the newborn will present 
with a clinical picture similar to hepatic hemangioen- 
dothelioma already discussed. Pancreatic AVMs [40, 
41] are usually associated with Osier-Weber- Rendu 
syndrome. Splenic vascular malformations are usu- 
ally asymptomatic and found incidentally at autopsy. 
They can also present with splenomegaly, pain, bleed- 
ing, portal hypertension, and hypersplenism [42]. 
Vascular malformations of the kidney are rare. 

Pulmonary AVMs can occur sporadically (15%) or 
as part of the autosomal dominant disorder (60%- 
90%) known as Osler-Weber-Rendu syndrome or 
Hereditary Hemorrhagic Telangiectasia (an autoso- 
mal dominant disease characterized by telangiecta- 



Perciitaneon:. Ma:'.a£e:''.eiil of Hemangiomas ..in.:. V'ascinar M..i liV-iiii:":! .-.:■: 





Fig US. a AVM of the left hand (hypothenar eminence) with 

distended, palpable and pinsatite vessels. The light hand is 
shown for compaiiso::. lv Angiographic evaluation dem- 
onstrating a la:ge nidus involving t:ie :".ypoihenai" eminence 
with enlarge;", feeding vessel 1 ; a nil draai.ng veins. d,e Angio- 
graphic images de:v..: iistiai.ng microcadierer snpeise lectio:' 
of the nidus by different feeders to a'lovy for alcohol enibo- 



sias, recurrent epistaxis, and a family history of tel- 
angiectasia). Pulmonary AVMs are multiple in up to 
55%, bilateral in 40%, and occur mainly in the lower 
lobes. The AVMs are usually simple with a single 
feeding artery (80%) but may be complex with mul- 
tiple feeding arteries (20%). Patients present with 
symptoms of hvpoxia due to arteriovenous shunting 
(e.g. dyspnea and cyanosis), paradoxical emboliza- 
tion resulting in CVA, TIA, or brain abscess and/or 
congestive heart failure [50, 51]. 



1.2.1.2 

Diagnostic Imaging 



The characteristic imaging find ings of AVMs include 
dilated feeding arteries and dm mi tit; veins. On CT 



i enhance after the adrr. 

contrast while on MR imaging 
the vessels are seen as multiple prominent flow voids 
onTl-and T2 -weighted spin echo sequences [5]. The 
vessels will be bright on gradient echo sequences. 
Unlike a hemangioma, there is no parenchymal 
mass and the nidus is usually not visible. Various 
signal changes indicative of blood products may 
be seen if the AVM has bled. Associated soft tissue 
(edema) and bony changes may also be seen. History 
and physical examination will usually suffice in the 
diagnosis of AVMs affecting the extremities with 
imaging performed to document the extent of the 
lesion. CT imaging is superior to MR in the delin- 
eation of pulmonary AVMs. Visceral lesions may 
be investigated with CT and MR imaging. Angiog- 
raphy is usually not required for diagnosis, but is 



F. M..irsholk>ckand M.John; 



performed for treatment planning, at the time of 
percutaneous embolization, or if the diagnosis is in 
doubt. Angiography is superior to delineate the nidi 
and also clearly demonstrates the large feeding ves- 
sels and early draining veins (Fig. 1.6b,c). 



1.2.1.3 
Treatment 

The vast majority of AVMs will cross surgical planes 
to involve the deep tissues, making surgical resec- 
tion impossible due to the risk o] siijnihcani hemor- 
rhage or of damage to associated tissues or organs. 
Excisional surgery becomes more feasible after suc- 
cessful embolization. Percutaneous management of 
AVMs is difficult, with complete lasting obliteration 
of the AVM usually not possible even with current 
techniques. Percutaneous treatment is performed 
mainly to control symptoms such as pain, distal 
ischemia leading to ulceration, hemorrhage, and 
congestive cardiac failure [5]. Amputation may be 
the necessary end result in cases of extensive AVM 
of the extremity when embolization fails to control 
the symptoms [43]. 

For those patients where treatment is ind tented, 
an initial diagnostic arteriogram is performed. 
Although treatment of the lesion might be per- 
formed at that time, the diagnostic and therapeutic 
procedures may be performed separately, to mini- 
mize contrast volume and to ensure that appropriate 
equipment is available. In young children, the pro- 
cedures might be combined to decrease the number 
of times the patient needs to be placed under general 
anesthesia. Superselective arterial embolization is 
usually performed using microcatheter techniques. 
The aim is to selectively embolize the feeders to 
the nidus without compromising blood supply to 
essential nearby structures. This often proves to be 
difficult and time-consuming because the major- 
ity of AVMs will have numerous feeding arteries 
and draining veins. Occluding a feeding vessel too 
proximally will only lead to development of new 
feeders and is thus ineffective. This will ultimately 
lead to recurrence and distal ischemia as new ves- 
sels are recruited [44]. Additionally, proximal occlu- 
sion may make subsequent attempts at embolization 
more difficult. 

In order to attempt embolization of the nidus 
within an AVM, various agents such as PVA par- 
ticles, absolute alcohol, and tissue adhesives (glue) 
have been used. PVA particles are available in sizes 
ranging from 50 um to 1000 (im. They are useful 



when multiple micro fistulous connections exist 
within the nidus. The size of the particles must be 
larger than the connections to avoid escape into the 
venous system. The effects of PVA particles are usu- 
ally temporary with a high rate of recurrence due 
to subsequent development of collaterals compro- 
mising future percutaneous management [44]. PVA 
particles are therefore best suited for preoperative 
embolization to minimize bleeding during surgical 
resection [45]. 

Absolute alcohol is a very effective embolization 
agent because it destroys the walls of the blood ves- 
sels by inciting a strong inflammatory reaction. In 
order to maximize the effect on AVMs and simulta- 
neously prevent effects on vital structures, alcohol 
is delivered directly into the nidus by superselec- 
tive catheterization or via direct puncture ([47,48], 
Fig. 1.6d,e). Occluding inflow arteries or outflow 
veins maximizes the effect on the nidus. The objec- 
tive is to confine the injected alcohol within the 
nidus. Inflow occlusion can be achieved with the 
use of balloon catheters. If inflow occlusion is not 
possible, then outflow occlusion can be achieved 
with the use of orthopedic tourniquets, blood pres- 
sure cuffs (inflated to supra systolic pressures}, or 
manual compression depending on the location of 
the lesion [44]. Contrast is injected into the nidus 
during inflow occlusion until the draining veins 
are seen. This reflects the volume of alcohol needed 
to fill the nidus without spilling into the draining 
veins. Follow ing emit i tit us ton of alcohol (retained 
for several minutes within the lesion prior to releas- 
ing the inflow or outflow obstruction), contrast 
injection should be repeated to follow the effects 
of alcohol, with stasis within the nidus being the 
ultimate goal. Some authors have advocated that 
alcohol should be the only agent used in the treat- 
ment of AVMs and that alcohol therapy can be 
curative [44]. While we agree, it is important to 
consider the risk of necrosis of nearby vital tissues 
and of the skin need to be considered when alcohol 
is administered by a percutaneous or endovascular 
route. Also, the risk of systemic toxicity increases 
in doses above 1 ml/kg or if a volume greater than 
60 ml is used. Complications can be as high as 15% 
of patients treated with absolute alcohol [44]. Most 
complications are self-limiting or may be success- 
fully treated (e.g., with skin grafting in the case of 
skin necrosis); however, neurologic complications 
can be permanent. Some au thors advocate that gen- 
eral anesthesia should be used when embolization 
will be carried out using alcohol, due to its possible 
local and systemic effects [25]. We agree that gen- 



Percutaneous Mar.aeriy.eiil of Heni.ing.omas an.". Vvi:cul.ii Malformations 



eral anesthesia should be used in children when 
embolization is performed with absolute alcohol. 
In adults, by keeping the patient awake with con- 
scious sedation, the local effects of absolute alcohol 
can be assessed clinically (e.g. assessing for neu- 
ropathy in the extremities during absolute alcohol 
therapy). 

Tissue adhesives work by forming a cast within 
a blood vessel resulting in occlusion. It is available 
as an injectable liquid that immediately polymer- 
izes when it contacts ionic fluids such as blood. It 
can also be diluted or modified to polymerize after 
variable periods. Tissue adhesives are of value in 
the treatment of very hiab-ilo'.v AVMs '.vliere ins ton! 
polymerization is advantageous to avoid entry 
into the draining veins. The disadvantage of tissue 
adhesives is that, unlike absolute alcohol, they may 
not totally destroy the nidus resulting in eventual 
recanalization. Coils and detachable balloons pro- 
duce too proximal an embolization and should be 
avoided in the treatment of extremity AVMs unless 
the arteriovenous connections are quite large and 
glue is unavailable [46]. When a transarterial route 
to the nidus is not possible, the nidus can be directly 
injected with absolute alcohol or glue [47, 48] or 
accessed via a transvenous route [44, 49]. 

Approximately 80% of pulmonary AVMs will 
be of the simple type with a single feeding artery 
making them amenable to percutaneous emboliza- 
tion with coils or detachable balloons (Fig. 1.7a,b). 



Cure of up to 84% ot pulmonary AVMs with a single 
procedure has been reported [50-52]. 

Renal AVMs are rare and are typically small in 
size but can be quite large. Percutaneous emboli- 
zation has been documented in the treatment of 
symptomatic lesions (hematuria, congestive cardiac 
failure, or hypertension). Percutaneous emboliza- 
tion using coils, Gelfoam, i'VA particles, and glue 
(N-butyl- cyan oner via lei lias been reported [53-55], 
Uterine AVMs have successfully been embolized 
with PVA particle's, Gelfoam, sine, ami coils [?(■], 



1.2.2 

Arteriovenous Fistulae 

Arteriovenous fistulas (AVFs), like AVMs, are high- 
flow vascular malformations but consist of a single 
macrofistulous communication between an artery 
and a vein (Fig. 1.8a-c). They are not common in 
childhood and are considered to be posttraumatic in 
origin. They are, however, still found in the absence 
of a history of trauma [26]. Like AVMs, they can 
produce CHF due to arteriovenous shunting. Small 
AVFs may close spontaneously and larger lesions 
can enlarge over time presenting in the extremity as 
a pulsatile lesion with a palpable thrill and audible 
bruit. Angiography easily demonstrates the single 
large interconnection. Visceral AVF are usually iat- 
rogenic and can present with hemorrhage. 




Fig 1.7a,b. Left pulmonary 
AVM years 



with HereJii.nv Hrmory.'.aeii" i'-Lnpe.a 
::.:.;"'.■;':":': I !.:■!!■:. b y.ii.Yc-iviiil ■;.:■! I eiv.oo.i^cio:'. .:■[' .; neu 



isi.i. a Form a: ion of ;'.rv. : p aim onarv 
pulmonary AVM 



F. Marshalleck and M. Johnson 




1.2.2.1 
Treatment 

AVFs are amenable to percutaneous endovascular 
embolization with coils and detachable balloons 
[57-59] or ethanol [60]. The aim is to embolize the 
macrofistulous connection or the immediate drain- 
ing vein. When this is not possible, a stent graft or 
covered stent can be percutaneously placed within 
the feeding vessel to cover the AVF [61]. If the feed- 
ing vessel can be sacrificed, it can be occluded using 
coils, detachable balloons, or glue [26]. In occluding 
large AVFs, migration of a deployed device can be 
prevented with the use of balloon catheters, snares, 
and tourniquets. 



1.2.4 

Lymphatic Malformations 

Lymphatic malformations (formerly known as cystic 
hygroma or lymphangiomas) are ma Id eve lop me nts 
of the various components of the lymphatic system. 
They are localized malformations affecting the vari- 
ous layers of the subcutaneous tissue and exhibit low- 
flow characteristics. They are usually multiloculated 
and are divided into microcystic (cysts <2 cm in dia- 
meter), macrocystk", and mixed categories. 



1.2.3 

Venous Malformations 



The management of VMs 
in the next Chapter. 



1.2.4.1 

Clinical Presentation 

Lymphatic malformations (LMs), like other vascu- 
lar malformations, are commonly present at birth 
with a predilection for the head, neck and axilla 
(Fig. 1.9). They affect both sexes equally. They are 
less common than venous malformations and are 
usually subcutaneous [1, 5, 68]. 



P emit. 1 , neon:. M.n'.a£r:''.ent of Heni.ing.omas .ni.". Vvi:cul.ii Mjlrormalio: 




1.2.4.2 

Diagnostic Imaging 

MRimagingotmaaocystkLMsshowsmultipIelarge 
cysts isointense Co muscle on Tl-weighted images and 
hyperintense on T2-weighted images, with periph- 
eral rim enhancement, septal enhancement, or no 
enhancement with gadolinium (Fig. l.lOa-e). There 
may be fluid-fluid levels and signal characteristics 
presenting blood products (Fig. l.lla-c). Edematous 
changes maybe seen in the surrounding subcutane- 
ous tissue [5]. Osseous distortion and overgrowth 
are best demonstrated on CT. The microcystic vari- 
ety demonstrates similar characteristics; however, 
the hyperintensity on T2-weighted images is more 
diffuse due to the presence of microcysts. Cysts are 
also readily demonstrated by ultrasound with low- 
flow dynamics seen on Doppler imaging [5]. 



Fig 1.9. Lymph a tic malformation of the i 
by Dr. Phillip John, MD) 



jck. (Image provided 1 .2.4.3 

Treatment 



They can enlarge significantly and extend 
through various tissue planes to dissect into the 
mediastinum thus making surgical removal diffi- 
cult. Mass effect can lead to airway compromise and 
osseous overgrowth [1], LMs can occur with other 
malformations such as capillary and venous malfor- 
mations. The most superficial form of LM is referred 
to as lymphangioma circumscriptum (LC) and con- 
sists of visible thin-walled clear lymphatic vesicles 
on the skin that, on occasion, exude lymph. If hem- 
orrhagic, the LC may become pink in appearance. 
It is seen most commonly at the shoulder, buttocks, 
neck, or mouth. Optimal treatment, to control per- 
sistent leakage of lymph and for cosmetic reasons, is 
operative resection [1]. 

The classic LM presents at birth as a soft translu- 
cent mass at birth that, unlike some venous malfor- 
mations, cannot be manually decompressed. Also, 
unlike venous malformations, LMs do not enlarge 
during the Valsalva maneuver. They may be asso- 
ciated with overlying lymphangioma circumscrip- 
tum. When complicated by hemorrhage, the mass 
becomes firm. LMs can also become secondarily 
infected causing them to become tender, warm, and 
erythematous [62]. They frequently enlarge during a 
viral illness. When the LM is extensive and becomes 
secondarily infected, the result can be airway com- 
promise [63, 64]. Other complications include SVC 
obstruction, chylothorax, pulmonary hypoplasia, 
and death [65-67]. 



Treatment is indicated to relieve symptoi 
pain or airway compromise or to improve c 
caLly. The macrocystic form of LM responds favor- 
ably to surgery and sclerotherapy with sclerotherapy 
having similar success rates but less morbidity [68], 
Sclerotherapy is performed using direct percutane- 
ous puncture using a variety of agents, e.g. ethanol 
[68], doxycycline [69-71], bleomycin [72], Ethibloc 
and OK-432 [73-76]. Bleomycin, a chemotherapeutic 
agent, may be used for sclerotherapy of LM, but has 
systemic side effects such as pulmonary fibrosis. 
OK-432 is a superantigen produced from Streptococ- 
cus A and is not yet available in the USA. Ethibloc 
is composed of Zein protein, contrast and ethanol 
and is also not available in the USA. Doxycycline 
has been used to treat microcystic LM, in neonates 
where the use of alcohol is limited due to the patient's 
weight and in the treatment of very large lymphatic 
malformations in which a large volume of sclerosant 
is needed. It is available in powder form (100 mg) 
and is mixed with saline in a concentration of 10 or 
20 mg/ml. Volumes up to 100 ml can be used. Doxy- 
cycline produces pain during injection, is nontoxic, 
and may result in only mild adverse reactions, e.g. 
fever [68]. The most commonly used sclerosant for 
macrocystic LM is ethanol with a maximum dose of 
1 ml/kg or a maximum volume of 60 ml [5]. 

Depending on the size of the cysts, their contents 
can be drained using single or multiple angiocath- 
eters, a catheter with multiple side holes, or a pig- 



F. Marshalleck and M. Johnson 




Figl.lOa-e. Unilocular lymphatic in.il for in at ion involving the i 
tongue demonstrating: a low density on CT, b increased signal 
weighted images due to its proteinaceotis content, c high signal 
weighted images, and d rim enliajwme:'/ with gadolinium ad] 

e Contrast injection during sclerotherapy demonstrating a singl 



tail catheter. The volume aspirated will del 
the volume of sclerosant to be used. Various authors 
report using a volume of sclerosant equal to 30%- 
100% of the aspirated volume [70]. Contrast injec- 



tion confirms the location of the needle and catheter. 
The contrast is aspirated and then the sclerosant is 
instilled. To minimize the use of contrast and con- 
comitant potential dilution of the sclerosant, access 



PerciitJiifoii:; M.n'.:]gr:''.enl of Heni.ing.omas .in.: VVi:aila< M.ilroinijiiioi 




Figl.lla-c. Lynipj'.'itic uuilfo: ai.Uion aa\eno: ::■ die light parotid gland 
de niois si:'Li:ing: a [liuiliple fltiid/rhiid level? on Tl MR-weighleo images, 
b Tl MR-\veiglueo iinagrs and e MR nii.iges with g,idoli:in-:i5 



into the cyst can be achieved using sonographic 
guidance. CT can also be used to guide sclerotherapy 
[69]. The sclerosant is injected with a tiny amount 
of contrast with subsequent CT scanning to confirm 
opacification of the cysts. Some authors will mix 
the sclerosant with a small amount of contrast and 
utilize fluoroscopy during injection to evaluate for 
venous escape [26]. When alcohol is the sclerosant 
of choice, it is instilled, leftto dwell for up to 15 min, 
and then drained with subsequent needle removal. 
In order to increase dwell time when doxycycline is 
used, aspiration of the sclerosant is not performed. 
A pressure dressing is applied to minimize leakage 
iollowuig the procedure. 

Sheils etal. describes a coaxial technique for large 
cysts using a coaxial system consisting of a 14G angio- 
catheter and 5F pigtail catheter. The cyst is accessed 
with the 14F angiocatheter and the 5F pigtail catheter 
is advanced through the existing angiocatheter into 
the cyst. Contrast is injected to opacify the LM. After 



the contrast is aspirated, l%Lidocaine is injected and 
left to dwell for 10 min. After aspiration, 3% sodium 
tetradecyl sulfate, a detergent sclerosant, is injected 
and left to dwell for 1-2 min. After aspiration, alco- 
hol is injected and left to dwell for 15 min. At each 
step, 50% cyst volume is utilized. After the alcohol is 
aspirated, the catheter is connected to a fackson Pratt 
suction bulb system and suction is performed for up 
to three days while the patient is on oral antibiotics. 
Pigtail catheter drainage can be followed up with 
repeated injections of the sclerosant until there is no 
further drainage. Ultrasound evaluation performed 
one month following sclerotherapy has shown com- 
plete ablation in 95% of patients where catheter drain- 
age has been used [70]. 

Fluid aspiration and contrast injection may not be 
feasible when treating the microcystic form of LM. 
Ultrasound imaging is therefore useful to monitor 
the injection of microcysts. Due to the local effects 
of alcohol such as tissue necrosis and nerve injury, 



h M.ir~h.iU.:k -mi V. !■ 



doxycycline is usually the sclerosant of choi 
treatment of microcysticLM [45]. 






1.2.5 

Capillary Malformations 

Capillary malformations (C.Ms), because of the: 
appearance, have traditionally been referred to as 
port wine stains and incorrectly as capillary heman- 
giomas. They represent maldevelopment of capillar- 
ies and present as well demarcated skin discoloration. 
CMs initially have a pink or red color and become 
more purple as children age. CMs frequently occur 
with other vascular anomalies. InKlippel-Trenaunay 
syndrome, they occur on the trunk or lower extrem- 
ity and are associated with limb hypertrophy and 
widespread venolymphatic malformations. In Sturge- 
Weber syndrome, the CM is typically in the VI (first 
division of the trigeminal nerve) distribution on the 
face and is associated with underlying ophthalmo- 
logic and leptomeningeal VMs and CMs. 

Treatment options for CMs include mainly pulsed 
dye laser therapy and also corticosteroids, inter- 
feron, and surgery [77-79]. The interventional radi- 
ologist may be called upon only to treat the associ- 
ated lesions such as VM and LM in order to alleviate 



Complications of Embolization 
and Sclerotherapy 



The 



utaneous treatment of a: 

nbe quite challenging. Emboliz 



al- 



Preprocedural Preparation 



The indications for treatment are clearly delineated. 
Treatment may be performed only to alleviate symp- 
toms, since in many cases a cure is not possible. 
The risks and benefits are clearly explained to the 
patient. Lab studies are performed as in many inter- 
ventional procedures to test clotting parameters and 
renal function. When small toatl malformations are 
treated with agents other than ethanol, the proce- 
dure can be performed using conscious sedation. It 
is generally recommended that when a significant 
volume of alcohol is to be used in the treatment of 
a large AVM, especially in patients with pre-existent 
cardiac compromise, that general anesthesia is per- 
formed. Pulmonary arterial pressures maybe moni- 
tored. Some authors advocate hydration before the 
procedure to protect the kidney from the effects of 
hemolysis [68]. A Foley catheter may be inserted. 
Proper equipment (sheaths, microcatheters, wires, 
and embolic agents) is essential. A nti- inflammatory 
medications such as corticosteroids are given just 
before and continued after the procedure to mini- 
mize swelling. Antibiotics (e.g. cephalexin) are usu- 
ally given for 10 days after sclerotherapy of LMs due 
to the risk of infection. Antiemetics, antibiotics and 
analgesics may also be administered especially for 
transarterial embolization cases. 



format i( 

can fail if the nidus cannot be reached or if the 
malformation is not adequately treated. Additional 
complications include embolization to distal tis- 
sues or organs, thrombosis of normal vessels, 
nausea/ vomiting, pain, fever, swelling, and post 
embolization syndrome. Although absolute alco- 
hol can be effective in the management of vascu- 
lar malformations, the complication rate can be as 
high as 10%-15%. Local effects of alcohol include 
tissue necrosis, neuropathy, and skin ulceration. 
Systemic effects include CNS depression, hypogly- 
cemia, systemic hypertension, pulmonary hyper- 
tension, cardiac arrhythmias, bradycardia, pulmo- 
nary vasoconstriction, disseminated intravascular 
coagulation due to fibrinogen consumption [80], 
hemoglobinuria, pulmonary embolism, and car- 



Post-Procedural Care and Foliow-Up 

In addition to routine vascular postprocedural 
care, several post-embolization-specific procedures 
should be employed: The treated extremity should 
be elevated to alleviate edema. Pain control is usually 
necessary and may require intravenous narcotics: A 
patient-controlled-anesthesia (PCA) pump works 
very well for the majority of patients. Whenever 
absolute alcohol is used, the overlying skin should be 
observed for blanching or blistering, which, if pres- 
ent, suggest skin injury. Mild cases can be treated 
with topical antibiotics or Silvadene cream. Plastic 
surgeons should be consulted early, as necrosis might 
ensue, and skin grafting might be required. Neuro- 



Percutaneous Manageir.enl of Heni.ing.omas jii.: Vascular Malrbrmaiioi 



T.ifle 1.2. - 



ia Is and equipment 



IR clinic with ancillary surf (clerks, nurses, phvsi. 

Portable Doppler ultrasound machine, to allow evaluation of vascular status 

lar lesions 

Imaging equipment and expertise: MRI/MRA, CT/CTA, US with color flow, h 



5ners), and time tor p::y: 

ior to and following 

h-speed DSA 

lary vein or artery at 

ice, e.g., in the 



>t be advanced thro-: eh small feed ins vessels to lesion 



el appropi iaie guidewne. The transcend guidev 



Cook Micropuncture sheath: A very versatile sheath which can be u 
angiography and/or embolization, or, as described above, as a direc 
Caih.elers 

• Standard array of 4F and 5F catheters 

• Hydrophilic catheters: Way be necessary if standard catheters a 

• Microcatheters: The Transit catheter (Cordis) is quite useful 

• Guidewires: Standard array: if microca dieter is used, will n- 
a good choice 

• Balloon catheters: E.g., die Cool. Over-1 he-wire Fogany bafoon caiheie:: Sue. catheters i:.)n he used as proximal occlu- 
sion catheters, throng;; which microca dieters can be a. mm need, and through which alcohol (e.g.! can be infused. The use 
of proximal occlusion as wi;h these devices is especially vadiaKe in Ireaiment of high -flow lesions. 

Blood pressure cuffs: Pediatric, standard, and large. Siipra-ysiolic inflation of Bf- 1 cuffs can be used ■: as can direct pressure 
and/or tourniquets) to prevent outflow rrom lesions to be emboli zed or sclerosed. 
Embolization materials 

• Absolute alcohol 

• Glue 

• +/- Sotradeehol (may be or" some value in dealing very sroerficial lesions, as it may be less iikeiy :c- cause skin necrosis) 

• Coils: Althoug.i usually not necessarv and ustiady conlra indicated in the treat men: of AV.Yls and V'V Ms 'because coils .its: 
unlikely to be successful in the treatment o: I hose lesions and ihey interfere widi subsecuenl access), coils are invaluable 
in I he treatment of PAV Ms and AVFs. A wide range of coils should be available for such lesions. 

• Balloons: May be necessary in ihe treatment o:' high -flow lesions 

Sifvacene cream: lii ihe unfortunate evenl that a person develops s^m breakdown after a procedure, appropriate would care 



Point 1: Be prepared. Ascertain that :lie pal lent and hi -/her family understand what to expect from Ihe procedure; ensure that 
adequate analgesia is available: ensure lhal af necessarv equip mem ( inc.nding ultrasound I is readily available. 
Point 2: 1 >o:t : lose access. Always, use a sheadi when approaching a lesion from the ins.de. Co-ax irf access :. sheath ami ca I heier, 
or sheath a no catheter and microca dieter) is mandatory. Similarly although yon ciin treat a lesion widi direc: injection through 
.: lire." le, :. Ilex it !e she.: I.:, e.g., ihe aF inner no it ion 01" a C. :■..■.. 41- yicropuncrare ;i :; ns. t.. :■ 11. ■. ..:na:o-r nrovi..ies oeiter srabililv. 
Point 3: Be prepared for ad outcomes, both successful and otherwise: e.g., prompt discussion w.li: a plastic surgeon can be of 
s benefit in patients who- develop ssin breakdown. 



logic examination of the treated extremity is essential 
to evaluate for neuropathy. Most peripheral malfor- 
mations will visibly swell soon after treatment, reach 
a peak a few days later, and subside over the next few 
weeks. The degree of expansion is expected to subside 
over the next two weeks. Overnight stay is indicated 
if large lesions are treated especially if airway com- 
pression can occur. ICU admission may be necessary 
with intubation to protect the airway. In the presence 
of hemoglobinuria, the urine needs to be alkalin- 
ized with sodium bicarbonate to protect the kidneys 
from crystallization [68]. When pigtail catheters are 
placed e.g. in LMs, the patient are hospitalized for 
a few days. Follow-up appointments may be made 
in 4-6 weeks to evaluate response to treatment and 
determine if repeat embolization or sclerosis is nec- 
essary. Lesions are followed clinically and with MR 
iiiuu-iii:: ns indicated. 



Conclusion 

As discussed, the most useful classification of vas- 
cular malformations to date is the one by Mulliken 
and Glowaki. The majority of vascular malforma- 
tions can be diagnosed clinically with MR1 now the 
gold standard to delineate the extent of the lesion or 
in cases where the diagnosis is in doubt. 

The majorities of hemangiomas are best left alone 
and will regress either spontaneously or with medi- 
cal treatment. End ova scu lar embolization can be 
helpful in the cases of noninvoluting hemangiomas, 
Kapositorm hemangioendotheliomas, and hepaiio 
hemangioendotheliomas. This can prove to be quite 
challenging. The aim is not to cure but to stabilize 
deleterious effects such as hemorrhage and throm- 
bocytopenia. 



F. Marshalleckand M.John; 



embolization of high-flow mal- 
j proper planning and the use 



Endo\ 
form at io 

of microcatheter techniques. Although alcohol 
may produce the best result, it should be used with 
extreme caution due to its local and systemic effects. 
Although a cure may not be possible, relief of symp- 
toms can be achieved in the majority of patients. 

Percutaneous sclerotherapy of low-flow vascular 
malformations can be technically straightforward, 
but requires pre-procedure and post-procedure 
planning in order to avoid serious complications. 



1. Mulliken ]B, Young A i [ ; 'f S: Vjslii1:3j' bi::h marks: heman- 
giomas and mal for ma: ions. Saunclers, Philadelphia 

2. Mulliken I, Glowaki I i:9B2; Hemangiomas and vascular 
malformations in infams and children: a classification 
based on endoihelia! characteristics. Plas: jieconstr Surg 
69:412 

3.Mulliken, JB, Fishman SJ, Burrows PE (2000] Vascular 
anomalies. Curr Probl Surg 37:517-584 

4. Wilt PD, Mulliken JB ( 1 999) Hemangiomas and vascular 
[•.; a [formation-:. Publicaiions com mi: lee, Clef: Palate Foun- 
dation 

5. Burrows PE, Laor T, Paltiel H et al. (1998) Diagnostic Imag- 
.ng in li:e ev .illation of vase ill;'.: bnoh marks. Pediatr '. term 
16:455-438 

6.Frieden I], Reese V, Cohen D (1996) PHACE syndrome. 
The association of posterior fossa brant malformalions, 
1: em. angiomas, arter;;:. an:' makes, coa re ration -:A~ I he aorta, 
cardiac dejects and rye aPnoimalities. A:ch Permalol 
132:307-311 

/.Gangopadhyay AN, Sinha CK, Gopal SC et aL (1997) The 
role o: steroids in childhood hemangioma: a I year review. 
Int Surg 82:49-51 

8. Bennett ML, Fleischer AB, Chamlin SL et al. (2001} Oral 
corticosteroids are effective for cutaneous hemangiomas: 
.in evidence based evaluation. Arch Dermatol ] 37:1208— 
1213 

9.AkyuzC,YarisN, KutlukMT et ak(2001) Management of 
cutaneous hemangiomas: a reirospective analysis of 1109 
cases .Hid comparison o: convention;:, prednisolone with 
high-dose meilivipjeonisolone literacy l-'edia:r Hematol 
Oncol 18:47-55 

O.Deb G, Donfresco A, De Sio L et al. (1996) Treatment of 
Item angiomas in infants and babies with interferon a I fa -2a: 
preliminary results. Int | Pediatr Hematol Oncol 3:1-16 

l.EnjolrasO.RkheMC.MerlandJJetal. (1990) Management 
of alarming hemangiomas in infancy: a review of 25 cases. 
Pediatrics 85:491 

2. Ezekowitz RAB, Mulliken JB, Folkman J (1992) Interferon 
i!lpha-2a therapv for lire threatening hemangiomas of 
infancy. N Engl J Med 326:1456-1463 

3.Barlow CF, Priebe CL, Mulliken JB et al. (1998J Spastic 
am leg i a as .■ coma licai.oi: !;i>rle:o:t alaiia- 
of hemangiomas of infancy | Pediatr 1 32:527-530 

4.Achauer MB, Chang CJ.Vander Kam VM (1997) Mai 



ment of hemangioma in infancy: review of 24a- patients. 
Plast Reconstr Surg 99:1301-1308 

5. Chiaverini C, Kurzenne J Y, Rogopoulos A et al. (2002) Non- 
invola:ing congeniial r.rmangi' ■:::.:: 2 cases. Ann I 'ermatol 
Neverol 129:735-737 

S. Leikensohn JR, Epstein LI, Vasconez LO (1981) Superse- 
lective embolization .uvi surgery of noiiinvoluimg hem- 
angi:::m;:s and A-V malfo:m;:l.ons. Phtst Reconstr Surg 
68:143-152 

7. Hosono S, Ohno T, Kimoto H et al (1999) Successful trans- 
c:::ai:eo::s emh'j-ta a an ill r.emangi' -ma associated 

will: high out pul card .a c failure and Kasabacfi-Mniit: syn- 
drome in a neonate: a case report 

S.Zukerberg LR, Nickoloff BJ, Weiss SW (1993) Kaposi- 
form Hemangioendothelioma of infancy and childhood, 
.)n aggressive neop.asn: associated will: Kas.:bach-\!er- 
ritt syndrome and lymoi:ang;oma:esis. An: : Surg Pathol 
17:321-328 

3. Sarkar M, Mulliken IB, Kozakewich HP et al. (1997) Throm- 
bocv lope in c coagulopathy i [•" lsanach-Meritt phenom- 
enon) is associated wit it Kaposiform Hemangioeitdothe- 
lionu mid ii'": will: commoi: .maiiiile hemangioma. l-lasl 
Reconstr Surg 100:1377-1386 

3. Vin-Christian K, McCalmont TH, Frieden IJ (1997) Kaposi- 
form hemagiontdolheloma. An aggressive, .oca.ly mvasive 
vascuhtr tumor that can mimic hemangioma of inntney. 
Arch Derm 133:1573-1578 

I. Shin HY, Ryu KH, Ahn HS (2000) Stepwise multimodal 
approach in die treatmen: ::■!" Kasaoitch-Merritt svndrome. 
Pediatr Int 42:620-624 

2.Haisley-Royster CA, Enjolras O, Frieden IJ et al. (2002) 
Kasanach- Merrilt phenomenon: a retrospective sludv 
of treatment with vincristine. I Pesliat: Hematol Oncol 
24:459-462 

!.OginoI,TorikaiK, Kobayasi Set al. (2001) Radiation ther- 
apy for life- or function- llneatening mfan: hemangioma. 
Radiology 218:834-839 

1. Drolet BA, Scott LA, Esterly NB et al. (2001 ) Early surgical 
intervention in ;: pa:ie:i: with Kasabach Mniitt phenom- 
enon. J Pediatr 138:756-758 

5. Yakes WH Kossi P, Odink H (1996) Arteriovenous malfor- 
ntatiirn management, Giidiovasc Interv Radiol 19:65-71 

^ Armstrong DC, Brugge K (2000) Selected interventional 
::!i>h. p i1-!i pi ; ot pediatric head and necs vascular lesions. 
Neuroimagng Clin North Am 10:271-292 

7 Vmt". W'i- .004: zjioovascula: managemen: o:' malignanl 
pcuiLiiiiL hemangion:;: and kaposiform hemangioendc'- 
thelioma. Presented at the 2004 SIR 29th annua] scientific- 
meeting 

i. Daller JA, Bueno J, Gutierrez J et al. (1999) Hepatic Heman- 
gioendothelioma: clinica. experience and :::;:;;agen:enl 
strategy. J Pediatr Surg 34:98-106 

3. McHughK, Borrows PE (1992) Infantile hepatic hemangio- 
endoli'.eliomas: significance of poioal venous and systemic 
collateral supply. J Vase Interv Radiol 3:337-344 

D.Kassarjian A, Dubois J, Burrows PE (2002) Angiographic 
c.assificaiion o:' Hepauc Hemangiomas ,n Imams. Radiol- 
ogy 222:693-698) 

1. LarcherVF, Howard ER, Mowat AP (1981) Hepatic heman- 
g.o:::;:: diagnosis and management. Arch Ins Child --<'i7-]4 

2. Warman S, Bertram H, Kardoff R et al. (2003) Interven- 
tion.'., therapy of ! ma mile Hepatic Hemangioma. ) Pediatr 
Surg 38:1177-1181 



Percutaneous Management of Heni.ing.omas ami Vascular Malformations 



3. Stanley P, Grinnell VS, Stanton RE et a 1. ( 1 983) Therapeutic 

embolization of .mainile hepatic heju.hiig.-.jma with b-'V'A. 
AIR 141:1047-1051 

4. PeusterM.Windhagen-MahnetB, Fink Fetal. (1993) Inter- 
ventional therapy for :: l- n : .i 1 1 u i ■ ■ = n i.l ■.:■ ; h e , i ■.:■ j :i ;"i of die liver 
in a newborn infant using a central venous appro.; oil. Z 
Kardiol 87:832-836 

5. Burke DR, Verstandig A, Edwards O et al. (1986) Infantile 
li e in ;i ng j .:■ =? n ... ■:■■: h c. i out::: a ngioi: rapine U.) lines a:' J factors 
determining l he efficacy of hepatic arterial embolization. 
Cardiovasc Interv Radiol 9:1 54-1 57 

6.Enjolras O.Mulliken ]B (1997) Vascular tumors and vascu- 
lar malformations, new issues. Adv Perinatal 13:375-423 

7.Beggs I, Garvin DD (1983) Pelvic arteriovenous malforma- 
tion in a man. Br J Clin Pract 37:186 

S.Neifeld JP, Doppman JL, Chretien PB (1975) Congenital 
pelvic arteriovenous fistulas: report of a case and review 
□f literature. J Urol 114:648 

9.Flye MW, Jordan BP, Schwartz MZ (1983) Management of 
congenital arteriovenous malformations. Surgery 94:740 

0. Brinley JL, Palubinskas AJ ( 1 977) Congenital arteriovenous 
malformation of the pancreas. Br J Radiol 50:219 

l.Mizutani N, Masudo U, Naito N et al. (1981) Pancreatic 
arteriovenous mo J formal ion in a panent with gasno-intes- 
tinal hemorrhage. Am I Gastroenterol 76:141 

2.Pinkas J, Djaldetti M, DeVries A et al. (1968) Diffuse angi- 
omatosis wiih Hypersplenism: Splenectomy followed bv 
polycythemia. Am ] Med 45:795 

3.Frieden I, Enirolas O, iisierly N r Voosular birthmarks and 
oilier abnormalities o: blood vesse.s and lymphatics, chap 
20 Iwivw.i', a rcour:- internal ion a I.com,'e-books/por75i> l ''.pdf 1 

-.. Voge.zmie ^ :200 in High -flow AVMs: moo em therapy with 
absolute alcohol. Preseniecl at die 200? Midwest Institute 
for Interventional Therapy :MIIT! conference 

5. Burrows f'E (coordinator; Vascular anomalies: interven- 
tions and patient management, -'reseiiied oi the J004 olR 
29th annual scientific meeting 

6. Kaufman SL, Kumar AAJ, Roland J MA (1980) Transcatheter 
emboloiherapv in ike management of congenital arteriove- 
nous malformations. Radiology 137:21-29 

7.¥akesWF, PeosnerPH, Reed MDetal. (1996) Serial embo- 
li zati 'ins of an exlremity arteriovenous malformation with 
alcohol via direct percutaneous puncture. A;R 146:1038- 
1040 

8. Doppman ]L, Pevsner P (1983) Embolization of arteriove- 
nous malformations bv direci percutaneous nunc lure. AIR 
140:773-778 

9.Abe FJ, TenBroek FW, van Schaik JPJ et al. (1997) Trans- 
venous embolization of an arteriovenous malformation 
of the mandible via a femoral approach. Pediatr Radiol 
27:855-857 

O.White RI, Pollack JS, Wirth JA (1996) Pulmonary arterio- 
venous malformations: diagnosis and transcatheter embo- 
lotherapy. JVIR 7:787-804 

1. Pollack J, White R (1996) Pulmonary arteriovenous mal- 
formations. SCVIK svllabus Ihoracic ami visceral vascular 
malformations 

2. Lee DW, White Jr. RI, Egglin TK et al. (1997) Embolother- 
apy i:t large pulmonary arrerionevous malformations: long 
term results. Ann Thorac Surg 64:930-939 

3.Clouse ME, Levin DC, Desautels RE (1983) Transcatheter 
embololhera.'v tor congenital renal arteriovenous m.ilfo- 
mations. Long term follow- up. Urology 22:3<a0-365 



Nakai M,Nakamura N, Suzuki Yet al. (20-33 1 Transcatheter 
arterial embolization with n- butyl 2-cyanoac.rylaie ihysto- 
acryl) for renal arteriovenous malformation: case report. 
Hinyokika Kiyo 49:51-53 

Morita T.Uekado Y, Kyoku et al. (1989) Transcatheter arte- 
rial embolization in pa i inns w.tit renal arteriovenous mal- 
formation: a reporl of two cases. Honyokika Kiyo 35:1 761- 
1765 

Ghai S, Rajan DK, Asch MR et al. (2003) Efficacy of emboli- 
zation in traumalic uterine vascular malformations. I Vase 
Interv Radiol 14:1401-1408 
Ricolfi F.Valiente E, Bodson F 
fistulae complicating central vi 
of end ovascu'ar treatment based on a series ot seven cases. 
Intensive Care Med 21:1043-1047 

DeSouza NM, Reidy JF (1992) Embolization with detach- 
ao.e balloons - applications ouisicle the head. Clin Radiol 
46:170-175 

Herbreteau D, Aymard A, Khayata MH et al. (1993) Endo- 
vasctdar treatment of arteriovenous fistulas arising from 
branches of the subclavian artery. 1 Vase Interv Radiol 
4:237-240 

Yakes WF, Luethke JM, Merland JJ et al. (1990) Ethanol 
embolization of arteriovenous fislulae: a primary mode of 
therapy. J Vase Interv Radiol 1:89-96 
Sprouse LR 2nd, Hamilton IN Jr (2002) The endovaseular 
treatment of a renal arteriovenous fistula: placement of a 
covered stent. J Vase Surg 36:1066-1068 
Ninh TN, Ninh TX (1974) Cystic hygroma in children: 
report of 126 cases. J Pediatr Surg 9:191 
Sumner TE, Volberg FM, Riser PE et al. (1981) Medistinal 
cystic hygroma in children. Pediatr Radiol 1 1:160 
Grosfeld JL, Weber TR, Vane DW (1 982) One stage resection 
for massive cervieomediastinal hygroma. Surgery 92:693 
Groves LK, Effler DB (1954) Primary chylopericardium. N 
Engl J Med 250:520 

Stratton VC, Grant RN (1958) Cervieomediastinal cystic 
hygroma associated widi chylopericardium. Arch Surg 

Csicsko JF, Grisfeld JL (1974) Cervieomediastinal hygroma 
with p ti] mo n a rv .tv nop. as. a in the newborn. Am I ids Child 
128:557 

Burrows PE, Mason KP ;2004! Percutaneous treatment 
of iow flow vascular malformations. I Vase Interv Radiol 
15:431-450 

MolitchHI, UngerEC.WitteCLetal. (1995) Percutaneous 
scleroiherapv of Ivmph angiomas. Radiology : ; '4:3-i3-347 
idiei.s Wti.Sc.erot'ierapy of Lymphatic Malformations, k re- 
sented :tl the 2:1:04 annual scientific meeting of the Society 
of Interventional Radiology 

C.ertcke KR 1 : 3S L) : I >oxycyclnie as a sclerosing a gen I. Ann 
Pharmacother 26:648-649 

Sung MW, Chang SO, Choi JH et al. (1995) Belomycin 
scleroiherapv in patients with congenital lymphatic mal- 
formation of the head and neck. Ant I utclarngol l(':236- 
241 

Giguere CM, Bauman NM, Sato Y et al. (2002) Treatment 
ot lymphangiomas with 0\ -432 ! Pic ib anil i sclerotherapy: 
a prospective multi-institutional tri.i. . Arch Otolaryngol 
Head Neck Surg 128:1137-1 144 

Laranne J, Keski-Nisula L, Rautio R et al. (2002) OK-432 
therapy for lymphangiomas in children. Eur Arch Oto- 
Rhino-Laryngol 259:274-278 



F. Marshalleck and M. Johnson 



75. Luzatto C, Midrio P, Tcliapr.'.sr.Mn Z et al. (2000} Sclerosing 
treatment of lymphangiomas with OK-432. Arch Dis Child- 
hood 82:316-318 

76. Smith RJ, Burke DK, Sato Y et al. (1996) OK-432 therapy 
for lymphangiomas. Arch Otolaryngol Head Neck Surg 
122:1195-1199 

77.Lanigan SW (2001) Treatment of vascular nevi in children. 

Hosp Med 62:144-147 
78. Lam SM, Williams EF (2004) Practical considerations in 

:he treat men; of capillary vascular :i; informations, or port 

wine stains. Facial Plast Surg 20:71-76 



79. Richards KA, Garden [M (2000) The pulsed dye laser for 
cutaneous and nonvascular lesions. Semin Cutan Med Surg 
19:276-286 

80. Mason KP, Neufeld EJ, Karian VE et al. (2001) Coagulation 
abnormalities in pediatric and adult patients after sclero- 
tlicrapy or embolization of vascular anomalies. Radiology 
217:1359-1363 

81. Hammer FD, Boon LM, Mathurin P et al. (2001) Ethanol 
i oleic ih era py of venous m;'. I formations: evaluation of sys- 
temic ethanol contamination. J Vase Interv Radiol 12:595- 
600 



2 Predominantly Venous Malformation 



Introduction 21 

Genetics 21 

Clinical Features 21 

Fix j I ,ii].i I ' l t r" l i -s e Veil on s Yolloriuaiions 11 

Associated Syndromes 22 

Blue Rubber Bleb Nevus Syndrome 22 

[vtuc'xuuii'.eous i-omikol Ve:'.o'.:f M.dfcim.Lilio 

Glomovenous Malformations 22 

Maffucci's Syndrome 22 

Coagulopathies 22 

His top atho logy 22 

Imaging 23 

Plain Radiograph 23 

i '■ 'ppler Ultrasound 23 

Computed Tomography 24 

Ivi.igntuC Resonance jii.ig.ng 24 

A! ;e: iography 24 

I 'irect Percutaneous Venography 25 

Peripheral Venography 25 

Management 26 

Medical Treatment 26 

Sclerotherapy 26 

Technique 27 

Sclerosing Agents 28 

Laser 29 

Surgery 29 

Pharmacologic Treatment 29 

intensive Care Unit Management 29 

Follow-up 31 

Outcome .'1 

References 31 



Introduction 

Vascular anomalies are divided into vascular 
tumours and vascular malformations. Vascular 
malformation classification is based on the anoma- 
lous channels: arterial, venous, lymphatic, or cap- 
illary. The most frequent vascular anomalies are 
venous malformations. The incidence is estimated 
to be around 1 in 10,000 [1]. The reader must be 
made aware of the numerous confusing n 
such as glomangioma, cavernous haemangio 
and haeman^ioma that have also been used inp 
literature. 



Genetics 

Venous malformations are sporadic in most cases 
but can be inherited. A locus for autosomal-domi- 
nant multiple cutaneous and mucosal venous mal- 
formations, VMCM1, was identified on chromosome 
9p21 [2-4]. A mutation was found in the endothelial 
cell-specific receptor tyrosine kinase TIE-2 [5]. This 
mutation is likely to occu 



ular malforma- 



Clinical Features 



Professor oi Radiology. Pediatric and Interventional Radi- 
ologist, Deportment of Medical Imaging, Hopit.il Ste-itisline, 
3175 Cote Ste-Catherine Road, Montreal, Quebec H3T 1C5, 



Generally, ven 


ous malformations are noted at 


birth but can 


also appear during infancy. They 


involve any tis 


sue or organ in all anatomic loca- 


tions. Venousn 


alformations grow with the patient. 


Exacerbations 


can occur during puberty or preg- 


nancy. Indeed 


hormonal modulations of venous 


malformations 


can be seen during the menstrual 


cycle or unde 


r anovulant therapy. Surgery or 


trauma can als 


o lead to the progression of venous 



ormal, 

.. Hemato- 
must include 
and d-dimer 



malformations. At clinical exam, venous r 
formations appear as soft tissue lesions that 
compressible, and increase with Vali 
vre or gravity. The overlying skin c; 
bluish, or purple in colour. The lesii 
not pulsatile. Bleeding i 
logic evaluation is important an 
complete blood count, fibrinogei 
dosage. Localized intravascular cons 
be observed, especially in extensive venous mal- 
formations [6], Coagulopathy can be exacerbated 
by sclerotherapy or surgery [7-9]. Extensive facial 
venous malformations are frequently associated 
with intracranial developmental 
lies [10]. 



Focal and Diffuse Venous Malformations 



2.5.3 

Glomovenous Malformations 

Glomovenous malformations are venous malforma- 
tions associated with glomus cells. The smooth layer 
is formed by glomus cells, which are smooth muscle 
precursor cells [11]. Glomovenous malformations 
frequently recur. Sclerotherapy can be useful for 
their treatment. 



2.5.4 

Maffucci's Syndrome 

Maffucci's syndrome consists in the association of 
venous malformations and multiple enchondro- 
mas. Intraosseous venous malformations as well as 
enchondromas are responsible for the bony defects 
[12]. 



Venous malformations are either focal or diffuse, 
and may involve the skin, the mucosae, the mus- 
cles, the bones. In cases of extensive venous mal- 
formations, associated distal sites of involvement 



Associated Syndromes 

2.5.1 

Blue Rubber Bleb Nevus Syndrome 

Blue rubber bleb nevus syndrome, a rare disorder, is 
characterized by continuous development of multi- 
ple focal venous malformations of the skin, muscu- 
loskeletal tissue, and mucosa throughout the body, 
including the gastrointestinal tract. These venous 
malformations can be treated by sclerotherapy or 
surgery. In some cases of blue rubber bleb nevus, 
it has been shown that the mutation may occur on 
chromosome 9p. 



Coagulopathies 

Localized intravascular coagulopathy is reported in 
venous malformations. Most patients with venous 
malformations have no clinical signs or symp- 
toms at presentation. A chronic form of consump- 
tive coagulopathy is however possible as shown by 
positive d-dimer levels, and normal or low platelets 
and fibrinogen values. Consumptive coagulopathy-' 
must be corrected prior to initiating the treatment 
of vascular malformations. Low- molecular- weight 
heparin and elastic stockings are recommended [8]. 
Administration of cryoprecipitate, platelets, or fresh 
frozen plasma to patients with chronic coagulopathy 
has proved to be helpful before performing sclero- 
therapy or embolization in order toobtai 
ful thrombosis [9]. 



Histopathology 



2.5.2 

Mucocutaneous Familial Venous Malformations 



we due to the abnormal 
i wall. Microscopy shows 



The characteristics of 
venous malformations are the s 
blue rubber bled nevus syndroir 

of gastrointestinal involvement. 



Venous maltormatio 
development of the 

multiple thin-walled vessels with flattened endothe- 
familial lial cells, lacking proliferative I eat u res [1] (quiescent 

s those of the endothelium}, and wall smooth muscle deficiency. 

pt for the lack The deficient smooth muscle layer leads to the ina- 
bility of the affected veins to constrict normally, 



!- : red om mainly Veo.ous Malformation 

inducing stagnation ol blood, thrombosis, throm- 2.8 
bolysis, swelling, and pain [13]. Venous mnlform.i- Imaging 
Cions do not exhibit up-regulation of angiogenic 
factors or matrix enzymes [14]. 2.8.1 

Plain Radiograph 




Fig. 2.1. Sixteen-year-old male. Lateral X-ray of the left wris 
shows a ventral sect liss-;e mass associated with iypical phle 
oclilh in a case of vetoes malformation 



Plain radiographs can show phleboliths (Fig. 2.1) 
that are suggestive of venous malformation but 
rarely present. Soft tissue swelling and bone deform- 
ity can be seen. 



2.8.2 

Doppler Ultrasound 

Doppler ultrasound is the initial modality of choice 
to differentiate venous malformations from other 
vascular abnormalities. Ultrasound is performed 
with a high-frequency linear array transducer (5-12 
MHz}. On grey-scale images, venous malformations 
appear as compress ihlt-ltypoedioic or heterogeneous 
lesions [15, 16] (Fig. 2.2a-c). Calcifications (demon- 
strated in less than 20% of cases), are quite specific 
of venous malformation; anechoic channels can be 
seen. Doppler examination displays a monophasic 
low-velocity flow (Fig. 2.3). In 20% of venous mal- 
formations, no flow can be demonstrated. Dynamic 
manoeuvres such as Valsalva or manual compres- 
sion are sometimes necessary to induce a visible 
Doppler flow. 



LfeC: 




Fig. >.2a-c. Six-year-old boy with a stable soft tissue mass of 
the right temporal region. \> I 1 ? shows a:i heterogeneous hvp- 
oechoic well-dekmiled superficial lesion containing anechoic 
structures, b The same lesion is compressible and seems nor 
vascularized, c Low velocity flow is detected when the com- 
pression is released (venous malformation) 












4VK9 




Fig. 2.3.! : op.\er UJ i:'. a:io:he: pa:ient wilh .1 s;:p eiTiCi.il veno 
in aifoi:':.u :o:~. shows lypica! venojs flow on pulse;". Poppler 



2.8.3 

Computed Tomography 

Computed tomography (CT) demonstrates the exten- 
sion of venous malformations but the contrast resolu- 
tion is less than with MRI. The lesion is hypodense 
or heterogeneous with a slow contrast enhancement 
after injection of contrast material (Fig. 2.4a,b). Phle- 
boliths, when present, are easily seen. 



or fast spin-echo Tl-weighted sequences and T2- 
weighted sequences with fat suppression (Fig. 2.5a- 
d). T2-weighted gradient-echo sequences can be 
useful to demonstrate calcification or hemosiderin. 
On gradient-echo sequences, the absence of intra- 
vascular signal suggests a slow-flow malformation 
[17]. Tl-weighted sequences with fat suppression 
and gadolinium injection have to be performed to 
evaluate the perfusion of the malformation. 3D-FISP 
(Fast Imaging with Steady Precision) phlebography 
sequence allows the evaluation of draining veins 
[18]. On Tl-weighted sequence, venous malforma- 
tions are hypo- or isointense. The signal is heteroge- 
neous when haemorrhage or thrombosis is present. 
On T2 -weighted sequence, the venous malformation 
is hyperintense. Areas of hyposignal are related to 
thrombosis, phleboliths, or septa, most evident on 
gradient images. Small fluid-fluid levels can be 
seen. After sclerotherapy, the lesion becomes het- 
erogeneous on both Tl- and T2-weighted sequences. 
Post gadolinium sequences are useful to evaluate 
the residual perfusion. MRI is very helpful for the 
delimitation and assessment of the extension, but is 
not specific of venous malformations. MRI results 
have to be correlated with clinical findings and Dop- 
pler examination to reinforce the diagnosis. 



2.8.4 

Magnetic Resonance Imaging 



Magnetic resonance imaging (MRI) 
lodality to evaluate the extension of v; 



2.8.5 
Arteriography 



ularmal- Arteriography of the affected limb i 



t clinic ally 
formations. The MRI protocol includes spin-echo useful, showing either noanomaliesorthe late op.Kiti- 




Fig. 2A. a Axial unenhanced c 


ervical CT of a 


i i-year-old female shov. 


s a slightly i'.ypodense mas 


s ■.".out;'. in: rig .\ calcified phle- 


bolith in the left parotid regio 


n.b After contra 


st injection, the venous 


ma 1 forma tion sligi'.dy enh;'. 


aces, remains hypodense and 


is be;ter delineated 











H'eduniiiianily Wr.oiis M;;!jb:';".\'.ti::i 




Fig. 2. 5., i ; :■ 'i ■.■.■.,: I ".". v.-, r ig:i>d MM ,.:.:w- •-..i.:i.v \\y(v: i:v.;j".- : r :'."..: iv \:.::\r i .iz.'. : -..:".i.i." ii^reps. b In short ti 
(STIR) sequence, the well-delineated mass is hyper intense, c In T2- weigh ted image, the lesion remain: 

d Coronal 11 -weigh .cd scjii after g:i.:cjn:iini injcij; ion .-,ai fat ^1 in nil ion allows skgi-.l m-J h^erogeiieotii enhancement 



cation of venous spaces with or without dysplasic 

nels.However, ;)[is;ii'j;]M|'liy is sometimes perfori 
rule out the presence of an arterial microfistula 



2.8.6 

Direct Percutaneous Venography 



:han- obtained, the opitcil icat ion with a low osmolarity 
tedto iodinated contrast is recorded, as a phlebogram. 
Three different patterns can be seen: a cavitary 
pattern with late filling of normal draining veins 
(Fig. 2.6), a spongy appearance with tiny cavities 
and late venous drainage (Fig. 2.7), and the dysmor- 
phic veins pattern (Fig. 2.8). 



Direct percutaneous phlebography is useful in some 
atypical venous malformations to allow a precise 
diagnosis and mapping of the malformation. Under 
ultrasonography, direct puncture of the malforma- 
tion is performed with a 20- or 21-gauge needle. 
The needle is connected to a syringe through an 
extension tubing and is progressively withdrawn 
while applying slight suction. Once blood return is 



2.8.7 

Peripheral Venography 

This technique is useful in the presence of complex 
venous malformations to evaluate the communica- 
tion between the dysmorphic veins and the normal 





Fig. 2 A Percutaneous Venography opacifies 
l.-le tilling .it :'•:■! ni.i! ;i -.lining wins 



■vith Fig.2.7.PeiY'.i;iinecus phlebography o\ a large spongy vi 
malformation of the right iliac fossa 



Management 

A multidisciplinary approach is essential to plan 
a treatment strategy specific to each patient. The 
modalities of treatment depend on: the localization, 
the size, the extension, the functional repercussion, 
and the aesthetic impact of the venous malforma- 
tion. The goal is to lessen the symptoms and improve 
the functional consequences. The management of 
venous malformations includes compression, resec- 
tion, and obliteration of the channels lumen by scle- 
rosing injection or laser photocoagulation. 



2.9.1 

Medical Treatment 




Asymptomatic venous malformations should be 
treated conservatively. Counselling regarding the 
hazards of puberty, oral contraceptive medication, 
and pregnancy has to be provided. Extensive arm 
or leg venous malformations should be treated with 
elastic stockings. Ant i-int lain ma toiy drugs are help- 
ful during the symptomatic thrombosis-related epi- 
sodes. Cyclooxygenas [ '-2 (COX-2) inhibitors (such as 
Celebrex) have also proved to be effective against 
pain. Low- molecular-weight heparin may also be 
used to decrease the disseminated intravascular 
consumption and is recommended in the preopera- 
tive preparation prior to resection. Corticosteroids, 



interferon, and other antiangiogenic drugs have 
proven useless in the treatment of venous malfor- 

[mruoiis. 



2.9.2 
Sclerotherapy 

Sclerotherapy, with or without surgery, is the treat- 
ment of choice for symptomatic venous malforma- 
tions. Sclerosants destroy the vascular endothelium 
through different mechanisms depending on the 
agents: chemical agents (iodine or absolute alcohol); 



H'edoninianilr Ver.ous Mi'.ooinwtio 



osmotic effect (salicylat 
detergents (morrhuate sc 
canol, and diatrizoate soi 



2.9.2.1 
Technique 



or hypertonic saline); 
m, sotradecol, polido- 
Ti)[13](Fig.2.9a-d). 



Sclerotherapy is performed under fluoroscopic con- 
trol. The puncture with a 24 or 22 Cathlon or 20- 
gauge Teflon intravenous cannula or other needle 
system can be done under ultrasound, CT, or MRI 
guidance. It is necessary to ve no graphic ally assess 
the draining veins before injecting sclerosing agents 



in order to prevent complications. Indeed, an appro- 
priate opacification is essential to evaluate the pat- 
tern of the lesion, its volume, and the draining veins. 
The amount of agent injected is estimated by the ini- 
tial amount of contrast medium needed to opacify 
the lesion before the visualization of the draining 
veins. Additional sclerosant can be needed in order 
to obtain a firm lesion on palpation or to dry up 
the blood return on aspiration of the cannula. A 
tourniquet can be useful. An automated orthopaedic 
tourniquet inflated to a pressure below systolic arte- 
rial pressure, provides the most effective method of 
control [13]. The cuff pressure can be adjusted with 
test injections of contrast medium until the drain- 



1 


in 


1 


• 



(ur-year-old boy with v. 
r venous m a Ifor nation. 
] important regression 



ins malformation of the thigh, a Coron.U an J b axial : iSt MRI show a well delimitated 
x months after three sessions of sclerosing ireaiment wi:h alcohol c coronal and d axial 



ng veins no longer till. After injection of moder- continuous pulmonary artery pre 

ite amounts of absolute ethanol into sequestered during the procedures [23, 26]. 

'enous malformations of the limb, the tourniquet 

s kept inflated for 20-30 min, to minimize the risk Detergent-Type Sclerosants 

)f egress of ethanol or clot into the draining veins. 

t is wise to release the tourniquet slowly [13]. The The detergent- type of sclerosants . 

ourniquet use is controversy. Some groups prefer clerol (polidocanol l%-3%, sodiui 

o use amanual compression wirh a gradual decom- fate (STS), sodium morrhuate, and ethanolamine. 

iression to avoid pulmonary emboli. Theses detergents can be used in liquid or foam. 



in ito ring 



icludes: aetoxis- 
tetradecyl sul- 



Aetoxisclerol (Polidocanol) (3%) 



Ethanol (95%-98%) 

Absolute ethanol is the most frequently used agent, 
and is also the most potent and destructive on the 
vascular endothelium. Ethanol causes an instant 
precipitation of endothelial cells proteins and rapid 
thrombosis. Injection: An undiluted form of etha- 
nol, or opacified ethanol with oily contrast medium 
(9:1 or 10:2), or opacified ethanol with metrizamide 
power under fluoroscopy monitoring [19]. Dose: The 
total dose of lml/kg (or 60 cc) per session should 
never be exceeded [13]. Ethanol blood levels cor- 
relate directly with the amount of ethanol injected. 
Complications: Ethanol is the most effective sclero- 
sant available but it also results in the most serious 
side effects. The most common complication of eth- 
anol is local tissue injury, such as skin necrosis (in 
10%-15% of cases) [13] or peripheral nerve damage 
(in approximately 1% of cases) [13, 17, 20, 21]. Most 
complications are transient, but permanent injury 
has been reported [20, 22, 23]. Complications rate 
for ethanol embolization ranges from 7.5% to 23%. 
Severe complications have been reported (cardiac 
arrest, pulmonary embolus) [23, 24]. Yakes et al. 
[23] reported four cases of cardiopulmonary col- 
lapse in out of over 50,000 embolizations or scle- 
rotherapy procedures. The mechanism remains 
unknown and may include pulmonary vasospasm, 
pulmonary embolism and direct card io toxicity. 
Central nervous system depression, hypoglycemia, 
hypertension, hyperthermia, haemolysis, pulmo- 
nary embolism, pulmonary vasospasm, cardiac 
arrhythmias, and electromechanical dissociation [9, 
13, 17, 21] have been reported in the literature [23, 25, 
26]. Continuous cardiovascular monitoring during 
the procedure is paramount. When large malforma- 
tions are being treated, some have advocated that 
such procedures be performed under general anaes- 
thesia. Some authors also recommend the use of 



This agent is used for the small venous malforma- 
tions. Polidocanol is effective by altering the vascu- 
lar wall. The emulsion o( netoMsdero! would allow 



/eins, thanks to 
on could direct 
is, sparing then 
ii Some authors 



for the visualization of the draining 

the bubbles; accordingly, this emuls 

the compression to the involved veil 

the normal adjacent veins. Injectior 

used it mixed with a lidocaine soluti 

pain after injection. Dose: The quantity is lcc per 

cavity [25] up to a total of 6 cc of polidocanol with 

0,2-1,0 cc of 1% lidocaine solution. Complications: 

Skin necrosis, sciatic neurolysis, and infections were 

reported in6-8% of cases [25, 27]. One cardiac arrest 

was also reported [28]. 

Sodium Tetradecyl Sulfate 
(Sotradecol-Elkins-Sinn, Cherry Hill, New Jersey) 

Sodium tetradecyl sulfate damages the endothelium 
resulting in thrombosis and fibrosis. This agent can 
be used in a liquid solution or by creating foam with 
air. The main difference between liquid solution and 
foam is the long life of the foam in the vein and 
by the clear separation obtained between the blood 
and the sclerosant [29]. Injection: We mix 5 cc of 
sotradecol with 2 cc of Lipiodol and 5-10 cc of air. 
Tessari described a stable and compact foam qual- 
ity using two plastic syringes and a three-way stop- 
cock, mixing ratio for sclerosant to air is 1:4 to 1:5 
[30-32]. The foam is obtained by mixing the agents 
(STS or polidocanol: air) through multiple passages 
between two syringes. Dose: The maximum dose is 
not well established. Complications: Skin necrosis 
was reported by O'donovan [33] in three out of 
15 patients. 

Ethanolamine Oleate 

Ethanolamine oleate is a mixture of 5% eth- 
anolamine oleate (Keuk Dong, Inchon, Korea} and 
iodized oil (Lipiodol) (ratio 5:1-5:2); the amount 



i-T^d'.'iiii^.iiiilv Veii'.'iii W; , .j"'"::'\':ti::' 



used ranges from 2 to 20 ml within 1 to 10 sessions. 
This salt of an unsaturated fatty acid has been 
used as a sclerosing agent because it has excellent 
thrombosing properties [34]. Approximately 50% 
of oleic acid combines with serum proteins within 
30 minutes [21]. This can cause renal toxicity in 
association with a marked intravascular haemo- 
lysis and hemoglobinuria and hepatotoxicity [34]. 
To prevent these complications, haptoglobin can 
be administrated intravenously during and after 
the injection of the sclerosant into the lesion [35]. 
Effective in 92% (23 out of 25) of cases, Konez et 
al. [36] reported the conjoint use of coils emboliza- 
tion as well as inflated balloons within the internal 
jugular vein in cases of cervicofacial venous mal- 
formations in order to prevent the systemic passage 
of the sclerosant. Trismus in two patients, abated 
completely within 1 week. 

Ethibloc (Ethicon, Hamburg) 

Ethibloc is not available in the United States. Ethi- 
bloc is a mixture of zein (corn protein), alcohol, 
and contrast medium. The sclerosing effect is due to 
the giant cell inflammatory reaction. Injection: The 
product is available in a preloaded syringe. Compli- 
cations: No significant lasting complication, except 
for the extrusion of the agent in 10% of cases [25]. 
Dubois et al. [37] reported 74% of good or excellent 
results in 28 out of 38 patients. 

Histoacryl 

Histoacryl is a biological product that undergoes 
polymerization on contact with an ionic substance, 
inducing then a permanent occlusion. Mostly used 
as a pre-surgical step, it was reported for the treat- 
ment of venous malformations of the orbit [38]. 
Delay in healing up may be due to the extrusion of 
the Histoacryl when left in place. 



In the presence of rapid venous drainage 
venous spaces, placement of coils can be useful to 
retain sclerosing agent and avoid pulmonary embo- 
lus, particularly in a venous malformations close 
to normal veins. Coils can be delivered directly 
through the access needle into the venous spaces 
or via the femoral or jugular vein (Fig. 2.10a-e). For 
limb superficial venous malformations, a periph- 
eral intravenous catheter can be useful to perform 
a phlebography during sclerotherapy for assess- 



ing the ischemic changes. Applying cold sterile 
saline onto the surface of the skin to induce local 
vasoconstriction seems to reduce the risk of skin 



damage [13]. 



2.9.3 
Laser 

Effective intralesional laser therapy was reported 
by Derby and Low [39] in cases of facial venous 
malformations. Pulse dye laser operation has been 
found to be most successful in removing hundred of 
lesions in cases of blue rubber bleb nevus syndrome, 
without recurrence [40]. 



2.9.4 
Surgery 

In the majority of cases, successful sclerosing treat- 
ment will obviate the need for surgery [41]. Surgery 
alone is only effective in well-defined and easily 
accessible lesions of moderate size in which the 
anatomy will allow for a maximal functional resto- 
ration. Serious complications such as bleeding and 
nerve injury can occur with surgery. 



2.9.5 

Pharmacologic Treatment 



Corticosteroids, interferon, other antiangiogenic 
drugs are useless in cases of venous malforma- 



Intensive Care Unit Management 



Airway obstruction in the case of neck lesions, 
and compartment syndrome in extremities lesions 
should be carefully observed and managed accord- 
ingly (tracheostomy, fasciotomy). Systemic corti- 
costeroid (dexamethasone 0.1 mg/kg intravenously 
every 8 h), and ice packs can be helpful to n 
the swelling. Gross hemoglobinuria c 
ally occur and is managed by hydratationand urine 
alkalization (95% dextrose and water mixed with 
75 mEq/L of sodium bicarbonate], administered at 
twice the maintenance rate. Urine is monitored visu- 
ally and usually clears within 6 h. 




Table 2.1. Matenols for sclerotherapy 

• Puncture needles 

• Butterfly needle: 25G or 

• Jelco 20G, 22G, 24G 

• Sclerosing agents 



HOW tO US? iOLMClflCJ 



m) 



Ethanol 7 cc ethanol '.villi irw.i'izaniide powdei i 3.75 g) or Max: 1 cc/kg 

With nonionic contrast medium 

_ l % Sodiu:v. Tel:.idecy. Wiih ivietriz.imide powder or Max:30 cc/session 
Sulfate (Sotradecol] Diluted contrast medium 

Foam = Mix: Maximum: 

(Fig. 2.111 • 5 cc Sotradecol not well established 

• 2 cc Lipiodol Our recommendation lii cc/sessio. 



i-redomiiianily Venous iv!;:!jb: :":• .;tj;:- 





2,11 
Follow-up 



Incase of skin necrosis, weekly surveillance is recom- 
mended, to exclude superimposed infection. When 
necessary, the referral to a specialized wound centre 
is mandatory for possible debridement and/or skin 
grafting. In the absence of complications, a 6-week 
post-procedure follow-up is indicated to evaluate the 
need for other si 



2.12 

Outcome 



The reported outcome of endovascular treatment of 
venous malformation has been extremely variable 
from one series to another. No series has been reported 
based upon strict MRI documentation and sufficient 
follow-up. No predictors of success have been clearly 
outlined. Overall, it appears that favourable results 
may depend on the extension of the venous malfor- 
mations and the number of sclerotherapy sessions. 
Recurrence due to recanalization of treated venous 
malformations is more likely in cases of diffuse 
involvement with associated coagulopathy [13]. 



l.VikkulaM, Boon LM, Mulliken :B [ii-D-l) yo.ecula: genet- 
ics of vascular malformations. Matrix Biol 20:327-335 

2. Boon LM. Mulliken JB, Vikkula M et al. (1994) Assignment 
■.'if .i loci.!-: :'■■[■ ,1,-,m jj-.j n l.v iiioeiiieo vtr.o:< mii!forma:ions 
to chromosome 9p. Hum Mol Genet 3:1583-1587 



3. Erouillard P, Vikk'.ila M (J. 00.31 Vascular malformations: 
localized defects :n vascular morphogenesis. Clin Genet 
63:340-351 

4.Gallione CJ, Pasyk KA, Boon LM et al. (1995) A gene for 
kimiii.il venous mjlformalions maps to chromosome '■p 
in a second large kindred. J Med Genet 32:197-199 

5. Vikkula M, Boon LM, Carraway KL 3rd et al. (1996) Vascu- 
l..ir dvsiv.oipi'.ogenesis causeo by .in activating mutation in 
the receptor tyrosine kinase TIE2. Cell 87:1181-1190 

6.Mazoyer E, Enjolras O.Laurian Get al. (2002) Coagulation 
aono;mali;ies assoc.iaieo with .occ-nsive veii::'US iiialfoima- 
iLons of the limbs.: different sot son fiom Xasabacli-Meiiit; 
syndrome. Clin Lab Haematol 24:243-251 

7.Aronoff DM, Roshon M (1998) Severe haemorrhage com- 
plicating Uie Kjooel-Trenaunav-VVebei' syndrome. South 
MedJ91:1073-1075 

8. Enjolras O, Ciabrini D, Mazoyer E et al. (1997) Extensive 
pine venous malformations in die upper or loner limbs: a 
review of 27 cases. J Am Acad Dermatol 36:219-225 

9.Mason KP, Neufeld EJ, Karian VE et al. (2001 ) Coagulation 
aonoimaliiies in peduiiic ;md adul: panenis afier sclero- 
iher.ipy o: embolization of vascular anomalies. AJR Am f 
Roentgen 177-13=9 13^? 

3. Bouko uz V, Hnjolras (), < iu;chard ;i' el al. ( 1 996) Cerebral 
developmental ivr.ous amimakos is^'.iaind wi;h head 
and net* vtn:iui — aiTi::— .ali:ini. A INK Am ; NcTuradiol 
17:987 944 

l.Brouiliard I', Boon I .VI, Mulliken ,B et aL(2002) Mutations 
in a mivel Ijl:i::, )>kimul;n, are ri p s;'i!::s:hlc fur j{li>rr.u 
venous mnl'::rm i:i.>:.s ("(ikiman^iurr.is''). A:- | Hum 
Genet 70*66 8/4 

2.Mullikvn .«, hihman S.', Burrows I'h (2000) Vaicuiar 
anomalies. Curr Prohl Surg 37:51 7 >R4 

3. Burrows Pb, Mason KP (2004) Percutaneous treatment 
of low flow vascular malformations. I Vase Inteiv Kadioi 
15:431-445 

4. Takahashi K, Mulliken JB, Kozakewich HP et al. (1 994) Cel- 
iula: markers loh dis:ing'.:;sli ihe pluses of heiiiang.oma 
diirii'.g infancy and childhood. ■ Clin lures: 33:2357-J3(j4 

5.Paltiel HJ, Burrows PE, Kozakewich HP et al. (2000) Soft- 
tissue vascular a 11 ma lies: utility of Us for diagnosis. Radi- 
ology 0:4:747-754 

S.Trop I.Dubois J, Guibaud L et al. (1999) Soft-tissue venous 



malformations in pediatric and young adult rat ients: di.it- 
nosis with Doppler US. Radiology 212:841-845 
Siegel MJ (2001) Magnetic resonance of musculoskeletal 
soft tissue masses. Radiol Clin North Am 39:701-720 
Li W, David V, Kaplan R et al. (1998) Three-dimensional low 
ttofe gadolinium-enhanced peripheral MK venography | 
Magn Resort imaging 3:630-633 

Suh ]S, Shin KH.Na )B et al. (1997) Venous malformations: 
sclerotherapy with :i mixttue of elhanol and lipiodol. '2ar- 
diovasc Intervent Radiol 20:268-273 
Lee BB, Kim DI, Huh S et al. (2001) New experiences with 
absolute ethanol sclerotherapy in die mami genie nt of ,i 
comp>x :'::■[" n : of congenital ven.ot.s malformation. ; Vase 
Surg 33:764-772 

SukigaraM.OmotoR.MiyamaeT (1985) Systemic dissemi- 
nation ■:.( e lit a noli mine o.eate after auectioii sc.erotiirrapv 
for esophageal varices. Arch Surg 120:833-836 
Rautio R, Saarinen i, Laramie I et al. 12004! Endovascular 
treatment of venous malformations .n extremities: results 
of sclerotherapy and the otiaaly or life after I rea intent. Acta 
Radiol 45:397-403 

Yakes WF, Engelwood CO, Baker R (1993) Cardiopulmo- 
nary col. apse: sequelae of ethan.ol emoololherapv. Ra.no.- 
ogy 189:145 

Hanafi M, Orliaguet G, Meyer F et al. COS] ! Embolie pul- 
monaire ::u coins de la sclerotherapie percutanee d'un 
angiome vemeux sous anesihesie generate c.tez u:i enn'.nr. 
Ann Fr Aries th Reanim 20:556-558 

Blum L, Gallas S, Cottier JP et al. (2004) Sclerose percu- 
tanee drs maJoimahons veiner.ses stiperfic.edes: rtiide 
retrospective de 68 cas. J Radiol 85:107-116 
Hammer FD, Boon LM, Matfiurin P et al. (2001) Ethanol 
sclerotherapy or venous malformations: evaluation of sys- 
temic ethano. contamination. 1 Vase interv Raoio. I 2:?^5- 
60027. 

Cabrera J, Cabrera J Jr, Garcia-Olmedo A et al. (2003 I Treat- 
ment of venous malformations wilh sclerosant lii nucro- 
foam form. Arch Dermatol 139:1409-1416 
Marrocco-Trischitta MM, Guerrini P, Abeni D et al. (2002) 
Reversible cardiac arrest after pokdoc.inol sclerotherapy of 
peripheral venous malformation. I ■ermatol '-urg 23: i io-l :<:■ 



29. Frullini A (2002) Sclerosing foam in 

rent varicose veins. In: Henriet IP led ) Foam scleroiherapv 
s:..r.e :■[ Ihe arl. r_.o : ;i.oiis f'hlebologiques Franca.ses, Pans, 
pp 73-78 

30.Tessari L (2000) Nouvelle technique d'obtention de la 
sclem-mousse. Phiebog rapine 53:1 29 

31.Tessari L. Cave7.ii A, Frullini A 12001 ! Preliminary experi- 
ence with a new sclerosing foam in the treatment of vari- 
cose veins. Dermatol Surg 27:58-60 

32.Rabe E, Pannier-Fischer F, Gerlach H et al (2004) Guide- 
lines for sclerotherapy of varicose veins (ICD 10: 183.0, 
183.1, 183.2, and 183.9). Dermatol Surg 30:687-693 

33.0'Donovan )C, Donaldson JS, Morello FP et al. (1997) 
symptomatic hemangiomas and venous malformations in 
infants, children, and voting adults: Ireatment with percu- 
taneous mieciion of sodium tetradecv! sulfate. A|R Am I 
Roentgenol 169:723-729 

34. Choi YH.HanMH, O-Ki K et al. (2002) Craniofacial cav- 
ernous venous malformations: percutaneous sclerother- 
apy with use of ethanolamine oleate. I Vase Interv Radio! 
13:475-482 

3?. Hashizume M. ivitano rt.Yamaga H et a..l 1 '"vi) Ha mot loo in 
[0 protect against renal damage from ethanolamine oleate 
sclerosant. Lancet 2:340-341 

36.KonezO, Burrows PE, Mulliken JB (2002) Cervical venous 
ma I formal ions: \!R1 fea I tires and i liter vention.i. sua teg.es. 
interventional Neuroradiology 3:227-234 

37.Duboia J, Sebag GH, De Prost Y et al. (1991) The treat- 
ment of soft tissue venous malformation in children: 
percutaneous sclerotherapy with Et hi bloc. Radiology 
180:195-198 

38.Gobin JP (2001) The sclerotherapy position in recurrent 
varicose veins treatment. Int Angiol 20(2 Suppl 1):336 

39. Derby LD, Low DW (1 997) Laser treatment of facial venous 
vascular malformations. Ann Plasl y.ug 3^:37! -37? 

40.Olaen TG, Milroy SK, Goldman L et al. (1979). Laser sur- 
gery for blue rubber bleb nevus. Arch Dermatol 11 5: SI - 
82 

41.Hein KD, Mulliken JB, Kozakewich HP et al. (2002) Venous 
ma I tor ma uons of skeletal muscle. Plast Reconstr Surg 
110:1625-1635 



Trauma and Iatrogenic Lesions 



Recognition and Treatment of Medical Emergencies 
in the Trauma Patient 



Lucy Wibben 



* and Melhem J. Sha 



Inlioduction 35 

Airway Compromise 35 

Recognition 35 

Treatment Adjuncts 36 

Definitive Control 37 

Circulatory Shock 38 

Recognition 38 

Resuscitation 39 

Pitfalls of Resuscitation 40 

Cookbook: Recognition and Tiealmenl of Medical 

Emergencies in the Trauma Patient 40 

References 41 



ist may occasionally be the only physician avail- 
able to provide timely recognition and prompt, sys- 
tematic treatment of life-threatening emergencies. 
Adequate treatment of these emergencies is essential 
for optimizing the care of the trauma patient. This 
chapter will discuss recognition and treatment of 
two common life-threatening emergencies that the 
interventional radiologist may be the first physi- 
cian to encounter in the trauma patient. Definitive 
treatment will be presented for completeness and 
further study. 



Introduction 

The role of interventional radiology (IR) is expand- 
ing in the care of the acutely injured polytrauma 
patient. The interventional radiologist is often asked 
to perform therapeutic embolization of active bleed- 
ins from mlTnnhrlominal solid organ injuries and 
pelvic arterial injuries to provide initial control 
[1-3]. These patients are often accompanied by a 
traumatologist while in the interventional suite. 
They are often ventilated and actively receiving 
blood products. When these patients are critically 
ill and potentially unstable, the interventionist can 
delegate their critical care to the traumatologist. 
However, the challenge to the interventionist occurs 
in the setting of the "presumed stable" trauma 
patient with compensated shock. The intervention- 



Airway Compromise 

3.2.1 

Recognition 

The airway of an acutely unstable patient should be 
addressed first. The American College of Surgeons 
addresses the airway as the initial step of the primary 
survey in the Advance Trauma Life Support Protocol 
for evaluation of trauma patients (Table 3.1) [4]. The 
primary survey addresses all life-threatening inju- 
ries in an expedient manner by systematic review of 
all the systems. The primary survey protocol also 
works well to address any acutely decompensating 
patient. Airway emergencies in a previously stable 
patient can occur as a result of on-going cervical 
edema or hematoma from soft tissue injury or frac- 
tures. It can also accompany mental status deterio- 



*,MD 



L.A.WlBBENV 

University of 

Radiology, 200 Hawkins D: 

M. Shahafuddin.MD 

University :n Iowa Hospi litis and Clinics. Pepa? intent of Rad 
ology,200 Hawkins Dr., 3957 ]PP, Iowa City, IA 52242, USA 



and Clinics, Department of 
ira City, IA 52242, USA 



Table 3.1. The primary survey of the trauma patient. Fro 
[37], with permission 

A Airway maintenance with ceivical spine protection 

B Breathing and ventilation 

C Circulation with hemorrhage control 

D Disability: Neurologic status 

E Exposure/environmental control: 

Completely undress the patient, but prevent hypothermi 



L. VVibbeaniryer and M. Sh.i rafted in 



ration, circulatory decompensation, or administra- 
tion of procedural conscious sedation medications. 
It is therefore imperative that the interventionist be 
familiar with the signs and symptoms of impending 
respiratory collapse and the treatment. 

Airway compromise may be insidious in onset. 
Recognition of the signs and symptoms of airway 
obstruction can enable controlled airway access 
and prevent a respiratory arrest. Table 3.2 lists the 
signs of impending respiratory embarrassment sec- 
ondary to airway obstruction. Stridor or inspiratory 
wheezing heard over the cervical region is a sensi- 
tive marker for impending respiratory compromise. 
Signs of increased labored breathing should also 
be observed. Flaring of the nasal alae, retraction of 
cervical soft tissues, and retraction of the ribs on 
inspiration all are signs of increased work of breath- 
ing. Paradoxical movement of the chest (abdominal 
distention on inspiration) is an ominous sign sug- 
gesting impending airway compromise. 

Adequate oxygenation and ventilation should be 
assessed. The pulse oximeter data is readily avail- 
able to the interventionist during the procedure 
and can provide quantitative data reflecting the 
patient's ability to oxygenate. Saturations above 
95% are usually adequate to ensure sufficient oxy- 
genation [4]. However, due to the sigmoidal shape 
of the oxyhemoglobin desaturation curve, factors 
that shift the curve to the left such as hypothermia, 
alkalosis, and a decrease in 2,3-diphosphoglycer- 



aldehyde (old banked blood) may ii 



oxyger 



unloading from oxyht 
patients should r> 
oxygen by nasal cannula or m 
ation, adequate ventilation cai 
accessed by arterial blood gas ; 
ing normal carbia. Inadequate 
ation or ventilation accompanit 
of labored breathing should prompt c 
i ■ :> r intubation. 



efficient 
iglobin. Therefore, 
^ceive supplemental 
ask. Unlike oxygen- 
i only be accurately 
iiia lysis demonstrat- 
or marginal oxygen- 
id by increased signs 



3.2.2 

Treatment Adjuncts 

Several options are available lor the initial man- 
agement of suspected airway obstruction. If neuro- 
logic deterioration secondary to closed head injury 
or administration of sedative agents is suspected, a 
soft plastic nasopharyngeal airway (trumpet) can be 
inserted (Fig. 3.1} [5-7]. Its counterpart the oropha- 
ryngeal should be avoided in the conscious patient 
because of excessive irritation of the upper airway. 
The nasopharyngeal airway will lift the tongue and 
accompanying soft tissues off the back of the airway, 
enabling ventilation. In addition, a gentle chin lift 
or jaw thrust can alleviate upper airway obstruction 
(Fig. 3.2). These maneuvers alone may be sufficient 
if only upper airway obstruction is the problem. It 
should be remembered that extension of the neck 



: 3.2. Symptoms and sign- of [es^i:\r.o:y obsinKkon g:adra according to sever- 
lodified from [38,39], with permission. First published in the British Medical 



Stage 


Signs and Symptoms 


1. Mild or Potential Obstruc 


on Hoarseness 




Cough 




No stridor at rest 




Stridor on moderate exertion 


2. Moderate Obstruction 


I'iyv. ■■:•■?;: 




Rib retraction on inspiration 




Use of accessory muscles of aspiration 




Stridor on slight exertion 


3. Severe Obstruction 


Apprehension 




Ke<: Issues? 




Sweating and pallor 




Increased blood pressure and heart rate 




Paradoxical movement of chest 




Stridor at rest 


4. Total Obstruction 


Slowed respirations 




Marked cyanosis 




Impaired consciousness 




Slowing heart rate 




Hypotension 




No longer stridorous 



Recognition and :re.iunem of Hec.;.i. Emergencies in the Trauma Patient 





Fig. 3.1. The mechanis:r. •:■: orophai 'y:;ge:il obstriicr io:i in the Fig. 3.2. Manual in -line stabiliziitio: 

supine position and die projet pi a cement of die nasop:'.a:yn- ah way is opened with the two-han 

geal airway. Modified from [42], with perm is si on. G:-pvi ight, ing technique 
n Heart Association 



s pi' 'vided as :iie oadeir.'s 
d jaw thrusi airway ope:i- 



should be avoided in all t 



til thei 



a patients 

cervical spine is radiographically and clinically 
cleared. These patients will almost invariably have 
hard cervical collars tor ned-. immobilization, and 
these should never be removed withi 
sulfation with the trauma ten in. 



3.2.3 

Definitive Control 



it propel 






If the patient continues to show sigi 
obstruction or compromise, despite tht 
a nasopharyngeal airway or manual airway opening 
procedure, a definitive airway needs to be estab- 
lished. Trauma patients have challenging airways 
due to the need to maintain manual in-line neck 
stabilization (MILS) and the presence of the cervical 
collar. Normally patients are placed in the sniff- 
ing position with the head and upper neck fully 
extended and the occiput slightly elevated. This 
position aligns the oral, pharyngeal, and laryngeal 
airways. MILS places the head in the neutral posi- 
tion, displacing the larynx anteriorly and impairing 
visualization [8,9]. The anterior faceplate of the cer- 
vical collar restricts mouth opening, further com- 
plicating access to the larynx [9]. Removal of the 
anterior faceplate as long as .MILS is maintained is 
safe and will facilitate intubation. When available, 
an experienced provider should attempt definitive 
control of a the airway. However, as the intervention- 
ist may occasionally be the only one immediately 



available, a systematic approach to the definitive 
airway will be presented. 

A systematic approach to the airway consists of 
assessment of the level of airway difficulty, prepara- 
tion of the patient, collection of airway equipment 
and pharmacologic, medication, and development 
of a back-up plan. A back-up plan is essential prior 
to any intubation attempt. Preparation is critical to 
avoid failed attempts resulting in airway trauma 
impairing subsequent laryngoscopies. 

The first step, assessment of the level of airway 
difficulty, is critical. An unanticipated difficult 
airway can lead to a no intubation-no ventilation 
crisis where only a surgical airway can correct the 
situation. It is estimated that anywhere from 1.5%- 
8.5% of airways are diilicult [10]. I 'espite a number 
of scoring systems that rate airway difficulty, only 
50% of these difficult airways can be accurately pre- 
dicted [11-13]. In general, patients who are difficult 
to intubate often have limited mouth opening, pro- 
truding teeth, receding chins, short necks, or lim- 
ited neck extension. The existing difficult airway 
scoring systems take into account these factors. If a 
difficult airway is anticipated, other options such as 
an awake intubation, Laryngeal Mask Airway, gum 
bougie, or a lighted stylet can be considered [7,14]. 

Patients need to be preoxygenated prior to intu- 
bation. Preoxygenation washes out the nitrogen and 
ensures adequate oxygenation if prolonged laryn- 
goscopy is needed. In the spontaneously breathing 
trauma patient, this is done by giving them high 
flow oxygen via a non-rebreathing mask or a bag 



L. VVibbenniryer nnJ M. '•iiaiiif.uldin 



valve mask device. In the case of the no n- ventilating 
patient, their ventilation must be provided through 
manual compression of a bag valve mask device. 
This is best done using a two-person technique. The 
mask is placed over the bridge of the nose with the 
thumbs and then secured over the mouth and chin 
area with the index and middle fingers. The ring and 
little fingers support the jaw in the jaw thrust posi- 
tion. An additional person is needed to compress the 
ventilating bag. 

Necessary equipment and pharmacologic medi- 
cation then needs to be made available to ensure 
smooth intubation. Essential equipment is listed 
in Table 3.3. Pharmacologic medications consist 
of hypnotic and paralytic agents and are listed in 
Table 3.4. All patients should have cardiopulmo- 
nary monitoring with cardiac, blood pressure, and 
pulse oximeter monitors. Intravenous lines should 
be checked for their patency. The choice of seda- 
tive or hypnotic agents will depend on the patient's 
hemodynamic status, with etomidate and ketamine 
usually preferred in the hemodynamically unstable 
patient. The administration of a paralytic agent has 

Table 3.3. Essen:!;!! ;n: \\~\y mjn; ! .geine;':". equipment. Modifirci 
from [40], p. 6, with permission 

• Supply of 100 c '" oxygen 

• Bag valve device 

• Suction equipment 

- Suction catheters 

- Large-bore tonsil auction apparatus (Y;mknuer) 

• Stylet 

• Oral airways 

• Nasal airways 

• Laryngoscope handle ,ind baices 
(curved, straight, various sizes) 

• Endotracheal iubes (various sizes i 

• Tongue depressors 

• Syringe tor cuff inflation 

• Tape 

• Tincture of benzoin 



Table 3 A Pharmacologic therapy tor intubation. Modified 
from [41], with pt 



< 60 si 



been demonstrated to help with successful intuba- 
tion [15,16]. However, the choice of a paralytic will 
depend on the difficulty of the airway and the abil- 
ity of the airway provider. A depolarizing paralytic 
(succinylcholine) is ideal as it results in rapid induc- 
tion and has the shortest half-life of all the paralyt- 
ics. However, its use should be restricted in patients 
with known neuromuscular disease, massive mus- 
cular trauma, burn injuries over 24 hours old, or 
suspected elevation in serum potassium. Non-depo- 
larizing paralytics should be reserved for patients 
with easy airways and experienced airway provid- 
ers, since paralysis can last up to 120 minutes. 

Conscious trauma patients are intubated by the 
technique of rapid sequence intubation (RSI) with 
MILS. Unconscious patients can be intubated with- 
out medication. RSI calls for administration of a sed- 
ative or hypnotic agent followed by a paralytic agent. 
Another team member provides MILS by cradling 
the trauma victim's neck between their arms, effec- 
tively preventing neck extension (Fig 3.2). Exter- 
nal cricoid pressure (8 kg of pressure applied with 
three fingers over the cricoid ring) is applied prior 
to the administration of medication to decrease the 
risk of aspiration. The patient is then intubated, 
and verification of correct positioning of the tube 
is performed by physical exam and disposable cap- 
nography to assess for exhaled carbon dioxide. Fur- 
ther discussion of intubation techniques is beyond 
the scope of this chapter; the interested reader is 
referred to several references [14,17-19]. 



0.1-0.3 mg/kg 30-60 sec 6-15 rain 

1-1.5 mg/kg 30-60 sec 5-10 rain 

0.4 mg/kg 60-90 sec 75-1 00 rr 

0.3 mg/kg 60-90 sec > 120 mil 

0.9-1.2 rag/kg 60 sec > 60 rain 



Circulatory Shock 

3.3.1 
Recognition 

Once an adequate airway and breathing of the 
trauma patient is achieved, the primary survey of 
the trauma patient addresses the circulatory system. 
A number of perfusion endpoints must be analyzed 
to determine whether the patient is adequately 
resuscitated or declining into circulatory shock. The 
predominate cause of shock in the trauma patient is 
under-perfusion secondary to bleeding. The treat- 
ment of shock is to replace the volume lost. By and 
large, the treatment of shock in the trauma patient 
has remained unchanged for the past few decades. 
The recognition of shock, however, can be chal- 
lenging. Blood pressure and pulse are neither sensi- 



Recognition an;; . re.iunem of Hec.;.i. Emergen ci- 



five nor specific markers for the diagnosis of early 
hemodynamic shock [20-22]. In fact, hypotension 
can Lie a late finding in shock after which circula- 
tory collapse can occur. In general, the signs and 
symptoms of shock are directly related to the blood 
volume lost [4]. The American College of Surgeons 
divides hemorrhage into four categories (Table 3.5). 
Most patients tolerate class-1 hemorrhage or 10% 
blood volume loss with little change in vital signs, 
due to a number of compensatory mechanisms, 
the earliest of which are tachycardia and narrowed 
pulse pressure (the difference between the sys- 
tolic and the diastolic pressures). It loss continues, 
the patient will demonstrate a decrease in cardiac 
output and blood pressure. As a result pallor, cool 
extremities, delayed capillary perfusion, decreased 
urine output, and mental status changes (agitation 
and anxiety) may develop. Although these signs may 
occur before, they are usually minutes ted in i.idulls 
following around 20'-'('--40 IJ r> blood volume loss and 
therefore are late markers of shock. Children, on 
the other hand, may not manifest these signs until 
around 40% volume loss. Children with hypotension 
and tachycardia are significantly volume-depleted 
and can rapidly decompensate. 

Biochemical markers may better quantify the 
initial and on-going magnitude of the shock state 
[22-26]). Both the base deficit and serum lactic 
acid level measure the acidosis produced by the 
anaerobic state during inadequate delivery of sub- 
strate to tissues [2~]. Shock impairs nutritive blood 
flow to tissues, shifting cellular metabolism into 
the less efficient anaerobic glycolysis pathway. The 
formation of ATP from ADP is slowed, resulting in 
accumulation of hydrogen ion (H + ) in the cytosol 
and extracellular fluid. This accumulation of the 
H + in the cytosol is quantified by the base deficit 
n the arterial blood gas. Base deficit 



(the quantity of strong acid or base that would be 
required to titrate the patient to a normal pH assum- 
ing aPaCO 2 of40 mmHgand a hemoglobin of 5 g/dl) 
reflects the metabolic component of shock. Initial 
and subsequent base deficit measurements provide 
markers to access the severity and recovery of the 
shock state. Initial base deficit < -6 has been asso- 
ciated with the need for transfusion [25]. Similar 
to the base deficit, lactic acid accumulates in the 
shock state. Under anaerobic conditions, pyruvate 
accumulates and is dehydrogenated in the cytosol 
to lactate. Lactate buffers H 4 ", resulting in lactic acid. 
Serial measurement of serum lactate can also help 
achieve a better follow-up of the compensated shock 
state and reduce the need for on-going resuscitation. 
Persistent serum lactate elevation following resus- 
citation in trauma has been reported to portend a 
worse outcome [23]. 



3.3.2 
Resuscitation 



Once shock is identified, resuscitation needs to be 
instituted to prevent on-going perfusion mismatch, 
which can lead to multiple system organ failure and 
death. Although there is some controversy regard- 
ing resuscitating penetrating trauma victims to 
normal blood pressure before surgical control of 
their bleeding, resuscitation to normal volemia is 
the current pnradigm in bluntly injured patients 
[28]. In penetrating trauma victims, normotension 
may lead to worsened hemorrhage, whereas blunt 
trauma patients often have accompanying head inju- 
ries that can be significantly worsened by hypoten- 
sion. Trauma patients in shock are first resuscitated 
with crystalloid, namely Lactated Ringers or normal 
saline. ATLS protocol dictates a 2-liter rapid bolus 



Table 3.5. Estimated fUiic ami iMood losses based on 


hemorrhagic shock se 


verity class on patient's 


inidal presentation. From 


[42], with permis 




















Class I 


Class II 


Class III 


Class IV 


Blood loss (ml) 






Up to 750 


750-1500 


1500-2000 


>2000 


Blood loss (% blood vi 


>lnme) 


Up to 15% 


15%-30% 


30%-40% 


>40% 


Pulse rate 






< 100 


MOO 


> 120 


>140 


Blood pressure 






Normal 


K : :■ [" m ..i 1 


i ■e.:r-;i ,; ;d 


', -ecie.ised 


Pulse pressure 






Normal or increases 


Decreased 


i 'ecreased 


recreated 


(mmHg) 














Respiratory rate 






14-20 


20-30 


30-40 


>35 


Urine output 






>30 


20-30 


5-15 


Ned. id: .- 


(ml/h) 














CNS/mental statu 






Slightly anxious 


Mildly anxious 


Anxious, confused 


Confused, lethargic 


Fluid replacemen 


t(3:l 


rule) 


ill vsiaKoid 


'..'iVsLlllOld 


Crystalloid and blood Crystalloid and blood 



L. VVibbeaniryer nnJ M. 5l"i.n I ]f'.:cidin 



in an adult and a 20cc/kg bolus in a child. Non- 
responders or transient hemodynamic responders 
usually have lost > 25% of their blood volume or 
have on-going bleeding and need to receive blood. 
As typing and cross-matching require time, type O 
negative blood is preferentially transfused. It is cru- 
cial to remember that the ability to provide adequate 
resuscitation is also dependent on catheter dynam- 
ics. ATLS protocol dictates the placement of two 
large-bore (14-16 gauge) peripheral catheters. With 
short intravenous tubing (< 3 feet) and 300 mmHg 
external compression, these catheters can provide 
flow rates of 249-500 cc/min [29]. Central access is 
required if peripheral access is inadequate. The loca- 
tion of the placement of catheters is also important. 
The lower extremities should be avoided if intraab- 
dominal injury is suspected. 



3.3.3 

Pitfalls of Resuscitation 

Tlie two main pi I hills ot rapid resuscitation and mas- 
sive transfusion are hypothermia and coagulopathy. 
Along with acidosis of the shock state, hypothermia 
and coagulopathy compose the "triangle of death" 
well known to the trauma surgeon [30,31]. When 
these three conditions are present in the emergency 
room or operating room, only stabilizing procedures 
are performed and the patient is transferred to the 
intensive care unit for resuscitation. With resolution 
of the triad, the patient is returned to the operating 
room if necessary. 

Hypothermia in the trauma patient is nui hi facto- 
rial, resulting from exposure to cold environment, 
bleeding, and infusion of cold fluids. Mild to mod- 
erate hypothermia (34°C to 30°C) can be associated 
with coagulopathy that can impair the patient's 
response to ongoing resuscitation and at times be 
refractory to treatment [32]. During massive resus- 
citation, hypothermia can be avoided by admin- 
istration of warmed fluids, either by means of an 
in-line warmer, or rapid infuser. The ambient room 
temperature should be maintained at 21°C. Addi- 
tionally, patients can also be actively warmed by one 
of the commercially available convective blankets. 

Coagulopathy can also be multifactorial in the 
multiple injured trauma patients. In addition to 
hypothermia-related coagulopathy, massive resusci- 
tation and massive transfusion are other important 
causes. Massive resuscitation can lead to throm- 
bocytopenia, prolonged prothrombin times, and 
decreased fibrinogen [32,33]. The incidence of coag- 



ulopathy during resuscitation is variable, and there- 
fore its treatment remains controversial. Although 
some formulas have been proposed for replacement 
of coagulation factors and platelets based on the 
number of units of blood received, several studies 
have failed to show their reliability [34,35]. Without 
obvious microvascular bleeding, many recommend 
that fresh frozen plasma and platelet replacement be 
guided by laboratory abnormalities [33,36]. 



Cookbook: 

Recognition and Treatment of Medical 

Emergencies in the Trauma Patient 

Susgect Airway Compromise 

• Perform Primary Survey 

• Assess Airway for Patency 

• Open Airway 

- Airway Opening Procedure 

- Insertion of Naso- or Oropharyngeal Airway 

• Prepare for Definitive Airway 

- Assessment of Airway Difficulty 

- Preoxygenation 

- Collection of Equipment 

- Development of a Back-up Plan 

• Provide Definitive Airway-Intubation 

• Confirm Placement of Airway 

- Auscultation 

- Capnography 

- Chest Radiograph 

Suspect Ciratl.iton Compromise 

• Perform Primary Survey 

• Assess Circulation 

- Look for signs of perfusion abnormalities 
- Hypotension, tachycardia, diaphoresis, 

agitation, pallor 

- Obtain biochemical markers of perfusion 
abnormalities (base deficit and lactic acid} 

• Provide Resuscitation 

- Crystalloid (Ringer lactate) bolus 

- Packed red blood cells for no n- res ponders 

• Avoid hypothermia 

- Warm room to 20°C 

- Warm fluids 

- Warm patient with a convective blanket 

• Assess for secondary coagulopathy during 
massive resuscitation 

- Obtain blood for PT, INR, PTT, Fibrinogen 



Recognition and . reatmenl of Mefhi.il Emergence: 



niicipated dif- 
.lagement (see 



Pondeknger RF, Trotteur G et al. (2002) Traumatic inju- 
ries: radiological hemostatic intervention ai admission. Eur 
Radiol 12:979-993 

Velmahos GC, Toutouzas KG et aL (2002) A prospective 
study i'n I he safety and efficacy of angiographic emboli za- 
tion i"o: pelvic ai;d visceral Minnies. I Trauma Injury Infect 
Grit Care 53:303-303 

Santucci RA.Wessells H et al. (2004) Evaluation and man- 
a gem en; of renal injuries: consensus statement of I he renal 
trauma subcommittee. BJU Int 93:937-954 
American College o:' Surgeons i I 997; Advanced iraun'.a life 
support manual, nth edn. ACS, Chicago 
Kaur 5, Hearc SO (1996) Airway management and endo- 
tracheal intubation. In: Rippe IM, Irwin RS, Fink MP, Cera 
FB teds: Intensive care medicine, vol I . Little Brown. New 
York, pp 1-15 

Hillman DR, Piatt PR et al. (2003) The upper airway during 
anaesthesia. Br I Anaesth 91:31-39 

Nolan JP, Wilson ME (1993) Orotracheal intubation in 
patient- with potential cervical spine injuries. An indica- 
tion for the gam eiasi.c bougie ; -re comment !. Armies the si a 
48:630-633 

Heath KJ (1994) The effect of laryngoscopy of different 
cervical spine immobilisation techniques 
Anaesthesia 49:843-845 
Crosby ET, Cooper RM et al. (1998) The u 
ficall airway wait re com men canons ho: i 
comment). Can J Anaesth 45:757-776 
Oates JD, Macleod AD et al. (1991) Comparison of two 
nre ih:: as for predicting cl it'll cult ; in u bar ion :. ser comment). 
Br ] Anaesth 66:305-309 
ll.Sawa D (1994) Prediction of difficult tracheal intubation 
(see comment). Br J Anaesth 73:149-153 
Rosenblatt WH :2l.:.4: Preoperative planning of airway 
managiriiient in critical care patients (see comment). Crit 
Care Med 32 [Suppl 4] 

Blanda \1, Callo 'JE :2C0ai Emergencv airwav manage- 
ment Emerg Med Clin North Am 21:1-26 
C 1 1 swell IC, Parr M J et al. (1994) Emergencv airway man- 
agement ai oaiients wilh cerv.oa. -pine iniinies (see com- 
ment). Anaesthesia 49:900-903 

Li J, Murphy-Lavoie H et al. (1999) Complications of emer- 
gency imubaiion wilh and wilhout paralysis. Am 1 Emerg 
Med 17:141-143 

Behringer EC i2032! Approaches [.:■ managing the upper 
airway. Anesthesiol Clin North Am 20:813-832 
Orebaagh SL f 200 J ) Pirllcuk airwav management in the 
emergency department. ) Emerg Med 22:31-48 
Butler KH, Clyne B (2003) Management of the difficult 
airway: alternative airwav leohniques and admncts, Emerg 
Med Clin North Am 21:259-289 

luna GK, Eddy AC et al. (1989) The sensitivity of vital signs 
in io enti lying major thoracoabdominal hemorrhage. Am 1 
Surg 157:512-515 

Thompson D.Adams SL et al. (1990) Relative bradycardia in 
patients Wilh isclaieol penetrating aodommal irauma and 
isolated extremity trauma. Ann Emerg Med 19:263-275 
Wilson M, Davis PP ei a I. (2303 i Diagnosis and monitoring 
of hemorrhagic shock oaring the initial resuscitation of 
multiple trauma patients: a review. J Emerg Med 24:413- 
422 



22. Abramson D, Scalea TM et al. (1993) Lactate clearance and 
survival following injury. 1 Trauma iniury Infect Crit Care 
35:584-588 

23. Manikis P, lankowski S el al. ( 1995) Correlation of serial 
blood lactate levels no organ failure and mortality after 
trauma. Am ] Emerg Med 13:619-22 

24. Davis JW, Parks SN et al. (1996) Admission base deficit pre- 
dicts transfusion requirement and risk or" complications 
(see comment). J Trauma Injury Infect Crit Care 41:769- 






25.Davis )W, Kaups KL et al. (1998) Base deficit i 
io pH in evaluating clearance of acidosis after 
shock. J Trauma Injury Infection & Crit Cai 



26.Mullins R (2000) Management of shock. In: Mattox KL, 

Feliciano DV, Moore EE (eds) Trauma. McGraw-Hill, New 

York,pp 195-232 
27.Bickell WH, Wall MJ Jr et al. (1994) Immediate versus 

delaved fluid resuscitalion for hypotensive patients wilh 

penetrating icrso iniuries (see comment). N Engl 1 Meil 

331:1105-1109 
28.Millikan JS, Cain TL et al. (1984) Rapid volume replace- 
ment for hypovolemic shock: a comparison of lech manes 

an..: equipment. | Trauma Iniury Infect Crit Care 24:42S- 

431 
29. Stone HH, Strom PR et al. (1983) Management of the major 

coagulopathy with onset during laparotomy. Ann Surg 

197:532-535 
30.Danks RR (2002) Triangle of death. How hypothermia 

acidosis and coagulopathy can adversely impact Irauma 

patients. ) Emerg Med Serv 27:61-66 
31.Ferrara A, MacArthur JD et al. (1990) Hypothermia and 

acidosis worsen coagulopatiiv in lire pal lent recti ning mas- 
sive transfusion. Am J Surg 160:515-518 
32.Faringer PD, Mullins RJ et al. (1993) Blood component 

supplementation during massive iransfusion of AS-1 red 

cells in trauma patients. ; Trauma Injury Infect Crit Care 

34:481-485 
33.Harrigan C, Lucas CE et al. (1985) Serial changes in 

primary hemostasis after massive transfusion, surgery 

98:836-844 
34.Wudel JH, Morris JA Jr et al. (1991) Massive transfusion: 

outcome in blum trauma patients. ] Trauma Injury infect 

Crit Care 31:1-7 
35. Reed RL 2nd, Johnson TD el al. (1992) The disparity 

between hypothermic coagulopathy and clotting -ladies. 

J Trauma Injury Infect Crit Care 33:465-470 
36. Bell RM (2000) Initial assessment, in: Mattox KL, Feliciano 

DV, Moore EE (eds) Trauma, 4th edn. McGraw-Hill, New 

York 
37.Mattox KL, Feliciano DV, Moore EE (eds) (2000) Trauma, 

4th edn. McGraw-Hill, New York 
38. Robinson RJS, Mulder DS, Forbes JA (1964) Airway control. 

Br Med J 1:369 
39.Rippe JM, Irwin RS, Fink MP, Cerra FB (1996) Intensive 

care medicine, 3rd edn. Little Brown, Boston 
40.Hartmannsbruber (2000) The traumatic airway: the anes- 

ihes.ologist's role in the emergency room, int Anesthesiol 

Clin 38:87-104 
41. Cummins RO (ed) (1994) Textbook of advance cardiac life 

support. American Heart Assoc ia iron, Dallas, Texas 



4 Visceral and Abdominal Solid Organ Trauma 



Gary Siskin and Jafar Golza 



Splenic Artery Embolization 43 
Introduction 43 
Patient Selection 43 

The Role of Abdominal CT 44 
. Diagnostic Angiography 44 

Splenic Artery Embolization 45 

Technique 45 
; Results 47 
■ Post-Embolization Follow-up 48 

Hepatic Artery Embolization 48 

Patient Selection 4S 



lagnos: 



49 



Hepatic Artery Embolization 49 

Technique 49 

Results 49 

Post-Embolization Follow-up 51 

Renal Artery Embolization 51 

Patient Selection 5J 

[ 'iagnosis of Renin Vascular Injury 52 

Renal A: tery Embolization 52 

Technique 52 

Results 52 

Post-Embolizatio 

Cookbook 54 

References 55 



Follow-up 



Splenic Artery Embolization 

4.1.1 
Introduction 

During the past 10-15 years, there has been a clear 
trend among trauma surgeons toward the use of bed 

rest and observation for hemodynamically stable 






G. Siskin, MD 

Albany Medical College, Vascular Radiology,. 

Scotland Avenue, Albany, NY 12208-3479, USA 

J. GOLZARIAN, MD 

Professor of Radiology. I a:ector. Vascular anc Interventional 
Radiology, University of Iowa, Department of Radiology. 
200 Hawkins Drive, 3957 fPP, Iowa City, IA 52242, USA 



patients after blunt injury to the spleen [1], How- 
ever, well-defined criteria are still not available to 
apply to these patients in order to determine who is 
best suited for nonoperative management and who 
will ultimately require surgery. We do know that 
liemodviiaiiiically unstable patients with multisys- 
tem trauma and significant intraperitoneal bleeding 
will likely not be successfully managed with obser- 
vation and splenic salvage. All other hemodynami- 
cally stable patients are now considered candidates 
for nonoperative treatment consisting of observa- 
tion with serial physical examination, frequent 
hematocrit determinations, bed rest, and limited 
oral intake [2]. 

The trend toward nonoperative management 
after splenic injury has significant benefits for this 
patient population. These include eliminating the 
risks for immediate and late complications asso- 
ciated with open surgery as well as preserving the 
immunologic functions provided by the spleen [1, 
3-5]. Immunologically, the spleen is best known for 
its filtering function, which serves to remove partic- 
ulate antigens, bacteria, and old erythrocytes from 
circulation [6]. In addition to this, the spleen pro- 
duces mediators such as immunoglobulin M, tuft- 
sin, and properdin [6, 7]. The importance of these 
functions manifests itself after splenectomy with 
a significant post-surgical infection rate, which is 
what initially prompted surgeons to increase their 
interest in splenic salvage [6-9]. 



4.1.2 

Patient Selection 

With the benel its ot splenic salvage rarely questioned, 
it makes sense that this is most desirable option for 
most, it not all, lieinodynamically stable patients. 
However, it is still not known with certainty which 
patients will be success! tillv managed with nonopera- 
tive management and which will ultimately require 
surgery despite initially being considered candi- 
dates for observation. Patient age, injury severity 



C-. Siskin ;i:id 1. L,olz;i:i,in 



score, neurologic status, grade of injury on abdomi- 
nal CT, and quantity of hemoperitoneum have all 
been cited as factors that may affect the success of 
nonoperative management [10-15]. Hemodynamic 
stability as a reflection of injury severity is likely 
the most important factor to consider; Peitzman et 
al. found that nonoperative management was more 
likely to be successful in patients with higher blood 
pressure, higher hematocrit levels, less severe injury, 
and smaller quantity of hemoperitoneum [15]. Early 
on, it was recommended that patients older than age 
55 be excluded from nonoperative management of 
blunt splenic injuries [10, 16]. However, more recent 
data suggest that the decision to pursue nonoperative 
management should be based on clinical and hemo- 
dynamic factors, and not necessarily on age [11, 17]. 
Despite the benefits of splenic salvage and the 
trend towards nonoperative management for these 
patients, there are those that advocate a more aggres- 
sive approach to the treatment of splenic injuries. 
These investigators cite the potential for delayed 
rupture or progression of injury that may require 
urgent transfusion or emergency splenectomy. 
Federle et al. did note in the review of their insti- 
tutional experience with splenic trauma patients 
that 15% of patients selected for nonoperative man- 
agement initially ultimately required surgery, most 
commonly within 48 hours of presentation [18]. One 
problem with managing patients with delayed rup- 
ture or injury progression is the difficulty utilizing 
techniques to preserve splenic tissue, such as par- 
tial splenectomy and splenorrhaphy, depending on 
clinical severity [19-22]. 



4.1.3 
Diagnosis 

4.1.3.1 

The Role of Abdominal CT 

Abdominal CT has become an integral part of the 
evaluation of patients experiencing blunt traumatic 
injury. For stable patients with splenic trauma, 
CT is needed before a decision is made to proceed 
with nonoperative therapy because it is important 
to identify and characterize not only the degree of 
splenic injury, but also any concomitant abdominal 
injury [19, 23, 24]. There have been several CT-based 
classification systems tor splenic injury, based on the 
morphologic grade of splenic injury and the amount 
of intraperitoneal hemorrhage, to guide therapy and 
predict success for nonoperative management [18, 



25-29]. One example of a grading system for splenic 
injury is the modified criteria of the American Asso- 
ciation for the Surgery of Trauma (AAST), which 
grades splenic injuries from 1 to 5 on the basis of 
increasing severity of parenchymal damage based 
on radiologic assessment [18, 27] (Table 4.1). Despite 
the widespread use of this and other classification 
systems, their use remains controversial since sev- 
eral studies have shown that they do not necessarily 
correlate well with clinical outcome [26, 27, 30-32] 
and may not provide sufficient information regard- 
ing vascular injuries to be useful [23, 33]. However, 
Federle et al. studied this issue and found that CT 
can effectively recognize vascular injury and active 
extravasation from splenic arterial branches [18]. 

In 2000, Shan mug an ath an et al. evaluated the 
potential role of contrast-enhanced spiral CT in pre- 
dicting the need for splenic angiography and embo- 
lization by correlating them with results from subse- 
quent splenic angiograms [19]. In their study, contrast 
extravasation at CT was highly predictive of the need 
for embolization, regardless of the CT grade of injury. 
The presence of a pseudoaneurysm or arteriovenous 
fistulas on CT, both of which are seen as focal areas 
of high attenuation, was a less sensitive finding, with 
only 58% of patients with these CT findings requir- 
ing embolization or splenectomy. The angiograms 
of the remaining 42% of patients with CT findings 
suggestive of a pseudoaneurysm or arteriovenous 
fistula did not reveal any focal vascular abnormali- 
ties. These false-positive CT findings were attributed 
to islands of enhancing splenic parenchyma sur- 
rounded by low- attenuating splenic lacerations or 
contusions and intact intiasplenic vessels traversing 
parenchymal lacerations simulating hemorrhage sur- 
rounding a focal pseudoaneurysm [19]. The finding 
of a splenic vascular lesion on contrast- enhanced 
spiral CT was 83% sensitive in predicting the need 
for splenic angiography and subsequent embolization 
or surgery. Using this approach, 94% of patients were 
successfully selected at presentation for conservative 
management. This study demonstrated thattheuseof 
contrast- enhanced spiral CT and splenic angiography 
improves early diagnosis and potentially increases [he 
number of patients with blunt splenic injuries who are 
treated successfully without surgery. 



4.1.3.2 

Diagnostic Angiography 



The role of diagnostic angiography in t 
tion of patients experiencing blunt splei 



il Solid Organ Trauma 



Table 4.1. AAST Splenic Injury Scale 
Grade Type of Injury D esc rip Lion 



H em ;i :o:";": 
Laceration 
Hemato::\a 
Laceration 
H em ;i :o:";": 

Laceration 
Laceraiion 



Subcapsular, <10% surface area 
Capsular tear, < = 1 cm parenchymal depth 

Subcapsular, :0-.~0% surface area, inlraparenchynul, <5 cm in chameter 
1-3 cm parenchymal depth that ckvs not Involve a trabecular vessel 
apsular, >50% surface area or rxpancjng; :up:ured subcapsular or 



parenchymal hematoma; intjjparenchymal, >? cm or expanding 

:>3 cm parenchyma! depth or involving Trabecina! vessels 

li'.volvii'g segmental ■ u hilar vessels prod; cing maior devai 

(>25% of the spleen) 

Completely shattered spieen 

Hilar "vascular injury that cevascularizes the spleen 



remains controversial. While some believe that all 
patients with splenic injuries should undergo diag- 
nostic angiography, others tire more selective and 
prefer angiography lor patients with high-grade 
parenchymal injury, vascular injury, or a large 
volume of hemoperitoneum on admission CT scan 
[6, 16, 23, 33-35]. It has been well established that 
selective splenic angiography is successful at iden- 
tifying vascular injuries, with positive findings 
including active extravasation of contrast and the 
presence of a pseudoaneurysm [33, 36]. It is the 
early work of Sclafani et al. and Hagiwara et al. 
that supports the angiographic evaluation of stable 
patients with CT evidence of splenic injury [23, 33]. 
A positive angiogram is a strong predictor of nonop- 
erative failure, necessitating the use of embolization 
or surgery to allow for continued splenic viability 
[1, 23, 33, 37-39]. Conversely, a negative angiogram 
can successfully predict the success of observation 
for these patients [33, 39]. 

However, others consider the use of angiogra- 
phy in all patients with documei 
is aggressive. Sclafani et al, 
incidence of negative angiogra 
lation [39]. Dent et al. recommend angiography 
for patients with persistent tachycardia despite 
fluid resuscitation, splenic vascular blush on CT, 
severe splenic injury on CT, or decreasing hema- 
tocrit that cannot be explained [40]. Using these 
criteria, angiography was only necessary in 7% of 
patients with blunt splenic injury and ultimately, 
the success of nonoperative management was sim- 
ilar to studies in which there was more liberal use 
of angiography. Similarly. Lit: et al. argue against 
the use of routine angiography since most patients 
with blunt splenic injury can be successfully man- 
aged by bed rest and observation [6, 7]. These 



I splenic injury 
ported a 70% 



indings therefore need to be considered when 
xposing patients to the inherent risks and cost of 
ngiography [12]. 



4.1.4 

Splenic Artery Embolization 

The use of splenic artery embolization was origi- 
nally described in 1973 in patients with hypersplen- 
ism [41]. In 1981, Sclafani first reported the splenic 
artery infusion of Pitressin, Gelfoam pledget embo- 
lization, and coil occlusion of the proximal splenic 
artery to treat splenic injury [36]. Since that time, 
the findings of many studies have supported the use 
of splenic artery embolization in patients managed 
nonoperatively after blunt splenic trauma [2, 12, 19, 
23, 33, 37, 39]. In general, embolization is reserved 
for patients with documented vascular injury at 
the time of diagnostic a ngiography, but the success 
and ease of performing this procedure has led some 
to utilize embolization in all patients with higher 
grade injuries [2]. 



4.1.4.1 
Technique 

It must be stated that the goal of embolization in the 
setting of splenic trauma is to reduce arterial flow 
to the spleen so that hemostasis can take place at 
the site of arterial injury. However, the importance 
of maintaining splenic viability must not be forgot- 
ten and therefore, continued perfusion of splenic 
parenchyma from collateral vessels must also be 
maintained during a successful embolization pro- 
cedure. 



C-. Siskin ;i:id 1. L.olz;i:uin 



Technically, an abdominal aortogram is recc 
mended prior to selective splenic artery catheteri 
tion in order to visualize the exact point of origii 
the splenic artery and to demonstrate the 
or absence of variant anatomy. Most interventional- 
ists then favor either a superselective approach with 
distal embolization of the injured splenic artery 
branches, or a relatively nonselective approach 
with proximal embolization of the main splenic 
artery, utilizing the angiographic appearance of the 
splenic vasculature to make that decision. While 
one can certainly make the argument that patients 
with active branch vessel extravasation should have 
clinical signs that should have necessitated imme- 
diate surgery, the expanding use of angiography 
and embolization at our institution frequently puts 
us in the position of treating patients that seem to 
require distal embolization for adequate treatment. 
In these patients, it often makes intuitive sense to 
select and embolize the injured branch vessels, a 
feat made easier by advances in microcatheter and 
microcoil technology [6]. In these patients, however, 
consideration must be given to the balance between 
superselective control of bleeding, the fluoroscopy 
exposure time required for selective catheterization, 
and the volume of splenic parenchyma put at risk for 
ischemia and subsequent infarction (Fig. 4.1). 

An alternative to superselective catheterization 
of splenic artery branches is the use of proximal 
splenic artery embolization [23, 33, 39]. In this 
technique, the main splenic artery is embolized, 



beyond the origin of the dorsal pancreatic artery 
but proximal to the splenic hilum, with appro- 
priately sized coils [33]. The reason behind this 
technique is to decrease perfusion pressure to the 
spleen while maintaining the ability of the spleen 
to receive arterial inflow from collateral vessels, 
including gastric, omental, and pancreatic vessels 
[1,42]. Therefore, the splenic artery should be mea- 
sured prior to introducing coils and care should be 
taken to use coils that are large enough so that they 
do not migrate into the hilum of the spleen since 
the risk of splenic infarction, 
of splenic perfusion was dem- 
onstrated by Hagiwara et al. who demonstrated 
preservation of splenic function by arteriography 
and scintigraphy in patients who underwent proxi- 
mal splenic artery embolization [23]. 

Patients with multiple vascular injuries are can- 
didates for either proximal coil embolization or 
combined therapy, which utilizes selective catheter- 
ization and embolization of the most significantly 
injured vessels followed by proximal coil emboliza- 
tion [2]. In these patients, distal embolization since 
this may require extensive fluoroscopic exposure 
times to catheterize multiple small vessels with 
subsequent infarction of too large a percentage of 
splenic parenchyma. Haan et al. found no differ- 
ence in failure rate when proximal embolization 
was compared with more selective, distal emboliza- 
tion. Interestingly, however, the largest failure rate 
was found when both techniques were utilized in 




Fig 4.1. a Selective spit 
b Selective splenic an 
splenic parenchyma 



'sm .iiiiii":g fioia ;ii": upper po.e airnjiii ;'.:ie: Mi::': trauma. 
pole briiadi of the splenLi" ;i:rciv supplying the injured 



Visceral and Abdominal Solid Organ Trauma 



the same patient, possibly due to the fact that thet 
patients had the most severe injuries [2]. 



4.1.4.2 
Results 

Sclafani et al. were the first to report that the use 
of splenic angiography and embolization expanded 
the number of patients that could be managed non- 
surgically [33]. In 1995, Sclafani et al. reported 
a nonoperative success rate of 83%, with a splenic 
salvage rate of 88% by performing angiography 
on every patient with a splenic injury who did not 
require urgent operation and proximally emboliz- 
ing the main splenic artery of the 40% of patients 
who had evidence of splenic vascular injury on angi- 
ography [39]. Hagi war a etal.embolized 15 patients 
due to extravasation within or beyond the splenic 
parenchyma or disruption of terminal arteries with- 
out extravasation [23]. In this study, it was noted 
that patients requiring embolizing required larger 
volumes of fluid for resuscitation than those who 
were not embolized, implying that the angiographic 
findings indicating the need for embolization cor- 
related with a greater degree of hemodynamic insta- 
bility at presentation, which may therefore be an 
additional indication for embolization. In addition, 
the CT grades for patients who did not undergo 
embolization were significantly lower than those for 
patients who were embolized. 

In 1998, Davis et al. evaluated their experience 
with 524 consecutive patients with blunt splenic 
injury over a4 J ,■> year period [37]. A focal area of high 
attenuation on CT, confirmed with angiography to 
represent a pseudoaneurysm, was seen in 26 patients; 
20 of these patients were successfully embolized and 
did not require further surgery while 6 patients were 
not embolized due to either free arterial extravasa- 
tion, early filling of the splenic vein due to a traumatic 
AV fistula, and multiple pseudoaneurysms. Given the 
fact that the appearance of some pseudoaneurysms 
will be delayed, Davis et al. emphasized the impor- 
tance of obtaining follow-up CT scans inpatients that 
are being managed nonoperativeiv [37]. 

HAANetal. [12] more recently reported their expe- 
rience with the use of angiography in stable patients 
with CT-proven blunt splenic injury. In their series, 
352 patients presented after blunt splenic trauma 
with 64% requiring immediate surgery. The remain- 
ing 36% of patients underwent splenic angiography. 
Patients with negative angiograms were observed. 
Embolization was performed in 32% of those under- 



going angiography due to positive findings includ- 
ing contrast extravasation, arteriovenous fistula, or 
pseudoaneurysm. Of these, 8% required laparotomy 
(2 for bleeding and 1 for abscess formation), for a 
splenic salvage rate of 92%, despite the presence of 
high-grade injuries in many of these patients. 

Sekikawa et al. reported on factors that support 
use of anonoperative approach using splenic artery 
embolization [34]. They found that injury severity 
score and shock index had no significant correlation 
with outcome, implying that more severe injuries 
do not necessarily predict failure of nonoperative 
management with embolization. However, hemody- 
namic instability (as evidenced by low hemoglobin, 
low hematocrit, and low blood pressure) was associ- 
ated with a poor clinicaloutcome after embolization. 
Based on their data, a patient's age, in addition to the 
presence of head or solid abdominal organ injury in 
hemodynamically stable patients, should not pre- 
clude the use of embolization although concomitant 
pelvic injury was associated with a poor clinicalout- 
come. Importantly, the time interval from injury to 
arrival at the hospital and from arrival at the hospi- 
tal to the start of the embolization procedure did not 
significantly affect clinical outcome. 

The experience of Haan et al. [12] confirms that 
embolization improved splenic salvage rates for all 
CT grades of injury, including grades 3-5. This was 
true even with patients that were at least as severely 
injured based on transfusion requirements, injury 
severity score, length of stay, and ICU stay [12]. Their 
data also offers additional support for the use of 
embolization in patients with concomitant neuro- 
logic injury, stating thai angiographv may be asso- 
ciated with a statistically and clinically significant 
decrease in mortality in patients with neurologic 
injury when compared with operative therapy [12]. 
They went on to theorize that patients with neuro- 
logic injury pursuing nonoperative management are 
spared the risk of intraoperative or postoperative 
hypotension. It has been stated that a single episode 
of hypotension can increase neurologic mortality by 
50-80% and intraoperative hypotension during sple- 
nectomy is a frequent occurrence [12, 43]. Therefore, 
utilizing angiography and embolization for these 
patients potentially avoids this risk and may lead to 
less secondary brain injury and lower mortality. 

Haan et al. also reported the results of a multi- 
center review of patients undergoing splenic artery 
embolization for splenic trauma and this represents 
the largest collection of splenic artery emboliza- 
tion patients studied to date [2]. In this review, 140 
patients undergoing splenic artery embolization at 



ind 1. GolzLiriiUi 



one of four institutions over a 5-year period were 
retrospectively evaluated. They found that splenic 
artery embolization is used for patients with high- 
grade splenic injury with mi average grade of injury 
of 3.5 in this series. Despite significant injury sever- 
ity, splenic salvage of patients selected for emboliza- 
tion was 87% in this series. This series documented 
a failure rate for embolization of 10% with 17% of 
patients demonstrating active contrast extravasa- 
tion failing [2]. In this series, patients with AV fis- 
tulae had the worst outcome and they hypothesized 
that proximal coil embolization may not be enough 
in these patients since the drop in perfusion pressure 
may not be sufficient to provide hemostasis [2]. This 
same multicenter center found a 19% rate of major 
complications and 23% rate of minor complications. 
Taking overlap of these patients into account, there 
was a 32% complication rate. The most common 
complication, seen in 60% of patients, was blood 
loss or continued blood loss. True infection is rare 
but significant splenic induction was common, with 
a rate of 20-27% [2]. The vast majority of patients 
experiencing splenic infarction, however, were 
asymptomatic and able to continue being managed 
nonoperatively. 



4.1.4.3 

Post-Em bo lization Follow-up 

Following nonoperative management and splenic 
artery embolization for blunt splenic trauma, most 
patients typically require continued observation in 
a monitored setting since there continues to be a 
risk of bleeding or sepsis. It is generally accepted 
that these patients should be followed with abdom- 
inal CT until resolution of the injury is seen [44]. 
Killeen et al. performed a retrospective study 
to evaluate the CT findings after splenic artery 
embolization in 53 patients [1]. They found that 
splenic infarcts described as small, multiple, and 
peripheral were seen in 63% of patients after a 
proximal splenic embolization while infarcts were 
seen in 100% of patients after a distal embolization. 
The infarcts seen after both proximal and distal 
embolization tend to resolve without sequelae [1]. 
In these patients, the infarcts tended to be larger, 
single, and located just distal to the embolization 
material. Gas within splenic parenchyma was seen 
in only 13% of patients but was shown to more 
commonly occur when Gelfoam was used as the 
embolization agent [1]. The presence of gas, how- 
g because it is difficult to exclude 



a splenic abscess clinically or based on CT find- 
ings [1]. 

Haan et al. further explored the CT finding of 
air within areas of infarction after splenic artery 
embolization [45]. They found that air in areas of 
splenic infarction was associated with infection in 
only 17% of patients but that this rate increased 
to 33% in symptomatic patients [45]. Clearly, this 
implies that air is not pathognomonic of infection 
and that further investigation must be performed in 
these patients prior to splenectomy. While asymp- 
tomatic patients can likely be observed, patients 
with symptoms and a minimal amount of air should 
be treated with antipyretics. Aspiration and percu- 
taneous drainage should be considered, as should 
splenectomy, in patients with large areas of infarct 
air and/or severe symptoms [45]. 



Hepatic Artery Embolization 



4.2.1 

Patient Selection 



r similar to how splenic injuries are being 
treated, there has been a trend in recent years towards 
the nonoperative management of hepatic injuries, 
especially in hemodynamic ally stable patients [46- 
52]. Given its large size, the liver is obviously suscep- 
tible to injury in association with blunt trauma. In 
addition, iatrogenic trauma due to biopsies and other 
procedures can lead to vascular injury potentially 
requiring embolization as treatment. The vascular 
injuries seen are similar to those seen with other 
solid organ injury', including arterial laceration with 
extravasation in addition to pseudoaneurysm forma- 
tion but unique to the liver is the potential for fistulas 
to develop between any of the intrahepatic vascular 
structures and the biliary system. 

Almost two decades ago, Meyer et al. recom- 
mended the following criteria for nonsurgical 
management of blunt liver trauma: hemodynamic 
stability, CT scans showing simple parenchymal lac- 
erations or intrahepatic hematoma, <250 cc of free 
intraperitoneal blood, no other significant intraab- 
dominal injury, and availability of close monitor- 
ing [53]. The absence of associated injury must be 
determined with the use of abdominal CT, since it 
has been estimated that 35% ol patients may have 
associated injuries requiring surgery [52]. When 
these criteria are utilized, it has been estimated that 



il Solid Organ Trauma 



80-90% of all blunt liver injuries may be manage 
without Laparotomy [54, 55], Certainly, emboliz; 
tion and other interventional radiologic procedure 
have contributed to this success [56]. 



4.2.2 
Diagnosis 

For years, contrast enhanced CT has been the imag- 
ing technique of choice to evaluate hemodynami- 
cally stable patients with blunt abdominal trauma. 
It is considered a reliable way to detect hepatic inju- 
ries and has therefore been used to grade injuries 
(Table 4.2) and assess whether surgery or more con- 
servative treatment will be necessary and successful 
[57]. Importantly, the severity of hepatic injuries may 
be underestimated on CT scans but the decision to 
pursue nonoperative management has been shown to 
not be dependent on the grade of liver injury [50, 54]. 
Successful management of lieinoolvnamtcallY stable 
patients of all grades has been demonstrated [55, 
59-61]. In general, if the patient is considered stable 
after resuscitation and the hepatic injury is either 
isolated or in combination '.villi other injuries that do 
not themselves require surgery, then a nonoperative 
approach to management is utilized [61]. 



4.2.3.1 
Technique 

Flush aortography i'< rvpk ally required before selec- 
tive catheterization of the common hepatic artery, 
similar to that required before splenic artery cath- 
eterization. Even more so in the case of the hepatic 
arterial circulation is the potential for variant 
anatomy. Once the origin of the hepatic artery has 
been identified, selective catheterization can be per- 
formed. Whereas the goal in splenic artery embo- 
lization is to potentially embolize proximally and 
preserve flow to the spleen via collateral circulation, 
a more distal and selective embolization is often the 
goal when it comes to the treatment of hepatic arte- 
rial injury. However, in some situation, the embo- 
lization of the right or left hepatic artery might be 
necessary. In those situations, Gelfoam emboliza- 
tion can be performed with larger particles and 
torpedoes (Fig. 4.2). Fortunately, the contribution 
of the portal vein to the hepatic circulation enables 
the liver parenchyma to tolerate hepatic arterial 
embolization without significant ischemic injury. 
Given the more distal nature of this embolization, 
microcatheters and microcoils are often required 
for these procedures, although success has been 
reported using a variety of embolic agents including 
Gelfoam, polyvinyl alcohol, and coils (Fig. 4.2). 



4.2.3 

Hepatic Artery Embolization 

Embolization has been shown to be an effective 
treatment to offer patients that respond to resusci- 
tation but rapidly become unstable if resuscitation is 
withdrawn [61]. While it certainly would be appro- 
priate to operate on these patients, an understand- 
able desire to avoid the potential complications 
associated with surgeryhas increased the utilization 
of embolization in this setting [61]. 



4.2.3.2 
Results 

Reports dating back to 1977 have demonstrated the 
effectiveness of emlxiliz.it ion for patients experienc- 
ing blunt hepatic trauma [62-65]. Hagiwara et al. 
[66] demonstrated that nonsurgical management 
of Grade III or IV us ins ansiosrapiiy and Gelfoam 
embolization was successful in allpatients.Inalater 
study, Hagiwara et al. [67] prospectively evaluated 



Table 4.2. Mirvis Computed Tomographic Scan Hepatic imury Severity Scale For Blunt Hepatic 
Trauma [69] 



■Capsular avulsion; superficial lacerationis) ■■■.'] cm d^ep. or sr-bcapsuhr hematoma 
<1 cm maximal llucknrss, pei i porta I bio cm : in eking only 

r 1 ;; renchym.i I laceration! s! 1 -3 cm deep: cea:ra' o: subcapsular hematoma(s) 1-3 err 
diameter 

Lacerationis! >3 era deep: cemral or subcapsular hematoma !3! >3 cm la diameter 
Massiw ceiili'jl/s'.'.bcjpsiila:' hematoma >I0 c:v. in diameter, lobar us 
ir devascul.ai/ahon 



G. Siskin and J. Gol: 




Fig. -1 .2 a I. 41 year- dd ::;:.a with abdominal -.in.:', pelvis trauma, n Contrast-enhanced CT of ihe upper abdomen oemonsliates 
a hematoma tin," ,r.t.v c bleeding : r r . \ > 1 1 : .11 the 1 igbi hepatii oaivi'.ohvn'.a. b Cell .;■." trunk angiogram shi ws ;l'.e :'.i iei i,i. .tS [■:■:'. 
i slip ft iifciin) with early venous drainage i.bi,i:k .n :■■:■•■. 1. c Selective mioiooiithete: plaoement in [he feeding artery and er.ibo liga- 
tion with coj.s is performed. Angiogram ."em- 'lis I iir.es die persistence of extravasation ianp n i. d Hryaiie angiogram, oblique 
view, after embolization of another fertimg artery deirr 'itsi rates ihe extravasation iiinoiij. e 10 minutes aflrr einnoiizatio:;, 
there ivas a rr-ers! stent extravasation from iiepati-." artery. 7 lie right heratic arteiy is their emhohzed nsn'.g '..edoum toipedoes. 
f Contrast enhanced CT obtained 14 hours a iter embolization shows no more bleeding 



patients with Grade III-V injury who were treated hemodynnmiaUly stable patients in whom contrast- 
with angiography and embolization. They con- enhanced CT shows extravasation of contrast, even 
eluded that embolization should be used to manage when the injury is severe. Since embolization obvi- 



lii] Solid Organ Trauma 



ously has no effect on bleeding from juxtahepatic 
venous injuries, Hagiwara et al. advocate the use 
of surgery in these patients. They found that all 
patients with juxtahepatic venous injury required 
>2, 000 ml/hour of fluid for resuscitation. There- 
fore, they consider the combination of a Grade IV 
or V lesion and fluid requirements >2,000 ml/hour 
to maintain normotension an absolute indication 
for surgery. The success of hepatic artery emboliza- 
tion was also demonstrated by Pachter et al. [59], 
Carillo et al. [60], Mohr et al. [68], and Sugimoto 
et al. [69], who successfully performed embolization 
on patients after blunt liver trauma who were con- 
sidered stable after resuscitation. Ciraulo etal. [61] 
evaluated their experience utilizing embolization in 
patients with severe hepatic injury and found that 
embolization with Geltoam and coils was success- 
ful in patient whose stability was maintained only 
by aggressive continuous resuscitation. In these 
patients, embolization resulted in a reduction from 
continuous resuscitation to maintenance fluids, 
which inferred an improvement in hemodynamic 
stability. 



4.2.3.3 

Post-Embolization Follow-up 

Once the decision has been made to pursue nonop- 
erative management for patients with blunt hepatic 
trauma, an additional decision concerning follow 
up imaging needs to be made as well. Typically, 
follow-up CT scans have been performed to detect 
complications, to document healing of the liver 
injury, and to guide patients' activity restrictions 
[54]. However, some have advocated a selective 
approach to the performance of follow-up CT scans 
[70]. Ciraulo et al. [71] reviewed their experience 
in 95 patients with blunt hepatic injury to determine 
if follow-up scan altered management or discharge 
decisions. They found that follow-up CT scans did 
not alter the decision to discharge stable patients 
with Grade I— I II injuries. Cuff et al. [54], agreed 
with this finding and concluded that follow-up 
CT scans may not always be necessary in patients 
being treated nonoperatively. In their study, only 
two patients were found to have significant CT find- 
ings during their hospitalization: one patient had 
a bile leak and biloma while another patient had a 
hepatic artery to portal vein fistula. Both of these 
patients had Grade IV injuries and both continued 
to be managed nonoperatively. Based on this study, 
Cuff et al. concluded that follow-up CT scans are 



y in stable patients with Grade I, II, or 
III injuries. Instead, the need for follow-up imaging 
should depend on the results of serial clinical evalu- 
ations with CT scans indicated in stable patients 
with persistent abdominal pain, sudden change 
in clinical examination, unexplained tachycardia, 
fever, jaundice, or decreasing hemoglobin [54]. 

Complications alter embolization include delayed 
hemorrhage, hepatic necrosis, infection/sepsis, and 
biliary fistula. They manifest with abnormal physi- 
cal findings, elevated liver function tests, or find- 
ings on imaging studies such as ultrasound or CT 
(Fig. 4.3) [56, 61, 68]. Fever is common after hepatic 
artery embolization, occurring in as many as 69- 
100% of patients [60, 68]. Hagiwara et al. found 
bilomas in 4 of 54 patients and these were associ- 
ated with pseudoaneurysms in 3 of these 4 patients 
[66]. They propose that the presence of bile delays 
healing of liver injury and causes an inflamma- 
tory reaction that can ultimately lead to rupture 
of blood vessels and delayed hemorrhage. Mohr et 
al. reported on five patients with hepatic necrosis 
after embolization, all of whom required operative 
debridement or resection with one of these patients 
ultimately dying after the procedure [68]. 80% of 
the patients with hepatic necrosis also experience 
infarction of the gallbladder that required chole- 
cystectomy. These patients required embolization 
of right hepatic artery branches during their initial 
procedure. Mohr et al. also noted a 2.i% incidence of 
biliary leakage alter emboli zation and these patients 
typically required biliary drainage lor a median of 
1 month. In total, Mohr found that 21% of patients 
required a return to the operating room for hepatic 
complications while Knudson et al. reported a rate 
of 18% [72]. Of note, Carrillo demonstrated the 
importance of other interventional radiologic tech- 
niques such as percutaneous drainage and biliary 
drainage in managing several of these complica- 
tions, contributing to the continued nonoperative 
management of these patients [60]. 



Renal Artery Embolization 

4.3.1 

Patient Selection 

As stated throughout this chapter, there is a continu- 
ing movement towards nonoperative management 
for patients experiencing significant solid organ 



ind 1. Golzanan 




Fig. 4.3. CT scan o: lire liver 1 week ,ii':e: lirpatie artery embo- 
lization as treat mm; for .; Liaun^ili." liver I Liberation. The CT 
reveals a subcapsular fluid ja Ilea ion and an inliapjienihy- 
raal, air-containing abbess in die righl lobe of the liver 



parenchyma, grading the severity of injury [84] 
and confirming the presence of active bleeding and 
other peritoneal or retroperitoneal injuries [85]. It is 
effective at diagnosing injuries ranging from contu- 
sions and hematomas to shattered kidneys (Fig. 4.4) 
or avulsion of the renal ped icle [86, 87]. CT find ings 
indicative of an arterial occlusion include a lack of 
renal enhancement in the presence of a normal renal 
contour, rim enhancement, central hematoma, and 
abrupt cut-off of an enhancing renal artery [88]. 
Segmental arterial injury should be suspected when 
an area of decreased parenchymal enhancement 
corresponds to an area perfused by one of the seg- 
mental arteries [83]. 



4.3.3 

Renal Artery Embolization 



injury after trauma. This has been seen concerning 
the nonoperative management for patients experi- 
encing renal vascular trauma as well [73]. While 
some believe that surgical exploration is warranted 
because it ultimately improves the nephrectomy 
rate [73-75], other believe that nonoperative man- 
agement is more effective at preventing the need 
for nephrectomy [76-80]. Accepted indications for 
surgery include avulsion of the renal pelvis, inju- 
ries to the vascular pedicle, and life-threatening 
hemodynamic instability while embolization can 
be considered in patients with continuous hema- 
turia or massive hemorrhage due to renal vascular 
injury [81]. 



4.3.2 

Diagnosis of Renal Vascular Injury 

The diagnosis of renovascular injury may be diffi- 
cult in some patients, especially because hematuria, 
which is thought to be present in most patients, can 
be absent in up to 33% of patients with injuries to the 
renal artery [82]. This is why imaging is often nec- 
essary in these patients. In addition, patients with 
renal vascular injuries typically have other signifi- 
cant intraperitoneal and retroperitoneal injury [83], 
supporting the need for imaging evaluation. These 
other injuries can lead to the elevated injury sever- 
ity scores and increased transfusion requirements 
often seen in these patients [83]. 

At the present time, contrast-enhanced spiral CT 
is the best imaging modality tin assessing the renal 



4.3.3.1 
Technique 

As has been the case with the embolization of other 
solid organs as described in this chapter, it is rec- 
ommended that these procedures start with a non- 
selective abdominal aortogram. Variations in the 
number of arteries supplying one or both kidneys 
are numerous and therefore must be documented 
before attempts at selective catheterization are 
made. In addition, the angle of origin between the 
abdominal aorta and renal arteries will help guide 
catheter selection for catheterization. Aortography 
is also important tortile out traumatic disruption or 
dissection of the renal artery before selective cath- 
eterization is attempted. Embolization is typically 
performed as distal as possible, or as close as possi- 
ble to the site of arterial injury, in order to minimize 
the amount of devascularized renal parenchyma 
after the procedure. This typically requires the use 
of microcatheters and microcoils (Fig. 4.4). 



4.3.3.2 
Results 

Renal vascular injuries, caused by both blunt and 
penetrating trauma, can be effectively treated with 
arterial embolization from a supers elective catheter 
position, resulting in organ salvage and tissue pres- 
89-92]. Hagiwara et al. evaluated 46 
patients with evidence of renal injury on 
inal CT [93]. Twenty-one of these patients 
ide 3 or higher injuries and underwent angi- 



*$<kV- A 



■ 








a-g. 22 year-old female with a car accident, a Enhanced CT dem- 
• retroperitoneal Kind collection and partial light kidney fracture 
iiTJ iow). b Slice cauda! to image a shows a hyprrden.se retropei iloneal 
b!ee-.i tijt: and the transecied srginent .if the right kidney thai is taking tip 
contrast : ni row j. 1 1 relayed piiase of an aoitogram demonstrate normal 
iiTphiogiam ii! the left side but a small nephrogi am in the rig:: I kidney 
fiTi'iiiu'). (I Catheterization of The rem: I arterv feeding the superior part of 
lhe right kidney with norma, nephrogram, e Catheterization of the renal 
branch of transected segment of the right kirin.ev demonstrates an arte- 
rial transection ; biii.:k .incur j and an arte: ial extravasation i wit ire iirmu). 
f Angiogram obtained after selective coil embolization proximal to the 
transected vessels. Note the opacihcation of the pyexcakcea! system o:' the 
superior fragment ■ ■: right kidney (,-p ere ■.■. j.g Enhanced CT afle: emboliza- 
tion shows no extravasation with stagnation o:' contrast from emboliza- 
tion procedure in the retroperitoneal collection (arrow) 



0. Siskin ;i:id 1. i.".olz;i:;an 



Table 4.3. Organ Injury Score lor Blunt Renal Trauma* 

Grade Type [ icsirsj.it ion of Trsuinu 

1 Contusion Hermit'.:.! is;, imaging studies normal 
Hematoma Subc, , .. l su..i< hematoma (nonexpanding) 

2 Hematoma Periien-.il liema:om:; (contained, noneNpanomgi 
Laceration Cortical laceration (<l cm) without urinary 

3 Hematoma Perirenal hem a: cm:: (contained, nonexpandmgi 
Laceration Cortico:-:edullarv laceration detpei I linn 1 cm without 

4 Laceration Cortici'iiiedulkuy laceration deeper I:".;";:: : cm with collecting system injury 
Vascular Injury to main renal artery or vein w;lh conia.uied Hemo: rhage 

5 Laceration Comp!e-elr s!"ia:lere.:. .-i.dney, ureleropelviceal avulsion 
Vascular Avulsion of renal hik.im. ,".evascula:i^:ng ^.idnev 



. no:re\pand:ng perirenal hcina.loma 



ntlie. 



n for the Surgery of Trauma (AAST) [99]. 



ography. One patient had venous extravasation 
and ultimately required surgery; this was the only 
patient in this series who required surgery. Eight 
patients demonstrated arterial extravasation and 
were treated successfully with embolization. Suc- 
cess with embolization has been demonstrated in 
hemodynamics lly unstable patients presenting with 
gross hematuria, active bleeding, and symptoms of 
shock. These patients are at risk for significant mor- 
bidity and mortality with surgery supporting the 
use of embolization during their care. It has been 
shown that in these patients, embolization can help 
control bleeding without nephrectomy [94]. 

Embolization is also well suited to patients that 
are initially stable after trauma but develop delayed 
bleeding over the course of days, weeks, or months 
[86]. In these patients, the delayed bleeding is most 
likely due to the formation of a traumatic pseudoa- 
neurysm or arteriovenous fistula, which is more 
common in patients experiencing penetrating 
trauma than blunt trauma [95]. Pseudoaneurysms 
form after blunt trauma due to rapid deceleration- 
induced injuries to renal arteries [96, 97]. As they 
form, pseudoaneurysms can contact the collecting 
system, which can lead to the delayed hematuria 
often seen in these patients [95]. These pseudoan- 
eurysms can be successfully treated with selective 
embolization. 



4.3.3.3 
Post-EmboEization Folio 1 



-up 



There are potential complications associated with 
the surgical management of patients with renal vas- 
cular injuries. These include azotemia and persistent 
hypertension, which may possibly require nephrec- 
tomy for management [83, 98, 99]. At the present 
time, significant and persistent hype 



not been reported after superselective renal embo- 
lization [86]. The post-embolization syndrome that 
is commonly seen after solid organ embolization for 
other indications and consists of pain, leukocytosis, 
and fever, is uncommon after selective renal artery 
embolization in the setting of trauma [100]. 



Cookbook 

Splenic Artery Embolization 

• Proximal Embolization: In the absence of active 
contrast extravasation, the splenic artery is 
proximally embolized. We typically utilize 
either a 5F Cobra catheter or a 5F Omni-2 cath- 
eter to catheterize the celiac axis. Depending 
on the tortuosity of the vessel, we then either 
use the Cobra catheter or a microcatheter with 
a 0.021" inner luminal diameter for more selec- 
tive catheterization. Once the catheter is in 
place, just distal to the dorsal pancreatic artery, 
coils are deposited. The size of the coils chosen 
depends on the size of the vessel. 

• Distal Embolization: If active extravasation is 
present, we find it desirable to selectively cath- 
eterize and embolize the injured branch vessel. 
This requires the use of a microcatheter (0.021" 
inner luminal dia.) and appropriate guidewire. 
Once the microcatheter is as distal as it can be, 
microcoils (either straight or helical, depend- 
ing on vessel size) are used for embolization. 

Hepatic and Renal Artery Embolization 

• In patients with hepatic or renal arterial injury, 
our goal is to embolize as distally as possible, 
with the intent of sparing as much parenchyma 
as possible. This typically requires the use of 
microcatheter and microcoils as above. 



il Solid Organ Trauma 



l.Killeen KL, Shanmuganathan K, Boyd-Kranis R et al. 

(2001) CT findings .ifier embolization for blunt splenic 

trauma.] Vase Interv Radiol 12:209-214 
2.Haan ]M, Biffl W, Knudson MM et al. (2004) Splenic 

embolization revisited: j multicenter review. J Trauma 
56:542-547 

3. Chaudry IH, Tabata ¥, Schleck S et al. (1980) Effect of 
splenectomy on reticuloendothelial function and sur- 
vival following sepsis. ] Trauma 20:649-656 

4. Rabbinate CD, Frumenti JF (1977) Splenectomy and 
subsequent mortality in veterans or the 1939-1945 War. 
Lancet 2:127-129 

5. Green JB, Shackford SR, Sise M) et al. (1986) Late septic 
complications in adults following splenectomy for 
trauma.] Trauma 26:999-1004 

6. Liu PP, Lee WC, Cheng YF et al. (2004) Use of splenic 
artery embolization as .in adjunct to nonsurgical man- 
agement of blunt splenic injury. I Trjuma 56:768-773 

7. Knudson MM, Maull KI. (1999) Nonoperative manage- 
ment of solid org;:: 1 , muities: past, present, and future. 
Surg Clin North Am 79:1357-1371 

8. King H, Shumacker HB Jr (1952) Splenic studies 1: sus- 
ceptibility to infection after splenectomy performed in 
infancy. Ann Surg 136:239 

9. Pachter HL, Grau J (2000) The current status of splenic 
preservation. Adv Surg 34:137-174 

10. Godley CD, Warren RL, Sheridan RL et al. (1996) Non- 
operative manage; 1 .: en I of oil::'/ splenic injury in adults: 
age over 55 years as ;'. power fill indicator for failure. J Am 
Coll Surg 183:133-139 

1 l.CocanourCS, Moore FA, Ware DNetal. (2000) Age should 
not be a consideration for nonoperative management of 
.v. ml so.ef.ic ::'. u:y. I Ttauma 4S:ouo-fl 1 

12. Haan J, Scott ], Boyd-Kranis RL et al. (2001 ) Admission 
angiography for bin::: splenic injury: advantages and pit- 
falls. J Trauma 51 :1 1 61 -1165 

13. Lucas CE (1991) Splenic trauma: choice of management 
AnnSurg213:98-112 

14. Koury HI, Peschiera JL, Welling RE (1991) Non-operative 
management of blunt splenic trauma: a 10-year experi- 
ence. Injury 22:349-352 

15. Peitzman AB, Heil B, Rivera L et al. (2000) Blunt splenic 
injury in adults: muiti-insiitutioo.al study of the Eastern 
Association for the Surge: y of Trail ma. J Trauma 49:177- 
189 

16. Smith )S Jr, Cooney RN, Mucha P Jr (1996) Nonopera- 
tive management of the ruptured spleen: a revalidation 
of criteria. Surgery 120:745-751 

17. Albrecht RM, Schermer CR, Morris A (2002) Nonopera- 
tive management of blunt splenic injuries: factors influ- 
encing success in age '■■."■? years. Am Surg bS:22 7-2 30 

18. Federle MP, Courcoulas AP, Powell M et al. (1998) Blunt 
splenic injury in adults: clinical and CT criteria for man- 
agement, with emphasis on active extravasation. Radiol- 
ogy 206:137-142 

I °. Siianmiiganathan !<. Ivfrvis SE, Royd-Kranis R et al. 
UCO'Vi Nonsurgical management of o.mit s_-.r:;ic ,:• u:y: 
use of CT criteria lo select patients for splenic arteri- 
ography and potential endovasciLar therapy. Radiology 
217:75-82 

20. Molin MR, Shackford SR (1990) The management of 



snlen 
843 



a system. Arch Surg 125:8' 



21. Feliciano PD, Mullins RJ, Trunkey DD et al. (1992) A 
decision analysis of traumatic splenic injuries. J Trauma 
33:340-348 

22. Godley CD, Warren RL, Sheridan RLetal. (1996) Nonop- 
erative management of blunt splenic iniury in adults: age 
over 55 years as a powerful indicator of failure. J Am Coll 
Surg 183:133-139 

23. Hagiwara A, Yukioka T, Ohta S et al. (1996) Nonsurgical 
management of patients wiih Mi:::: splenic injury: effi- 
cacy of transcatheter arterial embolization. Am j Roent- 
genol 167:159-166 

24. Gavant ML, Schurr M, Flick PA et al. (1997) Predicting 
clinical outcome of nonsurgical management of blunt 
splenic injury: using CT to tevea. abnormalities of splenic 
vasculature. AJR 168:207-212 

25. Buntain WL, Gould WR, Maull KL (1988) Predictability 
of splenic salvage by computed tomography. J Trauma 
28:24-29 

26. Mirvis SE, Whitley NO, Gens DR (1989) Blunt splenic 
trauma in adults: CT- based classification and correlation 
with prognosis and treatment. Radiology 171:33-39 

27. Umlas SL.Cronan J] (1991) Splenic trauma: can CT grad- 
ing systems enable prediction of successful nonsurgical 
treatment? Radiology 178:481-485 

28. Moore EE, Cogbill TH, Jurkovich GH et al. (1995) Organ 
injury scaling: spleen and liver ! I 9 , 14 revision). J Trauma 
38:323-324 

29. RescinitiA, F link MP, Raptopoulous V et al. (1988) Non- 
operative treatment of adult sp lento trauma: development 
of a computed tomographic scoring system that detects 
appropriate candidates for expectant management. J 
Trauma .18:828-831 

30. Kohn JS, Clark DE, Isler RJ et al. (1994) Is computed tomo- 
gtaphic graomg of s:\rt: ic iniury ttsefu. in the nonsurgi- 
cal management of blunt trauma? i Trauma 36:385-390 

31. Becker CD, Spring P, Glattli A et al. (1994) Blunt splenic 
trauma in adults: can CT findings be used to determine 
the need for surgery: AJR 162:343-347 

32. Sutyak JP, Chiu WC.D'Amelio LF et al. (1995) Computed 
tomography is inaccurate :n estimating the severity of 
adult splenic injury. J Trauma 39:514-518 

33. Sclafani SJA, Weisberg A, Scalea T et al. (1991) Blunt 
splenic injuries: nonsurgical itealment with CT, arteri- 
ography, and transcatheter arterial embolization of the 
splenic artery. Radiology 181:189-196 

34. Sekikawa Z, Takebayashi S, Kurihara H et al. (2004) Fac- 
tors affecting clinical outcome of patients who undergo 
trans catheter arterial embohsation in splenic injury. Br J 
Radiol 77:308-311 

35. Gaunt WT, McCarthy MC, Lambert CS et al. (1999) Tra- 
ditional criteria for observation of splenic trauma should 
be challenged. Am Surg 65:689-691 

36. Sola fa n: SIA ! 19S1 ) The role of angiographic hemostasis 
in salvage of the injured spleen. 1'iag Radiol 141:645- 
650 

37. Davis KA, Fabian TC, Croce MA et al. (1998) Improved 
success in nonoperative management of blunt splenic 



ij. Siskin ;l:ld 1. L.olzaiian 



James CA, Emanuel PG, Vasquez WD et al. (1996) Embo- 
lization of splenic artery branch p sen do aneurysms after 
blunt abdominal trauma. I Trauma 40:835-837 
Sdafani SJA, Shaftan GW, Scalea TM et al. (1995) Non- 
operative salvage of computed tomography-diagnosed 
splenic injuries: utilization of angiography for triage and 
embolization for hemosiasis. 1 Trauma 39:P1 S-827 
Dent D, Alsabrook G, Erickson BA et al. (2004) Blunt 
splenic injuries: high nonoperative management rate 
can be achieved with selective embolization, j Trauma 
56:1063-1067 

Maddison F (1973! Embolic therapy of Hypersplenism. 
Invest Radiol 8:280-281 

Anderson JH.VuBan A, Wallace Setal. (1977) Transcath- 
eter splenic arterial occlusion: an experimental study in 
dogs. Radiology 125:95-102 

Pietropaoli JA, Rogers FB, Shackford SB et al. (1992) The 
deletrious effects of intraopeiative hypotension on out- 
come in severe head injuries. J Trauma 33:403-407 
Cocanour CS, Moore FA, Ware DN et al. (1998) Delayed 
complications of nonoperative management of blunt 
adult splenic trauma. Arch Surg 1998; 133:619-625 
Haan J, Bochicchio G, Kramer M et al. (2003] Air follow- 
ing splenic embolization: in fee den or incidental finding! 
Am Surg 69:1036-1039 

Hiatt JR. Harrier HD, Koeing BV et al. (1990] Nonopera- 
tive management of maior blum liver injury with hemo- 
peritoneum. Arch Surg 125:101-103 
Pachter HL, Spencer Fc, Hofstetter SR et al. (1992] Signifi- 
cant trends in the treat me:'/ o: hepalic trauma. Ann Surg 
215:492-502 

Hammond JC, Canal DF, Broadie TA (1 992] Nonoperative 
management of adult blunt hepatic trauma in a munici- 
pal trauma center. Am Surg 5B::"51 -5S5 
Meredith )W, Young JS, Bowling] et al. (1994) Nonopera- 
tive management of blunt hepatic trauma: the exception 
or the rule? J Trauma 36:529-535 

Croce MA, Fabian TC.Menke PG et al. (1995) Nonopera- 
tive management of blunt hepatic trauma is the treat- 
ing:/ of c!i o;. :e fo: h em o.. ivnam.ca.lv- -/able patients. Ann 
Surg221:744-755 

Knudson MM, Maull KI (1999) Nonoperative man- 
agement of solid organ imuiies. Surg Clin North Am 
79:1357-1371 

Durham RM, Buckley J, Keegan Met al. (1992) Manage- 
ment of blunt hepatic inuuies. Am I Surg 164:477-481 
Meyer AA, Crass RA, Lim RC et al. (1985] Selective non- 
operative management of blunl liver injury using com- 
puted tomography. Arch Surg 120:550-554 
Cuff RF, Cogbill TH, Lambert PJ (2000] Nonoperative 
management of blunt liver trauma: the value of follow- 
up abdominal complied tomography scans. Am Surg 
66:332-336 

Brasel KJ, DeLisle CM, Olson CJ et aL (1997) Trends in the 
management of hepatic injury. Am I Surg 174:674-677 
Harper HC, Maull Ki ! I'm ) Transcatheter arterial embo- 
lization in blunt hepatic trauma. South Med J 93:663-665 
Becker CD, GaJ I, Baer HU.VockP (1996] Blunt hepatic 
trauma in adults: correlation of CT injury grading with 
outcome. Radiology 201:215-220 

Mirvis SE, Whitley NO, Vainwright JR et aL (1989) Blunt 
hepatic trauma in adults: CT-hased classification and cor- 
relation with prognosis and tieaiment. Radiology 11:27-32 



59. Pachter HL, Knudson MM, Esrig B et al. (1996) Status 
of nonoperative management of blunt hepatic injuries 
in 1995; a multiceme: experience with 404 patients. J 
Trauma 40:31-38 

60. Carrillo EH, Spain DA, Wohltmann CD etal. (1999) Inter- 
ventional techniques are usertil aciuncts in nonoperative 
management of hepatic inuuies. I Trauma 46:619-624. 

61. Ciraulo DL.Luk S, Palter M et al. (1996) Selective hepatic 
arterial embolization of grade IV and V blunt hepatic 
injuries: an extension of resuscitation in the nonopera- 
tive management o\ Ira u ma tic hepalic injuries. J Trauma 
45:353-358 

62. Rubin BE, Katzen BT (1977) Selective hepalic artery 
embolization to control massive hepatic 1 \tmnrrr.3(;c 
after trauma. AJR 129:253-256 

63. Sclafani SJR ! 1 9S5) Angiographic Lu:::r:il »f :nlraperi 
toneal hemorrhage caused by injuries to the liver and 
spleen. Semin Intervent Radiol 2:1 39 1 47 

(■4. Hashimoto S, Hiramatsu K, Ido K «: ai.( 1990) Kxpa::dm(j 
role of emergency embolization ir. the management "f 
severe blunt hepatic trauma. CarLiiwa^ ntcrver" Kadml 
13:193-199 

65. Bass EM, Crosier JH (1977) Percutaneous control of post- 
traumatic hepatic hemorrhage by geltoam embolization. 
J Trauma 17:61-63 

66. Hagiwara A, Yuldoka T, Ohta S et al (1997) Nonsurgi- 
cal management of patients ivith blunt hepatic injury: 
efficacy of transcatheter embolization. Am T Radiol 
169:1151-1156 

67. Hagiwara A, Murata A, Matsuda T et al. (2002) The effi- 
cacy and limitations of transarterial embolization for 
severe hepatic injury. J Trauma 52:1091-1096 

68. Mohr AM, Lavery RF.Barone A et al. (2003) Angiographic 
embolization for live: injuries: low mortality, high mor- 
bidity. J Trauma 55:1077-1082 

69. Sugimoto K, Horiike S, Hirata M et al. (1994) The role 
of angiography in the assessmenl of blunt liver injury. 
Injury 25:283-287 

70. Allins A, Ho T, Nguyen TH et al. (1996] Limited value of 
routine follow up CT scans in nonoperative management 
of blunt liver and splenic injuries. Am Surg 62:883-886 

71. Ciraulo DL, Nikkanen HE, Palter M et aL (1996) Clinical 
analysis of the utility o: repeal computed tomographic scan 
before discharge in oh.: nl hepatic utility. [ Trauma 41 :sl L - 
824 

72. Knudson MM, Lim RC, Olcott EW (1994) Morbidity and 
mortality following ma: 1 .':' penetrating liver injuries. Arch 
Surg 129:256-261 

73. Nash PA, Bruce JE.McAninchJW (1995) Nephrectomy for 
traumatic renal injuries.] Urol 153:609-611 

74. Cass AS, Luxenbeig Mil 9 S3) Conservative or immedi- 
ate surgical management of blunt :enal injuries. J Urol 
130:11-16 

75. Carroll PR, Klosterman PW, McAninch JW (1988) Surgi- 
cal management of renal trauma: analysis of risk factors, 
technique and outcome. ) Trauma 28:1071-1077 

76. Thompson IM, Latourette H, Montie JE et aL (1977) 
Results of non-operative management of blunt renal 
trauma.] Urol 118:522-524 

77. McGopnigal MD, Lucas CE, Ledgerwood AM (1987) The 
effects of treatment iA :ena. trauma on renal function. ] 
Trauma 27:471-476 

78. Husmami DA, Morris JS (1990) Attempted nonopera- 



Visceral and Abdominal Solid Organ Trauma 



tive management of blunt renal lacerations extending 
through the corticomedullary amnion: the short-term 
and long-term sequelae. J Urol 143:682-684 

). Alt man AL, Haas C, Dinchman KH et al. (2000) Selective 
nonoperative manage::'. em .:■[" blunt grade 5 renal injury. 
I Urol 164:27-30 

1 Danuser H, Wille S, Zoscher G et al. (2001 ] How to treat 
blunt kidney ruptures: primary open surgery or conser- 
vative treatment with deferred surgery when necessary; 
Eur Urol 39:9-14 

i. McAninch J W, Carroll PR, Klosterman PW (1991) Renal 
reconstruct ion after unary. I "Jrol 145:932-937 

!. Grablowsky OM, Weichert RF, Goff JB et al. (1970) Renal 
artery thrombosis following blunt trauma: report of 4 
cases. Surgery 67:895-900 

i. Carroll PR, McAninch JW, Klosterman P et aL (1990) 
Renovascular trauma: risk assessment, surgical manage- 
ment. Ant outcome, i Trauma 30:547-555 

1. Moore EE, Shackford SR, Pachter HL et al. (1989) Organ 
injury scaling: spleen, liver, .;;"id kidney.) Trauma 29:1664- 
1666 

i. Becker CD, Mentha G, Schmidlin F et al. (1998) Blunt 
abdominal trauma in adults: role of CT in the diagnosis 
and management of visceral injuries. II. Gastrointestinal 
tract and retroperitoneal organs. Eur Radiol 8:772-780 

j. Dinkel HP, Danuser H.Triller J (2002) Blunt renal trauma: 
minimally invasive managemenl with microcatheter 
embolization - experience in nine patients. Radiology 
223:724-730 

'. KnstianssonA, Pedersen J (1993) Management of blunt 
renal trauma. Br J Urol 72:692-696 

1. Sclafani SJA.Goldstein AS, Panetta T et al. (1985) CT diag- 
nosis of renal pedicle injury. Urol Radiol 7:63-68 

1. Fisher RG, Ben Menachem Y, Whigham C (1989) Stab 



wounds of the renal artery brunches: angiographic diag- 
nosis and treatment by embolization. AJR 152:1231- 
1235 

90. Corr P, Hacking G (1991) Embolization in traumatic 
intrarenal vascular injuries. Clin Radiol 43:262-264 

91 . Velmahos GC, Chahwan S, Falabella A et al. (2000) Angio- 
graphic embolization for intra peritoneal and retroperito- 
neal injuries. World J Surg 24:539-545 

92. Velmahos GC, Demetriades D, Chahwan S et al. (1999) 
Angiographic embolization for arrest of bleeding after 
penetrating trauma to the abdomen. Am I Surg 178:367- 
373 

93. Hagiwara A, Sakaki S, Goto H et al. (2001] The role of 
interventional radiology in the management of blunt 
renal injury: a practical protocol. } Trauma 51:526-531 

94. Baron BJ, Scalea TM, Sclafani SJ et al. (1993) Nonopera- 
tive management of blunt abdominal trauma: the role of 
sequential diagnostic peritoneal lavage, computed tomog- 
raphy, and angiography. Ann hmerg Med 22:1556-1562 

95. Miller DC, Forauer A, Faerber GJ (2002) Successful angio- 
embolization of renal artery pseudoaneurysms after 
blunt abdominal trauma. Urology 59:444xiii-444xv 

96. Jebra VA, El Rassi I.Achouh PE et al. (1998] Renal artery 
pseu do aneurysm after blunt abdominal trauma. J Vase 
Surg 27:362-365 

97. Swana HS, Cohn SM, Burns GA et al. (1996) Renal artery 
pseu do aneurysm after blunt abdominal trauma: case 
report and literature review. I Trauma 40:459-461 

98. Grant P, Gifford RW, Pudvan WR et aL (1971] Renal 
trauma and hyper reus ion. Am I Cardiol 27:173-176 

99. (.lounger 1-11 :. 1 973 ) Hyper tens. on resuliing from segmen- 
tal renal artery infarction. Urology 1:189-190 

100-LarsenDW, Pentecost MJ (1992) Embolotherapy in renal 
trauma. Semm intervent Radiol 9:13-18 



5 Embolization and Pelvic Trauma 



Jeffrey J. Wong and Anne C. Robi 



iiili'O'.kiiiii.'i! .ii'.'.l Background 59 
Penetrating vs Blunt Trauma 59 
Causes and Epidemiology 59 
Sou ['tis or" Bleeding: Arterial vs Veno 



■;. Vlii 



60 



5.1.4 Why Not Surgery? 60 

5.1.5 Role oi Interventional Radiology -0 

5.2 Presentation 60 

5.2.1 Shock/Active Hemorrhage 61 

5.2.2 Pelvic Fracture & Plain AP Radiographs 61 

5.3 Guiding/Directing Therapy 
and the Laparotomy 61 

5.3.1 Diagnostic Perilor.riil Aspiration, Ultrasound, 
and Contrast-Enhanced CT 61 

5.3.2 External Fixation 62 

5.3.3 Laparotomy 62 

5.4 Endovasciiln: Therapy 63 

5.4.1 Access to Vessels 61 

5.4.2 The Initial l-v^ic A; ;e: iogram and Further 
Studies 63 

5.4.3 Angiographic Appearance c\ Hemorrhage 64 

5.4.4 Embolization Agents 64 

5.4.5 What To Do If No Bleeding Is Seen? 
The "Check" Arteriogram 66 

5.5 Complications 66 

5.6 Conclusion 67 
Cookbook: 66 
References 57 



Introduction and Background 



5.1.1 

Penetrating vs Blunt Trauma 



Like all traumatic inju 
can be classified into e 
Penetrating mechanisn 
gunshot wounds biopsi 



;s, trauma to the pelvis 

ter penetrating or blunt. 

such as stab wounds, 

, or surgical or percuta- 



J. J. Wong, MB ChB, BMedSc; A. C. Roberts, MD 
University of California, Si;:'. I 'iegc Medical Center, Divi 
Vascular and Interveiv.ioi'.a] Radiology, 200 West Arbor 
San Diego, California, 92103-B756, USA 



neous spine procedures, may directly injure pelvic 
organs, nerves, or the blood vessels. Blunt trauma 
exerts its effects through vessels being sheared 
against fixed ligamentous structures and avulsion 
of vessels attached to displaced bony pelvic struc- 
tures. Pelvic fractures also damage adjacent pelvic 
or retroperitoneal structures. The interventionist's 
primary concerns are vascular injuries, notably 
hemorrhage from the branches of the internal iliac 



5.1.2 

Causes and Epidemiology 

Pelvic fractures account for 3% of all skeletal inju- 
ries and are associated with a substantial mortality, 
with reported figures varying from 5% to 60% [1- 
21]. Mechanisms for pelvic fractures include motor 
vehicle accidents (57%), pedestrians hit by motor 
vehicles (18%), motorcycle accidents (9%), falls (9%), 
crush injuries (4%), and sports/recreational mecha- 
nisms (3%) [22]. Pelvic fractures are grouped based 
on the direction of the causative force. These forces 
include lateral compression, anteroposterior com- 
pression, vertical shear, and combinations of these 
three [23]. Most injuries to the infrarenal aorta are 
caused by seat belts compressing the lower abdomen 
in the anteroposterior aspect during car accidents. 

A closed stable fracture with stable vital signs 
offers the best prognosis, while patients with an 
open fracture and hemodynamic instability have 
a higher mortality. This latter subgroup only rep- 
resents l%-2% of all pelvic injuries seen in Level 1 
trauma centers [24] but no other skeletal injury car- 
ries such a high mortality rate. 

As hemorrhage is the most common treatable 
cause of death in this population of patients, it is 
imperative that vascular injuries are treated swiftly 
ively. Clarke et al. [25] suggested that the 
by 1% every 3 minutes 
hemodynamically unstable. 



risk of mortality 
that a patient ren 



:. Wong ;ind A.G.Roberts 



5.1.3 

Sources of Bleeding: 

Arterial vs Venous vs Marrow 

Direct bleeding from the fractui 
bones or from injured pelvic 
can cause pelvic bleeding. The anatomy of the ret- 
roperitoneum and its contents provides a natural 
tamponading effect on the fragile venous plexus 
adjacent to the pelvic bones and osseous bleeding. 
However, in the event of pelvic disruption and an 
unstable pelvic fracture, this tamponading effect is 
lost. A 3-cm diastasis of the symphysis pubis will 
increase the potential volume of the pelvis from 4 
liters to 8 liters [26]. The lack of valves between the 
inferior vena cava and the pelvic veins allows for 
potential catastrophic blood loss if the retroperi- 
toneum is violated. Pelvic fixation, be it invasive 
or noninvasive, will re-establish and maintain the 
tamponading effect, thus controlling bleeding from 
the venous and osseous structures. A dog model has 
been used to show that ligation of multiple pelvic 
arteries had no effect on the pelvic venous pres- 
sure [27]. Because the source of pelvic hemorrhage 
is from venous structures 80%-90% of the time 
[28,29], there is a strong argument for starting with 
pelvic fixation and intrapelvic compression. Clini- 
cally, it is not possible to determine the source of 
pelvic hemorrhage without the aid of radiographic 
studies. Thus there remains much debate as to 
whether a patient who has presumed pelvic hem- 
orrhage undergoes angiography or pelvic fixation, 
since fixation can only temporize small vessel 
hemorrhage [28, 30]. If the bleeding source is arte- 
rial, angiography and embolization is necessary. 
Miller et al. [31] showed that if patients present with 
hypotension from a pelvic fracture, poor response 
to resuscitative efforts indicates the presence of 
arterial bleeding in over 70% of patients. Mean- 
while, responsive patients are unlikely to have arte- 
rial bleeding, with a negative predictive value of 
100%. 



pathways, as well as large hematomas obscuring 
the surgical field further complicate the technical 
aspect of surgical control of arterial hemorrhage. 
An obscured field of view increases the likelihood 
of complications such as nerve injury. The primary 
operative option for controlling pelvic bleeding 
consists of packing and correction of coagulopathy. 
Temporary aortic clamping has been suggested to 
improve access to the site of bleeding [32]. 



5.1. 5 

Role of Interventional Radiology 

The interventional radiologist has become the cen- 
tral figure in treating traumatic pelvic and retro- 
peritoneal arterial hemorrhage, and with impressive 
results. Angiographic embolization has a success rate 
between 85% and 100% when bleeding sites can be 
identified [33, 34]. The first-line therapy for an unsta- 
ble patient with a pelvic tract ure should be immediate 
angiographic evaluation and embolization. 



Presentation 

All trauma patients should be initially assessed fol- 
lowing standard Advanced Trauma Life Support 
(ATLS) guidelines, the details of which cannot be 
covered in this text. However, as interventional 
radiologists, we are most concerned with signs and 
symptoms that will raise our suspicion of pelvic 
fractures and hemodynamic compromise. As the 
presentation of patients with life-threatening hem- 
orrhage can vary from exsanguinating shock to 
subtle innocuous signs, a detailed knowledge of the 
presentations of pelvic trauma is necessary. 



5.2.1 

Shock/Active Hemorrhage 



5.1.4 

Why Not Surgery? 






Evidence ofshockconsists of hypotension, tachycardia 
(pulse > 100 beats per min), tachypnea, cool exlremi 
ties, low urine output, and a progressive decline in 
cal approach to retroperitoneal bleed- the level of consciousness. A normal blond pressure 
ing is not a treatment for retroperitoneal pelvic may be misleading in young patients, because the 
bleeding. Dissection into the retroperitoneum and blood pressure may be maintained by a compensatory 
pelvis results in the loss ot (lie internal compression increased heart rate. In these patients, hypotension 
effect provided by adjacent anatomic structures, occurs only when this mechanism fails and usually 
resulting in increased bleeding. The rich collateral indicates impending cardiovaskul > . •■! . •■■ 



Embolization and Pelvic Traum 



The hemoglobin level is not a specific marker of 
hemorrhage as, in the acute stages, the hemoglobin 
or hematocrit is likely to be normal. A few hours 
are required before extravascular flu ids equilibrate 
with the blood and for laboratory values to reflect 
blood loss. It is imperative to obtain two intrave- 
nous accesses with large bore cannulas and start 
an infusion of intravenous fluids as soon as pos- 
sible. O-negative blood should be readily available, 
and blood samples should be sent early for cross 



Guiding/Directing Therapy and the 
Laparotomy 

Once the suspicion of pelvic hemorrhage has been 
raised, one must systematically follow a protocol 
that will ensure injuries and sources of bleeding 
are addressed in order of gravity. Heetveld et al. 
[39] published an evidence-based algorithm for the 
management of hemodruainlcalh unstable pelvic 
fracture patients (Table 5.2). 



5.2.2 

Pelvic Fracture & Plain AP Radiographs 

The suspicion for a pelvic fracture should start with 
a history of a high-energy impact such as a motor 
vehicle accident or a fall from a substantial height. 
Careful inspection may reveal leg length discrepan- 
cies (Roux sign), flank ecchymosis (Grey-Turners 
sign}, blood at the urethral meatus, rectal bleeding 
and/or vaginal bleeding. Hematomas observed on 
the proximal thigh superficial to the inguinal liga- 
ment or over the perineum are also suggestive of 
a pelvic fracture (Destot's sign). Tenderness with 
gentle pressure on the iliac wings bilaterally also 
supports the diagnosis of a pelvic fracture. Care 
must be taken not to apply too much pressure as 
this may aggravate hemorrhage in an as-yet-undi- 
agnosed unstable fracture. Rectal examination may 
reveal a high-riding prostate or a large hematoma 
or palpable fracture line (Earle's sign). Examina- 
tion of the lower limbs may also show neurovascular 
deficits. 

Most patients will usually have plain films of 
the chest and pelvis, regardless of their hemody- 
namic situation. Anteroposterior (AP) pelvic films, 
although only 68% sensitive for diagnosing all frac- 
tures [35], will reveal and define any large fractures 
which would raise our suspicion of retroperitoneal 
bleeding. It has been shown if an unstable fracture 
pattern is seen or suspected, the probability of pelvic 
arterialbleeding is approximately 52% [11, 14,34, 19, 
21, 36, 37]. Niwa et al. [38] showed that AP pelvic 
films can be useful in predicting hemorrhage sites 
based on the location and severity of the fracture. 
Interestingly, stable pelvic fractures have shown to 
be more strongly associated with abdominal bleed- 
ing rather than pelvic bleeding [21]. 

Kane's classification of pelvic radiograph's 
helps to convey the gravity of the bony injury (See 
Table 5.1). 



5.3.1 

Diagnostic Peritoneal Aspiration, Ultrasound, 

and Contrast-Enhanced CT 

The initial examination of the patient may elicit 
signs of peritoneal irritation on abdominal pal- 
pation. As the laboratory and plain film inves- 
tigations fail to address the abdominal cavity, 
these clinical signs will point the trauma team 
to proceed with further investigations. An acute 
abdomen in the setting of hemodynamic instabil- 
ity should result in either a diagnostic peritoneal 
lavage (DPL) or focused abdominal sonography for 
trauma (FAST). These investigations will reveal 
blood/free fluid in the abdominal cavity suggest- 
ing intra-abdominal hemorrhage and, in the pres- 
ence of hemodynamic instability may indicate the 
need for a laparotomy. 

It is not recommended that hemodynaiuieally 
unstable patients undergo a computed tomography 
(CT) scan. However, in those patients stable enough 
to be transported, contrast-enhanced CT of the 
abdomen and pelvis can help distinguish actively 
extravasating contrast material from clotted blood 
[40]and thus guide surgical or angiographic ther- 
apy. 

In the hemodynamically ualMe patient, CT is the 
preferred method of investigation [41]. Cerva et al. 
[42] in 1996 showed that using angiography as a gold 



Table.". 1. Kjne's c]jss:i'li.":U'i::ii] of pelvii fruciine 
K.ine's d.iisi- Pefinkion 



Fracture of only 1 pelvic bor 



Single breaks in the ring near the pubic sym- 
physis or a sacroiliac joint 

Double breaks in the ring 
AiViahJjr fractures 



'.. Wong 'in J A. G. Roberts 



Table 5.2. Algorithm for the mana; 
unstable pelvic fracture patients 



caliy unstable patient with a pelvic fracture 




Laparotomy 



on catheter angiography. This posed a challenging 
therapeutic dilemma. As she was hemodynamically 
stable and there were no signs of hypovolemia, the 
eventual decision was not to embolize. The patient 
returned to the intensive care unit and did well. 



5.3.2 

External Fixation 



:al. [39] suggest 

i tamponading 

i open surgical 

e that may be 




Following DPL or FAST, Heetveld e 
using external fixation to provide 
effect. Internal pelvic fixation is ar 
procedure that consumes precious ti 
better spent on laparotomy or angiography. Nonin- 
vasive methods ol pelvic fixation include a pelvic 
sling, pelvic binder, C-clamp, military anti-shock 
trousers (MAST), and external fixation. The latter 
two options are most relevant to the intervention- 
ist because they impair access to the femoral arter- 
ies [44]. A hole can be cut with scissors into the 
upper edge of a pelvic binder to allow access into 



standard, contrast enhanced CT was 85% specific, 
84% sensitive and 90% accurate. This study used 
10-mm collimation at 20-mm intervals. However, 
a more recent study by Pereira et al. [43] found a 
sensitivity of 90%, specificity of 99% and accuracy 
of 98 with helical CT using 10-mm intervals with 
a pitch of 1.0:1 to 1.5:1. They suggested its use as 
a method for screening polytrauma patients with 
pelvic fractures to accurately identify patients who 
would benefit from emergent angiographic embo- 
lization. It is important that patients who may be 
going on to angiography not receive oral contrast 
material that would interfere with the angiographic 
evaluation. 

With improving technology resulting in faster 
image acquisition and higher-resolution images, 
contrast-enhanced multi-detector CT has become 
increasingly accurate and may even surpass conven- 
tional angiography in sensitivity. In one study, four 
hemodynamically stable patients exhibiting con- 
trast extravasation on CT did not require emboliza- 
tion during hospitalization [43]. An example of this 
clinical scenario is seen in Fig. 5.1, which involved 
a hemodynamic ally stable trauma patient with vis- 
ible extravasation on CliCT which was undetectable 



Following Heetveld et al.'s guidelines [39], if evi- 
dence of intra-abdominal hemorrhage is found, a 
laparotomy is indicated. As a general rule, intra- 
abdominal hemorrhage takes priority over retroper- 
itoneal pelvic bleeding. So, in the face of detected 
abdominal free fluid, a pelvic fracture and hemody- 
namic compromise or catastrophic exsanguination, 
a laparotomy is first-line therapy. If, after achiev- 
ing abdominal hemostasis [inn a- abdominal repair], 
retroperitoneal pelvic bleeding is detected and 
hemodynamic instability continues pelvic packing 
with large sponges should be performed and the 
patient be taken to angiography. Eastridge et al. 
[21], however, suggest considering angiography over 
laparotomy with unstable pelvic fractures, despite 
the presence of hemoperitoneum. 

Ligation of the internal iliac artery has been 
shown not to lead to satisfactory reduction in bleed- 
ing [45-48]. This is thought to be due to the rich col- 
lateral blood supply to the pelvic region and the loss 
of the tamponading effect of the retroperitoneum. 
Following surgery, angiography is warranted if the 
patient requires transfusions of 4 units or greater in 
24 hours or hemodynamic instability persists [36, 
49]. 



Embolization and Pelvic Trauma 




Fig5.1a,b. Fl1 : :v.vt.'.:-..aI lei'.o'.e v.;.< .hi lesuajriec c.r.vei ;:'.\v.|yec. in :■. roiid r i l : . : :: ■_" accidem Ij .ivelma lit 55 mpri. She wiis 
hemO';iyn.imiciilly s:a'rC.e when the mi;ial pelvic CT was performed, a coivrast-enhaiiced CT of the pelvis show? I'ighi pelvic 
fri;c lures and ac iive contrast extravasation ii<n-nu) with mass effect ■:■:: the bladder. This hncn'.g p:c:v.p:ed angiographic stuck 
of ihe internal time arteries, b Selective angiogram ot" me r;ehi intern.; I iliac shows no contriisl exiravasiili on. As :he patient 
was hemodynamically stable, no embolization was performed. 



5.4 

Endovascular Therapy 

Once abdominal hemorrhage has been ruled out 
by FAST, CT, or DPL with continued hemodynamic 
instability or if contrast extravasation is demon- 
strated on CT the patient should be transferred to 
the angiographysuite.lt is imperative that the resus- 
citative process is not impeded by this transfer, and 
that a full complement of clinical staff accompany 
the patient during angiography and embolization. 

It is important that angiographic evaluation is not 
delayed, since patients who have experienced signif- 
icant blood are at risk ot becoming coagulopathy. 
It is vital, therefore, to have a variety of blood prod- 
ucts available, and coagulation/hematologic studies 
should be performed before, during, and after the 
resuscitation phase. 



5.4.1 

Access to Vessels 

A femoral approach is preferred. If the side of the 
pelvic injury is known, the contralateral femoral 
artery should be used. This is because it is easier to 
catheterize 2nd- and 3rd-order vessels on the con- 
tralateral side, over the aortic bifurcation. If bilat- 
eral femoral artery access is impaired, an axillary or 
brachial approach can be used. An upper extremity 



approach is sometimes useful if an external pelvic 
fixator has already been placed or there are bilat- 
eral pelvic injuries. Ultrasound guidance may be 
helpful in patients with large hematomas involving 
the groin. 



5.4.2 

The Initial Pelvic Arteriogram and Further 

Studies 

The angiographic search for bleeding should start 
with a pelvic arteriogram with a 5 F pigtail cath- 
eter positioned above the aortic bifurcation. The 
most commonly injured vessels include the supe- 
rior gluteal, internal pudendal, obturator, infe- 
rior gluteal, lateral sacral, and iliolumbar arteries. 
The arteries that are injured tend to be associated 
with the bony injury; thus sacral fractures and 
sacroiliac joint disruption are associated with 
superior gluteal, iliolumbar, and lateral sacral 
arterial injury. Fractures of the pubic ramus and 
acetabulum tend to cause injury to the internal 
pudendal and obturator arteries. Therefore, one 
should pay attention to the plain films or CT to 
direct the angiographic evaluation. If no bleed- 
ing is observed, selective right and left internal 
iliac arteriograms should be performed. Oblique 
views frequently help "open up" the branches of 



the 



:. Wong ;ind A.G.Roberts 



Access to either the contralateral or ipsilateral 
internal iliac arteries can be facilitated using a Walt- 
man's loop technique with a Cobra 2 catheter; alter- 
natively, a long reverse curve catheter can be used. 
Care must be taken not to catheterize too distally so 
as to ensure visualization of the lateral sacral and 
iliolumbar arteries [50]. Carbon dioxide offers an 
alternative contrast agent that has the benefits of no 
allergic reactions, nephrotoxicity, or volume limi- 
tations, low cost and flexibility of use with differ- 
ent sized catheters [51-53]. High-pressure contrast 
injections should be avoided since they may poten- 
tially dislodge newly formed clots and result in loss 
of hemostasis [54]. 



Angiographic Appearance of Hemorrhage 

See Table 5.3 for the angiographic manifestations of 
vessel injury [54]. Care must be taken not to confuse 
the normal uterine blush or bulbospongiosal stain 
'.villi the blush associated, with contrast extravasa- 
tion (Fig. 5.2). Reported causes of false negative arte- 
riograms include intermittent vasospasm, spontane- 
ous vaso-occlusion of a bleeding artery by thrombus 
formation, venous bleeding that is not shown by 
arteriography, and technical difficulties selectively 
catheterizing the bleeding artery [54]. Arteriove- 
nous fistulas and pseudoaneurysm are recognized 
late complications of pelvic trauma. 



5.4.4 

Embolization Agents 

Embolization of visualized bleeding site(s) in the 
pelvis is typically performed using Gelfoam. Gel- 
foam pledgets have excellent properties for trauma 



use because they usually dissolve over several weeks 
and may allow for re canalization of the vessel fol- 
lowing healing. The size of the particles is also opti- 
mal because they are large enough not to invade the 
capillary bed, while small enough to prevent flow 
from collateral vessels and stop bleeding. Gelfoam 
powder, which has a much smaller diameter, should 
not be used for pelvic bleeding since it has the poten- 
tial to cause ischemia and has been implicated in 
nerve damage [55]. 

"Scatter" embolization using a Gelfoam 'slurry' 
provides a rapid solution in the face of multiple 
bleeding points and a hemodynamically unstable 
patient who cannot tolerate the time required for 
subselective catheterization. In this scenario, the 
catheter is placed proximally in the internal iliac 
trunk to allow flow of the embolic material to all 
bleeding points. If the patient is hemodynamically 
stable, further subselection can take place in search 
for the bleeder. However, it must be stressed that an 
ideal subselective embolization must take second 
place to the cardiovascular status of the patient. 

To protect the gluteal arteries from inadvertent 
embolization, spring coils can be placed at their ori- 
gins. Coils can also be added for larger lacerations in 
higher-caliber proximal vessels. In this scenario, Gel- 
foam may flow straight out of the vessel and into the 
pelvic cavity, but a large-caliber, proximally placed 
coil would not. Gelfoam may then be placed on top 
of the coil, which acts as a scaffolding, and if neces- 
sary, a second coil may be placed creating a "Gelfoam 
sandwich". Ideally, the injured segment should be 
crossed so a distal coil can be placed which will have 
the beneficial effect of preventing retrograde flow 
from collateral vessels beyond the injury. Coils can 
be used to treat AVFs and pseudoaneurysms, closely 
packed tip to and proximal to any observed lesion. 

Absolute alcohol and particulate polyvinyl alco- 
hol (PVA) emboli have no role in the trauma patient. 



Table 5.3. The angicgrapiiic m.iiihestaUC'ns of vessel injury [54] 

Angiographic manifestations of vessel injury 

Arterial cut-off 

Muni! irregularities or flap 

Laceration 

Thrombosis 

I ii&ed ion 

Free-flow contrast extravasation 

Stagnant intra parenchymal a or u:r. illation of contras 

Parenchyma I blush 

Stagnant arterial or venous flow 

I'MTuse v,i ;; i.-s c o n s l r i c : i i:-- :i 

Pseudoaneurysm 

Arteriovenous fistula 

Vessel displacement 



Angiographic nunifesiat. 
Free-flow contrast ei 
Stagnant intraparenchymal ;u 
Disruption of visceral contou 
Displaced organ 
intro parenchymal avascular z 



Embolization and Pelvic Trauma 




Fig 5.2a-d.Thirly-c>iie-yeai-o!d male involved m a motorcycle accidenl collision and s litre red multiple ink' lies inch, ding • ighi 
reran r tYaciure and .1 laceration 0; -he rigid bullock. The patient was iiypotensive and !■■:■ cletinne pelvic fractures were seen 
on plain iilm. a f-vivic CT demon si rated active extravasation 01' contrast from a rigid superior gluteal artery branch and an 
expanding rig 111 sice, b Ai :e: ial plus; angiogram ■ ■:' I lie 1 igb: inter 11 a I iliac demons: rates i.\\- ■ aivas of e.v.i avasation. c Venous 
pliase angi' 'gum snows retention 0:' conlias: in die s impeded area:;. (1 K:st-Gelloam embolization angiogram shows 1: 
of the distal branches oi" the superior gluteal artery. 



Unlike Gelfoam, which sprires the capillary bed ves- 
sels, alcohol causes sclerosis of all vessels it comes 
into contact with, including those at the capillary 
level. This results in irreversible end-organ ischemia 

Angiography may reveal injuries to larger more 
proximal vessels, such as the common iliac or exter- 
nal iliac arteries, traditionally treated surgically. 
However, with the advent of covered stents, inter- 



ventional radiologists may be able to offer a less 
invasive endovascular solution [42, 56]. In some 
unusual circumstances, temporary occlusion bal- 
loons can be used to obtain hemostasis allowing for 
surgical repair. 

Following embolization of a bleeding site, the 
internal iliac artery should be checked proximally to 
make sure there are no other bleeding sites that were 
not recognized earlier. Because of the rich cross- 



'.. Wo jig and A. G. Roberts 



pelvic collateral supply, the contralateral internal 5.6 

iliac artery should then be evaluated and embolized Conclusion 

as necessary. Once embolization is felt lo be com- 
plete, it may be reasonable to perform a final flush 
pelvic arteriogram. 



5.4.5 

What To Do If No Bleeding I s Seen? 

The "Check" Arteriogram 

Inthe setting of hemodynamic instability and unde- 
tectable extravasation from the pelvis on angiogra- 
phy, further investigation of other vessels includ- 
ing the lumbar branches, branches of the common 
femoral artery, superficial femoral artery, and pro- 
funda femoral artery should be performed. If there 
is potential for splenic, hepatic, or renal injury, these 
vessels should also be evaluated. If all other poten- 
tial arterial sources have been excluded and the 
patient remains hemodynamically unstable, then 
empiric embolization of the internal iliac arteries 
may be performed. 



Complications 



Endovascular therapy is now established as the 
treatment modality of choice for retroperitoneal 
and pelvic bleeding secondary to trauma. Despite 
evidence to support earlier involvement of the inter- 
ventional radiologist, some trauma centers still fail 
to consider angiographic study until much later into 
the resuscitative process. The adoption of an evi- 
dence-based trauma algorithm (Table 5.2) is vital to 
ensure rapid and decisive treatment. 



Cookbook: 




For selective angiogram 




• 5 French Sheath 




• Gtidewire (Terumo) 




■ Long reverse curve catheter (RUC 
excellent catheter for trauma as it i 


- Cook, Inc) an 

s for fibroid emboli- 


Alternative catheters 




• C2 catheter, can beused to as is, oi 

Waltraan Loop 


used to form a 



Although a post-embolizat ion arteriogram may show 
complete hemostatic control, a second embolization 
maybe needed. Vessels that had previously "clamped 
down'Vvasoconstricted due to shock may re-open fol- 
lowing reperfusion from the ensuing resuscitation 
and increased systemic blood pressure [54]. These 
vessels may have initially been injured, but were not 
detectable on initial arteriogram and therefore pro- 
vide a new source of hemorrhage. 

Embolization, by definition, reduces blood flow 
distally. Therefore, it is no surprise that distal necro- 
sis is a recognized complication of trauma emboli- 
zation. However, unintentional reflux of emboliza- 
tion material from the internal iliac into the femoral 
artery can cause inadvertent ischemia in the leg. 
Sciatic palsy with associated foot drop and sacral 
plexus palsy has been reported [57]. Embolization 
of the superior gluteal artery in a patient who will be 
subjected to prolonged bedrest may cause sacral and 
buttock ischemia leading to skin break down [58]. 
Sexual dysfunction seems not to be a complication 
of bilateral internal iliac artery embolization, but is 
more likely a result of nerve injury secondary to the 
fracture or pelvic trauma [59]. 



Embolization materials 

• Gelfoam - may be cons titu ted .'.:; pledgels. torpedoes, 
or slurry. Do net use Gel foam powder 

• Coils - may be : .:sed pari:c : .:larly to;- larger vessels 

• May use in comb: nation wit:: G e I fo a :r. for "Gel foam 
sandwich" 

• Very occasionally i"o: large vessel trauma, occlusion 
balloons or covered stents may be appropriate 



> Never forge: that r lei? selective embolization and a 
live patient :s preferable to a lechmc.'.l tour de force 
and a dead patient! 

> Long reverse curve ca:lie:er allows for :'. very fast way 
to access the interna! iliacs and to perform subselect:ve 
embolization cu:ck!y with .arge pieces of Gelfoam or 
large coils 

> Make sure if embolization done on one side, that the 
other iliac artery is evali.ir.ted to make sure no collat- 
eral flow 

> If :liac arteries are ok, but pat:eiil stil: ::emodynami- 
lm I iy unstable, check for bmbar bleeding or for bleed- 
ing from branches of femoral arteries 



Embolization and Pelvic Trauma 



Trunkey DD, Chapman MW et al. (1974) Mana^meni 
of pelvic fractures in blunt trauma injury. ] Trauma 
14(ll):912-23 

Kothenoerger D, Velasco R et al. i i97S) Open pelvic frac- 
ture: a lethal injury. I Trauma 18(3):184-7 
Rothenberger DA, Fischer RP et al. (1978) The mortality 
associated with pelvic fractures. Surgery ^4(3):356-61 
McMurtry R, Walton D et al. (1980] Pelvic disruption in 
the polytraumatized patient: .i managemem protocol. Clin 
Orthop(151):22-30 

Gilhland MD, Ward RE et al. (1982) Factors affecting mor- 
tality in pelvic fractures. J Trauma 22(8):691-3 
Richardson |D, Harty J et al. (1982) Open pelvic fractures. 
I Trauma 22(7):533-8 

NaamNH, Brown WH et al. (1983) Major pelvic fractures. 
Arch SurgllS(5):61G-6 

Mucha P Jr. and Welch TJ (1988) Hemorrhage in major 
pelvic fractures. Surg Clin North Am 68(4):757-73 
Panetta T, Sclafani SJ et al (1985) Percutaneous transcath- 
eter embolization for massive bleeomg from pelvic frac- 
tures. J Trauma 25(1 1 ):1021 -9 

Moreno C, Moore BE et .il. i I 936) Hemorrhage associated 
will: maior pelvic fracture: a multispeciaily challenge. ■ 
Trauma 26(ll):987-94 

Cryer HM, Miller FB et al. (1988) Pelvic fracture classifica- 
tion: correlation with hemorrhage. I Trauma _3i7j: : ;7.i- c : 
Evers BM, Cryer HM et al. (1989) Pelvic fracture hemor- 
rhage. Priorities in man a gem em. A:ch Surg 124(4):422-4 
Flint L, Babikian G et al. (1990) Definitive control of mor- 
tality from severe pelvic fracture. Ann Surg 211(6):703-6; 
discussion 706-7 

Poole GV.WardEFetal. (1991) Pelvic fracture from major 
blunt trauma. Outcome is determined by associated ami- 
nes. Ann Surg 213(6):532-8; discussion 538-9 
Gruen GS, Leit ME et al. (1994) The acute management of 
hemodynamically unstab.e mall ip.e trauma patients with 
pelvic ring fractures. J Trauma 36(5):706-1 



Poole GV and Ward EF (1994) Causes of mortality in 
patients with pelvic fractures. Orthopedics 17(8):691-6 
r.a ,; t ridge Bl ai":d Hargess AK :. I L)L, '7i Peaesiiian pelvic frac- 
tures: 5-year experience of a major urban trauma center, j 
Trauma 42 (4):69 5-700 

Bassam D, Cephas GA et aL (1998) A protocol for the ini- 
lia. management of unstable pelvic fractures. Am Surg 
64(9):862-7 

Hamill J, Holden A et al. (2000) Pelvic fracture pattern 
predicts pelvic arterial haemorrhage. Aust N Z ] Surg 
70(5):338-43 

Ertel W, Keel M et al. (2001) Control of severe hemorrhage 
using O-clamp and pelvic packing in multiply imured 
patients with pelvic ring disruption. ) Orthop Trauma 
15(7):468-74 

Eastridge B|, Starr A et al (2002) The importance of frac- 
ture pattern in guiding therapeulic decision-making in 
patients with hemorrhagic shoes ,m.- pelvic ring disrup- 
tions. J Trauma 53(3):44S-50: discussion 450-1 
Dalai SA, Burgess AR et al. (1989) Pelvic fracture in mul- 
riple traama: classification by mechanism is key to pattern 
of organ injury, resuscitative requirement, and outcome. 1 
Trauma 29(7):981-1000; discussion 1000-2 



23. Burgess AR, Eastridge B], et al. (1990) Pelvic ring disrup- 
tions: effective classification svstem and trealmenl proto- 
cols. J Trauma 30(7):848-56 

24.Tscherne HPT (1998) Unfallchirurgie: Becken und Acetab- 
ulum. Berlin, Springer 

25. Clarke JR. Trooskin SZ et aL (2002) Time to laparotomy for 
intra -abd omnia. meeding from i rat: ma does affect survival 
for delays up to 90 minutes. J Trauma 52(3):420-5 

ao.Agnew SO, ;]°94i Hemodynamicaily unstable pelvic frac- 
tures. Orthop Clin North Am 25(4):715-21 

27.Ger R, Condrea H et al. (1969) Traumatic intrapelvic retro- 
peritoneal hemorrhage. An experiment I -ttidv. | Surg Res 
9(l):31-4 

28. HuittinenVM and SlatisP (1973) Postmortem angiography 
and dissection of the hypogastric artery in pelvic fractures. 
Surgery 73(3):454-62 

29.Kadish L], Stein )M et al. (1973) Angiographic diagnosis 
a ltd treatment of bleed mg due to pelvic I rati ma. J Trauma 
13(12):1083-5 

30.Ben-Menachem ¥, Coldwell DM et al. (1991) Hemorrhage 
assooiaied with pelvic fractures: causes. a la gnosis, and emer- 
gent management. A] R Am I Roentgenol 1 57(5 ):1 005-1 4 

31. Miller PR, Moore PS et al. (2003) External fixation or arte- 
nogram in bleeding pelvic fracture: initial tiierapv guided 
by markers of arterial hemorrhage. 1 Trauma 54(3 ;:437— ij 

32.Buhren V and Trentz O (1989) [Intraluminal balloon 
occlusion of the aorta in traumatic massive hemorrhage], 
Unfallchirurg 92(7):309-13 

33. Mucha P Jr and Farnell MB (1984) Analysis of pelvic frac- 
ture management. J Trauma 24(5):379-86 

34.Agolini SF, Shah K et al. (1997) Arterial embolization is a 
rapid and effecl.ve technique lor controlling pelvic fracture 
hemorrhage. ) Trauma 43(3):395-9 

35.Gui!lamondegui OD, Pryor ]P et al (2002) Pelvic radiog- 
raphv in blunt trauma resuscital ion: a diminishing role. I 
Trauma 53(6):1043-7 

36. Velmahos GC, Chahwan S et al. (2000) Angiographic embo- 
lization for intraperitoneal and retroperitoneal imuries. 
World J Surg24(5):539-45 

37. Velmahos GC, Toutouzas KG et al. (2002) A prospective 
study on the safety -\v\\. efficacy -:A angiographic emboliza- 
tion for pelvic and visceral imuries. I Trauma 53(2):303-8; 
discussion 308 

38. NiwaT.Takebayasln Set al. ( a000) The value of plain radio- 
graphs in the prediction of outcome in pelvic fractures 
treated with embolisation therapy. Br J Radiol 73(873):945- 
50 

39.Heetveld MJ, Harris I et al (2004) Guidelines for the man- 
agement of haemodvnamically unstable pelvic fracture 
patients. ANZ J Surg 74(7):520-9 

40. Shanmuganathan K, Minis SE et al. (1993) Value of con- 
trast-enhanced OT in detecting active hemorrhage in 
palients with blunt abdominal or pelvic trauma. AJR Am I 
Roentgenol 161(1 ):65-9 

41. Pryor JP and Reilly PM (2004) Initial care of the patient 
with blunt polytrauma. Clin Orthop(422):30-6 

42.Balogh Z, Voros E et al. (2003) Stent graft treatment of an 
external iliac artery injury associated with pelvic fracture. 
A case report. J Bone Joint Surg Am 85-A(5):919-22 

43.Pereira S], O'Brien PD et al. (2000) Dynamic helical com- 
puted tomography scan accurately cetects hemorrhage in 
patients with pelvic fracture. Surgery 128(4):678-85 

44.Gansslen A, Giannoudis P et al. (2003) Hemorrhage in 



L Wong and A. C. Roberts 



oelvic fracr.ire: who jiecJ ■= angiography? Chit Opin ■: II j - j : 
Care9(6):515-23 

45. Ravitch MM i I 9'j4) Hypogastric Artery Ligation in Acute 
Pelvic Trauma. Surgery 56:601-2 

46. Seavers R, Lynch I et al. (1964) Hypogastric Artery Ligation 
for Uncontrollable Hemorrhage :n Acute Pelvic Trauma. 
Surgery 55:516-9 

47.Hauser CW and Perry IF Jr. (1965) Control of Massive 
Hemorrhage from Pelvic Fraclures by Hypogastric Artery 
Ligation. Surg Gynecol Obstet 121:313-5 

48.Saueracker A], McCroskey BL et al. (1987) Intraoperative 
nypogastric arierv emoc-jzaiion for life-ihreaiemiig oelvic 
i'.emonhage: a mv.immary report, i Trail ma 27: '. 3 :■;] 1 17- 
9 

50. Kerr A (2002 ) Pelvic and Obstetric Hemorrhage. Vascu- 
lar and Interventional Radio. ogy: Pr.nciples and Practice. 
Curtis JES, Bakal VV, Cynamon I. ano Sprayregen S. New 
York, NY, Thieme. 1:297-313 

49. Velmahos GC.Chahwan S et al. (2000) Angiographic embo- 
lization of bilateral internal iliac arteries to control life- 
threatening hemorrhage after bltml trauma lo the pelvis. 
Am Surg66(9):S58-62 

51.SatoMFH,HagiwaraAetal. (1991) Traumatic 1 

of the spleen diagnosed by COL PSA. lournal of Japa 
Association of Acute Medicine 2:728-732 



52. Hashimoto SHK, Sato M (1997) C02 as an intra -arterial 
digital subtraction angiography agent in -he management 
of trauma. Seminars in Interventional Radiology 14:162- 
173 

53. Hawkins IF )r„ Caridi JG.Weichmann BN, Kerns SR (1997) 
Carbon dioxide, o.gita. subtraction angiography m trauma 
patients. Seminars in Interventional Radiology 14:17:"- 
180 

54. Donde linger RF, Trotteur G et al. (2002) Traumatic inju- 
ries: radiological hemostaiic intervention ai admission. Eur 
Radiol 12(5):979-93 

55. Hare WS and Holland CJ (1983) Paresis following internal 
iliac artery emoo.izaiioii. kadiology 146(1):47-51 

56. Sternbergh WC, 3rd, Conners MS, 3rd et al. (2003) Acute 
bilateral iliac artery occlusion secondary to blunt trauma: 
successful endovascular treatment. ) Vase Surg 38(3):5B9- 



57. Perez JV, Hughes TM et al. i ISflSj Angiographic embolisa- 
tion in pelvic fracture. Injury 29(3):187-91 

58.Uflakcer R (2002) 8. Embolization in Trauma. Visceral and 
Nonvascular Percutaneous Therapy: A Teaching File, Lip- 
pi ncott Williams & Wilkins. 2:134-141 

59. Ramirez Jl.Velmahos GC et al. (2004) Male sexual function 
after bilateral internal iliac artery embolization for pelvic 
fracture. J Trauma 56(4):734-9; discussion 739-41 



6 Postcatheterization Femoral Artery Injuries 



Geert Maleux, Sam Heye, and Maria Thijs 



Introduction 69 

Incidence of Postcatheterization Vascular 

Injuries 59 

Pseudoaneurysm 69 

■Clinical Features 69 

Radiological Diagnosis 70 

Treatment 70 

Surgery 70 

Lllrc lhslti.1 nd -Guided ■'.■■ impression Repair 70 
■ Traiisciuhetet Endovascukir Techniques 71 
- Uittasouiid-g'.uced Thrombin Injection 71 

Arteriovenous Fistula 74 

Prevalence and Natural History 74 



lagnos: 



75 



Treatment 75 
Thromboembolic Lesions 
References 76 



Incidence of Postcatheterization 
Vascular Injuries 

Among these iatrogenic femoral arterial injuries, 
the formation of a pseudoaneurysm is the most 
common entity. The reported incidence of iatro- 
genic pseudoaneurysms ranges from 2% to 8% after 
coronary angioplasty and stent placement and from 
0.2% to 0.5% after diagnostic angiography [1]. These 
clear differences in complication rates are basically 
due to the use of larger sheaths and catheters and 
due to the aggressive postprocedure! anticoagula- 
tion therapy routinely used in interventional car- 
diological units [2]. Arteriovenous fistulas are less 
common and occur in about 1% of all percutaneous 
coronary procedures. Uncontrollable groin hemor- 
rhage, in situ arterial thrombosis, and peripheral 
embolization are rare entities; their incidence is less 
than 1%. 



Introduction 



The number of percutaneous femoral arterial cath- 
eterizations has increased exponentially in recent 
years with several million procedures performed 
worldwide annually. A direct consequence of that 
explosion in number of percutaneous diagnostic 
and interventional catheterizations is the increasing 
number of vascular complications due to the per- 
cutaneous creation of that vascular access mainly 
using the femoral artery. Potential complications are 
pseudoaneurysm, arteriovenous fistula, uncontrol- 
lable groin and/or retroperitoneal bleeding, in situ 
arterial thrombosis, and peripheral embolization. 
In order to deal with these complications, there is an 
increasing need for quick and optimal diagnosis and 
for efficient and, by preference, minimally it 
treatment. 



G. Maleux, MD; S. Heve; MD; M. Thijs; MD 
I'epjrtiiien: <■■:' Radiology. "Jniversiiy !-!ospil,il: 
Herestraat 49, 3000 Leuven, Belgium 



Pseudoaneurysm 

6.3.1 

Clinical Features 

Among iatrogenic femoral arterial injuries, the for- 
mation of a pseudoaneurysm is the most common 
entity. Clinicalsymptomsare pa in a nd swelling at the 
site of a recent arterial puncture, and physical exam- 
ination can reveal a palpable mass in case of a large 
pseudoaneurysm. In a study by Toursarkissian 
et al., monitoring patients with a pseudoaneurysm 
of less than 3 cm in diameter, spontaneous closure 
was noted in 86% of cases with a mean of 23 days 
[3]. Adversely Kent et al. found spontaneous clo- 
sure unusual in pseudoaneurysms larger than 1.8 
cm in diameter [4]. These contradictory findings 
can probably be explained by the anticoagulation 
status of the patient: spontaneous thrombosis of a 



pseudoaneurysm is probably unlikely in an antico- 
agulated patient. As the vast majority of patients 
presenting with a postcatheterization pseudoaneu- 
rysm in our institution is anticoagulated, we rou- 
tinely treat every pseudoaneurysm with a diameter 
of more than 1 cm. 



6.3.2 

Radiological Diagnosis 

Duplex ultrasound is a simple, cheap, and effective 
tool to correctly diagnose a pseudoaneurysm. Real- 
time ultrasound imaging shows an echo-poor soft 
tissue mass anterior to the femoral artery and distal 
to the puncture site. The surrounding fatty tissues 
canbeechogenic due to the hemorrhagic infiltration. 
Doppler evaluation shows the classic triad of swirl- 
ing color flow in a mass separate from the underly- 
ing artery, color flow signal in a track leading from 
the artery to the mass (pseudoaneurysmal neck), 
and a to-and-tro Peppier 1 waveform in the pseudoa- 
neurysmal neck [5] (Fig. 6.1). Additionally, duplex 
ultrasound is also the imaging tool of preference to 
guide treatment like compression repair or throm- 
bin injection, and it can also be used to perform 
follow-up studies after treatment. Of course, MR and 
CT imaging are also valuable tools, but these tech- 
niques are expensive and need additional contrast 
medium administration. Catheter angiography can 
also diagnose a pseudoaneurysm, but because of the 
invasiveness of the procedure, this technique is no 
longer considered a valuable option. 




Fig.6.1. Color Doppler ■.dtrasouid o: the groin shows a pseu- 
doaneurysm (iPsfiTfsfc) with Li diameter of 1 cm and color flow 
centrally in the pseud o.areuiysmjl carky. ; :uplex scanning of 
the pseudoaneurysnul nee It demonstrates a typical to- and - 
fro signal 



6.3.3 

Treatment 



6.3.3.1 
Surgery 



Surgery has been the classical treatment of iatro- 
genic groin pseudoaneurysms for many years but 
since the publication of new, less invasive tech- 
niques, the number of surgical corrections has 
diminished significantly in most institutions. 
Briefly, the surgical technique consists of opening 
the groin, dissection of the pseudoaneurysm and 
injured vessel above and below the puncture point, 
evacuating the surroundim; hematoma, ami sutur- 
ing the bleeding point with or without placement 
of any absorbable synthetic graft material over the 
bleeding point. Despite the significant decrease in 
number of surgically repaired pseudoaneurysms, 
there are still some strict indications for surgery 
(and these surgical indications are contraindica- 
tions for percutaneous repair}: 
a)rapidly expanding pseudoaneurysm due to con- 
tinuous bleeding, 
b)infected pseudoaneurysm, 

c) symptoms of compression of the pseudoaneurysm 
on surrounding tissues like the femoral artery 
(distal ischemia), femoral nerve (neuropathy), 
overlying skin (skin lesions), 

d) pseudoaneurysm not responding to percutane- 
ous treatment. 

The results ot surgical repair are nearly 100%, but 
this treatment is not free of morbid ity or even mortal- 
ity, in most cases due to the significant cardiac comor- 
bidity of the affected patients. In a cohort study of 
55 patients presenting with arterial injuries produced 
by percutaneous femoral procedures, Franco et al. 
reported nine postoperative wound complications, 
five myocardial infarctions, and two deaths [6]. These 
numbers result in a postoperative morbidity rate of 
25% and a postoperative mortality of 3.5%. 



6.3.3.2 

Ultrasound-Guided Compressio 






technique, first described by Fellmeth et al., 
placing an ultrasound probe directly 
over the neck of the pseudoaneurysm followed by 
downward pressure of the probe, which will result 
in occlusion of the neck of the pseudoaneurysm [7]. 
Duplex examination will dei 



Postcitbctenzaiion Femoral Artery Injui 



flow into the pseudoaneurysmal lumen. The pres- 
sure must be continued for at least 10 mimites and 
then controlled by duplex ultrasound. If there is still 
a residual flow into the pseudoaneurysm, continued 
pressure for 20 minutes is mandatory. This tech- 
nique, which was very popular in the last decade, 
has some important drawbacks. It is a time-con- 
suming and painful technique, mostly requiring 
oral or intravenous analgesics to avoid excessive 
patient discomfort. Additionally, the procedure 
can be contra indicated, not only when one of the 
above-mentioned indications for surgery is present, 
but also when there is an anatomy unsuitable for 
compression repair: when the neck, which must be 
compressed, is located above or near the inguinal 
ligament, no underlying tough structure is present 
that would enable occlusion of the neck when ante- 
rior compression is performed. Other disadvantages 
of ultrasound-guided compression repair are the 
limited success rate in anticoagulated patients and 
in patients presenting with large pseudoaneurysms 



6.3.3.3 

Trans catheter Endovascular Techniques 

Several transcatheterendovasculartechniques have 
been described to treat iatrogenic groin pseudoan- 
eurysms, all of them in the form of case reports and 
some small series. Basically, two main techniques 
should be mentioned, but they have only histori- 
cal value: coil embolization and placement of a 
stent-graft [10-12] across the pseudoaneurysmal 
is that these techniques have 



;ck. The i 
been omitted are the cost of the de 
sion of later femoral artery puncture in the pres- 
ence of an overlying stent-graft, potential metallic 
s tent-fractures, and the disappointing long-term 
results of primary and secondary patency rates of 
femoral artery stent-grafts [13,14]. The long-term 
outcome of subcutaneously placed vascular coils 



6.3.3.4 

Ultrasound -guided Thrc 



6.3.3.4.1 



Cope and Zeit first described the potential interest 
of thrombin as an effective embolic agent in 1986 



[15]. They reported the successful direct needle- 
injection of thrombin to thrombose peripheral pseu- 
doaneurysms such as common iliac, peroneal, and 
hepatic pseudoaneurysms. Despite this interesting 
report, it was not until a decade later that the first 
report of ultrasound-guided direct thrombin- injec- 
tion to close iatrogenic groin pseudoaneurysms was 
published, by Liau et al. [16]. 

6.3.3.4.2 

Biochemical Working Mechanism 

Thrombin is an active enzyme in the blood-clotting 
cascade. It is formed from prothrombin (factor II 
clotting cascade) and it converts inactive fibrinogen 
into fibrin, which actively participates in the for- 
mation of thrombus. When injecting thrombin into 
the pseudoaneurysmal lumen, thrombus formation 
will be clearly accelerated as the blood flow in the 
pseudoaneurysm is turbulent or even nearly static. 
These phenomena will lead to a high concentration 
of thrombin in the pseudoaneurysm over a period 
of time, long enough to activate the clotting cascade 
into a definitive direction of clot formation. This 
activation of the clotting cascade due to the injection 
of thrombin will not be restrained or stopped when 
the patient is antuoayiilaied by heparin or warfarin- 
derivatives or when the patient is anti-aggregated. 
However, Kruger etal. demonstrated the increase of 
thrombin-antithrombinlll complexes in the periph- 
eral circulation 2, 5, and 10 minutes after thrombin 
injection in the pseudoaneurysm, indicating that 
some amount of thrombin passed into the circula- 
tion [17]. No transcatheter occlusion of the neck of 
the pseudoaneurysm was performed during injec- 
tion. Possible pathways of this passage are a direct 
flow of thrombin from the lumen of the pseudoa- 
neurysm into the feeding artery and here binding 
to antithrombin III; another possible mechanism is 
the formation of thrombin-antithrombin III com- 
plexes in the pseudoaneurysm and then passage of 
the whole complex into the feeding artery. A third 
pathway is that thrombin is partially absorbed from 
the surface of the pseudoaneurysmal cavity into the 
venous drainage. This increase in thrombin-anti- 
thrombinlll complexes does not lead to a higher risk 
of peripheral clot formation, neither in the arterial 
nor in the venous system. Today, thrombin is avail- 
able in the form of human thrombin and as bovine 
thrombin. Due to production costs, human throm- 
bin is more expensive than bovine, but the latter is a 
non-human substance which potentially may induce 
allergic or even anaphylactic reactions [18]. 



6.3.3.4.3 

Technique of Percutaneous Embolization 

Thrombin injection can be performed in an inter- 
ventional suite or even in an ultrasound room, but 
precautions must be taken in order to avoid poten- 
tial infection during the procedure. Therefore the 
patient's affected groin must be cleaned and disin- 
fected with povidone-iodine and covered with a ster- 
ile drape. Before starting the procedure the distal 
pulses of the affected limb are examined. A steril- 
ized 7.5- or a curved 3.5-MHz array transducer is 
used to guide the whole procedure. A 3.5-MHz array 
probe can be helpful when treating an obese patient 
or when the pseudoaneurysm is surrounded by mas- 
sive hematoma or by massively infiltrated subcuta- 
neous fatty tissues. Although most interventional 
radiologists inject some local anesthetics before 
introducing the puncture needle, the embolization 
procedure can also be done without any anesthetics 
and without any discomfort for the patient [19]. A 
21-gauge puncture needle is clearly large enough to 
inject the thrombin, avoiding the use of larger (20- 
or 19-gauge) needles. Under ultrasound guidance 



using a freehand technique, the puncture needle 
(e.g., spinal needle) is placed in the middle of the 
pseudoaneurysmal lumen (Fig. 6.2a,b). Injection of 
thrombin is done safely when the ultrasound probe 
is directed longitudinally to the femoral arteries for 
having a correct view on the pseudoaneurysmal neck 
and lumen (Table 6.1}. After switching the gray-scale 
imaging to color Doppler imaging, thrombin can 
be injected very slowly and under continuous color 
Doppler control. After each injection of 0.10 ml of 
thrombin, the residual flow in the pseudoaneurysm 
is evaluated and when no more Doppler signal can 
be depicted, the injection is stopped (Fig. 6.2c). In 
the case of a multilocular (or complex) pseudoan- 
eurysm, repositioning of the needle into another, 

Table 6.1. My lookbook i material : for ultra sound -guided 
thrombin injection 

- 7.5 or 3.5 MHz array ultrasound probe 

- Povidone-iodine 

- 21-gauge spin.:! neer.le (Tej n m .:■ iiiiiope. i.iruveii, Refill in) 

- Human thrombin ( Tissual '. -uo, Baxter H viand I in inline-, 
Vienna, Austria) 





Fig.6.2a-c. a Color Doppler ultrasound shows color flow cen- 
trally in the pseudouiieurysniai cavity i,j. <;ci isk), which has a 
diameter of 1.7 cm. b Under grav-scale iLirasound the spinal 
needle {iiiroi; ) is positioned in the middle of the pse-;doun- 
eurysmal cavity, c Color Popper ukrasound after thrombin 
embolization shows uosence of color signal in the thrombosed 
pseudoaneurysm l -.utcrisk i. 7 lie Ivn'.oral a: ;eries remain nor- 
mally patent 



Postc.atlietenzadon Femoral Artery Injui 




Fig.6.3a-c. a Colo:' Poppler dtiasoand clemonstraies color 

riow in 7 fir nios: o;o\:mjl i [.:■ the fejnor.i I artery! pse.alo;" 

rysiii.il cavity i,isrri:.N"A) ,10c par:ial opacification of the distal 

m 1 )- b After piaiclmiiig die most proximal cavity 

iijg it spinal needle Unom, c ti'.rombni injection resulted 

complete occlusion of boili proximal iriirr-.'/.d) and distal 



still reperfused lobe can be necessary to completely 
close the pseudoaneurysm, but we advise starting 
the embolization procedure by puncturing the most 
proximal (to the femoral artery) cavity. In the major- 
ity ''I case 1 ; embolization ol 1 lie niO'Sl proximal cavity 
will lead to concomitant occlusion of the distal cavi- 
ties, as these are in direct connection with the proxi- 
mal one (Fig. 6.3a-c). Some residual flow signals in 
the neck of the pseudoaneurysm, but without any 
signal in the lumen, can be considered a successful 
embolization. After the procedure, physical exami- 
nation of the distal pulses is indicated to exclude 
distal embolization. 

Variants of the above-described technique are 
also mentioned in the literature, but they are more 
complexand more expensive and no longer promoted 
today. A technique of ultrasound-guided thrombin 
injection after transcatheter balloon occlusion of 
the neck of the pseudoaneurysm was promoted in 
the United Kingdom a lew years ago [20, 21] but this 
technique needs additional, contralateral puncture, 
contrast medium administration, and manipula- 
tion under X-ray guidance. Transcatheter injection 



of thrombin ii 

is another comple? 



ity of the pseudoaneurysm 
;dure with the same draw- 
the balloon occlusion technique. Another 
variant technique is the ultrasound-guided injec- 
tion of saline beneath the neck of the pseudoaneu- 
rysm [22]. This particular technique, described by 
GEHLiNGetal., should result in rapid occlusion of the 
neck and subsequently will thrombose thepseudoa- 
neurysmal cavity [22], Unfortunately no large series 
or confirmations from other centers are reported. 

6.3.3.4.4 
Results 



Immediate success defined as complete thrombosis 
ot the pseiidoaneiirysmal cavity following throm- 
bin injection is very high, and most series report 
an immediate success rate in between 90 and 100% 
(Table 6.2). Failure of percutaneous embolization 
can occur in multiloculated pseud oaneurysms, 
when one or more lobes are not punctured; Sheim an 
ars of failure [23]. They 



et al. sought the 

concluded that the volume of the p 






Table 6.2. Overview of results of published series on percuta- 
neous thrombin injection to tre.it iatrogenic, postci'.ti'.eteiiz.i- 
tion pseuclo aneurysms 



Author 


Nci-i^er of 

p..1i:cl";iS 


Technica 


1 Compli- 


KANGetal. [35] 


21 


95% 


_ 


Paulson et al. [8] 


114 


96% 


4 


Taylor et al. [36] 


29 


93% 


- 


Pezzullo et al. [37] 


23 


95% 


1 


LaPerna et al. [38] 


70 


94% 


1 


Gale et al. [39] 


20 


100% 


- 


Bhophy ei aL [40] 


15 


100% 


- 


MALECxetal. [19] 


100 


98% 


- 


Khoury et al. [25] 


131 


96% 


3/131 


Friedman et al. [41] 


40 


97.5% 


1 


Owen et al. [20] 


25 


100% 


1 


Matson el al. [21] 


28 


85% 


1 



the neck diameter, and the thrombin dose were not 
predictive criteria. They only found that failure of 
treatment may indicate an occult vascular injury 
and that surgical repair rather than reinjection 
of thrombin should be considered. Percutaneous 
thrombin- injection, even without additional trans- 
catheter occlusion of the pseudoaneurysmal neck, is 
also very safe; the reported complication rates vary 
between 0% and 5% (Table 6.2). Complications are 
rare and can occur immediately or long after the 
embolization procedure [24]. Distal embolization is 
reported and can be due to passage of clot into the 
arterial circulation, but most probably will occur 
due to needle misplacement in the femoral artery 
and subsequently direct thrombin injection into 
the femoral artery [25]. Another rare complication 
is allergic or even anaphylactic reaction, but only 
when bovine thrombin is used [18]. Mid- and long- 
term results of percutaneous thrombin injection are 
also very good. In a study by Maleux et al., 70% of 
previously occluded pseudoaneurysms disappeared 
completely whereas in 25% of cases a small, residual 
hematoma was tound; in 3.5°b a partial reperfusion 
of a previously thrombosed pseudoaneurysm was 
revealed by color-duplex ultrasound after a mean 
follow-up of 99 days [19] (Fig. 6.4). 

The simplicity and reproducibility of the proce- 
dure as well as the high efficacy and very low com- 
plication rate of percutaneous thrombin injection 
under ultrasound guidance have made it the treat- 
ment of choice for postcatheterization pseudoaneu- 
rysms in many institutions [19, 26]. 




Fig. 6.4. Ultrasound of the groin. : ;;': cays arte? thrombin iniec 
(ion reveals a small, resid 
(he femoral arteries 



6.4 

Arteriovenous Fistula 

6.4.1 

Prevalence and Natural History 

Iatrogenic, posk'.ithelenzation arteriovenous fistu- 
lae are by far less frequent than postcatheteriza- 
tion pseudoaneurysms. Kelm et al. and Perings 
et al. found an incidence of 1% in a prospective 
study including more than 10,000 patients who 
underwent cardiac catheterization [27,28]. This 
study also revealed five significant and independent 
risk factors for developing an iatrogenic arteriove- 
nous fistula: procedural administration of heparin 
> 12,500 IU, Coumadin therapy, puncture of the left 
groin, arterial hypertension, and female gender. 
Follow-up of these patients demonstrated that one- 
third of all arteriovenous fistulae closed spontane- 
ously within one year and the majority even within 
the first four months. Additionally, the authors 
found that the majority of patients who suffer from 
an iatrogenic arteriovenous fistula do not develop 
clinical signs of hemodynamic, significance during 
follow-up, and subsequently in most ca 
treatment is not needed. 



Postcatheterizador! Femoral Artery Injui 



6.4.2 
Diagnosis 

Auscultation of the groin classically reveals a (new) 
continuous bruit after sheath removal, and in most 
cases a concomitant hematoma and/or pseudoaneu- 
rysm can be found. The clinical diagnosis must be 
confirmed by duplex ultrasonography, which will 
show a triad of typical signs: (1) a colorful "speck- 
led" mass at the level of the puncture site, (2) an 
increased venous flow with a lack of respiratory 
variation and a pulsatile arterial component in the 
affected vein, and (3) decreased arterial flow distal 
to the suspected fistula. As for pseudoaneurysms, an 
arteriovenous fistula can also be detected by more 
sophisticated imagine tools like MR-,CT-and cath- 
eter angiography, but the standard imaging tool is 
still duplex- ultra sound. 



6.4.3 

Treatment 

According to the study results of Kelm et al., in 
the majority of patients suffering from an iatro- 
genic arteriovenous fistula, no invasive treatment 
is needed or even indicated [27]. In case of clinical 
symptoms due to the fistula, surgical repair, covered 



stents, or compression repai 
options [11, 12, 29]. The last 



: the therapeutic 
has a rather low 
rate. Covered stents seem to be an attrac- 
tive and minimally invasive alternative (Fig. 6.5a,b), 
although many questions about long-term patency, 
stent and graft fatigue still exist. Open surgical 
repair is very effective and durable, but is not free 
of perioperative morbidity and mortality. 



Thromboembolic Lesions 



In situ thrombosis of the common femoral artery 
due to catheterization or manual compression after- 
wards are rare lesions but, if diagnosed late, can have 
a tragic outcome. Caution must be the rule when 
puncturing and certainly when compressing too 
much and too long a graft (e.g., in patients with an 
aortofemoral graft). When performing catheteriza- 
tion in children or young adults, persistent spasms of 
the femoral or brachial artery can induce an in situ 
thrombosis and potentially provoke distal embo- 
lization of a part of the clot. Small-sized sheaths 
and catheters as well as administration of vasoactive 
drugs can avoid this complication. In situ throm- 
bosis of the punctured artery is also described in 




Fig.li5a,b. : : e.e;l:v T LUi£ic:\a?:v of the right femoral bifu 

ally from die de^y femoral artery, b After " 

USA], the arteriovenous Sftuh is completely excluded 



patients treated with closure devices at the end of an 
endovascular procedure [30-32]. In most instances 
these complications must be corrected surgically, 
although interventional, endovascular management 
such as transcatheter thrombolysis or wire recanali- 
zation and balloon dilatation can be successful [33, 
34] (Fig. 6.6a,b). 




Fig.6.6a,b.A 9-year-olri girl presentee, with progressive claudi- 
o.ii :■■'•!. V- 'iilaine stage _o. for iiie pas: 3 ill- 'in lis. ■? Ji e previous!!" 
underwent multiple oirili.;." catheterizations from the right 
groin, a Selective angiography o:' lire right femoral bifurca- 
demonslrates louil occlusion of the proximal part of the 
gin common femoral artery (linv-ns). b After wire recana- 
zation and bullion angioplasty (Wanda bakoon 5x40 mm, 
Boston Scientiiic. \a;ick, MA, USA: an acceptable patency 
of the common femoral ai lery is obtained. Clinical follow-up 
si: owed absence of cl.uidicauon and nc'tmal dis:al pulses 



l.Lumsden AB, Miller [M. Kosmsfci AS, Allen RC, Dodson 
TF, Salam AA, Smith RB (1994] A prospective evaluation 

of surgicallv treaiec. groin complications following percu- 
taneous cardiac procedures. Am Surg 60:132-137 
2.Topol EJ (1998) Coronary-artery steiv.i-i: ga. gins;, gorging, 
and gouging. N Engl I Med 339:1702-1704 



3.Toursarkissian B, Allen BT, Petrinec D, Thompson RW, 
Rubin BG, Reilly JM, Anderson CB, Flye MW, Sicard GA 
(1997) Spontaneous closure of selected iatrogenic pseud oa- 
fistulae. J Vase Surg 25:803- 



Kent KC, McArdle CR, Kennedy B, Bairn DS, Anninos E, 
Silkman "I :. : a 93 :■ A prospective study of the clinic. I out- 
come of femora, pseudoanearysms ar.d arleriovenous fis- 
tulas ino .iced by arteriai puncture. I Vase Surg i 7:125-1 33 
Abu-¥ousef MM, Wiese JA, Shamma AR (1938) The "to- 
anci-fro" sign: duplex I'opp.er evidence or" femoral artery 
pseud on neurysm. AIR 150:632-634 

Franco CD, Goldsmith ], Veith FJ, Calligaro KD, Gupta SK, 
Wengerter KK il'.^oj Managemenl of arterial injuries 
produced by percutaneous femoral procedures. Surgerv 
113:419-425 

Fellmeth BD, Roberts AC, Bookstein J], Freischlag [A, For- 
sythe JR. Buckner NK, Hye RJ (1991) Postangiographic 
femoral artery urines: nonsurgical repair wi;h US- guide.:, 
compression. Radiology 1 75:671-675 
Paulson EK, Nelson RC, Mayes CE, Sheafor DH, Sketch 
MH, KliewerMA(2001) Sonographically guided thrombin 
injection of iatrogenic femoral pseucioaneurysms: further 
experience of a single institution. AJR 177:309-316 
r!.se:'.l:e:g L, Pa;; I son ki<. Kkewer MA, Hudson Mi- : . I 'ekong 
PM, Carroll BA ll 1 '"'"''; Sonograpi'.ica.ly guided compres- 
sion repair of useudoaiieiirvsms: rurli'.er experience from 
a single institution. AJR 173:1567-1573 
Lemaire IM, Pondehnger RF 11994) Percutaneous coil 
emboiizaiion of iatrogenic femoral arteriovenous fistula 
or pseudo-aneurysm.Eur ) Radiol 18:96-100 
Waigand I, Uhlich F, Gross CM, Thalhammer C, Dietz R 
(1999) Percutaneous treatment of pseudoaneurysms and 
arteriovenous fis;ulas after invasive vascular procedures. 
Catheter Cardiovasc Interv 47:157-166 
Thalhammer C, Kirchherr AS, Uhlich F, Waigand I, Gross 
CM (ji'ii'ifii p ,;■.. tea Jieterizai ion r~ ■= e .1 .1 ■. ■ .i 1 1 ^ 1 1 1 " >" s. n l ;; and arte- 
riovenous fistulas: repair with percutaneous imp.aiiluiioi; 
of endovascular covered stents. Radiology 214:127-131 
Kessel DO, Wijesinghe LD, Robertson I, Scott DJ, Raat H, 
S;ockx k.Neveis.teeii A i 1 999) ^nuovasciilur s ten I -grans lor 
superficial femoral artery disease: results of ! -year to How - 
up. I Vase Interv Radiol 10:289-296 

Saxon RR, Coffman IM, Gooding IM. Xaiuzzi E, Ponec 
DJ (2003) Long-term results of ePTFE stent-graft versus 
angi. :plasly i:: i:ie :;ni. ■:■..■■:■'.■ p. i;e. I artery: single cenler 
experience from a prospective, randomized trial. I Vase 
Interv Radiol 14:303-311 

Cope C, Zeit 3 i 1 980) Coagulation of aneurvsms by direct 
percutaneous thrombin iniection. AIR 147:383-387 
Liau CS, Ho FM, Chen MF.Lee ¥T (1997) Treatment of iat- 
rogenic femora, artery pseudoaneurysm with percutane- 
ous thrombin injection. ) Vase Surg 26:18-23 
ivriigei [•'... Za'lino.ger \i. Soli n gen I- -I lyCossmann A, Schuke 
O, Feldmann C, Strohe D, Lackner K (2003) Femoral pseu- 
doaneurysms: managemenl with percutaneous Thrombin 
injections - success raies and effens •:■':: systemic coagula- 
tion. Radiology 226:452-458 

Pope M, Johnston KW (2000) Anaphylaxis after thrombin 
injection of a femora! pseudoaneurysm: recommendations 
for prevention. ) Vase Surg 32:190-191 
Maleiix G, Hendrickx S, Vaninbroulot I, Laerobt H.Thijs M, 
Desmet W, Nevelsteen A, Marchal G (2003) Percutaneous 



Postcatheterizaiion Femoral Artery Injui 



iniection of human thrombin to treat iatrogenic femoral 
psetidoaneurvsms: shell- and midterm ultrasound follow- 
up. Eur Radiol 13:209-212 

20. Owen RJT, Haslam P], Elliott ST, Rose JDG, Loose HW 
!2j""i Percutaneous ub I at ion of peripheral pseudoaneu- 
rysms using thrombin: a simpie .ind effective solution. 
Cardiovasc Intervent Radiol 23:441-446 

21.Matson MB, Morgan RA, Belli AM (2001) Percutaneous 
treatment or psetidoaneurvsms using fibrin adhesive. Br ■ 
Radiol 74:690-694 

22.Gehling G, Ludwig J, Schmidt A, Daniel WG, Werner D 
;ii003; Peripheral vascular disease. Percutaneous occlu- 
sion of fern or;:! artery pseudoaneurysm by par; -aneurys- 
mal saline injection. Caihct Cardiovasc I nl erven I sSirOO- 
504 

23.Sheiman RG, Mastromatieo M i 2 3331 iatrogenic femoral 
pseudoaneurvsms that are unresponsive to percutaneous 
thrombin injection: potential causes. AfR 181:1301-1304 

24.Kurz DJ, |ungius K-P, Lilscher TF (2003) Delayed femoral 
vein thrombosis after ultrasound-guided thrombin iniec- 
tion of a ;"■ ■?■ s t c .; r i j e : e i" i ^ :i : i ■. ■ :i pseudoaneurysm. I Vase interv 
Radiol 14:1067-1070 

25. Khoury M, Rebecca A, Greene K, Rama K, Colaiuta E, Flynn 
L. Be is k [l : ' : 'l i I nip lex sea nning-gu iced thrombin ntiec- 
tion tor the treatment of iatrogenic pseudo.'.neurysms. 1 
Vase Surg 35:517-521 

26. Morgan R, Belli A-M (2003) Current treatment methods for 
postcatrieterizurion pseudoaneu rysms. I Vase Interv Radiol 
14:697-710 

27.Kelm M, Perings SM, Jax T, lauer T, Schoebel FC, Heint- 
zen MP, Perings C, Strauer BE tiaaii Incidence and dirti- 
ed outcome of iatrogenic femoral arteriovenous fistuias. 
Implicit! ions for risk stratification and trea intent. ] Am Coll 
Cardiol 40:291-297 

28. Perings SM, Kelm M, Jax T, Strauer BE (2003) A prospec- 
tive study en incidence and risk factors of arteriovenous 
fislulae following Iransfemoral cardiac catheterization. Int 
( Cardiol 88:223-228 

29.6nal B, Kosar S, Gumus T, Ilgit ET.Akpek S (2004) Post- 
catrietenzation :'e[T.-.ii";:l arteriovenous fislulas: endovascu- 
lar treatment wiih stenl-grafis.Cardievasc in let vein kad.ol 
27:453-458 

30. Brown DB, Crawford ST, Norton Pl.Hovsepian DM (2002) 



A ii a i '.'■■£ i':i t'-Jiic follow-up after st.ture-meciialed femorai 
artery closure. I Vase Interv Radiol 13:677-680 
31.Nehler MR, Lawrence WA, Whitehill TA, Charette SD, 
Jones DN, Krupski WC (2001 ) iatrogenic vascular injuries 
from percutaneous vascular suturing devices, i Vase Surg 
33:943-947 

32. Abando A, Hood D, Weaver F, Katz S (2004) The use of the 
angioseal device for femoral artery closure. 1 Vase Surg 
40:287-290 

33. Geschwind JF, Dagli MS, Lambert DL, Kobeiter H (2003) 
Thrombolytic iherapy in lite setting of a nerial line- induced 
ischemia. I Endovasc Ther 10:590-594 

34. Gemmete JJ, Dasika N, Forauer AR, Cho K, Williams DM 
(2003) Successful angioplasty of a superficial femoral 
artery stenosis causeci by a suture- mediated closure deuce. 
Cardiovasc intervent Radiol 26:410-412 

35.Kang SS, Labropoulos N, Mans our MA, Baker WH (1998) 
Percutaneous ulna sound guides litioiv.oin iiiiection: a new 
method for I tea ting p'.-uo.uheterizal.on tern or a I pseudoaia- 
eurysms. I Vase Surg 27:1032-1038 

36. Taylor BS, Rhee RY, Muluk S, Trachtenberg J, Walters D, 
Steed DL, Makaroun MS (1999) Thrombin injection versus 
compression of femora, artery pseudoaneurysms. i Vase 
Surg 30:1052-1059 

37.Pezzullo IA, Dupuy DE, Cronan JJ (2000) Percutaneous 
injection oi thromb.n for the treatment of pseueioaiieu- 
rysms after calheterization: an alternative lo sonograph.- 
cally guided compression. AJR 175:1035-1040 

38. La Perna L, Olin JW, Goines D, Childs MB, Ouriel K (2000) 
Ultrasound-guided ' : stion for the treatment 

of postcatheteriz. ' :■■■.■•: neurysms. Circulation 

102:2391-1295 

39.Gale SS, Scissons Rl- |ones : , Sallea Cunha SX (2001 ) Femo- 
ral pset.doaneurysrn lh:i:*-r>in;o.l.oit. Am I Surg 181:379- 
383 

40.Brophy DP, Sheiman «:, Amatulle P, Akbari CM (2000) 
Iatrogenic femoral |"-t..d.Mriciirwiis: thrombin injection 
after failed US-guiLtd com pro*; on. Radiology 214:278- 
282 

41. Friedman SG, Pellento J S, Scher L, Faust G, Burke B, Safa T 
[11)1)1) Ultrasound-guided ti'.romnin imection is the treat- 
ment of choice for femoral pseucioaneurysnis. Arch Sure 
137:462-464 



7 Iatrogenic Lesions 



Michael D. Darcy 



Introduction 79 
P hysi op at ho lo gy 79 
Clinical Considerations 81 
Anatomy 82 

Technique? and Equipment S3 
Access Li rid Delivery 83 
Embolic Agents 85 
Alternative Techniques 87 
Ha'l::in !'a— panade 87 
Uncovered Stents 87 
■ Stent Crafting HS 
Results 89 
Hepatic 89 
:« .-Ml 90 

MisceiUntiiu* injuries 91 
Complications 91 
r iitti : e Developing:'.: .-rid Research 
Conclusion 94 
References 94 



Fortunately, the interventionist often has the ability 
to manage these complications, and embolization is 
one of the primary techniques utilized. 

Throughout the 1970s numerous case reports 
appeared demonstrating the ability to successfully 
embolize iatrogenic bleeding in a variety of organs 
[1-4]. While these reports demonstrated the proof of 
concept, these embolizations were done with large 6 
to 7 Fr catheters and often the arteries were occluded 
at a fairly proximal level. With the development of 
smaller catheters and improved embolization mate- 
rials, it is now possible to advance super- selectively 
and occlude the artery right at the site of injury. 
Since modern embolization techniques allow effec- 
tive control of bleeding while posing less of a risk 
to the target organ, embolization has become the 
method of choice lor managing many forms of iat- 
rogenic hemorrhage. 



Introduction 



Physiopathology 



Since the beginning ol both su rgery and percutaneous 
interventions, vascular injury lias nl'.vays been one of 
the potential complications. Bleeding can complicate 
a wide range of procedures from simple biopsies or 
venous access cases up to more complex angioplasty, 
drainage procedures, or surgeries. This can be a pri- 
mary injury that occurs at the site of the pathology 
that is being treated, such as arterial rupture during 
percutaneous transluminal angioplasty (PTA). Alter- 
natively, vascular damage can be a secondary effect 
such as when a hepatic arterial branch is injured 
along a percutaneous tract to the bile ducts (Fig. 7.1). 



M. D. Darcy, MD 

Professor of Radiology and Surgery, Division of Diagnostic 
Rodiology, Chief, !nlerve:i:ii;:i.il K.idioiogv Sec-ion, Washing- 
ton University School or Medicine. Mol.inc^iodt Institute of 
Radiology, 510 S. Kingshighway Blvd., St. Louis, MO 63110, 
USA 



Iatrogenic vascular injuries that require embo- 
lization are most often arterial in origin. Venous 
injuries rarely cause clinicallj' significant bleeding 
since local hematoma caused by bleeding from the 
injury will often compress the low-pressure vein 
and tamponade the bleeding. An exception to this 
is when a large vein is disrupted along a drainage 
catheter tract. In this setting, bleeding may wick 
along the catheter or enter the catheter itself if the 
catheter side-holes are inappropriately positioned 
in the parenchyma. Aside from the low frequency 
of major venous bleeding, venous bleeding is also 
difficult to diagnose arteriographically, plus access 
to the vein for embolization may be limited. For 
these practical reasons, embolization techniques to 
correct iatrogenic hemorrhage have focused on arte- 
rial bleeding. 

Iatrogenic lesions tend to be simple traumatic dis- 
ruptions of the artery, so unlike true aneurysms, the 




Fig. 7.1. a Right hepatic arteriogram in ,1 p l:. L l e j j t will', severe bleeding after 

percutaneous biliary drainage. A pseuacane'ai ysm is seen along the tract 

extravasating ;'<ir 'cir! inlo the biliary ,-a:heter. b Spot image 

ihowing an angioplasty balloon being ;:sed v: ta:'.:pom;de the bleeding. 

:n advanced into the pseudoaneurysm. c 

ows microcoils in place and no further 

bleeding 



defect usually extends through all layers of the arterial 
wall. The size of the defect may vary with the mecha- 
nism of injury. An artery damaged by a needle pass 
during biopsy may have a very focal wall defect. On 
the other hand, an artery split during a PTA may have 
a long linear defect. The size of the defect may alter the 
ease with which embolization can control the bleed- 
ing. The size of the defect can alter the presentation. 
Injury to a small hepatic branch may present remote 
in time from the original procedure that caused it and 
may present only with annoying persistent bleeding 
into the biliary drainage catheter without any signs of 
hemodynamic instability. A large rupture of a main 
hepatic or renal artery however wilt usually lead to 
immediate pain, tachycardia, and hypotension. 

The signs and presentation of an arterial injury 
will also vary depending on the location of the injury 
with respect to the surrounding tissue. If the damaged 
artery is deep inside a solid organ like the liver, the 
bleeding may be relatively contained by the surround- 



ing tissue and a pseudoaneurysm may form without 
signs of major hemorrhage (tachycardia, hypotension) 
being present. Instead the presentation maybe more of 
a chronic low-grade bleeding into a drainage tube or 
adjacent structure (e.g. into a calyx causing hematuria 
or into a bile duct causing hemobilia). In some cases 
intrarenal or intrahepatic pseu do aneurysms may even 
be found incidentally on cross-sectional imaging done 
for other reasons. However, if the injured artery is only 
surrounded by loose areolar tissue or fat, the bleeding 
may not be constrained and considerable hemorrhage 
may ensue. Aside from surrounding native tissue, one 
must remember that the bleeding may be partially 
tamponaded by the presence of the tube that caused 
the injury in the first place. In fact some pseudoaneu- 
rysms or even gross hemorrhage will not be evident on 
the initial arteriogram with the drainage catheter in 
place and will only become evident once the arterio- 
gram is repeated after removing the drainage catheter 
ov«r ag mdewire(Fi 5 .7.2). 






The location of the iatrogenic injury (i.e. how cen- 
tral or peripheral it is) will have a significant effect 
on the choice of therapy. A central lesion may be 
less amenable to embolization for several reasons. 
Embolization of a main arterial trunk may threaten 
the viability of the organ supplied by the injured 
vessel. It also may be difficult to embolize a main 
arterial trunk without risking nontarget emboliza- 
tion. In these settings, alternative techniques like 
stent- grafting may be more useful. 

Fortunately, injuries in solid organs like kid- 
neys and liver often tend to be in more peripheral 
branches. These lesions are often inaccessible to the 
surgeon for direct repair and so surgical therapy 
involves very aggressive approaches such as main 
arterial ligation or partial resection of the organ. 
This provides a unique advantage for embolother- 
apy since a peripheral, focal source of bleeding can 
often be embolized while sacrificing only a small 
portion of the organ. Thus repair by embolization 
will preserve a much greater percentage of the organ 
than would be possible with surgical repair. 



Clinical Considerations 

Some bleeding is natural after placing a catheter 
through a very vascular organ such as a kidney or 
liver. Therefore one of the first tasks is to decide 
when to proceed to arteriography. In some cases, iat- 



rogenic hemorrhage is profound with rapid onset of 
tachycardia and hypotension. In this setting imme- 
diate arteriography is clearly warranted. The pres- 
ence of pulsatile blood flow out of the tract during 
a tube exchange is another indication that angiogra- 
phy is needed. The decision is less clear when there 
is just low-grade continued bleeding such as bloody 
output from a nephrostomy catheter that fails to 
clear after a few days. 

One should first insure that the patient does not 
have a coagulopathy or thrombocytopenia that could 
account for the continued bleed ing. Venous oozing that 
might normally be inconsequential can become quite 
troublesome- m coagulopathy patients'. The patient's 
medication list should also be checked for anticoagu- 
lant or antiplatelet medications that may have acci- 
dentally not been stopped before the procedure. The 
other reason to carefully assess the coagulation status 
is that embolization has been shown to be less effec- 
tive in coagulopathy patients [5, 6]. This is because 
coils and other embolic agents do not by themselves 
provide complete occlusion to flow but rather pro- 
vide a substrate for the formal ion ot thrombus which 
occludes the vessel. If coagulation tests are normal or 
corrected and bleeding persists a week or more or if 
continued bleeding causes a significant drop in the 
hematocrit, then angiography may be indicated. 

The hemodynamic status of the patient should be 
carefully assessed. If the patient is clearly actively 
bleeding, one should proceed to angiography as 
soon as possible even if the patient is hypotensive. 




Fig. 7.2. n Kit: h; h^^iiit :.: ;e: iograni .-tie: fc:aiUi:'.eous b:jLi:y orajiiagc- shows sr\is:r. whejv the biliary catheter c: 
nirery (iinviri, but no b^eding o: p^v.idosincm vs:::. b After rei'.icvaig the biliary o.ulielc] over ■,-, guidtwue. re-pear nr 
shows extravasation or conlrasr alo:ig the tract (arrow) 



Waiting for the patient to stabilize may waste pre- 
cious time, and in fact it may not be possible to 
stabilize the patient until the bleeding is stopped. 
Although a few steps are necessary to try to stabilize 
the patient, resuscitation efforts should be carried 
on concurrently with the angiographic efforts to 
stop the bleeding. The patient should have several 
large-bore venous lines for fluid resuscitation and 
blood transfusion. Blood should be typed, crossed, 
and readily available. Drugs and equipment needed 
to start a vasopressor infusion should be at hand. 

Even if the patient does not appear to be actively 
bleeding at the start of the arteriogram, one must 
assume that they may develop significant blood 
loss during the case. Wire manipulation or pres- 
sure injections in the injured artery may stimulate 
increased bleeding. Also if biliary or nephrostomy 
catheter removal is necessary to demonstrate the 
pseud oaneurysm, massive bleeding may suddenly 
occur upon removal of the catheter. Therefore some 
of the same precautionary steps (large IVs, type 
and cross, etc.) should be taken with the patients 
with more chronic low-grade bleeding. One should 
remember to check the baseline hematocrit, since a 
patient who has been chronically bleeding may be 
starting out with a low hematocrit and may not tol- 
erate even a modest amount of bleeding. 

If the bleeding is secondary to an i mi welling cathe- 
ter such as a nephrostomy' or 1 bi I iary drain, one should 
determine whether the catheter is still necessary. If 
so, one may need to place a new catheter via a differ- 
ent access. At a minimum, you should be prepared 
to temporarily remove the catheter over a guidewire 
during the diagnostic arteriogram. The catheter may 
tamponade the bleeding, and extravasation may only 
be seen with the catheter removed. In a series of 13 
patients with severe hem.hlia alter biliary drainage, 
live(3S°b] ofthe vascular injuries could only be identi- 
fied after removing the catheter over a wire [7]. Main- 
taining guidewire access is crucial to allow replace- 
ment of the catheter or a PTA balloon to tamponade 
the bleeding once the diagnosis has been made. 

Depending on the organ being embolized and 
the level of embolization, some tissue ischemia or 
even necrosis may occur. Prophylactic antibiotics 
maybe indicated to prevent bacterial seeding of the 
infarcted tissue from developing into an abscess. 
This maybe more of a concern when the embolized 
artery is in an infected organ such as a renal pseu- 
doaneurysm that occurs after a nephrostomy done 
for pyonephrosis. Certainly if the patient has known 
bacteremia, antibiotic coverage should be started 
prior to the embolization. 



Anatomy 

Since vessels can be injured anywhere in the body, it 
is not possible to discuss the specific anatomy of all 
arterial beds. But there are some general anatomic 
assessments common to all regions and questions 
that must be answered prior to treating an iatrogenic 
vascular injury. 

The first important question is whether or not 
the injured vessel can be sacrificed. The answer to 
this partially depends on whether you would expect 
the tissue d istal to the target artery to remain viable 
or become ischemic after embolization. If sufficient 
collaterals are available, the tissue supplied by the 
target vessel may not be affected. So for example, 
embolizing a gastric branch to stop post-gastros- 
tomy bleeding (Fig. 7.3) is unlikely to cause any 
ischemia due to the rich collateral supply around the 
stomach. Tissue distal to the injured vessel may also 
be safe from ischemia if there is an alternate blood 
supply. For example, portal venous flow into the 
liver allows safe embolization of even major trunks 
of the hepatic artery. 

If good collateral perfusion is unlikely, one then 
must consider whether one can afford to let the 
tissue become ischemic. As an example, it would 
be very reasonable to embolize a peripheral renal 
artery branch that was injured during a biopsy 
and sacrifice a small section of renal parenchyma, 
since it would have negligible effect on renal func- 
tion. However if the main renal artery was ruptured 
during PTA, you would not want to embolize this 
artery except in dire situations since it would sacri- 
fice the entire kidney. 

Thorough understanding of the anatomy, in par- 
ticular potential collateral pathways, is also critical 
whenplanning at what level to embolize the artery in 
order to insure an adequate therapeutic result. If the 
target vessel is an end-artery (such as a renal arte- 
rial branch) it probably suffices to deposit emboli 
proximal to the injured arterial segment. However, 
if there are well known collateral pathways beyond 
the target artery, then it is necessary to advance 
the catheter beyond the point of extravasation and 
occlude the artery both distal and proximal to the 
injury. Otherwise collateral flow could lead to per- 
sistent bleeding. 

Knowledge of collateral pathways is also critical 
to allow the interventionist to check the appropriate 
vessels after what appears to be a successful embo- 
lization. Thus, after a gastroduodenal emboliza- 
tion for post-biopsy pancreatic head bleeding, it is 




Fig. 7.3. a Selective gasuc-duodenal ;u i^: iogram in a oatient with, tippe: gasty 'imrslii'a! bleeding aftei placement of a sii 
lumen gastrojejunostomy (which has backed up into the stomach), b Magnified view belter showing the bleeding [arr 
coming from the g,t si :cie; tin ostomy access si:e in the mid -body of the stomach, c ! :, ost embolization study arteriogram shot 
microcoil in the branch that was bleeding and no further extiavasado:'.. d Left gastric arte: iogram =■ li ■ 'Wing good perfusio: 
the mid -bod v th-is preventing gas:t ic ischemia 



important to do both a celiac arteriogram to look for 
pancreatic collaterals from the splenic artery and a 
superior mesenteric arteriogram to look for inferior 
pancreatico-duodenal collaterals. 



Techniques and Equipment 

Again given that iatrogenic injur i 
anywhere in the body, a complete dis 
specific techniques and equipment for all areas is 
not possible, but there are some general principles 
{Table 7.1). 



7.5.1 

Access and Delivery 

An arterial access sheath is crucial to i 
access in case the embolization catheter bet 
occluded. The sheath will also facilitate catheter 
exchanges. Typically a standard short sheath will 
work. A longer curved sheath like the Balkan sheath 
(Cook Inc.; Bloomington, IN) or a guiding catheter 
may be useful to engage the main arterial trunk, 
especially if additional support will be needed to 
ease passage of the embolization catheter across a 
severely angled or tortuous artery. A guiding cath- 
eter may also be useful for embolizations done 
from an axillary approach so that multiple catheter 



\i.D. rurcy 



Table 7.1. Cookbook: Sample - 1 11L ■■ ■ I . a;"^ t ; .:; n equipment list 
Embolization of large accessible artery 
5-Fr Cobra catheter 

0.035-11]. Rentsoii o: C-lidewiie to i]e.p eng.sge artery 
'Siaiiturco coils (size to maic;. resse.l 01' 'Selfcam olec.gei-; 
LLT wire to push coils 
Alternative cc-.tlieleis ."epenJing on vessel shape 

- 5-Fr Sos of Si in in on caiheie: to engage artery 

- Rosen gindewire .:i" siirf-sha:; giidem:e for exchange 

- MPA or Cobf.i catheter to advance :v.oie pe:ip:.efallv 

2. Embolization of small peripheral artery 

■ 5-Fr Cobra. Sos, o: Simmons c.flielef to engage 
arterial trunk 

• MassTransit of Kenegade micro catbe:er to advance 
peripherally 

• Transend guidevci:.: to guide- micro catheter 

■ 0.018-in.microcoils or PVA 

• 0.025-in. glidewire to push microcoils through 
microcatheter 



3. Direct puncture for visceral a 



urysni 



cl-g Micro-puncture needles 

0.018-in microcoils or thrombin (1,000-2,000 units) 

Alternatives 

- 22-gChiba needle and I [iron - , bin when smaller 
access desired 

- 18-g Trocai needle ic: beaer con;rol of needle and 
more choices of emnojc agent to use 

(can use larger 0.038-in. coils) 



exchanges are not done across the origin of the ver- 
tebral artery. 

The size and type catheter used for embolization 
must be tailored to the type of lesion to be emboli zed, 
the anatomy, and how peripheral the lesion is. If the 
size of the defect is huge las with wideneckpseudoan- 
eurysms) or if there is potential for the embolic mate- 
rial to migrate through the arterial defect (as with an 
arteriovenous fistula), then larger coils or large Gel- 
foam pledgets maybe needed. In that case a 5 Fr cath- 
eter is necessary to deliver the larger emboli. The 5 Fr 
catheter chosen will depend on the shape of the arter- 
ies. A Sos Omni (A ngio dynamics; Queensbury, NY) is 
a good initial catheter for selecting a variety of arterial 
trunks including renal, celiac, and mesenteric trunks. 
However the recurved shape may not allow the cathe- 
ter to be advanced more peripherally unless a very stiff 
wire is advanced out into the target artery. A Cobra- 
shaped catheter is sometimes less stable when engag- 
ing a main visceral trunk, but it tends to be easier to 
advance more peripherally. Alternatively a recurved 
catheter such as a Sos or a Simmons 2 can be used 
to securely engage the trunk and then pass a stiffer 
wire out into the periphery to allow an exchange for 



a st raighter catheter (e.g. an MPA catheter; Cook Inc.) 
that will track better peripherally. Although a hydro- 
philic coated catheter may be more easily passed out 
to the peripheral aspects of an artery, we do not tend 
to use hydrophilic catheters because in our experience 
coils tend to get stuck in hydrophilic catheters. 

For more focal defects in smaller arteries, a 3 Fr 
microcatheter such as the Mass Transit (Cordis; 
Miami, FL) or Renegade (Boston Scientific) has sev- 
eral advantages. Because they are small and very 
flexible, they can be advanced very peripherally into 
branches that would be too small for a larger 5 Fr 
catheter. More peripheral embolization minimizes 
the amount of tissue that must be sacrificed (Fig. 7.4). 
Microcatheters do have several disadvantages. They 
are more cumbersome to use, require a higher level 
of technical skill, and only allow delivery of smaller 
emboli which may not occlude the target artery as 
effectively. 

As an alternative to catheterization, an iatrogenic 
pseud oaneurysm can be directly punctured percu- 
taneously with a needle for delivery of the embolic 
agent. This is most commonly done for post-angi- 
ography femoral artery pseudoaneurysms, which 
are discussed more fully in another chapter. How- 
ever, direct puncture can also be used to access 
deep abdominal lesions that cannot be reached 
with an intra-arterial catheter due to tortuosity, 
small vessel size, or obstruction by emboli from a 
prior attempted embolization. This technique has 
been applied to various hepatic and splanchnic 
pseudoaneurysms using either thrombin or coils 
as the embolic agents [8-12]. The puncture is done 
with long 22-or 21-gauge needles when planning to 
use thrombin alone as the embolic agent. Although 
microcoils can be introduced through a 21-g needle, 
using an 18-g needle for access gives you the option 
of introducing larger 0.038-in. fibered coils. For 
deep intra-abdominal pseudoaneurysms, needle 
placement can be guided by ultrasound, CT, or fluo- 
roscopic visualization ol injected arterial contrast. 
Once the needle has been inserted, confirmation 
of proper position in the pseudoaneurysm is con- 
firmed by aspiration of blood and injecting contrast 
directly through the needle. Thrombin or coils can 
then be introduced directly through the needle into 
the pseudoaneurysm. 

In solid organs such as the liver, major iatro- 
genic bleeding is sometimes due to communication 
between the tract and a major venous structure. 
Arteriography will usually not reveal any abnormal- 
ity nor provide an access for therapy. In most cases of 
venous bleeding, simply leaving the drainage cath- 



Iatrogenic Lesions 




Fig. 7.4. a Right renal arteriogram showing a pse.;doane : arysm in a patient 
who developed perinephiic hemorrhage after :", biopsy h A microcaiheter 
has been advanced peripherally into [he specific branch [iUiou-'i leading to 
the pseud oaneurysm. c Mid-.ir:eri;i' phase of die post embolization arte- 
riogram shows good o reservation of the n 



eter in place or sometimes upsizing the catheter will 
effectively tamponade the smaller venous bleeds. If 
tamponade fails to control the bleeding because of 
the large size of the vein that was transgressed, embo- 
lization can be done via the tract itself (Fig. 7.5). The 
drainage catheter is first replaced with a sheath with 
a side arm adaptor or a Lieberman catheter (Cook 
Inc.) with an attached hemostatic valve with a side- 
arm adaptor (Merit Medical; South Jordan, UT). The 
sheath or catheter is pulled into the tract and con- 
trast is injected into the tract while acquiring digi- 
tal subtracted images. Once the venous connection 
is identified, coils are placed in the tract across the 
point where the tract and vein intersect. After tract 
embolization, it may not be possible (or desirable} 
to re-advance a drainage catheter down the tract. 
Therefore a new access for draining the biliary tree 
should be secured before starting the tract embo- 
lization. Although rarely needed in our practice, 
tract embolization has been effective at stopping the 
bleeding when this technique was used. 



7.5.2 

Embolic Agents 

Coils are possibly the most commonly used devices 
for embolization of iatrogenic bleeding because they 
are readily visible during fluoroscopy, they can be 
deposited very precisely, and they generally provide 
effective occlusion. Larger coils (0.035-0.038 in.) 
must be passed through 5 Fr catheters, but in our 
experience these coils occlude arteries more effec- 
tively than the smaller 0.025-in. coils or 0.018-in. 
microcoils. Thus 0.038-in. coils are preferred when- 
ever vessel size permits passage of a 5-Fr catheter. 
However, even 0.038-in. coils may not effectively 
occlude blood flow if the patient has a coagulopathy. 
One of the main functions of a coil is to provide a 
nidus for thrombus formation, if coagulopathy pre- 
vents thrombus formation, then blood may continue 
to flow around coils, especially if they are not tightly 
packed. Correction of the coagulopathy should be a 
priority but may not be possible. Alternatively Gel- 




Fig. 73. a Injection of contrast along ;i bikary tract showing major 
communication to hepatic veins iiii ivni. b The tract in the region 
of the venous com mtiiiicj lion was eniL"' elided with coils and a na so- 
biliary tube wns placed lb:' ci'ainage 



foam can be injected on top of the framework of 
coils; this will provide more complete obstruction 
to blood flow. 

Gelfoam is another commonly used agent but 
unlike coils it is injected and relies on flow direc- 
tion. This is useful when the catheter cannot be 
advanced close enough to the bleeding site to allow 
placement of a coil. It is a versatile embolic agent. 
It comes in sheets and is cut into appropriate-size 
pieces for each case, which allows the emboli to be 
easily tailored to the situation. For large arterial 
defects or bleeding from large vessels, Gelfoam can 
be cut into large torpedoes. For smaller vessels it 
can be cut into smaller cubes. To embolize multiple 
branches at once it can also be made into a slurry by 
rapidly injecting it back and forth through a three- 
way stopcock. Because it is a flow-directed embolic 
agent, careful fluoroscopic monitoring of injections 
is critical to avoid reflux into nontarget vessels. For 
this reason, Gelfoam must be suspended in contrast. 
Since Gelfoam dissolves after a couple of weeks, it 
has the theoretic benefit of allowing vessel recana- 
lization, although in some organs (such as kidney} 
the distal tissue has already infarcted long before 
the vessel recanalizes. 

Polyvinyl alcohol (PVA) is a semipermanent 
flow-directed injectable particulate agent that can 
also be useful for treating iatrogenic hemorrhage. 



Its main benefit over Gelloam 
the particles, making it e 



> the s 



1.1 Hers 



eof 



ise them through 
a microcatheter. Because of their small size they are 
mostly useful for bleeding from small arteries. With 
large arterial defects or arteriovenous fistulas, the 
PVA can just flow out through the defect or into the 
venous circulation. For iatrogenic bleeding, larger 
PVA particles (>500 microns} are typically used 
since smaller particles are more likely to travel into 
very peripheral arterioles and are more likely to 
cause tissue ischemia. 

PVA has several disadvantages. Like Gelfoam, 
the particles themselves are not fluoroscopically 
visible, and only the contrast they are suspended in 
allows you to monitor the injection. This is an indi- 
rect method of monitoring the embolization since 
the number or density of particles is not always 
uniform in any given injection. Care must be 
taken to avoid reflux and nontarget embolization. 
The particles also can occlude catheters requiring 
forceful and less controlled injections to clear the 
catheter, or the occlusion may even be firm enough 
to require removal of the catheter. Avoiding exces- 
sive particle density in the embolic mixture and 
frequent flushing with saline can help prevent this 
problem. PVA particles may also clump together, 
leading to premature occlusion of a vessel proxi- 
mal to the injury. Newer formulations of PVA engi- 



neered into more spherical shapes such as Contour 
SE (Boston Scientific} may reduce clumping and 
catheter occlusion. 

Thrombin has been used for embolization with 
increased frequency, especially for management 
of post-catheter ization pseudoanemysms [13,14]. 
Occluding pseud oaneurysms with thrombin was 
first described in 1986 by Cope and Zeit [15], and 
even in that initial report the technique was also 
used for an intrahepatic lesion, not just femoral 
lesions. Being a liquid agent it is readily delivered 
through even small-caliber catheters or needles. 
Usually only 1-2 ml of thrombin (1000 units/ml} is 
needed to thrombose a pseudoaneurysm. It must be 
injected carefully, since over- inject ion of the pseu- 
doaneurysm can lead to nontarget downstream 
thrombosis. Most often the effect of thrombin injec- 
tion is monitored in real time using color-flow ultra- 
sound. In addition to its use as the primary embolic 
agent, thrombin can be used to augment the efficacy 
of coils. In situations where it is difficult to control 
hemorrhage due to a high rate of blood flow, it can 
be useful to soak the coils in thrombin to increase 
their thrombogenic potential. 

The liquid tissue adhesive n-buiyl cyanona-vkite 
(NBCA) has been utilized in an increasing number 
of applications and was recently proposed for use in 
stopping active hemorrhage. In a series of 16 patients 
with arterial hemorrhage (one of which was an iat- 
rogenic injury), the authors reported being able 
to stop active bleeding in 75% of patients without 
any complications related to the embolic agent [16]. 
Although this is not a tremendous success rate, 10 
of the 16 patients had already previously failed prior 
embolization with coils or particles. 

NBCA does have some attractive properties. 
Since it is mixed with ethiodized oil, the mixture 
is radio-opaque, which should aid fluoroscopic 
control of the embolization. By varying the ratio of 
ethiodized oil and NBCA, the polymerization rate 
can be adjusted, thus providing the ability to cus- 
tomize how far peripherally the agent will penetrate. 
Finally, although it is a liquid, it does not penetrate 
out into the capillaries, and thus the risk of infarc- 
tion should be low. Further investigation into the 
use of this agent is warranted. 



7.5.3 

Alternative Techniques 



niques that stop bleeding without leaving behind 
emboli to occlude the lumen should be considered. 



7.5.3.1 

Balloon Tamponade 

Balloon tamponade is the simplest technique and 
involves inflating either a compliant occlusion bal- 
loon (Balloon Wedge Pressure Catheter; Arrow Intl.; 
Reading, PA) or an appropriately sized angioplasty 
balloon across the injured segment of artery. If using 
an angioplasty balloon, it is recommended that low- 
pressure inflation be done to avoid further tearing of 
t lie ai'teiy Some recommend using a balloon that is 1 
mm smaller than the size of the balloon that caused 
the rupture [17]. Balloon occlusion can be used as a 
temporary measure to stop hemorrhage while plan- 
ning definitive therapy, or in some instances it has 
been used as the definitive treatment. The theory 
behind balloon occlusion as the sole therapy is that 
the balloon will prevent continued extravasation 
and allow the perivascular thrombus time to orga- 
nize and seal the leak. 

There have been a number of reports of using 
temporary balloon tamponade to repair arterial 
ruptures both in the iliac and renal arteries [17-20]. 
With this technique, a balloon is left inflated across 
a ruptured artery anywhere from a few minutes 
up to an hour. Repeat arteriography is done after 
the balloon is deflated and if the leak persists the 
balloon is reinflated. With shorter inflation times 
this cycle may have to be repeated several times. 
One problem with this approach is that the tissues 
may not tolerate the ischemia for the length of time 
needed to finally stop the bleeding. This is why some 
authors only inflate for a few minutes at a time in 
order to allow reperfusion of the tissue in-between 
inflations, even though additional bleeding may 
occur during the deflation periods. Although longer 
inflation times maybe associated with less bleeding, 
intraluminal thrombus can form while the balloon 
is arresting blood flow [20]. 



In some cases the dai 
patency cannot be sac 



7.5.3.2 




Uncovered Stents 




Standard uncovered 


stents can occasionally be 


used to seal a vascula 


r defect [21] (Fig. 7.6). While it 


seems counterintuitrv 


e that a bare stent would seal 


an arterial leak, this 


can work if the defect runs 



laged artery is critics 

ificed. Thus alternate tech- obliquely through the arterial wall. In this setting, 




Fig. 7.(i. a Hepatic ;i:':ci'iijgi".i;Vi in ;'. pa:ie:i; who developed needing sev- 

jf'.e: J VV-',i.'L\r ope:a;ion. A nseudoaneurvsm iiinf". i is seen 

where the gastroduodenal artery was resected. l> A balloon occlusion 

catheter (iiiTpir! was inflated across die arterial derec- to tamponade 

bleeding until a decision was T.ace retarding cehniiive therapy, c A 

bare balloon expandable stent [iinvn-~) was placed across the arterial 

defect with plans to pass microc.oils throng;: the sir:'/; however, tins 

ladv si; owed I hat die ane::al d erect had been sealed by the 

overed stent alone 



the expanded stent forces together the two sides 
of the oblique tear, thus sealing the defect. This 
maneuver is a little risky since it can be difficult to 
tell whether the tear runs obliquely through the arte- 
rial wall. If it does not, placing a stent may simply 
hold open the defect and promote continued hem- 
orrhage. However if the stent by itself does not stop 
the bleeding, the stent can be used as a gate to trap 
coils out in the pseudoaneurysm. A catheter can be 
passed through the stent interstices allowing coils to 
be deployed out in the pseudoaneurysm. 



7.5.3.3 

Stent Grafting 

A potentially simpler and more secure method is 
deployment of a stent graft across the arterial defect. 
There are currently several commercially available 
stent grafts. Some, like the Fluency (Bard Periph- 
eral Vascular; Tempe, AZ) and the Viabahn (W.L. 
Gore; Flagstaff, AZ) have a polytetrafluoro ethylene 



(PTFE) layer that would seal the hole in the vessel 
wall. The Wallgraft (Boston Scientific) has a woven 
Dacron graft material and although this is more 
porous, it will still effectively seal a hole and prevent 
further bleeding. 

Currently most of these devices are fairly large 
and are best suited for repair of larger vessels such 
as subclavian, iliac, femoral, or splenic arteries 
(Fig. 7.7). There is also a smaller stent graft origi- 
nally designed for coronary applications, the Jostent 
( lomed; Helsinghoiy, Stvedt-n), Ilia! lias been i i s r- ■::! in 
smaller hepatic and renal arteries [17, 22-28]. Cur- 
rently this device is still in trial and is not readily 
available in the United States. It is important with 
all these devices to carefully match the device diam- 
eter to the vessel diameter. Choosing a device that 
is too small for the vessel will yield a poor seal and 
potentially could allow continued hemorrhage. On 
the other hand with the Viabahn, if the device is 
significantly over-sized for the vessel, some of the 
graft material will fold into the device lumen and 
may compromise blood Hove. 





Fig.7.7. a Injection of a s'jbchivijn she.\t:'. Uiai had been placed on the ward 
and was later found to have pu.s.il lie blooi flow coming out of the sheath. The 
sheath {arrow) enters die s : abclav;an ,ine:y and had been accidentally advanced 
up into the right vertebral artery, b Righi biach.ocephakc arteriogram after 
removal of the mis. "laced ■=:■_ eiii h shows :apid com;as: extravasation/ bleeding 
(arrow) from the subchivian .u te: y puncture site. A VV:ilJgrp.ft has been partially 
deployed prior to removing I lie sheath, c A fie: teposiitoning the Wallgraft, the 
arterial defect has been sailed. The gr.tfi does noi compromise the carotid but 
does occlude the vet trbr.il aitetv, which wns well c/erft.sed from the contralat- 



7.6.1 
Hepatic 

Hepatic iatrogenic bleeding can be divided into 
intrahepatic and extrahepatic varieties. Intrahe- 
patic bleeding and pseudoaneurysms can result 
from any of the interventional hepatic procedures 
including percutaneous biopsy, transjugular biopsy 
[29], percutaneous biliary drainage or stenting, and 
even transjugn btr int in hepatic portosystemic shunts 
(Fig. 7.8). Extrahepatic lesions are often postsurgi- 
cal in nature due to injury to a vessel or breakdown 
of a vascular anastomosis. Extrahepatic lesions 
may cause pain by mass effect and may be more 
likely to cause hemoperitoneum since they have 
less surrounding tissue to help contain the bleed- 
ing. Intrahepatic lesions are more likely to cause 
pain or hemobilia but may go undetected for a long 
time, with some patients not presenting until several 
months after the injury [30]. 

The locations also tend to dictate the therapy 
used. Since intrahepatic lesions often involve a 
peripheral branch, the artery can generally be sac- 



rificed. Thus when the lesion can be reached with 
a catheter, standard coils or Gelfoam embolization 
is usually the primary therapy. Embolization of 
intrahepatic lesions is usually highly successful. In 
one study [31], hemobilia was controlled in 100% of 
eight cases. Hidalgo et al. [30] controlled hemobilia 
in 11 of 12 patients, however, several of the patients 
had recurrent bleeding 2 weeks to 2 months later. 
Although Tessieret al. had a success rate of only 86% 
for embolization, they noted that mortality was only 
14% in patients treated with embolization but was 
25% after surgery [32]. Results of direct puncture 
have been very favorable with effective occlusion 
of the lesions; however there are no large series and 
only scattered case reports of treating hepatic and 
pancreatic lesions with this technique [8, 9, 33-35]. 
One recurrence 2 months after initially successful 
direct puncture embolization hs 
[8]. 

Extra-hepatic pseudoaneurysm 
treated with coil embolization sin 
fice a major branch to a lobe or evt 
Successful percutaneous thrombin 
anastomotic pseudoaneurysm has been described 
[12]. Recently, a number of case reports have been 
published on the use of stent grafts to repair these 



i been reported 



a the entire liver, 
l injection of a 




Fig. 7.8. a Arterial phase of a hep; 
study shows prominent oparifical 
shows rapid flow into the bile dui 
arterio -biliary fistula 



: arteriogram in ,1 .\me::: w:t!i hemobilia ? days after T!PS. b L : -. t e r passe of the same 
■n of the bile edicts, e Magitifie;: supei-selec live .i] :ec ii-gi ;"un Ui rough ;'. micro catheter 
; from this arterial branch, d Pos:-cmbolizaiic::i a: reriogr.ir.i show:; no further flow to t 



extra hepatic lesions [27, 28, 36, 37]. While successful 
in all cases, larger series are needed to validate this 
technique. 



7.6.2 
Renal 



Pseudoaneurysms or arteriovenous fistulas occur 
after 0.2-2% of biopsies in transplant kidneys. Simi- 
larly after percutaneous nephrostomy the incidence 
of significant arterial injury is around 1%. Although 
vascular injuries typically manifest within the first 
week or so, delayed presentations out to 21 months 
after initial nephrostomy have been reported [38]. 
Although most papers report only a few patients [39, 
40], embolization is well accepted as the preferred 



method to deal with vascular injuries after r 
biopsy and nephrostomies [41]. 

Technical success of embolization for 
vascular injury is quite high, around 95-100% [42- 
44] . Typically the recurrence rate is nearly 0%; how- 
ever, in one series a second embolization session was 
needed in2 (15%) of 13 patients to fully occlude arte- 
riovenous fistulas and achieve true technical suc- 
cess [44]. An analysis of the effect on renal function 
of selective embolization for traumatic renal lesions 
revealed that the mean volume ol infarcted kidney 
was only 6% (range 0-15%) and 1 weekpostemboli- 
zation the serum creatinine was normal in all their 
patients [42]. A series of renal transplants estimated 
that the maximal volume of infarcted kidney after 
embolization for biopsy-related injuries was always 
less than 30% [44]. Also, while renal function dete- 






riorated in three patients, the serum creatinine sig- 
nificantly improved in 10 of 13 (77%). 

The incidence of rupture after renal PTA has 
ranged from 1.6% to 5%. This is clearly a situation 
where traditional embolization is undesirable since 
it will lead to infarction of the entire kidney. How- 
ever, if the patient is not a surgical candidate and 
they have another well functioning kidney, embo- 
lization with sacrifice of the kidney can be used 
as a life-saving maneuver if no other options are 
available. Stent graft use in the renal arteries has 
been described in a number of small case reports 
[17, 22-26, 45, 46]. They have been mostly used for 
exclusion of renal aneurysms but have occasionally 
been used to treat ruptures. In a series of five renal 
ruptures [17], all were able to be managed nonsur- 
gically. Some were treated by balloon occlusion 
alone but one patient required a stent-graft. The 
stent-graft used in this setting was a home-made 
device of thin-walled PTFE mounted on a Palmaz 
stent. Another patient had a second bare stent 
placed within the original stent that caused the 
rupture, and this was followed by 2 minutes of bal- 
loon tamponade with successful sealing of the leak. 
There are no good series with long-term follow-up 
reported; however, a trial with 12 renal stent grafts 
showed reasonable patency with a restenosis rate of 
only 7.3% at 6 months [23]. 



7.6.3 

Miscellaneous Injuries 

Outside of the liver and kidneys, there are innu- 
merable other types of iatrogenic arterial injuries 
that can occur. Of course the commonest iatro- 
genic injury is post-catheterization femoral artery 
pseudoaneurysms, but this is discussed in another 

One type of injury that maybe increasing in fre- 
quency (due to the increased use of central venous 
lines} is damage to the subclavian or carotid arter- 
ies or branches during line placement. If the injury 
involves a small artery such as a thyrocervical 
branch, selective embolization will typically solve 
the problem. If the subclavian artery itself is punc- 
tured or has a catheter placed into it, management 
becomes more difficult. Surgical repair carries 
high risks and may even require a thoracotomy, 
whereas standard embolization is not practical 
because of the arm ischemia it would cause. Plac- 
ing a stent-graft is probably the preferred way to 
deal with this, assuming that the arterial defect is 



in a location that allows a graft to be placed without 
compromising the carotid artery. In fact, the first 
reported application of an intravascular stent graft 
was to close a large hole (10 Fr) in the subclavian 
artery caused by an improperly placed Port cath- 
eter [47]. 

Arterial rupture is a feared complication of iliac 
PTA but fortunately is uncommon, with a 0.2-0.4% 
incidence [18]. However when it does occur it can 
be life-threatening, with the patient rapidly becom- 
ing hypotensive. This kind of injury has tradi- 
tionally been managed surgically. Embolization 
is not typically done since it will likely make the 
leg severely ischemic. However embolization can 
be combined with a femoral- femoral cross-over 
graft. This type of graft has lower patency than 
a direct aortofemoral bypass, however it can be a 
useful option inpatients who are high surgical risks 
since it is an extra-abdominal operation and can be 
accomplished without general anesthesia. It does, 
however, require that there is good inflow into the 
opposite iliac artery. With the increasing availabil- 
ity of commercially available covered stents, stent- 
grafting is now considered by some to be the treat- 
ment of choice [48]. 

Embolization techniques have been applied to 
iatrogenic hemorrhage throughout the entire body. 
There are numerous case reports of embolization 
successfully terminating bleeding after a wide vari- 
ety of operation or procedures as varied as prosta- 
tectomy [49], orthopedic osteotomy [50], salpingo- 
oophorectomy [51], bone marrow biopsy [52], and 
pelvic abscess drainage [53]. Kwon and Kim [54] 
reported a particularly large series of 24 cases of iat- 
rogenic arterial injuries in the uterus after curettage 
or cesarean section. Gelfoam embolization of the 
uterine arteries successfully stopped bleeding in all 
cases. Interestingly, four patients desired to become 
pregnant after undergoing bilateral uterine embo- 
lization, and all four were able to deliver full-term 
babies. A theme running through all these reports is 
successful cessation of bleeding with no or minimal 
complications, enforcing the idea that embolization 
shou Id be the first approach to treat iatrogenic arte- 
rial hemorrhage. 



Arterial puncture site complications such as hema- 
toma, pseudoaneurysm, arteriovenous fistula, dis- 



Ml'', rurcy 



section, and arterial occlusion can all certainly 
occur but are not unique to embolization cases. 
Trauma from the catheters and guidewires can also 
cause spasm or dissection in the main trunk or 
branches leading to the arterial defect that one is 
attempting to treat. This can prevent being able to 
advance the catheter to the bleeding site, or if the 
catheter can be advanced beyond the spasm / dis- 
section there may be no flow to carry flow-directed 
embolic particles more distally. Spasm by itself may 
decrease blood flow to the arterial defect sufficiently 
that the bleeding will stop. However, this is a less 
secure method of managing the iatrogenic lesion. 
If the spasm resolves, bleeding may recur once the 
injured segment is again subjected to normal arte- 
rial pressure. Also any pseudoaneurysm or fistula 
would not be directly occluded and the permanence 
of the occlusion of that lesion would be questionable. 
If there are any collateral communications beyond 
the spasm or dissection, the pseudoaneurysm would 
likely remain patent. 

If spasm occurs, local injection of vasodilators 
(e.g. 50-100 microgram boluses of nitroglycerin} 
may relieve the spasm. The best approach is to try 
to avoid the spasm in the first place. Minimizing the 
manipulation of guidewires and being very gentle 
will help reduce spasm. Also if it is anticipated that 
advancing the catheter will be difficult, prophylac- 
tic boluses of nitroglycerin can be employed. If dis- 
section occurs, it may be possible to tack down the 
flap with a stent or PTA depending on the location 
and the caliber of the artery. 

As emboli are being delivered to the target, the 
next complication that may be encountered is non- 
target embolization. When using flow-directed 
emboli such as Gelfoam or PVA, overly vigorous 
injection can lead to reflux of emboli out of the 
target artery. The risk of reflux is increased as the 
end of the embolization approaches, since there will 
be increased resistance to flow in the target artery. 
The chance of particle reflux can be minimi/cd by 
gentle injection, having the emboli suspended in 
contrast, and careful fluoroscopic monitoring of the 
injection. 

Nontarget embolization can also occur when 
using coils. Rarely do coils migrate to a remote 
location, but they can cause undesirable occlusion 
of arteries or branches adjacent to the target vessel 
(Fig. 7.9). Having the catheter securely positioned 
well into the target artery before pushing out the 
coils will help prevent nontarget embolization. 
However it is not always possible to have the cath- 
eter advanced well into the target artery, especially 



if the injury is close to the vessel origin. Maintain- 
ing good manual control of the catheter close to the 
groin access will allow one to move the catheter in 
or out slightly to help form the coils in situ. A gentle 
tapping motion ofthe pushing wire will also help the 
coil to form in a tight configuration. Rapidly push- 
ing the coil out often elongates the coil and forces 
the delivery catheter back out ofthe target artery. 
Choosing appropriately sized coils is also critical 
to prevent this complication since pushing in over- 
sized coils will also tend to back the catheter out of 
the target artery. 

If a coil does get misplaced, retrieval with snares 
can be attempted. This can be quite difficult since 
the errant coil may often wedge itself into the periph- 
eral aspect of another branch. That plus the spasm 
that frequently occurs from excessive manipula- 
tion makes it difficult to open a snare sufficiently to 
get around the coil. An unusual form of nontarget 
embolization is delayed migration of coils from the 
original point of deployment into another structure. 
Coils placed in an intrahepatic pseudoaneurysm 
have been reported in two cases to migrate (pre- 
sumably via erosion of the adjacent structures) into 
the bile ducts [8, 55]. In these cases biliary obstruc- 
tion resulted and required percutaneous or surgical 
removal of the coils. 

Tissue infarction can range in severity from an 
expected inconsequential occurrence up to a fatal 
complication. When embolizing end-arteries such 
as renal branches, it is expected that the paren- 
chyma distal to the embolized segment will infarct. 
However if the embolization is done peripherally 
enough, only a small segment of the kidney will 
infarct with no effect on renal function, and some- 
times it will cause only minimal symptoms (pain 
and fever). In the liver, the consequences depend 
partially on the degree of intrahepatic collateral 
flow beyond the embolized artery. Hashimoto et 
al. [31] found that no hepatic infarction occurred 
when there was good collateral flow, but all four 
patients w it li poor collateral ( low dt-veloped infarc- 
tion. Three of these four had no symptoms and had 
infarcted segments seen on CT scans done to eval- 
uate transient transaminase elevations. However, 
one patient did progress to hepatic failure. Such 
severe ischemia is uncommon in embolization for 
iatrogenic bleeding but has been reported by others 
[30]. Clinically significant ischemia can be mini- 
mized by super-selective embolizations with 3 Fr 
catheters and by avoiding the use of very small par- 
ticles or liquid embolic agents that penetrate out to 
the capillary level. 




Fig. IS. a CT of a 38-year- ok. female who had rank pain after a pai tiai nephrectomy. A hematoma and a pseudoaneuiysm 
iiiMiiu 1 ) a iv seen, b Lefi renal arteriogram shows a pseucoai'.e'.u ysm near the origin o: the inle: ioi brunch of the rein I artery, 
c Coils were placed distal and into the pseud' '.lnetuvsm, ai'd an aticmp: was made to p'ac^ the final coils iust proximal to the 
pseud oaneuiysm. These last coils moved oc.i into ihe main renal ai ierv occiuding ah rena! artery ~ow. d An ai ie: iogram done 
3 months later shows some recanali nation tun 'Ugh iiie displaced coils with some perfusion to the kidney 



Future Development and Research 

Areas for potential research include development of 
new embolic a^etits and improving enisling embolic 
devices. Gelfoam and PVA are less than ideal embolic 
agents. Some of the newer embolic agents such as 
Embospheres (Biosphere Medical; Rockland, MA) 
maybe more easily injected through small catheters, 
but they also may be more prone to causing tissue 
ischemia since they can travel more peripherally. 
Studies will need to be done to see whether this 
agent is appropriate for treating iatrogenic bleeding 



and to determine what size spheres should be used. 
Liquid agents have some potential benefits in terms 
of ease of use and control. The initial studies with 
NBCA [16] are a step in the right direction, but some 
of the other new liquid embolic agents will also need 
to be evaluated. 

Nontarget embolization can ruin an otherwise 
good result and while coils are one of the most com- 
monly used embolic devices, they can be difficult to 
form properly. Better control over the coils is desir- 
able. The Guglielmi detachable coils (Target Thera- 
peutics; Fremont, CA) do provide the ability to redo 
the deployment if it is unsatisfactory, and they have 



night 



been applied to closure of iatrogenic vascular lesions 
[56]. However, they are expensive and the electro- 
lytic detachment is more complex than the use of 
standard coils. There should be investigation into 
new embolic devices with better control and possi- 
bly different shapes that might enhance both place- 
ment and control of bleeding. 

Although stent-grafting is an increasingly attrac- 
tive therapy for some cases of iatrogenic bleeding, 
smaller, better devices and more ready availability 
are necessary. Bifurcated or fenestrated devices sim- 
ilar to those developed for aortic aneurysms i 
widen the application of visceral stent-grafts, 
the presence of critical side branches s 
limits the use of stem-sratts. 



Conclusion 

Embolization has become one of the primary tech- 
niques for tre.nina utrogriik" bleed ma- However since 
by default it causes vessel occlusion, it is mostly appli- 
cable to small, less important arteries or in periph- 
eral branches that can be readily sacrificed. Proper 
technique generally allows a high degree of clinical 
success with minim;! I risk. With the ongoing develop- 
ment of stent shifts, end avascular treatment may also 
become the primary means of repairing larger, more 
critical vessels that must remain patent. 



References 

l.Tisnado J, Beachley MC, Amendola MA (1979] Transcath- 
eter embolization of traumatic renal ar:e:iovenous fistula. 
Urol Radiol 1:175-177 

2. Walter JF, Paaso BT, Cannon WB (1976) Successful trans- 
ca:he:er em ho 1. c control cc massive- aemaiobdia secondary 
to liver biopsy. Am J Roentgenol 127:847-849 

3.Gunther R, Jonas U, Jacobi GH (1977) Kidney damage 
during translumbar aortographv ; rea:ed by selective ■; .ur- 
eter embolisation. Rofo 126:426-429 

4.Athanasoulis CA, Waltman AC, Barnes AB.HerbstAL (1976) 
Angiegrapa i.: coiilioi of peiv.c bleeding from treated carci- 
noma of the cervix. Gynecol Oncol 4:144-150 

5.Encarnacion CE, Kadir S, Beam CA, Payne CS (1992) Gas- 
;rointes-inal bleeding: I : e.Uir.trnt wiili gasuoimestmal a r re- 
rial embolization. Radiology 153:505-508 

6.Schenker MP, Duszak R fr, Soulen MC, Smith KP, Baum RA, 
Cope C et al. (2001) Upper gasiicintesanal he:vi. :"i li.ia^ 
and Iranscaiheie: emboleclinapy: clinical and technical 
factors impacting success and survival. I Va:.c Inter v kaoiol 
12:1263-1271 



7.SavaderS|,TrerotolaSO,Merinen?A'enbruxAC,Osterman 
FA i 199 c: ) Hemobia.i are: percutaneous nansliepatic b.li- 
jry dra.nage: [real ment ivi:li tianscaiheler emhelelherapv. 
I Vase Inlerv Radiol 3:345-352 
8. Araoz PA, Andrews [C i2000j 1'ireci percutaneous embo- 
lization o\ visceral aicery aneurysms: techniques and pit- 
falls. I Vase Interv Radiol 11:1195-1200 
9.CapekP, RoccoM, McGahan], FreyC (1992) Direct aneu- 
rysm puncture and cod occlusion: a new approach to pen- 
pancreadc arterial pse.ideaiaeurvsms. I Vase Iraerv Radiol 
3:653-656 

lO.Kemmeter P, Bonne II B, VanderKolk W, Griggs T, van Erp 
I (J. ODD) Percutaneous thrombin injection o: sp.aiaci'.nic 
artery aneurysms: two case reruns. I Vase interv Radiol 
11:469-472 

ll.LukancicSP.NemcekAAJr.VogelzangRL (1991) Posttrau- 
matic intrahepatic arterial pseudoaneurysm: treatment 
with direct percutaneous puncture. 1 Vase interv Kadi':-! 
2:335-337 

12. Patel JV, Weston MJ, Kessel DO, Prasad R, Toogood GJ, Rob- 
ertson i (2003) Hepa:ic arleiv pseiideanec.iysm after aver 
ujnsphiaiati.'n Irealmcn' with pi-. ..'.ineous thrombin 
injection. Tr;.*'vplantalicm 75:1755 1757 

13.Brophy DP, Sheiman KG, Amatulle I', Altbari CM (2000) 
Iatrogenic f e ■ n - ■ r ^ I p^cu. I:\nn u"VMr:s "lr.imbin injection 
after failed '..'S ^juic ^urprL^iuvi. vidiology 2!4:.17S- 
282 

14.Morgan R, Belli AM (2003) Current treatment methods for 
pos lea I he lei .i..-.i... n ^-.TuCi-jiiicm i sjjjv. i \ ,:ic Imeiv Radiol 
14:697-710 

15. Cope C, Zeit R i 1 9Si:'j Coagulation of aneurysms by direct 
percuianeous thrombin injection. AIR Am j konvgenol 
147:383-387 

16. Kish JW, Katz MD, Marx MV, Harrell DS, Hanks SE (2004) 
N-t'uryl cyanoacrvlate embolization for control of acute 
arterial hemorrhage. ; Vase Interv Radiol 15:689-695 

17.Morris CS, Bonnevie G), Najarian KE (2001) Nonsurgical 
treatment of acute iatrogenic renal artery injuries occur- 
ring a leer :e:aal artery angioplasty ,w\:. s tenting. AJR Am I 
Roentgenol 177:1353-1357 

18. Cooper SG, Sofocleous CT (1998) Percutaneous manage- 
mem of angioplasty- related .li:,: artery i upr.ae waih pres- 
ervation of luminal patency by prolonged balloon tampon- 
ade. I Vase Interv Radiol 9:81-83 

19.|oseph N, Levy H. Lipman s i 1 ( !S7! A ngiop la sty- related iliac 
artery rupture: treatment by temporary bakoon occlusio:a. 
Cardiovasc Inlervent Radiol 10:276-279 

20. Smith TP, Cragg AH (1989) Non-surgical treatment of iliac 
artery rupture to I towing angle p. a sty. 1 Vase Interv Radiol 

21 . Kelly A 1 1 1 995) Case report: ih.ic artery rupture-percutane- 
ous treatment by stent insertion. Clin Radiol 50:876-877 

22.Bisschops RH, Popma JJ, Meyerovitz MF (2001) Treat- 
ment o: fibromuscular dysplasia and renal artery aneu- 
rysm with use of a stent-graft. | Vase Interv Radiol 
12:757-760 

23.Gaxotte V.Laurens B, Haulon S, Lions C, Mounier-Vehier 
C, Beregi JP (2003) Multicenter trial of the Jostent balloon- 
expaneao.e stenl-gran in renal and iliac artery lesions. ; 
EndovascTher 10:361-365 

24. Pershad A, Heuser R !2004) Renal anery aneurysm: suc- 
cessful exclusion with a slem graft. Catheter Cardiovasc 
Interv 61:314-316 



25.SchneidereitNP,Lee S, Morris DC, Chen JC (2003) Endo- 
vascular repair of a ruptured renal artery aneurysm. 1 
EndovascTher 10:71-74 

26.Tan WA, Chough S, Saito J, Wholey MH, Eles G (2001) Cov- 
ered stent for renal artery aneurysm. Catheter Cardiovasc 
Interv 52:106-109 

27.VenturiniM,AngeliE, SalvioniM.de Cobelli F, Trent in C, 
Carlucci M, et al. (2002) Hemorrhage from a right hepatic 
artery pseudoaneurysm: end ova sen la r treatment with a 
coronary stem-graft. I Endovasc The: 9:221-224 

28.SakaLH,UrasawaK,OyamaN,KitabatakeA(2004)Success- 
ful covering of a hematic a:: civ aneurysm with a coronary 
stent graft. Cardiovasc Intervent Hadiol 27:274-277 

29. Roche CJ, Lee WK, Duddalwar VA, Nicolaou S, Munk PL, 
Morris DC (2001! Intrahepatic pseudoaneiirysm compli- 
i"ati:ig iransiugular biopsy of ihe liver. AlhL Am J Roent- 
genol 177:819-821 

30. Hidalgo F, Narvaez JA, Rene M, Dominguez ), Sancho C, 
Montanva X : : 39." : Treat merit of hemobilia with selective 
hepatik artery embolization. I Vase Interv Hadiol :::7 t; 3- 
798 

3] . Hashimoto M. Akabane Y, Heianna ■. Tate E, Ishiyama K, 
Nishii T et al. (2004) Hepatic infarction following selective 
hepatic artery embolization with microcoils tor iatrogenic 
biliary hemorrhage. Hepatol Res 30:42-50 

32.Tessier D], Fowl RJ, Stone WM, McKusick MA, Abbas MA, 
Sarr MG e: al. !2r:03; Iatrogenic hepatic artery pseudoan- 
eiirysm s: an uncommon complication after hepatic, biliary. 
and pancreatic procedures. Ann Vase Surg 17:663-669 

33.Merhav H, Zajko AB, Dodd GD, Pinna A (1993) Percutane- 
ous transhepatic embolization of an inlra hepatic pseud oa- 
neurysm to I hawing liver biopsy in a liver transplan: patient. 
Transpl Int 6:239-241 

34. Mi'lonig G, Graziaclei I W, Ivaldenherger K Kornigsrainer A. 
lascbke W. V'ogel W i'2C04'i l-ercuta neons nianagement oi" a 
hepatic artery aneurysm: bleeding after liver Iransplanta- 
tion. Cardiovasc Intervent Radiol 27:525-523 

35. Chan RP, David E i'2004: Reperfusion o: splanchnic artery 
aneurysm fol. owing transcaiheler emnojzauon: treatment 
wit;: percutaneous thrombin mieclion. Cardiovasc Inter- 
vent Radiol 27:264-267 

36. Larson RA, Solomon J, Carpenter JP (2002) Stent graft 
repair of viscera! artery aneurysms. I Vase Surg 36:1 2::-5- 
1263 

37. Paci E, Antico E, Candelari R, Alborino S, Marmorale C, 
Landi E (2000) Pseudoaneurysm of the common hepatic 
artery: treatment with a stem-graft. Cardiovasc Intervent 
Radiol 23:472-474 

38. Kaufman JA, Edelstein RA (1994) Arteriocaliceal fis- 
tula from prolonged nephrostomy lube drainage. | Urol 
151:1616-1618 

39.Peene P.Wilms G, Baert AL (1990) Embolization of iatro- 
genic renal hemorrhage- following percutaneous nephros- 
tomy. Urol Radiol 12:84-87 

40. Ueda J, Furukawa T, Takahashi S, Miyake O, Itatani H, Araki 
Y (1996) Arterial embolization to control renal hemor- 
rhage in oanems wit.; p ere ina neons nephrostomy. Abdom 
lmaging21:361-363 



I.Zagoria RJ, Dyer RB (1999) Do's and don't's of percutane- 
ous nephrostomy. Acad Radiol 6:370-377 

1. Chalziioannoii A, brouiitz.os F. Primc-Ls F. Ma I agar: K, L : o:o- 
cleous C, Mounkis D et al. (2004! Effects of superseleclive 
embolization for renal vascular injuries on renal paren- 
chynia and function. Eur i Vase Endovasc Surg 2?:2fli-20o 

5. Perini S, Gordon RL, LaBerge JM, Kerlan RK )r, Wilson MW, 
Feng S et a I. ! lOQS! Transcatheter embolization of biopsy- 
related vascular iniury in the transpl an; liidney: immediate 
a :;d long-term outcome. I Vase Interv Radiol 9:101 1-1019 

). Maleux G. Messiaen T. Stccl.x L, V'anrentergl'.em Y. Wilms G 
(200?) Transca theier embolization of biopsy- related vascu- 
lar iniuries m rena. allografis. Long-term lech meal, clinical 
and biochemical results. Aeia Hadiol 44:13-17 

j. Liguori G.Trombetta C, Bucci S.Pozzi-Mucelli F, Bernobich 
E. Helgrano F (2002) l- : erciiluneoiis management of renal 
arterv aneurysm with a stent-grafi. I Urol I 07:2513-2510 

i.MajwalTK, Ismail A, Alaqtly R (2002) Renal artery steno- 

fistula.J Invasive Cardiol 14:411-413 
7.Becker G), Benenati )F, Zemel G, Sallee DS, Suarez CA, 

Roeren TK et al. (1991) Percutaneous placement of a 
balloon-expandable intraluminal grafi lor lite-threaten- 
,ng si.ioc.avia:; :::>na. hemorrhage. I Vase Interv Radio! 
2:225-229 

3. Allaire E, Melliere D, Poussier B, Kobeiter H, Desgranges 
K Heequemin | P (2003:1 ii ac artery rupture during balloon 
dilatation: what treatment? Ann Vase Surg 17:306-314 

J.Ibarra R, Magee C, Ferral H, Thompson IM (2003) Post- 
proslateetomy deeding managed by endc'vasetilar embo- 
lization.] Urol 169:276-277 

l.Rickman M, Saleh M, Gaines PA, Eyres K (1999) Vascular 

Bone Joint Surg Br 81:890-892 
I.Mariano ST, Stein B, Vine HS, Rosshirt W, Sussman SK, 
Ohl.i SK ■: -03H .: Angiographic diagnosis and Iransarterial 
embolization of iatrogenic ovarian, artery injury. ] Vase 
Interv Radiol 11:625-628 

2. Areliano-Rodrigo E, Real MI, Muntanola A, Burrel M, 
Rozman M, Fraire GV et al. (2004) Successful treatment 
by selective arterial embolization of severe retroperitoneal 
hemorrhage secondary to- boise marrow biopsy in post- 
polycythemic myelofibrosis. Ann Hematol 83:67-70 

S. Harisinghani MG, Gervais DA, Maher MM, Cho CH, Hahn 
PF. V'aignese I el a.. :2li.;3: Transgltneal approach for per- 
cutaneous drainage of deep pelvic abscesses: 154 cases. 
Rad i o logy 22 8 :7D1 -705 

4. Kwon I H, Kim GS :2002 ) Obstetric iatrogenic arterial in: lines 
of ihe uterus: diagnosis with US and treatment with trans- 
catheter arterial embolization, k:\diegrapliics 22:35-46 

s. Ozkan OS, Walser EM, Akinci D, Xealon W, Goodacre B 
(2002) Guglielmi detachable coil erosion into the common 
bile duct after embolization of iatrogenic hepatic artery 
pseudoaneurysm. J Vase Interv Radiol 13:935-933 

j. Angle IF, Mats urn::- to AH : McO-raw IK. Hagspiel KI' 1 , Spinosa 
D), McCullough CS (1999! Percutaneous embolization of a 
high -flow oanereai.c Iransplant arteriovenous fistula. Car- 
diovasc Intervent Radiol 22:147-149 



Visceral Aneurysm 



8 Embolization of Visceral Arterial Aneurysms 



Craig B. Gla 



Introduction 99 

t' :■ L l'. :: ■ p h y ■= : -.:i J .;: g y 100 

Clinical Considerations 

Anatomy 103 

Technique 104 

Hepatic 1 05 

Incidence !05 

Causes 105 

Risks Posed by the , 

Management 106 
.1 Anatomic/Physiologic Consider 
.2 Technique 106 
.3 Results of Embolization 106 
.4 Complications 106 

Splenic 107 

Incidence 107 

Risks Posed by the Aneurysm 

Management 107 
.1 Anatomic/Physioiogic Consider 
.2 Technique 107 
.2 Results of Embolization 108 
.3 Complications 109 

Mesenteric 109 

Incidence 109 

Causes 111 

Risks Posed by the Aneurysm 

Management 111 
.1 Anatomic/Physioiogic Consider 
.2 Technique 111 
.3 Results of Embolization 112 
.4 Complications 112 

Renal 112 

Incidence 112 

Causes 112 

Risks Posed by tht 



C. B. Glaibebman.MD 

Ins true tor, promoted to assista:'.: professor of radiology, Radi- 
ology. Division of interventional Kaoiology, Washington Uni- 
versity School of Medicine, Mallntckrod; Institute of Radiol- 
ogy, 510 S. Kings highway, St. Louis, MO 63110, USA 
M. D. Dahcy, MD 

Professor, Rjdiologv -.in-." '•urgeiv I o vision oi I" 1 i a gnostic Radi- 
ology, Chief, Intervention.! I Radiology Section, Washington 
University Sch' >ol ■ if Medicine, KLiKincltrodt Iiisiilute of Radi- 
ology, 510 S. Kingshighway Blvd. St. Louis, MO 63110, USA 



M.ui.'geinent 113 

Aii.ilomic/Phvsiologic Considerations 
.9.4.2 Technique 113 

.3 Results of Embolization 113 
.4 Complications 113 

Complications 114 

Future Development and Research 114 

Conclusion 115 

References 115 



Introduction 

Visceral arterial aneurysms (VAAs) are rare with 
incidence rates ranging between 0.01% and 0.2% at 
autopsy. However, they are important entities to rec- 
ognize due to disastrous outcomes that result should 
rupture occur. Most visceral aneurysms are asymp- 
tomatic and are either overlooked clinically or found 
incidentally. Roughly 25% of symptomatic patients 
with VAAs will present with rupture [1]. Depending 
on the literature, reported mortality rates of rup- 
tured aneurysms range from 10% to 50% [1-4]. It is 
important to note that aneurysm location contrib- 
utes to the varying morbidity and mortality. 

VAAs occur in the splenic, hepatic, superior mes- 
enteric, gastroduodenal, pancreaticoduodenal, and 
renal arteries. Classically, splenic arlerv aneurysms 
have been found to account for 60% of all VAAs. 
Interestingly, Shanley et al. reviewed the litera- 
ture from 1985 to 1995 and found that hepatic artery 
aneurysms were more common [5]. This finding 
may reflect an increase in the number of percuta- 
neous hepatic and biliary interventions being per- 
formed. Furthermore, the routine use of computed 
tomographic imaging after trauma also attributes to 
the increased discovery of hepatic artery pseudoan- 
eurysms. Because VAAs occur infrequently, it must 
be remembered that much of the current literature 
consists of retrospective reviews of small cohorts 
that span as many as 10 to 15 years of experience. 
Therefore, much of what has been reported is either 
anecdotal or based on collective case reports. 



lan and M. D. Darcy 



A brief review of our experience over the last two 
years demonstrates that our group has embolized 
21 VAAs. By far, the majority of cases (nine) were 
performed urgently for traumatic liver and splenic 
lacerations. This was followed closely by seven iatro- 
genic pseud oaneurysms that were caused by percuta- 
neous biopsy, biliary intervention, or from previous 
laparotomy. Three pseud oaneurysms resulted from 
inflammatory causes such as pancreatitis, divertic- 
ulitis, and peptic ulcer disease. Two aneurysms were 
treated in patients with aagiomvolipomas. A vari- 
ety of techniques including coil, Gelfoam, and PVA 
embolization were used. No stent grafts were placed 
in this time period. Immediate technical success 
was achieved in all cases; however, the long-term 
outcomes are currently unknown. 

Historically, visceral aneurysms have been treated 
surgically by resection, ligation, and bypass, as well 
as vein patch angioplasty. Today, the interventional 
radiologist is particularly well suited to perform 
e forms of treatment with high techni- 
s and less patient morbidity. VAAs have 
been treated with transcatheter techniques such as 
coil embolization, thrombin injection, or stent graft 
placement. Although the literature is relatively scant 
regarding the long-term outcomes of embolization 
or stent graft placement, the trend has been toward 
minimally invasive therapies. Preemptive treat- 
ment of these lesions with percutaneous methods 
has become more popular due to the high mortality 
associated with rupture and the reduced morbidity 
that embolization procedures offer. 



Pathophysiology 

The arterial wall is composed of three layers. The 
outer serosal covering is the adventitia, the muscu- 
lar middle layer is the media, and the inner lining is 
the intima. True aneurysms are distinguished from 
false or pseudoaneurysms based on which layers of 
the arterial wall are present in the aneurysm itself. 
In order to classify an aneurysm as being "true," it 
must be comprised of all three layers. Pseudoaneu- 
rysms have any combination less than all three of 
the arterial wall components. 

Aneurysms can be either saccular or fusiform. 
Saccular aneurysms are typically spherical in shape 
and have a small communication or "neck" arising 
from the parent vessel (Fig. 8.1). Fusiform implies 
longitudinal dilatation along the course of the 



artery. True fusiform 






s ofv 



place for a tusitorm 
aneurysm to occur is in the superior mesenteric 
artery distribution; it is typically the result of post- 
stenotic dilatation from atherosclerotic disease. 

More commonly, VAAs are saccular pseudoaneu- 
rysms resulting from an insult to the arterialwall. His- 
torically, one of the most common causes of saccular 
aneurysm formation has been from bacterial endo- 
carditis. Originally described by Osier, aneurysms 
caused by an infectious etiology have been termed 
mycotic. Direct infection of the vaso vasorum in the 
adventitial lining lias been postulated as a cause of 
mycotic aneurysm formation. The resulting inflam- 
matory response typically results in saccular pseu- 
doaneurysm formation. The incidence of this type 
of aneurysm has diminished over time due to ear- 
lier detection and treatment with antibiotics. Today, 
mycotic aneurysms in the presence of endocarditis 
have a high association with intravenous drug abuse 
and can occur in visceral and peripheral arteries. 

Similarly, adjacent intlammatorv changes such 
as pancreatitis can cause compromise of vessel 
wall integrity. Proteolytic degradation can occur if 
pancreatic enzymes come in contact with arteries. 
Gastroduodenal and pancreaticoduodenal pseu- 
doaneurysms are especially prone to rupture in the 
presence of duodenal ulceration, pancreatitis, or 
pseudocyst formation [6, 7]. These should be treated 
regardless of size. 




L' A 



Fig. 8.1. Large saccuhir pseudooneurysni .; rising from a sig- 
moid branch of the IMA demonstrating a short, well-defined 
neck. (Courtesy of Jennifer E. Gould, MD) 



Embolization of Vis! 



Other causes of saccular aneurysm formation 
include trauma and iatrogenic injury from percu- 
taneous or surgical interventions. Any focal insult, 
perforation, or laceration can lead to pseudoaneu- 
rysm formation. These aneurysms are often symp- 
tomatic due to hemorrhage, pain, and hypotension 
that occur. Iatrogenic injuries will be discussed in a 
separate chapter of this text. 

Multiple saccular microaneurysms are com moiilv 
seen with vasculitis caused by polyarteritis nodosa 
(PAN). Amphetamine abuse can also lead to multiple 
renal and hepatic microaneurysms similar to those 
seen with PAN. The small size and diffuse nature of 
these lesions often precludes embolization. 

Inherent weaknesses caused by acquired or con- 
genital etiologies may lead to VAAs. Congenital 
weakness of the arterial wall from a collagen dis- 
order such as Ehlers-Danlos or Mar fan's Syndrome 
can result in either saccular or fusiform aneurysms. 
Ansi ography should not be performed in patients 
with Ehlers-Danlos due to the high risk of arterial 
rupuire.Therek're.eniraolmihoii'.vould be extremely 
dangerous to execute in these patients. 

Fibromuscular dysplasia (FMD) is an inherent 
arterial wall abnormality that classically affects 
the media of the renal arteries and can be associ- 
ated with renal artery aneurysms. Several subtypes 
of FMD have been described and the disorder can 
affect other medium-sized vessels including the 
carotid, vertebral, brachial, and visceral arteries. 
For the angiographer, FMD has the classic beaded 
appearance often described as a "string of pearls." 
Both aneurysms and dissections can be seen with 
this disorder. The treatment for [-'MP is angioplnstv 
of the intraluminal webs, which results in signifi- 
cant remodeling. 

Renal artery aneurysms can also be seen in 
patients with an^iomyolipomas (AMI.s) (lig.8.2}. 
Classically, AMI.s occur in elderly females and 
patients wiih tuberous sclerosis. The enure lesion 
can often be cmboh/ed in addition to coiling the 
aneurysms, A combination of coils and PVA or 
simply ethanol infusion wtrh a balloon occlusion 
catheter can be performed as definitive treatment 
or if surgical resection is anticipated. 

Although atherosclerosis has been implicated, 
there is debate as to whether it is the cause or the 
result of aneurysm formation. The heavy calcifica- 
tion seen in splenic artery aneurysms may be due to 
altered hemodynamics. For example, splenic artery 
aneurysms can be seen with portal hypertension 
(Fig. 8.3). Saccular or "berry" aneurysms associ- 
ated with hypertension and atherosclerosis arise at 




Fig. 8.2. Seleiiive r;gln renal iiiiiiogrrun denianslialins; :. huge 
AML with aneurysms thai di-phces die kidney Literally and 
:aipe: ioi ly. Tae p.meiil has tuberous sclerosis 



branch points where intrinsic weakness of the wall 
may exist. 

Aneurysm configuration should influence 
treatment planning. Proximal and distal control 
should be obtained if the aneurysm itself cannot be 
occluded.lt is typically easier to occlude short necks 
seen with the saccular form; therefore many differ- 
ent treatment options exist. These will be discussed 
later in the chapter. Fusiform aneurysms may not 
allow for both proximal and distal control and are 
often better treated with surgical ligation or recon- 
struction. 



Clinical Considerations 



Ap propria 



patient work up inclui 

of current and past medical history, 
pertinent imaging, a limited physical exam with 
evaluation of the pulses, and basic laboratory 
parameters. Endocarditis, vasculitis, pancreatitis, 
prior trauma, and congenital arteriopathies such as 
Ehlers-Danlos are important entities to be aware 
of. Knowing the past surgical history and whether 
prior percutaneous biopsy was performed would be 
relevant for iatrogenic causes. Since asymptomatic 
patients have VAAs that are often discovered inci- 
dentally and symptomatic patients often have vague 



C. R. GkiibenuanandM. D. Dar 




Fig. 8.3. a Ceikk" .;:-giog:';iiii in ;i p.itienl wi;h pciLiil !;ype:t 
demonstrating a dist.ii sp'r:iic lii l^:y aneurysm, b Celiac angiogram 
from another patient with nr.iluple spienk" aneurysms associated 
with portal hypertension who ha- .mo er gone liver transplantation. 
Note the iarge hepa:io psc.idoaneuiysni ;usl medial to the upper 
pole of the right kidney, c ?eleo:ive oommon hepatic artery injec- 
tion in the patient from b 



abdominal pain, CT or MR imaging has typically 
been performed looking for other etiologies prior to 
referral to the Interventional Radiologist. Reviewing 
the images and examining the patient in the Inter- 
ventional clinic are important steps in formulating 
a successful treatment plan. A complete blood cell 
count, prothrombin time with INR, and a basic or 
complete metabolic profile should be available prior 
to the procedure. Acceptable lab value limits vary 
depending on the institution. 

Diabetics and patients with borderline renal func- 
tion should be prehydrated to reduce the chances of 
contrast-induced nephropathy. A recent randomized 
controlled trial suggested that prehydration with 
sodium bicarbonate is more effective than sodium 
chloride in preventing contrast-induced renal failure 
[8]. For those patients with contrast allergies, prophy- 



lactic steroids are typieallv administered at least 12 
hours prior to intervention. If access from a brachial 
artery is required, a pre-procedure neurologic exam 
including mental status is vital to document any 
change during or after the procedure since catheters 
will cross the origin of at least one cerebral vessel. 

Ideally, monitored conscious sedation should be 
administered by a registered nurse with intensive 
care experience or training. Continuous assessment 
of vital signs and oxygenation is important to main- 
tain a comfortable level ot sedation without respira- 
tory or cardiovascular suppression. Typically, fast- 
onset, short-acting agents are used. In our practice, 
we use a benzodiazepine such as Versed (Roche 
Pharmaceuticals, Manati, PR} and the narcotic 
analgesic Fent a nyl (Suhlmsaze; Abbott Laboratories, 
North Chicago, PL). 



Embolization of Vis! 



Patients with active bleeding should be vigor- 
ously transfused to maintain hemodynamic sta- 
bility, and there should be no delay in transporta- 
tion to the angiography suite for treatment. Severe 
coagulopathy should be corrected with fresh frozen 
plasma and platelet transfusion because hemor- 
rhage can persist despite a technically successful 
embolization. Coagulation factors are required 
to maximize the effectiveness of the emboliza- 
tion materials and are responsible for the ensuing 
thrombus at the site of embolization. Plasma can 
infuse throughout the procedure, and the need 
for transfusion should not delay treatment if the 
patient is actively bleeding. 

Periprocedural antibiotics should be considered 
if end-organ ischemia is a possibility. This is more 
common with the small permanent agents such as 
PVA and in organs where there is poor collateral flow. 
Tissue necrosis and hematoma can lead to abscess 
formation. When total occlusion of the splenic 
artery occurs and infarction results, patients should 
receive the pneumococcal vaccine. They should also 
take prophylactic antibiotics for future procedures 
as if they underwent a surgical splenectomy. 

At our institution, outpatients are typically 
observed overnight and hematocrit levels are 
checked to watch lor 1 postprocedural complications. 
Postembolization syndrome consisting of pain, 
fever, and nausea can be seen in the first 24 hours 
following intervention. Follow-up Doppler ultra- 
sound can be used to assess the success of emboliza- 
tion. However, the timing and frequency of reimag- 
ing are debatable. There are reports in the literature 
demonstrating that recanalization of treated aneu- 
rysms can occur. 

For those patients who present emergently and 
require fluid and blood product resuscitation, trans- 
fer to the intensive care unit for close monitoring is a 
must. If coagulopathy exists, or there is concern for 
further bleeding, the sheath can be left in the access 
site should there be need for repeat angiography and 
embolization. 



8.4 
Anatomy 

It is vital to understand the arterial anatomy and 
know the vascular supply distal to the planned 
embolization. In some VAAs, tissue ischemia can 
occur if the parent vessel is completely occluded. 
However, if good collateral flow exists, such as in 



the stomach and duodenum, permanent emboliza- 
tion of entire vessels can be performed with some 
degree of impunity. 

The hepatic and splenic arteries typically arise 
from the celiac axis, which has its origin at the T12/ 
LI level of the abdominal aorta. The three main 
branches of the celiac include the splenic, left gas- 
tric, and common hepatic arteries. The splenic 
arterv is typically large and tortuous and sup- 
plies small branches to the pancreas. The common 
hepatic branches into the gastioduode nal and proper 
hepatic arteries. There is significant variant anat- 
omy of the hepatic arteries that the interventionist 
should be aware of. The most common variation is 
the replaced right hepatic artery, which arises from 
the superior mesenteric artery (SMA). This occurs 
in 12%-15% of the population. Other less frequent 
variations include the replaced left hepatic from the 
left gastric artery (11%) and the completely replaced 
common hepatic from the SMA (2%). 

The gastroduodenal artery arises from the 
common hepatic artery and supplies branches to 
the pancreatic head via the superior pancreatico- 
duodenal arcade (SPDA) and greater curvature of 
the stomach via the gastroepiploic. It is an excellent 
collateral vessel connecting the celiac to the SMA if 
either one becomes occluded. 

The SMA arises at the LI level and supplies the 
small bowel via jejunal and ileal branches, the right 
and middle colon via the ileocolic, right and middle 
colic arteries, as well as the pancreatic head via the 
inferior pancreaticoduodenal arcade (IPDA). 

The inferior mesenteric artery (IMA) arises from 
the aorta at the level of the left pedicle of L3 and sup- 
plies the left colon, sigmoid, and rectum. It is fre- 
quently occluded in older populations. Collateral 
flow to this distribution can come from the mar- 
ginal artery of Drummond or from branches of the 
internal iliacs. 

The renal arteries originate from the aorta at the 
L2 level. A third of the population has multiple renal 
arteries. The main renal arteries are 5 to 6 mm in 
diameter and typically bifurcate into anterior and 
posterior divisions. There is further subdivision 
into segmental, interlobar, arcuate, and interlobular 
arteries before termination in glomeruli. Capsular 
and adrenal arteries take their origin from the main 

The concept of collateral and end-organ vascu- 
lar supply is vital to understand when considering 
embolization of visceral vessels. Choice of embolic 
agents for these vascular distributions and applica- 
tions is described elsewhere in this book. 



n and M. D. Darcy 



Technique 

After arterial access is obtained, a sheath with hepa- 
rinized saline flush through the side port should 
be placed to maintain access throughout the case. 
Typically a 5 or 6 French short vascular sheath such 
as a Flexor Check-flo (Cook I no, Bloomington, I N) 
is used. Sometimes sheath upsi/c or exchange is 
required based on the arterial anatomy or type of 
intervention to be performed. Hoi example, guiding 
catheters such as a Balkin Up and Over (Cook inc.) 
may offer the advantage of a more secure access 
around corners during coil deployment. Certain 
devices such as stent grafts may mandate the use of 
larger sheaths for delivery. 

Catheter selection is based upon individual pref- 
erence and experience, but is typically guided by 
patient anatomy. It is often wise to perform a non- 
selective aortogram with a pigtail catheter (Merit 
Med ical Systems Inc., Salt Lake City, UT) to identify 
vessel origins, assess patency, and look for potential 
stumbling blocks such as variant anatomy or ste- 
nosis from atherosclerotic disease. A reverse curve 
catheter such as a Sos (A ngiodynamics Inc., Queens- 
bury, NY} can often be used to select visceral and 
renal arteries. A floppy tipped wire, such as a Bent- 
son (Medi-Tech/Boston Scientific, Watertown, MA) 
is used to select visceral branches and advance the 
catheter into the origin to perform selective angi- 
ography. Other catheter considerations include the 
Simmons (Cook Inc.), Roche Celiac (RC-1) (Roche 
Inc., Indianapolis, IN), Cobra (Merit Medical Inc., 
South Jordan, UT), Rochelnferior Mesenteric (RIM) 
(Cook Inc.), or even a Multipurpose Angiographic 
Catheter (MPA) (Cook Inc.) if approaching from 
the arm. Often, the coaxial use of microcatheters is 
advantageous when attempting to occlude vessels as 
peripherally as possible. Being as selective as pos- 
sible is important when embolizing tissues that have 
poor or no collateral supply. 

In our experience, the Sos catheter can be used 
to select the origin of nearly every visceral vessel 
including the renals. Once selective angiography is 
performed and access is secured by passing a wire 
distally, the Sos can be exchanged for a more appro- 
priate catheter to fit the anatomy, or a microcatheter 
can be used coaxially through the Sos. Hydrophilic 
coated wires and catheters are useful for navigating 
tortuous vessels. The secondary curve of the Cobra 
catheter can help to select more peripheral branches 
as well as provide a "backstop" to keep it from buck- 
ling out of the origin of the desired vessel. 



Another trick is the formation of a Waltmanloop 
if a reverse curve catheter is unavailable or unsuc- 
cessful at cannulating the origin of visceral arter- 
ies. This is typically termed over the aortic bifurca- 
tion with the tip of a Cobra or hockey-stick catheter 
in the contralateral external iliac artery. Using the 
back end of a Bentson wire, the loop is formed by 
pushing cephalad and twisting. The back end of the 
wire must be positioned in the ipsilateral common 
iliac segment of the catheter to provide the stiffness 
required to advance the entire loop into the distal 
aorta. The wire is then reversed so that the floppy 
tip can be used to select the desired vessel origin. 
An alternative way to form a Waltman loop was 
described by Sh husky- Goldberg and Cope and uses 
a stitch and guidewire combination to physically 
pull the catheter into a reverse curve formation [9]. 

Prior to intervention, selective angiography of 
the target vessel is performed. Access is secured and 
tested bypassing a wire through the catheter to ensure 
that it does not buckle out of the origin and lead to 
nontarget embolization. Permanent occlusion is the 
goal of treatment for VAAs due to high morbidity and 
mortality should they rupture. Permanent materials 
include, but are not limited to, coils, thrombin, glue, 
and stent grafts. Distal to proximal embolization 
should be performed to prevent recanalization of the 
aneurysm from retrograde flow through collaterals. 
If the aneurysm neck is amenable, packing with coils 
can be performed. Care must be taken not to rupture 
the aneurysm with this type of intervention, which 
is far more challenging and time-consuming than 
smsplv embolm ni; the supplying vessel. 

If both distal and proximal occlusion cannot be 
obtained because the aneurysm neck is too close to 
a vessel origin or distal occlusion would result in 
undesirable end-organ ischemia, a bare metal stent 
can be deployed across the neck of the aneurysm. A 
guidewire is then passed between the interstices of 
the stent and into the aneurysm sac. Selective coil 
embolizationofthe aneurysm can then be performed 
using a microcatheter and microcoils. Alternatively, 
a stent graft can be considered for this situation. 

Due to the small size and tortuosity of the vis- 
ceral vessels, stent graft placement is often prohibi- 
tive. In the appropriate situation, stent grafts offer 
the benefit of occlusion of the aneurysm neck while 
maintaining distal perfusion [2]. Although an ele- 
gant solution, the anatomy must be such that there 
is enough of a landing zone proximal and distal to 
obtain an effective seal on both ends of the graft to 
exclude the aneurysm. Further device development 
and experience are needed to perfect this therapy. 



Embolization of Visceral Arterial Aneurysms 



If the aneurysm is amenable to direct percutane- 
ous puncture, another opt ion includes direct throm- 
bin injection or coiling via an 18-gauge needle. 
Simultaneous balloon occlusion of the aneurysm 
neck via arterial access can be performed to prevent 
nontarget embolization. 



twice as often as females, and patients are typically 
in their fifties. Hepatobiliary and intraperitoneal 
rupture occur with equal propensity. These lesions 
tend to be solitary although they can be multiple 
in conditions such as polyarteritis nodosa [4,12]. 
Multiplicity is also seen with amphetamine abuse 
and trauma. 



8.6.2 
Causes 



8.6.1 
Incidence 

Hepatic artery aneurysms comprise 20% of all 
visceral aneurysms [4, 10-12]. Eighty percent are 
extrahepatic and 20% are intrahepatic. The majority 
affect the common hepatic artery. Males are affected 



Mycotic aneurysms from bacterial endocarditis were 
initially the most common cause of hepatic artery 
aneurysm [11]. However, traumatic and iatrogenic 
causes are likely the most common etiology today. 
Medial degeneration and atherosclerotic changes 
have also been implicated. However, atherosclerosis 
is more likely the result rather than the cause of the 



First and Second Line Tools for Emblization 



Celiac (Splenic, Hepatic, GDA, Pancreaticoduodenal): 

• First choice: 

- 6 F sheath in common femoral artery 

- 5 F Sos to engage the celiac artery 

- 3 F microcatheie: :::d pidewire coaxially 

- microcoils apo:op:ia;i:ly size.". [.:■ til tlie :a:gel he.'a;ic 

• Second choice: 

- 5 F Sos to select the celiac 

- exchange groin shea;h to: .'- Rahsin sheath or guiding 
catheter to maintain access re the celiac artery 

- Bentson or hydrophilic wire to gain peripheral access 
using a 5 F Cobra calhele: io.ni be 4 F hydrophilic) 

- Coaxial use of .'. microcatheter or cirecl embolization 
through Cobra 

SMA 

• First choice: 

- 6 F sheath in common femoral artery 

- 5 F Sos to engage the superior mesenteric artery 

- 3 F microcaiheie: a:'.d piidewire coaxially 

- microcoils approp: lately sized ;o fit the target 

• Second choice: 

- 5 F sheath in the left brachial artery 

- 5 F MPA to engage the SMA 

- 3 F microcaiheie: a:'.d piidewire coaxially 

- microcoils approp: lately sized ;o fit the target 

IMA 

• First choice: 

- S F sheath 

- 5 F RIM catheter 



- 3 F microcaiheie: and p::deu : ire coaxially 

- microcoils appropi ;ilr'v sized ;o fit the target 

• Second choice: 

- 5 F sheath :n the let; orach ial artery 

- 5 F MPA or vertebral artery catheter (for the length) 
to engage the IMA 

- 3 F microcaiheie: and pi:deu : ire coaxially 

- microcoils appropi ;,\w',y sized ;o fit the target 

Renal 

• First choice: 

- 6 F sheath in common It:" oral artery 

- 5 F Sos to engage the renal artery 

- 3 F microcaiheie: and pi;deu : ire coaxially 

- microcoils appropriately si/.ed to :';l ;lie iarget renal 

• Second choice: 

- 5 F Cobra to select the renal artery 

- exchange groin shealh foi a Balk j:: sheath or guiding 
catheter to maintain access lo die renal artery over a 

- obtain peripher.il access with iheSFCobra 
(can be 4F hydrophilic) 

- Coaxial use of a microcaiheier or direct embolization 
through Cobra 

• Other options: 

- 500-700 or 700-900 micron 

PVA/embo spheres/Contour SE in vascular beds wltli 
good collateral flow 

- Stent-graft where amv.emy is favorable 

- Direct puncture with an 1 S-g needle foi coil, thrombin, 
or glue injection 



lan and M. D. Darcy 



aneurysm. Inflammatory processes and vasculitis 
also cause hepatic artery aneurysms. Polyarteritis 
nodosa, systemic lupus erythematosus, Takayasu's 
arteritis, and Wegener's granulomatosis have all been 
implicated in published case reports [11-13]. Congen- 
ital arteriopathies such as Marfan syndrome, Ehlers- 
Danlos syndrome, and hereditary hemorrhagic telan- 
giectasia can lead to aneurysm formation [14]. 



8.6.3 

Risks Posed by the Aneurysm 






ent with 
melena, 
present: 



Intrahepatic rupture can result in hemobili 
without biliary obstruction. These patients can pres- 
ight upper quadrant pain, jaundice, fever, 
hematemesis.Extrahepatic rupture usually 
:utely and can lead toexsanguinationdueto 
itraperitoneal hemorrhage. They can erode 
^structures such as the stomach, common 
bile duct, duodenum, or portal vein. Due to the high 
mortality from rupture, elective treatment of small 
asymptomatic aneurysms has been advocated. 



8.6.4 
Management 



iitfoadjai 



five angiography is performed. Portal vein patency 
should be confirmed prior to occluding any vessel 
supplying the hepatic parenchyma. Approach from 
the left brachial artery may be required if the celiac 
axis cannot be catheterized due to an unfavorable 
angle off the aorta. Furthermore, if the origin of the 
celiac is occluded, retrograde catheterization of the 
GDA can be attempted from the SMA. Hydrophilic 
or microcatheters are helpful to navigate tortuos- 
ity. Once securely positioned within the origin of 
the common or proper hepatic artery, the micro- 
catheter can be used to perform selective angiog- 
raphy and embolization. Distal to proximal coil 
deposition should be performed for small intra- 
hepatic aneurysms in peripheral vessels. In cases 
of multiple traumatic pseudoaneurysms, Gelfoam 
embolization can be used to temporarily tampon- 
ade bleeding. 

Treatment of saccular aneurysms that arise from 
the common, proper, or extrahepatic right and left 
hepatic arteries require more planning. Selective 
coil or percutaneous thrombin injection are options 
that can allow for continued perfusion distal to the 
aneurysm. Tortuosity and small caliber may pre- 
clude stent graft placement. The GDA can be sacri- 
ficed if absolutely necessary due to collateral flow 
from the SMA. 



8.6.4.1 
Anatomic/Physiologic Con: 

The liver 



i "dual" blood supply fromboth the 
■s and the portal vein. Although good 
collateral flow exists, necrosis can occur if enough 
arterial supply is occluded at the time of emboliza- 
tion. Therefore, it is wise to assess the direction of 
blood flow in the portal vein because hepatofugal 
flow may increase the risk of infarction. One should 
be cautious and superselective when embolizing in 
the presence of portal vein thrombosis. 

There have been reports of recanalization of 
pseudoaneurysms after percutaneous embolization. 
Follow-up ultrasound, CT, or MRI can be performed 
to assess success of embolization [4,15]. One must be 
aware of the relatively frequent anatomic variation 
seen in the hepatic artery distribution. 



8.6.4.3 

Results of Embolization 

Review of the relatively limited literature reveals 
that embolization with coils, Gelfoam, detachable 
balloons, or glue and placement of stent grafts 
is technically successful in 95%-100% of cases 



[6,10,16-22]. Failures were typically d 
when patients tended to rebleed. In 
follow-up imaging was performed, a s 
were found to recanalize [19,22]. In s 
recanalization, success] ul thrombosis 
percutaneously with glue using the co 
in the series by Parildar et al. [22]. 



8.6.4.4 
Complications 



i early 



8.6.4.2 
Techniqui 



:ally, the celia 
■r Cobra cathet 



axis is selected with 
■from the groin and n 



Complications of the embolization procedure 
include those of diagnostic angiography with the 
addition of aneurysm rupture, nontarget emboli- 
zation, ischemia or infarction, abscess formation, 
and rarely sepsis. In earlier literature, spontaneous 



Embolization of Vis! 



rupture occurred in as many as 44% of cases [23]. 
It is likely that this rate is much lower today due 
to earlier discovery with frequent and improved 
imaging. However, mortality associated with rup- 
ture is still nearly 35% [24]. Although small case 
numbers, rupture during manipulation has not been 
reported in the current literature [25-27]. In 1986, 
Uflacker reported successful treatment of 11 cases 
of visceral artery aneurysms by coiling feeding ves- 
sels both dista]]y and proximally, thus reducing the 
risk of rupturing the fragile pseudoaneurysm wall 
by directly coiling the sac [28]. 



8.7.1 
Incidence 



8.7.3 
Management 

8.7.3.1 

Anatomic/Physiologic Considerations 

The splenic artery arises from the celiac axis and 
is often tortuous. Therefore, glide wires and hydro- 
philic catheters are helpful in gaining peripheral 
access to this vessel. It has a long course from the 
aorta to the splenic hilum, making it one of the most 
amenable arteries for stent graft placement. It sup- 
plies branches to the body and tail of the par 
If necessary, complete occlusion of the main sple 
artery distal and proximal to the aneurysm m 
canbe performed. If the artery is completely thro 
bosed, collaterals can be parasitized resulting i 
splenic remnant or even hypertrophy of splenu 
after an embolization. 



Considered the most common, the splenic artery 
aneurysm has been reported to comprise approx- 
imately 60% of all VAAs [1,2]. This entity affects 
females four times as often as males. Typically seen 
in multiparous women, this aneurysm has a high 
propensity to rupture in the third trimester of preg- 
nancy [29]. Asymptomatic aneurysms can often be 
seen as round calcified masses in the left upper quad- 
rant on plain films and computed tomography. This 
type of aneurysm has been associated with portal 
hypertension. Many causes exist and include pan- 
creatitis, portal hypertension, endocarditis, cystic 
medial necrosis, iatrogenic, and collagen vascular 
diseases such as Ehlers-Danlos. 



8.7.2 

Risks Posed by the Aneurysm 

Life-threatening rupture, which commonly occurs 
in the third trimester of pregnancy, is a serious risk 
of splenic aneurysms. The small 2-3 cm asymp- 
tomatic lesions typically pose no immediate threat 
and can be observed with serial CT. There is some 
debate regarding the size of aneurysm that can be 
observed. However, patients who develop left upper 
quadrant or abdominal pain with no other identifi- 
able source would likely benefit from elective embo- 
lization even if in the 2-3 cm range. Although not 
defined, rapid interval growth should also be an 
impetus to embolize because the morbidity from 
rupture is significant. 



8.7.3.2 
Technique 

Typically a groin approach is used and the celiac 
axis is selected with a Sos catheter. Selective angi- 
ography is performed to lay out the splenic artery. 
A glidewire is then passed distally and either the 
Sos or a Cobra catheter is advanced. Embolization 
can be performed through the 5 French catheter at 
this point. If too tortuous, then a microcatheter can 
be passed coaxially. (Fig. 8.4) We use either a Mass 
Transit® (Cordis, Miami, FL) or Renegade® (Boston 
Scientific, Boston, MA) microcatheter. These cath- 
eters can withstand a power injection of 2-3 cc per 
second if needed. 

Coil embolization is technically easy and suc- 
cessful. Coils should be sized slightly larger than the 
vessel lumen. Smaller coils or Gelfoam can be used 
to sandwich or "nest" between the flanking coils. 
This will ensure a compact and occlusive embolus. 
Patients should be observed for splenic infarction, 
although short gastric or other small collateral ves- 
sels can perfuse portions of the spleen distal to the 
occluded main renal artery. 

A long guiding catheter can be used to secure 
access or to upsize from the standard 6F short 
sheath used in the common femoral artery, should a 
stent graft be chosen. The splenic artery is probably 
the most amenable to stent graft insertion due to its 
long length and relative lack of branch vessels feed- 
ing other organs. Although tortuous, the vessel will 
often straighten out when a wire is passed distally. 



n and M. D. Darcy 




r "i 




Fig.8.4. a Nonselective cej.ic imgiogiam m a patient with a large 
splenic pseud oaneurvsiv. ■liter trauma. Noie the marked vasospasm 
and contrast pooling i:i the lei'i upper quadrant, h Coaxial use of a 
micro catheter to obtain access distal to the neck of the pseud oan- 
eurysm despite the vasospasm, c h\ I ;::»-■ :p splenic angiogram after 
coil embolization of the splenic artery. No further contrast extrava- 
sation was noted and the patient's vitals stabilized. (Courtesy of 
James R.Duncan, MD) 



A good landing zone of at least 1 to 2 cm is required 
on either side of the aneurysm neck to obtain a 
seal. Self- expanding stent grafts such as the Via- 
bahn (Gore Inc., Flagstaff, AZ) are readily available, 
but are far too large for use in the visceral vessels. 
Smaller coronary stent grafts such as the balloon 
mounted Jostent'- 1 (Ahoott L;.i hoi-atones. Inc., Abbott 
Park, Illinois) have been used, but are not readily 
available or approved for this indication. Although 
delivery is more precise, balloon-mounted stents 
are less flexible. Flexibility is desirable for passing 
catheters through tortuous vessels. Currently, there 
is little published on stent graft use for VAAs, and 
more investigation i: 



8.7.3.2 

Results of Embolizatioi 



TV. Iin 



cess is high, approaching 100%. On the 
that the celiac axis is occluded or too 
stenotic, access to the splenic artery can be obtained 
by retrograde cannulation of the gastroduodenal 
artery from the SMA. The GDA is often hypertro- 
phied if the celiac has been chronically occluded. In 
this case, getting coils through multiple turns may 
not be possible. However, if the catheter tip is in a 
secure enough position, particles such as PVA or a 
Gelfoam slurry can be used to stop acute bleeding 
prior to taking the patient to the operating room. 



Embolization of Visceral Arterial Aneurysms 



8.7.3.3 
Complications 

Complications of the embolization procedure 
include those of diagnostic angiography" with the 
addition of aneurysm rupture, nontarget emboli- 
zation, splenic infarction, abscess formation, and 
rarely sepsis (Fig. 8.5}. Total splenic infarction can 
occur, which puts the patient at an increased risk 
of infection with encapsulated bacteria such as 
pneumococcus. Older literature suggests that bland 
splenic artery aneurysms rupture at a rate of approx- 
imately 2% [30]. However, in pregnant patients, rup- 
ture occurs in nearly every case with mortality rates 
for mother and fetus 70% and 95% respectively [31]. 
Obviously, any aneurysm in the pregnant female 
should be addressed since over 95% will rupture if 
left untreated [30,32]. 

After a retrospective review of ten years of expe- 
rience, Carr et al. found that 42% of their cases of 
VAAs presented with rupture [33]. Half of all the 
VAAs were splenic .irtery aneurysms. Of the splenic 
artery aneurysms that were observed, 33% went on 
to rupture. This is much higher than the previously 
reported 2% rupture rate and reflects the fact that 



the patients in the series by Carr et al. suffered 
from associated conditions such as hypersplenism, 
pancreatitis, abscess, and PAN. The overall mor- 
tality rate after rupture was 25% despite surgical 
intervention [33]. This would suggest that interven- 
tion should be pursued in cases due to associated 
conditions since a high rupture rate exists during 
observation. Incidental aneurysms can be watched. 
Again, no case reports of rupture during manipula- 
tion were encountered in the current literature. 



Incidence 



The superior mesenteric artery aneurysm is the third 
most common VAA but only accounts for 6% of all 
splanchnic aneurysms. It is typically associated with 
an infectious etiology such as endocarditis. Throm- 
bosis and dissection can be seen with these lesions 
and patients can present with symptoms of mesen- 




patienl fro::: !-':gure 4 ..". c ["!'.■."■ i"i ^ I :",i i - 

i:'.g ,i perisplenic abscess. The pa tic::: J eve loped fever and an 
elevated white cell count one week after embolization. Note 
arterial perfusion c:' a sp>::ic remnant medial to the abscess. 
b iIT image inferior to that o:' a demonstrating the coils n. 
the splenic hilum. Collateral Sow to the spleen despite com- 
plete occlusion of the splenic a: tery exisis. t Inferior aspect 
of I he abscess distinguishing kitinev from the residual spleen, 
i Courtesy of Jaines R. Duncan, MP i 



n and M. D. Darcy 



teric ischemia or intestinal angina. Aneurysms and 
pseudoaneurysms of the remaining mesenteric ves- 
sels are rare and present in descending frequency: 
celiac, gastroduodenal (GDA) (Fig. 8.6}, gastric, 



gastroepiploic, and pancreaticoduodenal. There is 
no significant gender predilection for mesenteric 
aneurysms, and the age range typically spans the 
sixth and seventh decades. 





Fig. 8.6. a Nonselective angiogram of the celiac axis, o'emonslraiing 
a pseudoiinenrysm of the l.F'A. b I'eiayed image ih.il shows persis- 
ieni coairast willun the pseudoLineurysm sac. t Follow-up subtracted 
image alhei coil cmooj'zaiion wi;h a niicroca;he : .er. No furthet hhing 
of the pser.doaneuiysni from ihe GHA is noted, d selective nonsub- 
tiacteti injection of a duodenal branch ciemonslrates rodateinl tilling 
of :he pseud OL\neu:ysm thai would lead to fr.r.hci henioi ih.ige. e Final 
subtracted .mage alter embolization oi ihe col.a;eral vesse! with PVA. 
Thrombosis of the feeding vvssels has been achieved 



Embolization of Visceral Arterial Aneurysms 



8.8.2 
Causes 

Generally, SMA aneurysms are mycotic, celiac 
aneurysms develop from cystic medial degenera- 
tion, GDA pseudoaneurysms occur in the presence 
of duodenal ulceration, and gastroepiploic and 
pancreaticoduodenal aneurysms arise secondary 
to inflammatory changes from pancreatitis. Other 
causes include polyarteritis nodosa, amphi 
abuse, and connective tissue disorders. 



8.8.3 

Risks Posed by the Aneurysm 



Ischemia from proximal thrombosis c 
lization and rupture are significant i 
these types of VAAs. Local infectic 
hematomas and generalized sepsis c; 
sequent bowel resection maybe requii 
from thrombosis or embolization is si 
cularization is not possible. 



distal embo 

1 of adjacent 
i occur. Sub- 
id if ischemia 



8.8.4 
Management 

8.8.4.1 

Anatomic/Physiologic Considerations 

Due to good collateral flow, complete occlusion of 
splenic, peripheral hepatic, and gastroduodenal 



arteries is well tolerated. Ischemia, stricture, and 
infarction of the bowel can result from emboliza- 
tion in the peripheral SMA and IMA distributions. 
Because of the rich arcade around the pancreatic 
head, the GDA can easily reconstitute the celiac axis 
via retrograde flow from the SMA. This is beneficial 
for preventing ischemia, but can allow persistent 
perfusion of aneurysms if both distal and proximal 
control is not achieved at the time of embolization. 
Often, pseudoaneurysm formation in the pancre- 
aticoduodenal, SMA, and IMA distributions is due 
to adjacent inflammatory processes such as pan- 
creatitis or diverticulitis. (Fig. 8.7) Hematomas in 
the mesentery can become abscesses. Even though 
microcoils are used in these instances, it is unwise to 
implant a stent graft into a potentially infected bed. 



8.8.4.2 
Technique 

Technique will vary significantly depending on the 
vascular distribution of the mesenteric aneurysm. 
Typically, celiac and proximal SMA aneurysms are 
best treated surgically. 

Although a common femoral approach is prefera- 
ble to that of the brachial artery, a lelt upper extrem- 
ity puncture may be required to negotiate the acute 
angles the visceral arteries take from the aorta. A 
Sos catheter works well from the groin, and an MPA 
is typically all one needs from the arm. Guiding 
catheters or sheaths offer support and directional- 
ity if access is difficult to maintain. 




Fig. 8.7. .i -r,::ivr .m ■;.■■.■>-" '' mi .i-r\v.- c--i.::v.\::c .■ p;iriido:i:":ei:rv>:v. of [.;; ■= i g m ■.:■■: o Lv:i;:cli of I lie !M.A dr.e k- -^ :vej : i .r , I i i i i - : . h :'ol«- 
'.::'' siibt:;icled .ii:.itie driiio:^ I idling cod occlusion o: both Lin; itches tli.ii were siipplv.iio. die ..cue psendo.ii'.eiirv-i-ii. No- isch.e:r.::] 
resulted due to coll.'.ter.i. rkuv from inter n.d ijjc LT.'.ndies vi.i I lie heniorrhoid.il .rrteries. (Courtesy of lean iter E. Gould, MD] 



lan and M. D. Darcy 



As long as there is no celiac, proper hepatic, or 
SMA origin occlusion, the GDA can be sacrificed. 
If the GDA is required to maintain perfusion of the 
liver or, if flow into the S.MA is dependent upon the 
celiac, then direct coiling of GDA pseudoaneurysms 
is preferable. This can be accomplished with stent 
placement over the aneurysm neck and microcoil 
deposition through the interstices via a microcathe- 
ter. A small-caliber stent graft such as a fostent could 
theoretically be used in this situation. However, use 
of this device is still not approved by the FDA. Appel 
et al. described the placement of a 26-mm stent graft 
for humanitarian treatment of ;i trail ma tic pseud oa- 
neurysm of the SMA [34]. 

The RIM catheter was specifically designed for 
the inferior mesenteric artery. This catheter typi- 
cally seats well in the origin, which arises at the 
level of the left pedicle of L3. A microcatheter can 
then be passed coaxially into the desired branch 
and microcoils deposited. Supers elective technique 
is desired. Although some collateral flow is supplied 
to the distal colon via internal iliac branches, care 
must be taken when occluding more proximal vas- 
cular territories. 



8.8.4.3 

Results of Embolization 

Results of transcatheter embolization of mesenteric 
aneurysms appear favorable in the literature, and 
technical success has been reported to range from 
75% to 100% [1,3,15,18,19,24-27,33,35-42]. However, 
many of these studies are not only retrospective and 
small, but the mesenteric VAAs are often lumped in 
with splenic and hepatic artery aneurysms, making 
it difficult to isolate effective treatment rates for 
SMA, IMA, and GDA aneurysms independently. The 
anatomy and location of the lesion will often dic- 
tate the success of embolization. Furthermore, some 
case series report the use of different methods, such 
as percutaneous thrombin or coil injection versus 
transcatheter embolization. 



thromboembolic phei 
manipulation. Smaller emboli not seen during t. 
embolization procedure may become evident a fi 
hours later and manifest as abdominal pain. Tr 
typically resolves with heparinization, pain ma 
agement, bowel rest, and time. If hemorrhage occu 
in the mesentery, abscesses can develop and perc 
taneous drainage may be required. 



8.9.1 
Incidence 

Incidence rates of aneurysm formation differ for 
the various etiologies of renal artery aneurysms. 
However, the literature suggests that incidence rates 
range between 0.015% and 9.7% [43], A classifica- 
tion scheme divided into saccular, fusiform, dis- 
secting, and pseudoaneurysms has been described 
by Poutasse [44,45]. The natural history of these 
aneurysms is not well defined in the literature. How- 
ever, there are small and large case series that dem- 
onstrate low rupture rates ranging from 0% to 14% 
[46-50]. Hubert et al. followed some patients with 
solitary aneurysms as large as 4.0 cm for as long as 
17 years without rupture [50]. Renal artery aneu- 
rysms that are treated surgically are approached by 
nephrectomy, ex vivo repair, and auto-transplanta- 



8.9.2 
Causes 

Typically pseudoaneurysm formation in the renal 
artery distribution Is iatrogenic or traumatic. Other 
causes of aneurysm formation include fibromuscu- 
lar dysplasia, polyarteritis nodosa, amphet 
abuse, angiomyolipoma in the presence or ab: 
of tuberous sclerosis, and neurofibromatosis. 



8.8.4.4 
Complications 

The major complicating factor of embolization 
in the mesenteric distribution is bowel ischemia 
and infarction. Ischemia can cause strictures and 
obstruction whereas infarction can lead to perfo- 
ration and sepsis from dead gut. Dissection and 



Risks Posed by the Aneurysm 



Rupture is 
pregnant v 
with splen 



rare risk of these aneurysms. However, 
nen are more prone to rupture just as 
artery aneurysms. Schorn et al. pro- 
ew of the literature regarding the 



Embolization of Visceral Arterial Aneurysms 



risk of rupture of these lesions [51]. They found one 
dated series that described the risk of rupture as high 
as 14%. However, many subsequent large autopsy 
series demonstrated no instances of rupture when 
renal artery aneurysms were present. In their review 
of the literature, they also discovered that noncalci- 
fied aneurysms were more at risk of rupture. 

In cases of FMD, distal embolization and dissec- 
tion can be seen. Malignant hypertension may be 
the presenting symptom as a result of embolization 
of clot from the aneurysm or occlusion from a dis- 
section flap. 



8.9.4 
Management 

8.9.4.1 

Anatomic/Physiologic Considerations 

The vascular supply to the kidney is considered end- 
organ, and infarction is common after emboliza- 
tion. Therefore, in patients with renal insufficiency 
or underlying diseases such as tuberous sclerosis 
or von Recklinghausen's disease, nephron-sparing 
procedures are vital. Superselective embolization is 
advisable in all cases of renal artery embolization 
unless partial or total nephrectomy is planned. 



8.9.4.2 
Technique 

The renal arteries arise at the L2 level from the 
abdominal aorta. A 10° LAO view during a flush 
injection of the aorta will often provide the best 
view of the origins. Careful examination for acces- 
sory renal arteries is necessary. A Sos or Cobra 
catheter will easily select the main renal ostium. A 
Rosen wire is an atraumatic guidewire that allows 
for secure exchange or upsizing to a guiding cath- 
eter or Ealkin sheath. Selective injection should be 
performed to identify the feeding vessel or vessels. 
This can be done through the sheath. 

Many embolization techniques can be used in 
this setting depending on the type, number, and 
location of the aneurysms. For example, aneurysms 
of the main renal artery may be amenable to stent 
graft placement, thus allowing distal perfusion to 
be maintained [52-56]. However, until stent graft 
placement is perfected, surgical repair by resection, 
aneurysmorraphy, ;ind aulotrfmsphntation is more 
inly performed in this setting. 



More peripheral aneurysms can be selectively coil 
embolized using microcatheter techniques. Both 
distal and proximal control is not always possible, 
and may not be necessary due to the vascular anat- 
omy. In the presence of multiple aneurysms from a 
lesion such as an angiomyolipoma, a combination 
of particle and coil embolization can be performed. 
If actively bleeding, the aneurysms are coiled first, 
followed by occlusion of the feeding vessels to the 
tumor with PVA or embospheres. Gelfoam can be 
sandwiched between coil nests to assist thrombo- 

[( surgica] resection is planned, ablation of entire 
lobar or main renal arteries can be performed with 
ethanol. This requires the use of a balloon occlusion 
catheter such as a single lumen Balloon Wedge-Pres- 
sure Catheter (Arrow International, Reading, PA) to 
prevent systemic spread of alcohol. Ethanol ablation 
should be performed within one or two days of the 
planned resection. This will help to avoid a pro- 
longed post-embolization syndrome, which can be 
quite uncomfortable for patients and can reduce the 
risk of abscess formation from the ensuing infarc- 



8.9.4.3 

Results of Embolization 

The technical success is quite high once the renal 
artery is securely accessed. If superselective tech- 
niques are used, very little ischemia results and 
there is very little chance of inducing renal failure. 
The small number of reported cases in the literature 
makes it difficult to assess the long-term s 
tiansauheter embolization. 



8.9.4.4 
Complications 

Although rare, dissection or perforation of the renal 
arteries and their branches can occur. Rupture may 
lead to rapid development of retroperitoneal hem- 
orrhage. Both dissection flaps and rupture can be 
immediately controlled with balloon tamponade. 
Although dissection flaps can often be tacked down, 
rupture typically requires emergent surgery. Even 
perforation of smaller branch vessels can occur if 
a guidewire is passed too far into the periphery. 
Hematoma and abscess can develop. 

Embolization of plaque from heavily calci- 
fied arteries or aorta is a risk when manipulating 



lan and M. D. Darcy 



sheaths or catheters in the origins of these vessels. 
Even though richly vascular, the renal parenchyma 
is prone to ischemia and infarction due Co its end- 
organ supply. Every attempt should be made to 
become as selective as possible Co preserve distal 
flow and prevent nontargeC embolization. 

Care must be Caken when embolizing wich abso- 
lute eChanoldue Co the vigorous thrombosis it causes 
by denaturing proteins. Echanol can cause seizures 
and inCoxication if ic reaches Che systemic circula- 
tion. It can also permeaCe Che tissues and cause injury 
lo ad i .ice nt structures such as bowel and nerves. 



Complications 

Access site complications include hematoma, pseu- 
doaneurysm formation, and arterial dissection. 
Although sometimes unavoidable, good technique 
should keep these complications at an acceptable 
level. Brachial punctures are slightly more prone to 
complicaCion, especially neural compression from 
hemaComa. Ic should also be remembered ChaC work- 
ing from the left arm entails a catheCer crossing Che 
origin of Che left verCebral artery, and a sheath will 
be nearly occlusive in the brachial arCery. If pos- 
sible, using a smaller sheath such as a 4 or 5 French 
system can help diminish post-procedure complica- 
tions. Microcatheters will work coaxially through 
4 French caCheters. 

Placement ol a vascular sheath serves two impor- 
tant purposes. It protects the access artery from 
injury if multiple catheter exchanges occur, and it 
maintains access to Che arCery should the working 
catheter become occluded. 

Aneurysm rupture during vigorous contrast 
mieifion or direct manipulation can occur. Emer- 
gent tamponade can be achieved by inflating a bal- 
loon either across the neck of Che aneurysm or in the 
origin of the feeding vessel. Having an appropriately 
sized balloon occlusion catheCer ready on Che back 
table can be useful should rupture occur during the 
procedure. Emergent surgery may be necessary if 
the bleeding cannoC be controlled or if permanent 
embolization of the parent vessel is not technically 
feasible. The ruptured aneurysm may continue to 
bleed if distal occlusion is not achieved. 

One of the feared complications of deploying 
coils, injecCing Chrombin or glue, or infusing par- 
ticulaCe embolics is nontarget embolization. SCrin- 
genC Cechnique Co ensure satisfactory positioning 



of catheters is vital to prevent nonCarget emboliza- 
Cion. Before embolizing, catheCer stability should 
be CesCed by passing a wire through it. Catheters are 
more apC Co buckle out of vessel origins and cause 
nonCargeC embolization or loss of access in the pres- 
ence of extremely torCuous courses. Should a coil 
become lodged in Che caCheCer, a forceful injecCion 
wiCh a 1- or 3-cc syringe can be aCCempCed Co torpedo 
it free. However, this maneuver may back the cathe- 
Cer out of the CargeC vessel. If nonCargeC embolizaCion 
of a coil occurs, a snare can be used in an attempt Co 
reCrieve ic if it lodges in an undesirable location. 

Sinceminiscule amount soi thrombin are required 
Co thrombose aneurysms, great care should be Caken 
when injecting this embolic. Distal embolization 
during thrombin injection usually resolves without 
significant sequelae. Although not always possible 
with VAAs, the use of real-time ultrasound can help 
Co moniCor Che progress of thrombosis. Obtain- 
ing arCerial access provides the ability Co balloon 
occlude the origin of the aneurysm when injecting 
Che aneurysm percutaneously. 



Future Development and Research 

Coil embolization has been one of the mainstays 
of VAA treatment and has been augmented wich 
direct glue or thrombin injection. Coil design has 
not changed sigmhauifly. However, extremely pre- 
cise deployment mechanisms such as electrolytic 
detachment are available today. 

Many different agents, both temporary and per- 
manent (Gelfoam, glue, thrombin, PVA, detachable 
balloons, and eChanol)canbe used depending on the 
situation and the desired end result. There has been 
no significant change ot these materials in the las! 
fewyears. Refinement of PVA into embolic "spheres" 
has been achieved, and is thought to provide a more 
uniform embolization rather than the clumping that 
can occur with regular PVA. A newer nonadhesive 
liquid embolic agent called Onyx (Micro Therapeu- 
tics. Inc., Irvine, CA) has been used in some neu- 
roinCervenCional and neurosurgery applications. 
Because Onyx is nonadhesive, care must be taken 
when using this product dueCoChepropensiCy of the 
embolic to migrate into Che parenC vessel. 

The fuCure may lie wiCh sCenC graft placement, 
which has been performed with good Cechnical suc- 
cess inChelasC four years, albeit in small case reporCs 
[16, 34, 52, 54-67]. However, no long-Cerm daCa exisC, 



Embolization of Visceral Arterial Aneurysms 



and further investigation is warranted. The capacity 
to occlude aneurysm necks while maintaining distal 
perfusion in a single step would seem to make stent 
grafts the ideal therapy. However, small vessel diam- 
eter, short landing zones, frequent bifurcations, and 
tortuosity provide ample hurdles for placement of 
stent grafts in the visceral arteries. Furthermore, 
the paucity of these cases makes it difficult to collect 
statistically significant data and gain experience in a 
short period of time. Therefore, there is little to drive 
the market to further develop this technology. 



8.12 
Conclusion 

Although quite rare, visceral artery aneurysms are 
life-threatening when hemorrhage occurs. Aggres- 
sive management by the interventional radiologist 
is paramount. Often, these patients are too sick for 
major revascularization procedures, making endo- 
vascular techniques a more desirable approach. Sur- 
gical morbidity and mortality is fairly high after 
aneurysm rupture. 

The asymptomatic lesions are being discovered 
more regularly due to the increased demand for 
crow-sectional imaging. The timing ol treatment 
depends onaneurysm size and location. For example, 
some authors have suggested treating asymptomatic 
splenic artery aneurysms while others have argued 
that observation is adequate [60, 68, 69]. However 
observation is the exception rather than the rule 
with visceral artery aneurysms. All splenic artery 
aneurysms in women of child-bearing age should be 
treated due to the high incidence of rupture in the 
third trimester. All mesenteric aneurysms should 
be treated due to a high likelihood of complications 
from rupture, thrombosis, or embolization. 

A variety of methods for embolization of VAAs 
are described in the literature. The type of treat- 
ment should be tailored to each individual case as 
the anatomy will typically dictate the therapy best 
suited. 

Despite the fact that minimally invasive embo- 
lization procedures have been performed for over 
twenty years, very few long-term data on VAA 
occlusion are available. Further investigation and 
development of newer techniques such as stent graft 
placement are needed. Coil, glue, thrombin, and 
particle embolization will continue to be effective 
methods for treatment ot visceral artery aneurysms 
in both the elective and emergent settings. Good 



technique and a firm understanding of the visceral 
vascular supply are vital, just as in any embolization 



i .Lookstein RA, Gullet [ (2004) Embolization of complex 

vascular lesions. Mi Sinai [ Med 71:17-28 
2. Larson RA, Solomon J, Carpenter JP (2002) Stent graft 

refill!' of visceral artery aneurysms. 1 Vase Surg, 3i>:1 JoO- 

1263 
a.Me'issano ij, Chiesa K i 1 99S) Success nil surgical treatmeni 

or" visceral artery aneurysms. After fa; aire •:■' a e [cutaneous 

treatment. Tex Heart Inst J 25:75-78 
4. Abbas MA ei al. iaSOai Hepaiic artery aneurysm: factors 

that predict complications, ] Vase Sarg . ! S:4I -45 

5. Shanley CJ, Shah NL.Messina LM (1996) Common splanch- 
nic artery aneurysms: sp.enic, riepatic.and celiac. Ann Vase 
Surg 10:315-322 

:::.] 'cshmukh H el al. !J004i Trans catheter embolization as 
primary treatment for viscera, pseudoaneiirysms at aan- 
..-'■-■•-■• -i ;,i;.- a on t.--. nte a :td imaging I'oi.ow up. Indian ■ 
Gastroenterol 23:56-58 

7.(irr JA e; al. (2000) Visceral pseudoaneurysras due to 
docysts: rare but lethal complications of 
pancreatitis. | Vase Surg 32:722-730 

B-MLTlen t | el aL (2004) Prevention of contrast-induced 
iu ph- p..'a* u :h sodium bicarbonate: a randomized con- 
trolled triiL. JAMA 291:2328-2334 

9.Shlansky-Goldberg R, Cope C (2001) A new twist on the 
Waltman loo:'' for uterine finroid embolization. J Vase 
I nter v R ad iol 1 2 :99 7- 1 000 

D.Saleum PF et a!. i:99.~i Hepatic artery aneurysm: an ever 
present danger. J Cardiovasc Surg (Torino) 36:595-599 

l.LalRBetal. (1989! Hepatic artery aneurysm. J Cardiovasc 
Surg (Torino) 30:509-51 3 

2. 0'DriscollD.OIliffSP.OlliffJF (1999) Hepatic artery aneu- 
rysm. Br] Radiol 72:1018-1025 

3. Chan R] et al. (1998) Segmental mediolytic arteriopathy of 
the splenic ,m.:. hepatic arteries mimicking systemic nec- 
roiizing vasculitis. Arthritis Si'. cum. 4! :9a 5 -93 5 

4,Erskine )M (1973! Hepatic artery aneurysm. Vase Surg 
7:106-125 

it. Messina LM, Shanley C) i 1 997) Viscera; artery anearysms. 
Surg Clin North Am 77:425-442 

6. Vent ur mi M el a 1. 1.1001 ', Hemorrhage from a right hepatic 
artery pseudoaneinysm: end ova sou la r treatment with a 
coronary stent-graft. ) Endovasc Ther 9:221-224 

7.ThomasDE,LeonLM(1998l Hepatic artery aneurysm rup- 
ture: case repor:, imag.ng findings, and .iteianire review. S 
D I Med 51:413-416 

B.Stambo GW. Hallisey M], Gallagher J) Jr (1996) Arterio- 
graphii" embolization of viscera! artery pseu do anearysms. 
Ann Vase Surg 10:476-430 

9.Sakm TA et aL (1992) Nonoperative management of 
visceral aneurysms and aseudoanei.rysms. Am ] Surg 
164:215-219 

0. Rokke O et ;;!. i 199:-; Hepatic artery aneurysm. ! diagnosis 
and treatment. Tidsskr Nor Laegeforen 1 16:487-489 

l.Reber PU et al. (1998) Life- threatening upper gastroin- 



n and M. D. Darcy 



testina! tract oleeding caused oy ruptured extrahepatic 
pseudoaneurysm after pancreatoduodenectomy. Surgery 
124:114-115 

22.Parildar M, Oran I.MemisA (2003) Embolization of vis- 
cera! psc i.i .:! i::-;i :ie'.: r y ^ ::: :; with niaunuiv. coils and X-n.itvi 
cyanoaci ylate. Abdom imaging 23:36-40 

23.Stanley ]C, Thompson NW, Fry WJ (1970] Splanchnic 
artery aneurysms. Arch Surg 101:689-697 

24. Busuttil RW, Brin BJ ( 1 980] The diagnosis and management 
of visceral artery aneurysms. Surgery 88:619-624 

25. Hossain A et a I. (201)1 I Viscer.il artery anearyims: experi- 
ence in a tertiary-care center. Am Surg 67:432-437 

26. Kasirainn K ei a 1. 12001 1 Endovascular management of vis- 
ceral artery aneurysm. ) Endovasc Ther 8:150-155 

27.Rokke O ei al. (1997! Successful management of eleven 
splanchnic artery aneurysms. Eur I Surg 163:411-417 

28.Uflacker R (1986) Transcatheter embolisation of arterial 
aneurysms. Br I Radiol 59:317-324 

29. Angela!-: i: El et al. :. 1 '■■''.' i Sp.en.ic artery aneurvsni nip: are 
during pregnancy. Obstet Gynecol Surv 48:145-148 

30. Stanley JC, Fry WJ (1974) Pathogenesis and clinical signifi- 
cance of splenic artery aneurysms. Surgery 76:898-909 

31. Barrett JM, Caldwell BH (1981) Association of portal 
hypertension .in.: ruptured splenic artery aneurysm in 
pregnancy. Obstet Gynecol 57:255-257 

32. Macfarlane|R,ThorbiarnarsonB( 1966) Rupture of splenic 
ar;ery aneurysm during pregnancy Am I Obstet Gynecol 
95:1025-1037 

33.Carr SC et al. (2001) Visceral artery aneurysm rupture. ] 
Vase Surg 33:806-811 

34.Appel N, Duncan JR, Schuerer DJ (2003] Percutaneous 
stent -gran treatment of superior mesenteric am: internal 
iliac artery pseudoaneurysms. ■ Vase Interv Raaiol 1 4:917- 
922 

35.Araoz PA, Andrews JC (2000) Direct percutaneous embo- 
li zanon of visceral artery aneurysms: techniques and pit- 
falls. [Vase Interv Radiol 11:1195-1200 

36.Carr SC et al. 1 i 996! Current management of visceral artery 
aneurysms. Surgery 120:627-633; discussion 633-634 

37.Gabelmann A, Gorich ], Merkle EM (2002) Endovascular 
treatment of visceral artery aneurysms. I Endovasc Ther 
9:38-47 

38.Lauschke H et al. (2002) Visceral artery aneurysms. 
Zentralbl Chir 127:538-542 

39. Masoia: iello S ei al. i l iKI 7! Aneurysms of tile splanchnic 
arteries. Minerva Chir 52:45-52 

40. \':: ■■::.: . r ei .1. i J 002 : Manageme-;'.: ofvi-oei al arteiv a net: - 
rysms. Retrospective study of 23 cases.Ann Chir 127:281- 
288 

41.Panayiotopoulos YP, Assadourian R, Taylor PR (1996) 
Aneurysms of the visceral and renal arteries. Ann R Coll 
Surg Engl 78:412-419 

42. Smith JA, Macleish DG, Collier NA (1989) Aneurysms of 
the visceral arteries. Aust N Z I Surg 59:329-334 

43. Martin RS 3rd et a I. :. i 98''i Ken:'., artery aneurvsni: selective 
treatment for hypertension and prevention of rupture. I 
Vase Surg 9:26-34 

44. Poutasse EF i 1 966) Renal artery aneurysms: iheir natural 
history and surgery. J Urol 95:297-306 

45. Poutasse EF (1975) Renal artery aneurysms. J Urol 113:443- 
449 

4'.'. Hennksson C et a I. i i^Sol Natural history of renal artery 



aneurysm elucidated ov repeated angiography and palho- 

anatomical studies. Eur Urol 11:244-248 
47. Hageman IH et ah i 1 978) Aneurysms of the renal artery: 

problems of prognosis and surgical management. Surgerv 

84:563-572 
48. Harrow BR, Sloane IA (1959! Aneurvsni of renal artery: 

report of five cases. J Urol 81:35-41 

49. McCarron JP Jr. Marshall VF, Whitsell JC 2nd (1975] Indica- 
tions for surgery on renal artery aneurysms. I Urol 1 14:177- 

50. Hubert JP Jr, Pairolero PC, Kazmier FJ (1980) Solitary renal 

artery aneurvsni. Surgery 88:557-5oo 

Sl.Schorn B et al. 11997; Kidney salvage in a case of ruptured 
renal artery aneurvsm: case report .in.: literature review 1 . 
Cardiovasc Surg 5:134-136 

52. Bruce M, Kuan YM (2002] Endoluminal stent-graft repair 
of a renal artery aneurvsm. j Endovasc Ther 9:359-362 

53.Liguori G et al. (2002 .: Perot: i a nee- .is management of renal 
artery aneurysm with a stent-graft. I Urol 167:2518-2519 

54. Pershad A, Heuser R 12004! Renal artery aneurysm: suc- 
cessful exclusion with a sleni graft. Catheter Cardiovasc 
Interv 61:314-316 

55.Rundback JH et al. (2000) Percutaneous stent-graft man- 
agement of renal artery aneurysms. 1 Vase interv Radio] 
11:1189-1193 

56. Schneidereit NP et al. (2003) Endovascular repair of a rup- 
tured renal artery aneurysm, j Endovasc Ther 10:71-74 

57. Marx M eta 1. 12002 i Treatment of a splenic artery aneurvsni 
with use of a stent-graft. J Vase Interv Radiol 13:1282 

58. Brountzos EN et al. (20"?! Pancreatitis-associated splenic 
artery pseud oanetirysm: endovascular treatment with self- 
expandable stent-grafis. Cardiovasc Imerveiit Radio! 2o:88- 
91 

59. Henry M et al. 12000! Percutaneous endovascular treat- 

menl of peripheral aneurysms. 1 Cardiovasc S::rg (Torino! 

41:871-883 
60.Arepally A et al. (2002) Treatment of splenic artery aneu- 
rysm with use of a stent-graft. 1 Vase interv Radiol 1 3:631 - 

633 
61. Atkins BZ, Ryan JM, Gray JL (2003] Treatment of a celiac 

artery aneurvsm with endovascular slem grafting - a case 

report. Vase Endovasc Surg 37:367-373 
62.AtarEetal.(2004! Percuianeotis treatment of a celiac artery 

aneurysm as.ng a stem graft, isr Med Assoc ) 6:370-371 
63. Millonig C, el al. (2004! Percutaneous management of a 

hepatic artery aneurysm: bleeding after liver iransplanta- 

tion. Cardiovasc Intervent Radiol 27:525-528 
64.Paci E eta I. (2000! -'seiideaneurvsm of the common hepatic 

artery: treatment with a stent-grafi. Cardiovasc Intervent 

Radiol 23:472-474 
65.Rocek M et al. (2002] Percutaneous treatme 

mesenteric artery pseudoaneurysm using a 

Am J Roentgenol 178:1459-1461 
66.Seriki DM et al. (2004) Endovascular stent j 

■of pseudoaneurysm of the super;. :■ 

diovasc Intervent Radiol 27:271-273 
67.YoonHKetal. (2001) Stent-graft repair of a splenic artery 

aneurysm. Cardiovasc Intervent Radiol 24:200-203 
6S.Trastek VF et al. i 19S2; Splenic artery aneurysms. Surgery 

91:694-699 
69.DaveSPetal. (2000) Splenic artery aneurysm in the 1990s. 

Ann Vase Surg 14:223-229 



;nt-graft.AJR 



artery Car- 



Venous Ablation 



Endovenous Thermal Ablation of Incompetent 
Truncal Veins in Patients with Superficial Venous 
Insufficiency 



Neil M. Khii 



[iilToduotion 119 

Pathophysiology mid Epidemiology 119 

Anatomy 119 

EVTA: Background 120 

Evaluation Prior to EVTA 121 

EVTA Technique 121 

Clinical Data 12 3 

Summary 125 

References 125 



Introduction 

Endovenous thermal ablation has become an 
accepted option to eliminate the reflux caused by 
incompetent saphenous veins. In this chapter, a 
review of the clinical problems and anatomy pre- 
cedes a review of this exciting new venous occlusion 
technique. 



Pathophysiology and Epidemiology 



of patients with all forms of CV1 have significant 
superficial venous insufficiency (SVI}. If present, 
treatment of CVI begins with elimination of this 
reflux. Reflux in the saphenous (truncal) veins is the 
most common cause of venous hypertension. Patho- 
physiologically significant reflux in the great saphe- 
nous vein (GSV) or in one of its primary tributaries 
is present in 70%-80% of patients with CVI. Small 
saphenous vein (SSV) reflux is found in 10%-20% 
and non-saphenous superficial reflux is identified 
in 10%-15% of patients [2, 3]. 

SVI is certainly the most prevalent medical con- 
dition treated by interventional radiologists. Up 
to 25% of women and 10% of men in the U.S. are 
affected, with 50% of people >50 years old having 
some form of SVI [3]. Most patients with SVI have 
symptoms, which include aching, fatigue, throb- 
bing, heaviness, and night cramps. A minority of 
patients develop skin injury from chronic venous 
hypertension, which includes eczema, edema, pig- 
mentation, lipodermatosclerosis, and ulceration. 
Heredity is the primary risk factor for developing 
SVI; 85% of patients are affected if both parents are 
involved, 47% if one parent is involved, and 20% if 
neither parent is involved [4]. Prolonged standing 
and multiparity increase the risk of expressing this 
heritable risk. 



Lower-extremity chronic venous insufficiency (CVI) 
is caused by venous hypertension [1]. Most patients 9.3 
develop venous hypertension from the hydrostatic Anatomy 
forces produced by reflux that results from primary 
valvular insufficiency [2] Venous obstruction, mus- 
cular pump failure, and congenital anomalies are 
much less common causes. In addition, 85%-90% 



N. M. Khilnani, MD; R. J. Mln, MD 

Cornel! Vasail-ar. Weill Median! College of Cornell Uiiiversiiy, 
416 East 55th Street, New York, NY 10022, USA 



The superficial ven< 
ties is composed of inn 
lecting veins, the saphe 
taries. The GSV begins 
portion of the foot, run 
leolus, and ascends the 
thigh to ultimately join the femoral 
le (saphe note moral junction, SFJ) 



system of the lower extremi- 
lumerable subcutaneous col- 
nous trunks and their tribu- 
; on the anterior and medial 
s anterior to the med ial mal- 
aspect of the calf and 
at the fossa 



meters below the inguinal ligament (Fig. 9.1). The 
GSV is adjacent to the saphenous nerve (sensory) 



N.M.Khilnani and R.J.Min 




Fig.9.1. Frontal and posterior diagrams 
of the lower extremi-y demonslriuing 
the great and small saphenous veins and 
their named tributaries. The two saphe- 
nous systems can be connected via the 
vein of Giacomini 



from about 6 cm below the knee to the ankle. The 
GSV and its major named branches run superficial 
to the deep and deep to the superficial fascia within 
the saphenous space. The GSV has two important 
and named tributaries above and below the knee: 
the anterior and posterior circumflex veins of the 
calf and thigh. In addition there are three smaller 
tributaries at the groin which are important in that 
they are often a source of recurrent varicose veins 
following their surgical ligation along with the GSV 
(high ligation). A common variant, the anterior 
accessory GSV (AAGSV), runs more laterally than 
the GSV within the saphenous space and often is 
usly termed a duplication of this trunk. This 
s frequently present and can be responsible for 
>n the anterior aspect of the thigh. 
An extrafascial tributary vein that communicates 
with the GSV but runs parallel to the GSV course 
and is relatively straight should be described as a 
superficial accessory GSV. 

The SSV begins on the lateral aspect of the foot, 
ascends posterior to the lateral malleolus and then 
up the midline of the calf, between the same fascial 
is the GSV. The SSV runs adjacent to the 
rve (sensory) from just below the popliteal 
crease to the foot. In about two thirds of cases, the 
SSV drains into the popliteal vein at or just above 
the popliteal crease. In about one third of cases it 
has a cephalad extension with or without a saphe- 
nopopliteal junction (SPJ) to ultimately drain into 



planes 



a posterior thigh perforating vein, ot 
terior circumflex vein of the thigh v 
Giacomini (Fig. 9.1). 



EVTA: Background 

Treatment of SVI is indicated for symptoms unre- 
lieved by conservative methods such as graduated 
compression stockings (GCS), exercise, and avoid- 
ing prolonged standing. It is also indicated for com- 
plications from chronic venous hypertension such 
as bleeding varices, superficial thrombophlebitis, 
and skin injury. Treatment begins with elimina- 
tion of any truncal incompetence. The treatment 
modalities available to accomplish this include high 
ligation, truncal vein stripping along with a high 
ligation, endovenous thermal ablation (EVTA), and 
duplex ultrasound (DUS)-guided sclerotherapv. 

In the last 5 years, EVTA has developed into a 
successful option for obliterating truncal incom- 
petence. Its main advantages over surgery are that 
there is no need for anesthesia or sedation and no 
recovery or down time following the procedure. The 
underlying mechanism of this procedure is to endo- 
vascularly deliver sufficient thermal energy to the 
wall of an incompetent vein segment to produce irre- 
versible occlusion. The first modern report of EVTA 



Endogenous Thermo] .-VlM.iiion .:■!" [iKcmpeiea/ 7nin.ro! Veins :n f';'.l ient:; wjrli y.-.-.'crn.-j.;l Venous Insufficiency 



was made using laser-delivered energy to ablate the 
saphenofemoral junction [6]. Since that time several 
devices have been approved by the U.S. Food and 
Drug Administration. The currently available tools 
utilize radiofrequency or laser energy of a variety of 
different wavelengths to deliver the required ther- 
mal dose. 

As mentioned, the goal of EVTA is to endove- 
nously deliver sufficient thermal energy to the wall 
of an incompetent vein to irreversibly occlude it. A 
catheter inserted into the venous system either by 
percutaneous access or by open venotomy delivers 
the thermal energy. The procedure canbe performed 
on an ambulatory basis with local anesthetic and 
generally require little or no sedation. The patients 
■ally fully ambulatory following treatment 

cose tributary and reticular 
is are treated separate!)' with 
such as compression sclero- 



and the 
The : 

veins and telangiectasi 
adjunctive therapi 



therapy or microphlebectomy. Some physicians will 
perform microphlebectomy for varicose veins at 
the same time as EVTA. Other physicians elect to 
perform phlebectomy or compression sclerotherapy 
of the varicose veins at a later time. Almost all phy- 
sicians will defer therapy of spider veins to a later 

In our practice, phlebectomy is generally recom- 
mended at the same time as EVTA when varicose 
tributaries are larger than 8-10 mm in diameter or 
when EVTA will occlude their inflow and outflow. 
If such veins are left untreated they may thrombose 
in 596-1096 of cases and become painful, erythem- 
atous, and possibly result in skin pigmentation. 
Also, if they thrombose, their complete eradication 
may be made more difficult and certainly will be 
delayed. For smaller varicose veins and all reticular 
and spider veins, we offer compression sclerother- 
apy beginning 4 weeks after EVTA. By this point 
the veins have substantially decompressed, making 
their eradication with injections easier. 



9.5 

Evaluation Prior to EVTA 

Treatment for patients with CVI begins with a care- 
ful history and directed physical exam. All patients 
with visible varicose veins or symptoms suggesting 
venous insufficiency should be evaluated with DUS 
[7, 8]. In patients with spider veins near the medial 
ankle (corona phlebectasia) or along the medial 



calf or thigh, a DUS of the GSV is recommended 
to identify truncal reflux. The goal of the DUS is to 
determine which veins are normal and which veins 
are incompetent to create a map of the pathways of 
reflux in a given patient. Such a map is necessary to 
define the best combination of treatments which are 
available. In some cases, it may be possible to iden- 
tify the abnormal vein segments by physical exam. 
However, the different pathways of incompetence 
overlap sufficiently such that reliance on physical 
examination alone will lead to frequent diagnostic 

Reflux in truncal veins must be treated prior to 
addressing any visible abnormalities. EVTA is a 
treatment option for endovenously eliminating such 
reflux. The indications for ablation of incompetent 
truncal veins are identical to those for surgical liga- 
tion and stripping. Absolute contraindications have 
yet to be identified for the laser procedure. For RF 
ablation, a pacemaker or implantable defibrillator 
is ;.i contraindication. Relative contraindications lor 
EVTA include absent pedal pulses limiting GCS use, 
liver abnormalities limiting local anesthesia use, 
pregnancy, nursing, and uncorrectable coagulopa- 
thies. 



EVTA Technique 

Once the pathways of incompetence are established, 
EVTA can be utilised to ireat incompetent truncal 
veins and straight segments of their tributary veins. 
In almost all cases venous access is directly into the 
vein to be treated or directly into one of its principle 
tributaries, if the tributary vein is straight. Gener- 
ally the access is at or just below the lowest level of 
reflux in the treated truncal vein as defined by DUS. 
This is generally recognized as the level where the 
diameter of the truncal vein decreases just periph- 
eral to a large incompetent tributary. In many cases, 
segmental great saphenous vein incompetence can 
occur in two separate portions of this vein. The 
reflux can completely spill out into a tributary vein 
only to re-enter the GSV at a lower level. It is impor- 
tant to determine if the intervening portion of the 
GSV is hypoplastic or normal. Hypoplastic segments 
occur commonly [9] and cannot be traversed by a 
guidewire. In this circumstance, venous access will 
be required into the most peripheral part of both 
segments and the segments are subsequently ablated 
sequentially. Normal intervening segments can be 



N.M.Khilmmi and R.J. Min 



crossed and allow the operator to treat all incom- 
petent segments of the GSV through one venous 

Prior to treatment, the treated veins are mapped 
and the courses of the treated segments as well as 
important landmarks such as junctions, venous 
aneurysms and large perforator inflows are drawn 
on the patient's skin with a surgical marker. The 
patient is then placed horizontal on the table allow- 
ing full access to the treated segments. In general, 
patients being treated for GSV reflux are placed 
supine. Patients being treated for reflux in the SSV 
are placed prone with their feet hanging off the end 
of the table to relax the calf. Posterior medial tribu- 
tary and Giacomini vein ablations may require more 
challenging positions. 

Venous access is accomplished with either a 19- or 
21-G needle using real-time US guidance and a one- 
wall technique. The incompetent truncal vein can 
become much smaller when the patient lies down. 
Placing the patients in the reverse Trendelenburg 
position and keeping the procedure room warm can 
dilate the vein to make access easier. Also, when 
the puncture is directed into a tributary vein or the 
AAGSV, care must be taken to avoid venospasm, 
which is much more common with missed punc- 
tures of these veins. 

The details of the laser and RF treatments vary at 
this point but the ultimate treatment goals and tech- 
niques are similar. With RF ablation, the RF cath- 
eter is withdrawn only after its tines are exposed 
to allow contact with the vein wall. The catheter is 
withdrawn maintaining the vein wall temperature 
at or above 85°C as measured by a thermocouple 
embedded in its tip. For laser ablation the fiber is 
withdrawn at a rate determined by the energy depo- 
sition per length of vein treated. For the remainder 
of this presentation on technique, the laser proce- 
dure will be discussed. The details of the RF venous 
ablation can be reviewed in the references cited in 
the results section. 

For laser EVTA, a 5F sheath is inserted through 
the entire abnormal segment and into a more central 
vein. A bare tipped laser fiber is inserted to the end 
of the sheath, which is then withdrawn exposing the 
tip of the fiber. The sheath and fiber are then with- 
drawn to place the tip at the staring point of the abla- 
tion. For the GSV this is usually about 1 cm below 
the SFJ and for the SSV about 2 cm below the SPJ 
where the SSV turns parallel to the skin just below 
the popliteal fossa (Fig. 9.2). With the laser, confir- 
mation of the position can be made with localization 
of the light which comes from the red aiming beam 



which can almost always be visualized through the 
skin. 

The most time consuming part of the procedure 
is the US-guided delivery of perivenous tumescent 
anesthesia (TA). TA is a form of local anesthesia 
delivery which was popularized by plastic surgeons 
which utilizes large volumes of dilute anesthetic 
solutions which are infiltrated to anesthetize large 
regions for treatment. TA can make EVTA pain- 
less obviating the risks and additional monitoring 
associated sedation or anesthesia. In fact, it can be 
argued that sedation adds risk to EVTA by blunt- 
ing the patients' response to pain making them 
potentially more susceptible to extravenous thermal 
injury as well as delaying immediate ambulation 
after the procedure. 

TA is also necessary for safety and efficacy ol 
the procedure as well. Large volumes are utilized to 
compress the truncal vein to maximize the transfer 
of energy to the surrounding vein wall. Even though 
venospasm frequently occurs soon after sheath inser- 
tion, it is usually still necessary to empty the vein 
further with TA to ensure adequate treatment. Also 
important is that the large volume of fluid around 
the vein is necessary to insulate the vein from the 
surrounding structures. This part of the procedure 




Fig. 9.2. Longitudinal view of the saphenofemoral junction 
(SFJ) during positioning of a Ltser fiber prior to EVTA. The 
left of the image is toward the padent's head. A thin arrow 
points to the tip pf the laser fi^er approximately 5-10 mm 
below die SF[ at die takeoff of the superficial epigastic vein. 
The (*) identifies the SFJ, FV the femoral vein, GSV the great 
viph.enous ven' ,;:'d J KG t!ir siipcrfkiol epigastric vein 



:i of [no;m.peic , ;i: ; run,",il Veins :n Pi'. I ient:; with x^'cinoj.;! VeiiOii; : ji-iiffi.riirii.w 



minimizes potential thermal injury to the skin or 
adjacent nerves or arteries. In practice a 1-cm-diam- 
eter cylinder of TA surrounding the treated vein and 

1 cm separation of the treated vein from the skin is 
adequate. 

For EVTA, we generally use 100-200 ml of a 0.1% 
lidocaine solution buffered with sodium bicarbon- 
ate. Utilizing these volumes we can get close to the 
4.5-mg lidocaine/!;!; dose without epinephrine and 
7-mg/kg doses with epinephrine. In plastic surgery 
these doses are routinely safely exceeded. The argu- 
ment for this safety is that the large volumes of fluid 
containing this drug are absorbed slowly avoid- 
ing high systemic levels. However, in an outpatient 
environment, it is best to avoid reaching these dose 
thresholds. In our practice, we also avoid the use 
of epinephrine to avoid any toxicity related to this 
drug. 

After placing the patient in a Trendelenburg posi- 
tion to further empty the vein of blood, the sheath 
and fiber are withdrawn as a unit through the treated 
vein segment as the laser is activated. With DUS, gas 
bubbles can be seen to emanate from the tip of the 
laser fiber which serves as additional confirmation 
of the tip position at the appropriate location. 

Suggested parameters vary slightly with the dif- 
ferent laser devices but are under the control of the 
operator. In our practice, using the 810-nm diode 
laser (Diomed, Andover, MA) 14-watt continuous 
mode is selected. The amount of energy necessary 
to effect reliable vein ablation seems to be an average 
of 80 J/cm throughout the treated segment [10]. The 
average pullback rate to accomplish this is about 

2 mm/s. In practice for the GSV we generally pull 
back at 1 mm/s for the first 10 cm or so f treatment 
since failures, if they occur, will happen at this loca- 
tion. We also pull back at this rate near the inflow 
of incompetent perforators or pudendal veins or the 
take off of large incompetent tributaries to maximize 
successful occlusion at these important locations. 
When treating the GSV or a superficial accessory 
saphenous vein when the vein is superficial, when 
treating the SSVorthe GSV below Boyd's perforator, 
we withdraw the fiber at 3 mm/s to minimize skin or 
nerve injuries. 

Following the procedure, patients should be 
placed into a Class II (30-40 mmHg) graduated 
compression stocking (GCS) usually for 2 weeks. 
The purpose of this is to keep the variceal tributar- 
ies as empty as possible in case they occlude to min- 
imize the amount of resultant thrombus. The GCS 
s the velocity of blood flow in the deep 
reasing the likelihood of deep vein throm- 



bosis. Anticoagulation with low-molecular-weight 
heparins is routinely used after EVTA in Europe but 
not in the US. 

After EVTA, most patients will develop an ecchy- 
mosis over the entire treated segment. This gener- 
ally develops the day after the procedure but fades 
by about 2-3 weeks. Most patients will comment 
about some mild discomfort over the treated vein 
which begins hours alter I he procedure but resolves 
within 24-48 h. The GCS helps minimize this dis- 
comfort; some patients use acetaminophen with 
good response. With laser ablation, patients will also 
develop a discomfort over the vein about 7-10 days 
after the procedure, which is generally described as 
being similar to a pulled muscle. This is most likely 
caused by transverse and longitudinal retraction of 
the vein as the acute inflammation transitions to 
cicatrization. This resolves with movement, occa- 
sional NSAID use and continued use of the GCS. No 
f unlit r treatment has bent n 



ryu 






Periodic follow-up DUS is suggested to monitor 
for the response to therapy. In general, at about 4 
weeks following EVTA, one will identify a smaller- 
diameter, thick-walled truncal vein likely the result 
of significant vein wall injury and its inflammatory 
response. Little or no lumen or intraluminal throm- 
bus is typically identified and no flow will be found 
in the treated segment. By 6-12 months the vein will 
continue to shrink in size so that in successfully 
treated cases the vein can no longer be visualized 
[11-13]. If the vein shrinks to this extent further 
follow-up is probably not necessary. 

Most patients will require adjunctive treatment of 
the branch varicosities. Compression sclerotherapy 
and micro-phlebectomy are the most commonly 
used techniques to accomplish this. Occasionally, 
deeper tributary veins may require DUS-guided 
sclerotherapy, and rarely, large variceal clusters will 
require conventional phlebectomy. 



Clinical Data 



The technical success of EVTA is defined as a pro- 
cedure with successful access, crossing the segment 
to be treated, delivery of tumescent anesthesia and 
delivery of thermal energy to the entire incompe- 
tent segment. Clinical success is defined as the per- 
manent occlusion of the treated vein segments and 
successful elimination of re!; 



N.M.Khihuni and R.J.Min 



an improvement in the clinical classification of the 
patients by at least one grade at a certain time inter- 
val after the procedure. As previously mentioned, 
in practice most patients will also be treated with 
either adjunctive micro-phlebectomy or compres- 
sion sclerotherapy and as a result the clinical success 
data for the different clinical reports can be con- 
founded by the success of the adjunctive procedures 
and by enthusiasm by which they are utilized by the 
treating physician. 

Duplex ultrasound is essential to document the 
permanent occlusion of truncal veins treated with 
EVTA [11-13]. A successful procedure will result in 
non- thrombotic circumferential vein wall injury 
from the highest level of the incompetent segment 
and through its treated course on early evaluation. 
On late follow-up the vein segment will ideally be 
obliterated and impossible to find or at least will 
be significantly smaller in cross section than prior 
to treatment and will have no flow throughout the 
treated segment. 

RF ablation was the first approved device for 
EVTA. Several single-center reviews have been pub- 
lished and are presented in Table 9.1. These stud- 
ies have consistently demonstrated a high degree 
of success in occluding the target vein. Complica- 
tion rates have been low, with paresthesias and skin 
burns being the most common and vexing problem 
with RF ablation. DVT is an uncommon problem 
in these series [19]. The imaging follow-up of these 
patients (median 25 months) has demonstrated 
truncal occlusion of the GSV in 90% with persistent 
patency without reflux of the SFJ in the overwhelm- 
ing majority of cases [12]. 

An industry-sponsored registry is being accu- 
mulated with published 2-year follow-up data [17]. 
The patients in this trial were selected to have GSV 
<12mm and without significant tortuosity. In this 
review 142 limbs were followed up at 2 years with 
DUS. Reflux was demonstrated absent in 90% with 
significant reductions in pain, fatigue and edema 
and 94.6% improvement in symptoms overall. There 



ilarization. Complications 
were minor, with DVT in 0.7%, PE in 0.3%, skin 
burns in 4.2% of the early cases and 0% after the use 
of TA, and paresthesias persistent at 2 years in 5.6% 
of patients (Table 9.1}. 

A single-center pilot randomized trial was per- 
formed early in the RF experience comparing pro- 
cedure related success and complications of ligation 
and stripping to RF EVTA of the GSV [20]. Fifteen 
patients were randomized to RF and 13 to surgery 
and all patients were followed for a mean of 50 days. 
The technical and clinical success and complica- 
tion rates were similar. The RF technique was per- 
formed without tumescent anesthesia and many of 
the described complication would likely have been 
avoided with its utilization. Using a visual ana- 
logue pain scale, clear advantages were noted for 
EVTA most marked from days 5-14 following treat- 
ment. Less analgesics and days off from work were 
required by the EVTA group. Health related quality 
of life assessments ultimately improved to a similar 
extent but the EVTA group reached maximum ben- 
efit earlier. 

Another small multi-center randomized trial 
comparing RF ablation to surgical stripping of the 
GSV with high ligation has been performed [21]. The 
data collected at a mean of 4-month follow-up and 
demonstrated that the recovery following RF abla- 
tion was shorter than following surgery with a sig- 
nificantly higher fraction of patients back to their 
usual level of activity at one day following the abla- 
tions. Of note was that the recovery was significantly 
quicker in those patients treated with RF ablation 
using only TA than in those treated with TA along 
will] any of he 1 1' torm of anesthesia. QOL ;is assessed 
using a standardized instrument was found to be 
significantly better immediately after RFA than 
after surgery, although by 4 months this difference 
was becoming significantly smaller. 

Endovenous laser ablation of the great saphenous 
vein, short saphenous vein and other saphenous- 
related trunks has been approved by the FDA. The 



Author 


Limb, 


hi .1 oiv- ■■.'." 


knvl'jdeii ve; 


j]/:\i: Ly '.'pea no reflux i 


! J ',:\is:he-Lis 


DVT 


Burns 


Ref# 


w„™ 


140 


™'r 2 


96% 




8%/l%@6mc 


is. 





14 


k iSTNt:. 


300 




97% 




N'R 


0.7% 


0.3% 


15 


GOLl'M AS 




6-24 mos 


68%/22% 










NR 


16 


Merchant 


232(5'] 2 mo 


!./ 12-24 1110 


s. 84%/6%@12 


mos., 85%/4%@24 mos. 


1 5%/6%@24 mc 


s. 1% 


2.1% 


17 


(VNUS registrv) 


142@24 mo 
















Wagner 


24 


3-12 mos 


21/21@3 mo; 


!. And 3/3 & 12 mos. 





1/24 





18 



is Thermal Ablation of [nccmpeie:'/ Trim.:;!! Veins :n f';'.l ients with v.ioerlicial Venous Insufficiency 



valuating the use of laser ablation of the GSV 



Limbs Follow-up Sue 



reported success rates reported in several single- 
center series laser ablation for GSV are presented in 
Table 9.2. In these series there were no restrictions 
to vein size or degree of tortuosity. These data have 
consistently shown successful nonthrombotic occlu- 
sion of the target truncal vein in >90% of cases with 
very rare recanalizations of previously occluded 
vein segments. Clinical improvement was noted in 
almost all of the cases with successful truncal vein 
occlusion. The incidence of DVT, paresthesias and 
skin burns was almost 0% in these series. Most 
patients have bruising over the region treated prob- 
ably related to the needle injections for tumescent 
anesthesia. Many also describe a pulling sensation 
about 1 week after the procedure which is thought 
to be secondary to the evolution of the inflamma- 
tory response from vein wall injury maturing into 
a cicatrization phase. Superficial phlebitis was 
reported in 5-12% of patients alter laser treatment 
noted primarily in cohorts of patients treated with 
delayed compression sclerotherapy rather than in 
those treated with immediate ambulatory phlebec- 
tomy of the larger tributary varicosities. 

Optimization ot the laser technique has prompted 
an evaluation of outcomes related to treatment 
parameters. Zimmett has suggested a lower rate of 
bruising and discomfort associate with continu- 
ous as opposed to pulsed laser energy delivery [27]. 
Timperman [10] has shown that the success of laser 
EVTA is unity when energy delivery is maintained 
on average as greater than 80 [/cm of vein treated. 
Kurt lie-]- optimization is likely to influence the out- 
come and side effects somewhat. However, given the 
high degree of success comparisons of large num- 
bers of patients will likely be necessary to establish 
any expected subtle difference. 

Although speculation exits regarding differences 
between laser and RF EVTA success and side effects, 
there is no significant published experience com- 
paring these two technologies. The important thing 
to recognize is that both of these technologies rep- 
resent exciting, minimally invasive, low-risk, quick- 
recovery options for patients with symptoms or 
complications ot superficial venous insufficiency. 



Re-canalization ot a treated vein presenting with in 
the first few months after EVTA likely results from 
insufficient thermal energy delivery. This is either 
because of excessively rapid pull back of the thermal 
device or inadequate TA resulting in poor transfer of 
thermal energy to the wall. Vein diameter probably 
has no bearing on the success assuming adequate TA 
is applied, regardless of whether RFA or laser is the 
source of this energy. Late recurrences can be related 
to re-canalization of a previously occluded vein but 
are more likely related to development of incompe- 
tence in previously untreated vein segments. 



Summary 

EVTA should be considered a scientifically accept- 
able option to eliminate truncal reflux. The pro- 
cedures can be performed without sedation in an 
ambulatory setting and are very effective, safe and 
associated with virtually immediate recovery. EVTA 
appears to be associated with a lower rate of recur- 
rent SVI due to a virtual absence of the high rate 
of groin neovascularity seen with high ligation and 
stripping of the GSV. EVTA procedures have already 
begun to supplant traditional surgery for truncal 
incompetence. 



References 

] . Nico hides AN', Huisein M!<, ?zendro G. 'Christopoulos 1 ', 
Vasdekis S, Clarke H (1993) The relation of venous ulcer- 
ation with ambulatory venous pressure iiie.isi;:e:v.enK 
JVasc Surgl7:414-419 

1. Labropoulos N, Peas K, Nicc.-.iudr:; AN. [.=■.■:": M. hL.miLiswami 
G,Vofteas N (1 996) The role of the distribution and anatomic 
extern of re:V.;x :n the development of s.ens aad sympicT.s 
in chronic venous insufficiency. ; Vase Surg 23:504-510. 

3.Labropoulos N '."linic.il correlation to various patterns of 
reflux. I Vase Surg 31:242-248 

4.Cormi-Thenard A (1994) J Dermatol Surg Oncol 20(5): 
318-326 



N.M. Khilnani and R.J.Min 



Muliane I \ ! 1 9:-2 i Varicose veins of pregnancy. Ani ! Obsiet 
Gynecol 63: 620-628 

Navarro L, Min R], Bone C (2001 ) Endovenous laser: a new 
minimally invasive method ot treatiiient for va noose veins- 
pi eii miliary observations using an 51 urn diode laser. Der- 
matol Surg 2 7(2):1 17-22 

Khilnani NM, Min Rl (2003) Duplex ultrasound for super- 
ficial venous insufficiency. Tech Vase Interv Ruciiolo:] 1 1- 
115 

Min RJ, Khilnani NM, Golia P (2003) Duplex ultrasound 
of lower extremity venous insufficiency. I V'.tso Interv 
Radioll4:1233-1241 

iati A, Mendoza E (2004) Eur J Vase Endovasc Surg 
28(3): 257-261 

Timperman PE, Sichlau M, Ryu RK (2004) Greater energy 
delivery improves treatment success of endovenous laser 
treatment o: incompetent saphenous veins. I Vase Inteiv 
Radiol 15:1061-1063 

Min RJ,KliilnaniNM, Gobi P (2003) Duplex u:1ra*mnd of 
lower extiemitv venous irisjllji:t*'ty. I Vise nitrv sadiul 
14:1233-1241 

Pichot O, Kabnick LS, Creton D. Mercahanl RP, Schullei 
Petroviae S, Chandler JG (2nn4) Duple* ultrasound scan 
I'lndnigs two years after j-jran' saphenous vein racliiifre 
quency obliteration. I Vas< ■•my .*"( I ,:IH9 |i;i 
Khilnani NM, Min RJ (200- : l-iKing of superficial venous 
insufficiency. Sem Interv K..;.i:il (;n press) 
Weiss RA, Weiss MA \1\ -'r ■! tc Mdiitrt; -tn.y 

encovenous occlusion u-.*'j; a ur-i;,-t radirlrc;. -tn.y 
catheter under duplex guui.vi.e '■: ehmira:e saphenous 
varicose vein reflux: A 2 yea: follow up. Der— iiol Su-j; 
23:38-42 

5. Kistner, (2003) Endovasc ar :ihhk p !2t:on nf the greater 
sapheii::'us vein: The Closure : ■■•..edjre I I'hlelvl '. 1 $..? 
333 

6. Goldman MP, Amiry S (2002) Closure of tic greater saphe 
nous vein with eno'oluminal raciiofiequency thermal heat- 
ing of the vein wall in combination with ambulatory pli lo- 
bectomy: 50 p.'.tienls with more than :'-:v.ontli follow- up. 
Dermatol Surg 28:29-31 



17. Merchant RF, DePalma RG, Kabnick LS (2002) Endovascu- 
Li :" obliteration oi saphenous reflux: a multicenter study, j 
Vase Surg 3(6):1 190-1196 

18. Wagner WH, Levin PM, Crossman DV, Lauterbach SR 
Cohen JL, Farber A.(2004) Early experience with radiofre- 
quency ablation, ot the greater saphenous vein. Ann V.isc 
Surg 18:42-47 

19. Merchant R )r., Kistner RL, Kabnick LS (2003) Is there an 
increased rislv lor 1">VT with Ihe VWIS procedure: I V'.tsc 
Surg38(3):628 

20.RautioT.Ohin-22n,PeralaI,OhtonenP,HeikkinenT,Wiik 
H, Karbalamen P, llaakipuro K.Juvonen T (2002) Endove- 
nojs i>h.i:« :.' is conventional stripping operation 

in (he treatment i:l' primary varicose veins: a random- 
ise lit comparison of rosis. I Vas..- Surg 
35:958 965 



l.Lurie R Cretin D. Kklof I 
O.SciuIler I'cjovrc S.Ses 

ized study of endovenous 
■e procedure) v 



Kabnik LS, Kistner RL, Pichot 
i C (2003) Prospective random- 
id iofrequency obliteration (c lo- 
tion and stripping in a selected 



patients population lliVOLVeS Study). | Vase Sing 33:207- 
214 

22. Min RJ, Khilnani N, Zimmet SE (2003) Endovenous laser 
treatment of saphenous vein reflux: long term results. I 
Vase Interv Radiol 14:991-996 

23. Proebstle TM, Gul D, Lehr HA, et al. (2003) Infrequent early 
recanulizaiion of GSV after endovenous laser treatment. I 
Vase Surg 38:511-516 

24.Sadiek NS, Wasser S (2004) Combine;; endovascular laser 
with ambulatory phlebectomy for the treatment of super- 
ficial venous incompetence: a 2-yea: perspective. I Cosmet 
Laser Ther 6(1): 44-49 

25. Todd K, Fronek H.Isaacs M, Mackay E.Pearson D () Endo- 
venous htse: treatment: a twelve month evaluation o\ 291 
put lents. I Vase interv k ao io Is upp lenient: 1^1 44 

26. Roisent.il M i I EVLT for Ihe incompetent greater and lesser 
saphenous veins; IV'IR supplemenl 521 I 

27. Zimmet SE, Min RJ (2003) Temperature changes in perive- 
nous tissue during endovenous laser tr 
model.J Vase Interv Radiol 14:911-915 



Embolotherapy Applications in Oncology 



10 Chemo-Embolization for Liver 



Christos S. Georgiades and Jean-Francois Gesi 



CONTENTS 

10.1 Introduction 129 

10.2 Clinical Considerations 130 

10.2.1 Patient Selection jn; Preparation 130 

10.2.2 Pathophysiology and Anatomical 
'Considerations 132 

10.3 Technique 132 
10.3.1 Procedure 132 
10. 3 .1 Recovery 135 

10.3.3 Follow-up 137 

10.4 Results 136 

10.5 Conclusion 139 



10.1 

Introduction 

Chemoembolizaticm is being used with increasing 
frequency in the treatment of solid hepatic tumors. 
Continued refinements of the technique and better 
designed studies have confirmed the survival bene- 
fit imparted bychemoembolization on patients with 
unresectable liver cancer, though still being consid- 
ered a palliative option. While no conclusive data 
exist as to a survival benefit for patients with hepatic 
tumors other than hepatocellular carcinoma (HCC), 
this procedure is rapidly gamins favor over other 
nonsurgical alternatives (see below). HCC is the 
most common solid, nonskin cancer in the world, 
with an especially high prevalence in Southeast 
Asia and Sub-Saharan Africa. The main predispos- 
ing factor in these geographic regions is Hepatitis B 
(the strongest risk factor associated with developing 
HCC), whereas in Europe and the USA the increasing 
incidence is attributable to a concomitant 



C. S. Georgiades MD, PhD 

Assistant Professor of xockobgy and y.irgei v. Johns Hopkins 
Medical Institutions, Blalock 545, 600 N. Wolfe Street, Balti- 
more, MD 21287, USA 
J.-F. Geschwind.MD 

Associate Pro] ess.: 1 : of ka^iologv, ?;::gerv aiv.i 0:- oology, lohr.s 
Hopkins Medical Institutions, Blalock 545, 600 N.Wolfe Street, 
Baltimore, MD 21287, USA 



in the incidence of Hepatitis C(three-fold from 1993 
to 1998) and alcoholic cirrhosis [1]. Epidemiologic 
analyses project increasing incidence in cirrhosis 
and HCC in the USA for the foreseeable future [2,3] 
and the trend is expected to eclipse the 70% increase 
in incidence of HCC over the last twenty years [1], 
Though less frequent, the incidence of cholangio- 
cellular carcinoma (CCC) in the USA is also on the 
increase with approximately 4,000 new cases per 
year. HCC and CCC comprise the overwhelming 
majority of primary malignant hepatic neoplasms. 
Secondary or metastatic hepatic neoplasms are espe- 
cial!)' common owing to the blood tilt rat ion In notion 
of the liver. With prolonged life expectancy, more 
people develop cancer and with improved cancer 
treatments patients live longer and thus have a 
greater chance of developing liver metastases. These 
factors have contributed to the ever-increasing inci- 
dence of patients with secondary hepatic neoplasms, 
especially in the Western World. Whether primary 
(HCC, CCC) or secondary however, hepatic malig- 
nancies carry a dismal prognosis with overall long- 
term survival percentage rates in the single digits. 
Both HCC and CCC are slow-growing tumors and 
even when the presence of risk factors initiates 
some sort of surveillance (cross- sectional imaging 
or tumor markers such as alpha-feto protein (AFP)) 
the majority of patients are unresectable at presen- 
tation. Of the 10%-20% of patients with HCC who 
are deemed resectable most will recur even with 
optimum treatment either due to residual tumor or 
metachronous lesions that eventually will develop in 
a cirrhotic liver. Liver involvement with metastatic 
disease from solid tumor imparts an equally dismal 
prognosis. A patient with resectable liver metasta- 
ses and controlled primary disease who can benefit 
from such aggressive treatment is the rare exception. 
Median survival for patients with unresectable liver 
cancer is disappointingly short irrespective of his- 
tology. Survival ranges from a short two months for 
patients with adenocarcinoma of unknown primary 
to a maximum median survival of 15 months for 
colon metastases. Carcinoid patients in particular, 



'._". S. Geoigiodes ond l.-H G esc hrdnd 



have very slow disease progression and despite the 
presence of liver metastases may survive for years. 
Survival for unresectable disease from hepatocellu- 
lar carcinoma, ehokuisioairdnoma, and metastases 
from pancreatic, breast cancer, and melanoma are 
between 4 and 9 months [3-9]. 

These disappointing results coupled with the 
poor response of HCC to traditional chemotherapy 
have provided the impetus for the development of a 
variety of nonsurgical techniques for the treatment 
of hepatic neoplasms (Table 10.1). Such techniques 
are generally divided into transarterial interven- 
tions versus percutaneous ones. The latter group is 
further subdivided into thermal ablation techniques 
versus chemical ablation techniques. 

Transarterial chemoembolization (TACE) has 
become the most popular locoregiona! technique 
for the treatment of unresectable HCC. Llovet etal. 
[10.] showed for the first time that TACE imparts a 
survival benefit to patients with unresectable HCC. 
It should be mentioned that some institutions do not 
routinely use chemotherapy during embolization 
for primary or secondary unresectable liver can- 
cers. Though data show such patients who receive 
TACE have a longer survival and/or better response 
than those receiving blunt transarterial emboliza- 
tion (TAE) only with kipiodol (Savage Laboratories, 
Melville, NY), Gelfoam (Pharmacia and Upjohn, 
Kalamazoo, Michigan) or particles (PVA by Boston 
Scientific or Embospheres by Biosphere Medical 
Corp., Boston, MA), the difference is not statistically 
significant [10-13]. The reason that the differences 
inpatient survival between chemoembolization and 
blunt embolization are not significant is probably 
the small number of patients in the relevant studies. 
Chemoembolization should reasonably be expected 
to effect greater response not only pathophysiologi- 
cally; studies (despite lack ot statistical significance) 
always show greater survival for patients treated 
with TACE versus TAE. 



Table 10.1. Nonsurgical 
Nonsui g ic.d 
Percutaneous 
Thermal ablation 



ns for unresectable hep.;l 
:-ioiiiii:'.g hematic neoplas 



10.2 

Clinical Considerations 

10.2.1 

Patient Selection and Preparation 

In addition to improved survival for patients who 
undergo TACE for unresectable disease, there have 
been reports of patients whose tumor has shrunk 
enough to permit resection or liver transplanta- 
tion and provide a chance for cure. Despite these 
benefits, TACE is considered a palliative treatment 
option. Surgical resection is the only procedure that 
can be performed with curative intention, therefore, 
TACE should be reserved for patients who are not 
surgical candidates. This is the only absolute con- 
traindication to TACE. Table 10.2 summarizes a list 
of relative contraindications. 

Prior to TACE all patients should undergo a 
gadolinium-enhanced [Omniscan, GE Healthcare, 
Princeton, NJ] MRI of the liver, preferably with per- 
fusion^ iff usion sequences (Fig. 10.1). CT, without 
and even with contrast cannot adequately delineate 
viable tumor and differentiate it from necrosis, scar, 
or inflammatory tissue. CT can follow the tumor 
response as far as size goes, but we believe this is 
inadequate, whereas enhancement on MRI perfu- 
sion imaging provides more accurate information 
on which part of the tumor is viable and which is 

Dual phase, contrast-enhanced MRI with perfu- 
sion sequence will not only delineate the extent and 
viability of tumor but also serve as a baseline study 
to plan future treatment. A simple dual-phase MRI 
or CT is acceptable as well but inferior to MRI per- 
fusion in quantifying viable tumor. In addition to 
information regarding tumor viability and morphol- 
ogy, cross-sectional provides information regarding 
the tumor's vascular supply and anatomy. For exam- 
ple, knowing the presence of portal vein thrombosis 

malignancies 

I nira- arterial 
lation TACE TARE 



m ;:":' 



RFA 



Cryo 



All of the cerci.it.;:' eons ieckn.iqces are Incited oy die size and number of the lesions i up to three lesions e.;cii measuring up :c 

■i cm i as wed as dieir loci; lion. Miochaphiag:r.atic lesions r.iav oe peiccianeecsly inaccessible, and lesions close to l. : :ge v.i-cula: 

structures respond ceo: ly to tliei ma I iiKade:'. techniques ; XrA, N'CT, Cryo. and LI PC). On ihe contrary, ' 

are not limited by the numner, size, or location of ihe lesions; rather by the hepatic function reserve 

TACE, trans-arteriai chemo-rniooiizaiion; TARE, irans-aicerkk rarho -embolization; MCT, microwave ci 

radio frequency ablation; L1PC. laser inteistida! photocoagulation; Cryo, cryo- an Lit ion; PEl, percuta: 

PAAI, percutaneous acetic acid inaction; PCI. perccianeecs chemotherapy injection 



arterial techniques 

is shown in Table 10.2. 
■guhidor. theracv; UFA. 
!ous ethanol injection; 



Chemo-Embolization for Liver 



Table 10.2. Contraindications 

Contraindications to TACE 
: . Borderline liver fundi on 
2. Total bilirubin > 4 mg/dl 
3.Albumin<2 

4. Encephalopathy 

5. Coagulopathy 

6. Poor general health 

7. Signifies] 
S.EIevaied 



Mitigating action 

1-3. Super selective TACE of :. hepanc segment may be considered 

4. TACE may be performed .f encephalopathy was minimal. 
Consider lactulose. 

5. Give FFP or platelets as indicated 
6,7. See below 
8. Mucormist p.o. and i.v. fluids if creatinine > 1.2 but < l.S. 

shu:'.:iiig 111 rough I he minor Avoic, "ACE [f Cr > 1.7 



Borderline liver function, eiscephaleprili'.y, .ind poor general heallh nre subjective factors, and liie complete clinical picuire 
T.ust be considered prior ;o deeding wheiher to proceed ivi:)i TAC1-1. Cl'.i.d-f'r.gh C .iver disease pjuenls show poor r espouse io 
TACE, and in general we avoid TACE in such patients. On the other hand, Child-Pugh A liver disease patients seem to obtain the 
T.axiiV.iiT. hei;ef: \ from TACE. exhioitmg :he longest - : n iv.vo I. Though we do use TACE for Child-Pugh K liver disease p.;t;eijis, 
their survival benefit is less tr.an ilia I of rl'.eir stage A counterparls.Toial bilirubin of -i mg/dl is c.irreniiv used ji our ins I it 1 .;; ion 
its Lie c.::-off. Wr have recemly .n creased tl"..s fro::i .1 ::ig,'d. without :;ny adverse effects. Uiipso.ished o : .\la jgjin f: e:v. our insti- 
:uti::ui have show: 1 . Mi a: port, 1 . 1 ve:n tliiomoosis Joes not increase ihe risl-: of complications, at leas: in Cl'..ld-A paiie:t:s. Hnally, 
.'.: :eiiove:'oiis sliunl .no ; li r" : i.: u :"i die i 1 1 n '.-."■ I ."an resu.i .:". :".■ ■::;.■ i gd emhu'ai.o:".. I'h.s .s ev ..." em on i.:e initial angiogram, and 
o lu nl Celfoam eniboliz.'.tion can eliminate :lie sriunoing and allow rlie pa:ien: to proceed wiih T AC 1-1. A: our institution arterial 
access is avoided if INR > 1.7 or APTT > 1.7 or platelet count < 50,000. 



and/or variant vascular anatomy may obviate the 
use of embospheres during treatment or reduce the 
procedure time and contrast load. Patients are pre- 
medicated depending on the tumor histology, renal 
function, and prior surgical and medical history. 
For example, patients with carcinoid will have to be 
premedicated with Sandostatin to prevent possible 
carcinoid crisis after TACE. At our institution we 
have thus far performed TACE in patients with car- 
cinoid metastases to the liver more than a few hun- 
dred times without witnessing any carcinoid crisis. 
Patients whose sphincter of Oddi function has 
been eliminated, i.e. hepatoiejii rtostomy, sphinc- 
terotomy patients or patients with percutaneous or 
internal biliary stents, are at high risk (> 50%) for 
developing a hepatic abscess after TACE. Stringent 
24-h bowel preparation and i.v. administration of 
broad- spectrum intravenous antibiotics prior to the 
procedure is recommended, but abscess formation 
is still likely. This is thought to be a result of coloni- 
zation of the biliary tree secondary to a resected or 
incompetent sphincter of Oddi. TACE causes isch- 
emia to the biliary tree which, unlike liver paren- 
chyma, is exclusively supplied by the peribiliary 
plexus via the hepatic artery. The necrotic tumor 
bed may become infected and abscessed, which can 
be particularly difficult to eradicate even with per- 
cutaneous drainage and i.v. antibiotics. This compli- 
cation should always be discussed with the patient, 
with anoverallriskof abscess formationof50%-70% 
mentioned. Laboratory values should be obtained 
prior to TACE, including: A comprehensive meta- 
bolic panel (NA, K, glucose, creatinine, BUN, total 




Fig. 10.1. Axial, venous phase, ga a o.inium -enhanced MR! 
of a patient with unresectable HOC showing a peripherally 
enhanced lesion huroii'sl in die medial segment of the left 
lobe. Solid, well demarcated lesions such as this ^nii respond 
better to TACE than do diffuse or multifocal lesions. Addition- 
ally, hyper vase u I ;ir tumors appear to respond better to TACE 
showing a higher degree of necrosis on follow-up MRI 



bilirubin, AST, ALT, alkaline rahephatase, albumin, 
total protein), hematology panel (hematocrit and/or 
hemoglobin, white blood cell count, platelet count, 
coagulation profile (INR, APTT) and tumor mark- 
ers (i.e. AFP for HCC, CEA for colon cancer). These 
values serve not only to ensure a safer procedure 
(i.e. normal coagulation) but also to allow for proper 
tollc.v-iip ol hepatic and renal I unction and n 
response (using tumor marker levels). Finally, s 
the procedure is performed under sedation, an 
NPO status is required. 



'._". 3. ije'.'igudes .in..: [.-!-'. Gesdiuiij.i 



10.2.2 

Pathophysiology and Anatomical 

Considerations 

Chemoembolmilion lakes advantage of the fact that, 
while normal liver parenchyma receives most of its 
blood supply (60%-80%) from the portal venous 
system, malignant hepatic tumors, whether pri- 
mary or metastatic, are nearly exclusively supplied 
by branches of the hepatic artery. Cancer angio- 
genesis (a.k.a. neovascularization) is a process by 
which neoplastic cells recruit new blood vessels in 
order to ensure adequate local oxygen tension. Due 
to their relatively higher metabolic demands cancer 
cells live in a nearly constant state of hypoxia. They 
respond by secreting chemntactic factors that pro- 
mote the formation of new blood vessels. Arterial 
epithelial cells are much more responsive to these 
factors, which explains why such malignant tumors 
are supplied by the hepatic artery. Chemoemboliza- 
tion then selectively targets the tumor while liver 
parenchyma is mostly (but not entirely) spared. The- 
oretically then, chemoembolization can be used to 
treat any solid malignant hepatic neoplasm whether 
primary or metastatic, solitary or multifocal, and 
irrespective of size. Of course exclusion criteria do 
exist and are described in detail below. Accessory 
and/or replaced right or left hepatic arteries are 
n (up to 20%-30%). Usually such variations 
sequential, but rarely they may render 
catheter-selection of the hepatic artery to be treated 
with the 5 Fr catheter difficult (see Technique sec- 
tion below). Even then a 3 Fr microcatheter is usu- 
ally a successful alternative. In addition, common 
origins ol the leit hepatic and left gaslric .'is '■veil m 
right hepatic and right gastric arteries maybe added 
complicating factors. Such associations are impor- 
tant insofar as they increase the risk of nontarget 
embolization, mainly the stomach, proximal small 
bowel, and/or pancreas. Though self limiting gastri- 
tis, duodenitis and pancreatitis have been reported, 
no deaths have yet been documented from nontarget 
embolization. Vigilance during the initial diagnostic 
arteriogram and intimate knowledge of the related 
vascular anatomy and its possible normal variations 
is a must in order to minimize the chances of com- 
plications. Recently, a few published reports have 
described extrahepatic supply of liver cancers. For 
example, intercostal or diaphragmatic arteries can 
be recruited by neoplasms. If repeated TACE fails to 
show the appropriate response or if a section of the 
tumor remains viable while the rest shows signifi- 
cant necrosis, search for such extrahepatic supply 



is indicated. Selective arteriograms of intercostal 
and diaphragmatic arteries or a 3-dimensional CT 
angiogram can determine whether indeed this is 
the case. Currently, portal vein thrombosis (PVT) 
(Fig. 10.2) is considered by most physicians as a con- 
traindication to TACE for fear of causing hepatic 
ischemia/infarction and possible decompensation. 
However, unpublished data from our institution 
confirm that for Child-Pugh A or B patients TACE 
is a safe intervention even in the presence of PVT. 
We had no mortality of significant morbidity for 
30 days post-procedure in these patients. In addi- 
tion we showed a survival advantage compared to 
historicalcontrols. 



10.3 
Technique 

10.3.1 
Procedure 

After a treatment plan is formulated based on image 
findings and the patient's clinical situation, informed 
consentis obtained. Informed consent should disclose 
the following risks: Injury to blood vessels and/or 
organs (which may require blood transfusion or sur- 
gery), anaphylactic reaction to contrast, worsening 
of renal function, infection, liver function worsening 
or liver failure and possibly death. Appropriate i.v. 
antibiotic prophylaxis is administered (cefoxetin 2 g 
i.v. once, at our institution) immediately pre-proce- 
dure. Preprocedure documentation of femoral, dor- 
salis pedis and posterior tibial pulses is mandatory in 
order to choose the appropriate access site (strongest 
pulse between right or left common femoral artery, 
CFA) and compare with the post-procedure exam for 
any changes. The patient is placed on the fluoroscopy 
table supine and both groins are prepared and draped 
in a sterile fashion. Sedation is provided according 
to the local nursing protocol (i.e. Versed and Fen- 
tanyl, supplemented as needed with Phenergan and/ 
or Benadryl). 

• Step 1-Obtaining vascular access. In most cases 
access using an 18 g, single-wall needle followed 
by an 0.035" guide-guide wire is successful. In dif- 
ficult cases a 0.018" micropuncture set can be of 
help, with or without the use of ultrasound guid- 

• Step2-Maintaining access. A 5-Fr short vascu- 
lar sheath providing access in the right or left 
(strongest pulse) CFA is used at our institution. 



Chemo-Embolization for Liver 




Fig. 10.2. Antero-posierior, riigii.il subtraction angiogram of 
the abdomen of" a patient with unreiiech.ible HO.] showing clot 
in portal veins. This arterial phase hepatic angiogram shows 
early arterial-port;'.! venous shinning through a hypervascu- 
lar tumor (white arrow). The shunting into the portal veins 
uncovered a ill ling defect (dor) in die main ! bhii:k onowlioiui) 
and left i whit? jiii>ir/i ( ':i,i) portal veins, embolization with 1 
cc of" Gelfoam slurry shut down the shunt while the tumor 
supplv was preseived. This allowed us to proceed with TACE. 
The presence of portal vein IhioinPosis should not be consid- 
ered by itself as a com::iii;dical.oii to TACE. "Unpublished data 
from our institution shows TACE to be a safe and effective 
procedure in Child-Pugh A and B patients 

A 4 Fr access set can be used in cases where less 
traumatic arterial access is needed (i.e. slightly 
abnormal coagulation profile, severe peripheral 
vascular disease), however smaller catheters may 
be a bit less controllable. 

• Step 3-Abdominul ;ioi Togi'am. (Optional). A flush 
aortogram via a multisideholed, pig-tailed cath- 
eter at the level of the celiac artery will delineate 
the vascular anatomy, tumor supply and provide 
a road-map for more selective access. For the most 
part this step am be skipped. In rare cases and after 
failing to easily select the SMA and celiac axis with 
a selective catheter (see step 4), which may suggest 
variant anatomy, one may revert to an abdominal 
aortogram. If performed, a 15 cc per second injec- 
tion for a total of 50 cc (15/50) is adequate. 

• Step4-Selective arteriograms. First, a 5-Fr glide 
catheter (Simmons-1 or Cobra glide-catheters, 
Terumo Medical, Somerset, NJ) is used to select 
the superior mesenteric artery (SMA) and per- 
form a prolonged angiogram that is carried well 
into the venous phase (Fig. 10.3). 



■ An injection rate of 6 cc per second for a total 
of 40 cc (6/40) is what we use. If the portal vein 
is proven patent on recent MRI, CT or previ- 
ous angiogram, the portal venous phase can be 
skipped, lowering the injection rate to 6 cc per 
second for a total of 20 cc (6/20). Then the celiac 
artery is selected (again Simmon's I or Cobra 
glide catheters) and a selective arteriogram is per- 
formed with an injection rate of 6 cc per second 
for a total of 20 cc (6/20). In most cases the celiac 
axis arteriogram will show the tumor blush to 
best advantage (Fig. 10.4). Prior to power-injec- 
tioii angiography using a mechanical injector, one 
should perform a gentle, short contrast injection 
by hand. This will serve many purposes: (1) it will 
ensu re the catheter is in the right position, (2) that 
its tip is not in a small side branch (inadvertent 
power-injection in a small branch may cause dis- 
section and/or thrombosis), and (3), it will give an 
idea on how fast (or slow) the flow is in the vessel 
of interest. The above-mentioned injection rates 
can be tailored to the flow observed within the 
vessel of interest. If for example the blood flow in 
the SMA appears to be very slow, instead of the 
usual 6/20 injection rate one can lower it to 5/15. 
Likewise, if the flow is fast or vessel caliber large 
the injection rate can be increased to 7/35. 




Fig. 10.3. Ante 
the abdomen 
phase shows ; 

') and left (Jong U 



or. digital subtraction angiogram o: 
.t with multifocal HCC in the venous 
ain (double arrows) as well as right 

row) portal veins. Though only a rela- 



tive contraindication, po:ta' vein thrombosis demands \ 
precautions to avoid complete cessation or" flow in the hepatic 
artery and to limit the use of embolization material (such its 
Gelfoam or particles] during TACE 



'._". 3. Geoigudes .in..: [.-!-'. GesdiHind 




Fig. 10.4. Antero-poslciior, digital subtraction angiogram i:'. 
the arterial phase during celiac artery contrast injection in 
a patient Willi HCC (same as rig 10. 1) shows an enhancing 
lesion (iicrpiri'i supplied by ijie let", brpatic ai ;ery. High tumor 
vascularity ("blush") is a marker for successful TACE 



■ Step 5-Selecting the finalcatheter position. A glide 
0.035" wire is advanced through the glide catheter 
followed by the catheter itself. Though one wants 
to be as selective as possible to avoid chemoembo- 
lizing normal liver, being too selective will result 
in parts of the tumor not being treated. In gen- 
eral, either the right or left main hepatic artery is 
the optimal position for the treatment catheter. In 
cases where there is tumor in both lobes, the one 
showing more tumor blush during the diagnostic 
celiac (or SMA) arteriogram should be targeted. If 
the 5 Fr glide catheter cannot be advanced to the 
desired location because of unfavorable anatomy, 
a 3 Fr microcatheter (Renegade, Boston Scientific) 
over an 0.018" guide-wire (i.e. Transend, Boston 
Scientific) can be used coaxially. If one is target- 
ing a peripheral solitary lesion, then one can be 
as selective as possible provided the whole lesion 
is targeted. In our experience the more selective 
the catheter is the higher the degree of necrosis. 
During this step, the radiologist may observe 
arteriovenous shunting through the tumor. If one 
proceeds with TACE, one risks nontarget embo- 
lization (lungs, since the most common type of 
shunting is hepatic artery to hepatic vein) and 
inadequate tumor treatment. Instead, the opera- 
tor should treat first with blunt embolization 
using Gelfoam until the shunting resolves. If at 
the end of blunt embolization the tumor is still 



enhanced by contrast then one can proceed with 
TACE. Otherwise a 2-week wait period is recom- 
mended at which time a repeat arteriogram will 
decide whether TACE is possible. 
» Step 6-Treatment. TACE is performed slowly by 
hand-injection under continuous, real-time fluo- 
roscopy to ensure there is no reflux of chemother- 
apy back around the catheter that may result in 
nontarget embolization. Though nontarget embo- 
lization of the contralateral hepatic artery may 
not be a serious problem, inadvertent emboliza- 
tion of the gasiroduodenal artery can have serious 
consequences including gastroduodenal necrosis. 
Single, double, or triple agent chemotherapy mix- 
tures are used with varying frequency depending 
on institutional preference. At our institution we 
use triple chemotherapy mixture composed of 
100 mg Cisplatin (Bristol Myers Squibb, Princ- 
eton, NJ), 10 mg Mitomycin C (Bedford Labo- 
ratories, Bedford, OH) and 50 mg Doxorubicin 
Hydrochloride {Adriamycin; Pharmacia- Upjohn, 
Kalamazoo, MI). Currently there is no data as 
to the relative efficacy of specific chemotherapy 
mixtures. The chemotherapy is mixed 1:1 with 
Lipiodol (Savage Laboratories, Malville, NY) (2:1 
if slow flow is noted in the artery to be treated}. 
Lipiodol has been shown to concentrate within the 
ulature and reside there for weeks, 
incentration of chemotherapy 
in the tumor by up to 100-fold compared to sys- 
temic chemotherapy. After chemotherapy infu- 
sion, 10-20 cc of nonbuffered lidocaine can be 
infused through the same catheter for pain con- 
trol. This provides not only immediate pain relief 
in case the patient complains of intra-procedural 
pain but has also been shown to help in the imme- 
diate post-procedural period and until patient- 
control anesthesia can be initiated (see below). 
Additional embolization with 150-300 microm- 
eter particles (PVA, Boston Scientific or Embos- 
pheres, Biosphere Medical, Boston, MA} until the 
flow in the treated artery is slowed down, further 
increases the chemotherapy residence time within 
the tumor, though data related to actual benefit 
are lacking. At our institution, complete embo- 
lization is avoided for two reasons: (1) we want 
to ensure patency of the artery for future treat- 
ments, and (2), in vitro experiments have shown 
that prolonged hypoxia induces selection for more 
aggressive neoplastic cells. Table 10.3 shows the 
list of materials requ ired for TACE and Table 10.4 
shows the possible complications and associated 
mitigating measures. 



Chemo-Embolizi 



Table 10.3. The basic materials n 



Aliein.- 



Obtaining arterial 
Maintaining arterial 



18-g single-wall needle (Cook") and 0.035" 

guide wire (Bentson, Cook) 

5 Fr, 11-cm vascular sheath : Cordis' 1 i 



IV: toiiiiii'.g aodc:'.:i:'.a! a h. htaail-rlusri cath^.er ; .Ang;.: ,"vn.' 

aortogram 

Pc: forming selective 5 Fr, hook, enci'.cle glide catheter (Sin 

SMA& celiac Terumo") 

arteriogram 

Selecting the final Simmon's 1 or Cobra (Terunio) glide t 

catheter position over 0.035" glide wire (Terumo) 



T:v.- 1 :•/. eni 



a-cc syringes and chemotherapy r« 

stop-cock 

Manual hemostasis 



Micropuncture set (Cook) 
4 Fr, 1 1 -cm vascular sheath (Cordis) 
ics") 4 Fr, Pigtail-flush catheter (Angiodynamics 

.on's 1, Cobra (Teiiiino i or Michelson (Angiody- 

heter 3 Fr miciocataeter i Renegade, Boston Sci- 
entific 3 ) coaxially through the 5 Fr or 4 Fr 
catheter over a 0.018" -wire (Transend, 
Boston Scientific) 

it 3-way 1-cc syringes may be necessary if high res] 
tance in micro catheter 

Closure device 



7 Finishing 

7 lie micro catheter ; step 5 ; :s used only if :be Simmons ] glide- cannot be advanced iai eiv aig:: to satelv miiise tae chemcaherapy 
mixture. Once ready v: infuse, the chemolhei aay and related supplies slv ■ li 1 J be manipiLa:eo at a table separate {:<:::: the main 
one, to avoid inadveitenl chemotherapy contamination or unrelated supplies. 
■■ Cook, hlooaiar.gLOn, IN: Cordis. Miaiv.i. !-L; A ngic-cy ramies, ^ueeiisouiy, NY: 7e; una:. Soi'.ieisel, Ml: K.:- J -^a:: Scientific, Boston, MA 



» Step 7-Finishing. A single high-resolution expo- 
sure of the liver is obtained to document distribu- 
tion of Lipiodol (along with chemotherapy). The 
catheter and sheath are removed and hemostasis 
is achieved with manual pressure or the use of a 
closure device. Peripheral pulses are rechecked 
and documented to make sure they are stable. 
Though exceedingly rare with proper technique, 
significant changes may signify complications 
such as access artery dissection or distal throm- 



10.3.2 
Recovery 



1 of the common femoral artery vas- 
cular sheath and proper hemostasis is achieved, 
the patient is placed on monitoring for 4-5 h and 
patient controlled analgesia (PCA) pump and i.v. 
hydration are initiated. At the end of the monitor- 
ing period and if no untoward events are noted 
the patient is sent to the floor. Routine nursing 
checks and care are adequate thereafter. P.R.N, 
medication should include (in addition to the 
morphine or fentanyl PCA pump), anti-nausea 
and additional pain medication for breakthrough 
pain. Hydration is critical not only because of 
the patient's NPO status prior to the procedure 
and possible nausea, but more importantly to 
mitigate the consequences of a possible tumor 



lysis syndrome such as acute renal failure. After 
the 4-h observation period the patient is encour- 
aged to ambulate, initially under supervision. 
Table 10.5 shows a sample admission order sheet. 
The use of arterial puncture closure devices can 
cut down the observation period to 2 h. As soon 
as the patient ambulates, the Foley catheter (If 
one was placed) is removed. At the same time p.o. 
intake is advanced as tolerated. When the patient 
is ambulatory a noncontrast CT of the abdomen is 
obtained to document the distribution of Lipiodol 
and the degree to which the tumor has taken up 
the chemoembolization mixture (Fig. 10.5). Uni- 
form Lipiodol uptake by the tumor correlates with 
a higher degree of necrosis compared to spotty 
or lack of Lipiodol uptake. Likewise, region with 
high Lipiodol uptake post-TACE correlate with a 
higher degree of necrosis on follow-up imaging 
examinations (Fig. 10.6). 

After a 24-h period of observation and symp- 
tomatic control, the patient is discharged to home, 
barring continued significant symptoms and with 
appropriate instructions to exercise vigilance for 
possible infection or groin hematoma. A sample dis- 
charge form is shown in Table 10.6. More than 90% 
of TACE patients are discharged to home after this 
period. Cases which require one more day of hospi- 
talization are rare and even longer stays are exceed- 
ingly rare. Discharge medications should include a 
7-day course of oral antibiotics (i.e., Ciprofloxacin 
250 mg p.o. bid) and P.R.N, pain medication. 



C. S. Georgia des and [.-!-. Citsc hwind 



Table 10.4. Possible complic: 



iated with TACE 



Nontarget embolizatio 

Post-embolization syndrome 
(Pain, fever, nausea /vomiting I 
Tumor lysis syndrome 



n of TACE Mitigating intervention/notes 



n, and gastric acidity reductic 



Pre- and post-procedure liyii j r:1 _■:■ n 
Follow creatinine 

LFT elevation Follow as outpalieni if mild and asymptomatic 

Liver failure Avoid TACE in patients with Child-Pugh C liver disease. Do not perform TACE from proper 

hepouc artery. Selec.l right or left. Mjv be irreversible and fatal 

In our experience at leas; halt I he p.meiiis unJei going TACE will exhibit svmpioms re I ale J 10 postembolizati on to n lesser or 
greater Jegree. The vast maionty i '.■■■ 90%) of patients will recover enough to be discharged the next morning. The rest may 
require a second hospital Jay. l-'ulahlies related to TACL are exceedingly rare anc. re late J to limited liep.ilic function reserve an J 
ensuing liver Jecompeitsalion i;nJ acute failure. It is thus crucial to document the patient's liver ftmciion reserve anc: follow 
the contraindications indicated in Table 10.2. 



n orders for patients after TACE 



Table 10.5. Sample aJm 

• Admit: (Attending ni 

• Diagnosis: Liver neoplasm, s/p chemoembolizalioii 

• Condition: (i.e. stable] 

• Vitals: Q 15 min x 4, then q 30 x 2, then hour x 2, then floor routine 

• Allergies: (i.e.NKDA) 

• Activity: Right (or left) leg straight x 4 h and head flat x 1 b. Tit en ambulate under supervision x 15 min. Then 

patient may walk but avoid stairs/straining 

• Nursing: Right tor left : groin cliecks q 1 :■ ntin x 4, tit en .: 30 min x J, tit en .: hour x 1 for possiole hem atom a /bleed 

• Diet: Advance as tolerated (tailor net patient, i.e. diabetic, low cholesterol etc) 

• iVF: 1 00 cc/ri x 1-i h. Lien J/c if pit Lent tolerates p.o-. intake I tailor per patient : 

• Meditations: Patient controliec. anesth.es. a pump I P ■: !! A pump, separate order sheet) 

- i i.e. femanvl JO meg i.v., .t I min, lock ■"■lit at maximum o limes per hi 

- Oxycodone 

- Tylox 1-2 tablets q 4-6 h prn breakthrough pain 

- Zofran 8 mg p.o. q8 h prn nausea 

- Ciprofloxacin 250 mg p.o. bid 

• Laboratories: None necessary (tailor to patient! 

■ Studies: CT abdomen without i.v. or oral contrast after patient able to ambulate 

urJers should be tailored according to specific patient needs. For example, medications should be compatible- with the pa item's 
a i.e iiiies: trie amount or i.v. fluids should take into account I he pal le ill's jejt.il function and carJiac status, and la o oratories should 
be used to follow suspected TACE toxicilies ! i.e. .^n elevated oikrubin should be followed, as should an elevated creatinine) 




Fig. 10J5. Axial, nonenhanced CT of a patient with HCC (same 
as in Figs. 10.1 and 10.4) obtained one day following TACE. 
shows the lesion retaining the radio-opaque Lipiodol {ai'ivv, ) 
in its most vascular regions. The retention of Lipiodol - and 
consequently c h em otiierapy since tiie two are mixed - can per- 
sist for weeks after TACE and cot relates with tumor necrosis 



Chemo-Enjbolizadon for Liv 





Fig.10.6a-c. Axial CT image of the liver of a patient with unresectable HCC a. The 
patient is one day post-TACE. Lio:oco, !i',!gh-iiensi:y regLor.?; i- - : een scattered through- 
out the large righ: i it ili mor. A :egi ;n of rel.'tive.v ;::g!ir:' Lipii -. I - ■ I npr;ikc iviiliin [lie 
turaor is noted {white iinvivhccut). Six-weel-: MR! fo.kiv-up b shows a partly necrotic 
tumor with cystic degeneration o: the l;:im:--:' corresponding 10 [.:e region of high Lipi- 
odol uptake (' iWu'rc iimiiic/il'l;:/). Gross palho.oeic.il specimen of die liver c confirms 
focal necrosis of the same region {white arrowhead) 



10.3.3 
Follow-up 



11 benefit, patients should Lie seen at reg- 
ular intervals, ,1 perfusion MRI of the liver obtained 
(Figs. 10.7 and 10.8) and compared to the baseline or 
previous MRI. Six-week intervals between successive 
TACEs is currently the standard at our institution. 
Prior to each TACE the MRI will establish tumor via- 
bility (or necrosis}. Necrosis is quantified as 0%-25%, 
25%-50%, 50%-75%, and 75%-100% based on perfu- 
sion (or contrast enhancement) MRI. If no residual 
viable tumor is noted, a follow-up MRI is scheduled 
for 6 weeks later and no TACE is performed. Lack of 
satisfactory response after one TACE does not pre- 
dict eventual response, and additional TACE should 
target the same tumor. We have observed many times 
patients having "failed" to respond to the first TACE, 
responding very favorably after the second or third 



TACE. Before TACE is called a "failure" we believe the 
patient should be treated at least 2-3 times targeting 
the same region. The emergence of any contraindica- 
tions to TACE between successive TACEs precludes 
repeat treatment; thus prior to each procedure the 
relevant laboratory values should be obtained and 
the patient re-evaluated. In some cases where the 
patient was precluded from having surgery solely due 
to tumor size, adequate reduction in size following 
TACE may render the patient operable. Though rare, 
we have had at least five patients in the last 2 years 
who became resectable or candidates for liver trans- 
plantation after TACE reduced the tumor burden or 
shrunk it away from vital structure such as portal/ 
hepatic veins. Thus continuous re-evaluation and 
consultation with oncology and surgery is vital so that 
such patients do not miss a chance for cure. Finally it 
should be noted that TACE-associated toxicity is sig- 
nificantly less than that of peripherally administered 



'._". S. Georgia des and [.-!-. i."iesc lnvind 



Table 10.6. Sample discharge ir 



is for patients after TACE 
.e all i.v. lines 

- Ciprofloxacin 250 mg p.o.bid x 7 days 

- Oxycontin 10 rag p.o. ql2 h prn pain 

- Oxycodone 5-10 rag p.o. q 4-6 h prn breakthrough rain 

- Zofran 3 nig p.o. q8 h prn nausea 

• Instructions: - No straining, stair clinibing, or driving x 48 h 

- If groin swelling or bruising, or fever, natisea/vomiiing. 
or worsening abdominal pain call (on call number) 

• Diet: - As tolerated 

• Follow ■:.-. \ - Con; r.isi -enhanced MKi of iiiv: .mo same cay cl.n.c appointment 

with (Intervemional Kadioiogisi! in 4-6 weeks 

• Send copy of discharge summary to ! referring physician's name) 

■' Ti'.ese are gener.'.l guidelines. Specific instructions should be tailored to patient's 
needs, allergies, and clinii 



• Discharge k 




Fig. 10.7. Axial. gadolinium enhanced MR! of the liver of a 
patient with HOC shows a Luge, peripherally enhanced lesion 
(arrows) replacing the right lobe. The patient was unresectable 
owing to the size ji the tumor. I 'ecision was made to treat the 
patient with TACE for palliative reasons 



Fig. 10.8. Axial, ga c cl in in m- enhanced MR! of the liver of the 
same patient as in Fig. 10.7 after two TACE treatments shows a 
dramatically decreased in size I timer with decreased enhance- 
ment (iirrpii-'s). Though in any tumors may not decrease in size 
significantly after TACE, most will show a degree of necrosis 
as measured on contrast-enhanced MR! 



chemotherapy (unpublished data, C. S. Georgiades, 
K. Hong, M. W. D'Angelo, J. F. Geschwind). Of 
course the main drawback of TACE is that it does not 
treat metastases outside the liver, which are in any 
case rare for HCC. For many secondary hepatic neo- 
plasms, liver metastases are indeed the life-limiting 
disease aspect and such patients may benefit despite 
the presence of extrahepntk metastases. 



10.4 
Results 



TACE is c 

able and o 



e of the most popular techniques avail- 
e that is rapid ly gaining favor. Despite the 



fact that it has been used world-wide for many years, 
there has been, until recently, a lack of prospective 
randomized trials regarding its efficacy. Because 
of this, the procedure generated considerable dis- 
cussion and dissent among physicians. Skepticism 
has been finally put aside by one randomized con- 
trolled trial and a recent meta-analysis that con- 
cluded that TACE significantly improves survival 
of patients with nonresectable HCC compared to 
nonactive treatment. Llovet et al. [10.] performed 
a prospective randomized trial that recorded a 1-, 
2-and 3-year survival of 82%, 63%, and 29% for 
patients undergoing TACE vs. 63%, 27%, and 17% 
for patients treated symptomatica lly. As a matter 
of fact, the trial was terminated early when the sig- 
ii! Meant survival benefit of the TACE group became 



evident. Cam ma etal. [14] performed a meta-analy- 
sis of randomized controlled trials looking at the 
two-year survival of patients with unresectable HCC 
who underwent TACE, transarterial chemotherapy 
(TAC) or transarterial embolization (TAE) versus 
nonactive treatment. Patients who underwent tran- 
sarterial treatment had significantly improved sur- 
vival with a pooled odds-ratio of 0.54 (95% CI). The 
TACE and TAE groups had an odds-ratio of 0.45 
(95% CI) and the TAC group an odds-ratio of 0.65 
(95% CI). 

Prospective, randomized trials investigating pos- 
sible survival benefits of TACE for metastatic neo- 
plastic disease to the liver are lacking, as are such 
trials for cholangiocarcinoma. From our own expe- 
rience however (accepted for publication, March 
2005, JVIR), patients with unresectable cholangio- 
carcinoma treated with TACE had a median survival 
of 23 months, compared to only 6 months for histor- 
ical controls receiving supportive treatment alone, 
9 months for those receiving systemic chemother- 
apy and 16 months for those receiving chemoradia- 
tion. Metastatic colon cancer to the liver has proven 
to be less responsive than hoped for to TACE, with 
reported median survival of 10-12 months. Still, 
patients with colorectal metastases to the liver resis- 
tant to systemic chemotherapy may obtain benefit 
from TACE [15-17]). Other metastatic neoplasms to 
the liver that are amenable to TACE include carci- 
noid, breast cancer, adrenal cancer, sarcomas etc. 
From our own experience, though technically fea- 
sible for all types of pathology, TACE appears to be 
more effective in tumors which are highly vascu- 
lar. Such observations have been reported by other 
authors as well [18] We have found sarcomas and 
carcinoid to be especially responsive but (as men- 
tioned above) colon cancer to be less responsive to 
TACE in keeping with being relatively less vascu- 
lar. In the latter case, we are currently investigating 
the efficacy of radioembolization (Yttrium-90, see 
Chapter 11) for colorectal metastases to the liver, 
and preliminary results appear to be more hopeful 
than those previously collected for TACE. 



10.5 
Conclusion 



With concerns regarding the survival benefits of 
TACE for patients with inoperable hepatocellu- 
lar carcinoma finally being put to rest [10,14], all 
such patients should be considered for TACE. The 



median survival of patients with inoperable HCC 
is 4-7 months (which can be extended with maxi- 
mal supportive care to approximately 10 months). 
TACE prolongs median survival to more than two 
years and, though rarely, converts some patients 
into candidates for resection or transplantation. 
Preliminary unpublished data from our institution 
confirm a significant survival advantage of TACE 
in patients with inoperable mass-forming cholan- 
giocarcinoma. Nearly uniformly fatal, this disease 
imparts a median survival of 4-8 months. Such 
patients who have been treated with TACE (Protocol 
identical to that of HCC) show an extended median 
survival up to 20 months. The benefit of TACE on 
patients with other histological types of inoperable 
liver cancer has not been evaluated by prospective, 
randomized trials. Given the low toxicity rate, lack 
of alternatives and encouraging results for HCC and 
CCC, it is reasonable to offer this treatment to such 
patients. Indeed, no doing so would deprive them 
of a reasonably expected survival benefit. Though 
TACE can be applied to any morphologic type of 
liver cancer (from solitary lesions to diffuse bilobar 
disease), it must compete with percutaneous ablation 
techniques for patients with up to three lesions each 
less than 4 cm each. These percutaneous ablation 
techniques (Table 10.1) under optimal conditions 
provide benefits similar to surgical resection but 
suffer from their own specific limitations. The most 
commonly used percutaneous ablation techniques 
areradiofrequencyablation(RFA) and alcohol injec- 
tion (PAI). In experienced hands these techniques 
have excellent results for up to three lesions with a 
maximum diameter of 3-4 cm each. However, their 
efficacy rapidly tapers for larger lesions. Table 10.7 
shows the typical response/ recurrence rates of HCC 
treated with RFA. Radioembolization with Yttrium- 
90 microspheres is an alternative to TACE as an 
intra-arterial treatment for unresectable HCC, CCC, 
or secondary liver cancer, but efficacy data are more 
scarce. TACE is an evolving technique and many 
questions remain unanswered. For example, what 
is the best chemotherapy "cocktail"? What TACE 
protocol yields maximum survival benefit? Can it 
be combined with systemic chemotherapy or other 
abl.ition techniques? Will the addition of novel phar- 
maceuticals (i.e. anti-angiogenesis drugs or drugs 
that target calnbolic pathways uniquely) improve 
survival? If so, what is the best regimen? We are 
years and many studies away from answering these 
questions, but the prospect of transforming inoper- 
able liver cancer into a chronic disease managed by 
a protocol of surveillance and regular ir 



C. S. Georgiades and I.-I-. G esc Ivwind 



is a matter of time. As treatment options change and 
as more and more patients receive multiple differ- 
ent treatments for their disease (i.e. RFA and TACE, 
or systemic chemotherapy and TACE) we must also 
evolve in how we view this disease. One of the salient 
points is the del in it ion ol response, which according 
to current criteria, depends on tumor size alone. The 
advancement of imaging methods and their ability 
to distinguish between necrosis and cystic change 
versus viable tumor has rendered this view anti- 
quated and inappropriate in our opinion. We believe 
the most appropriate method to measure response 
is to quantify the percent viable tumor remaining 
after treatment. Currently the best way to achieve 
this is by contrast-enhanced MRI or PET scanning 
(the latter somewhat limited by its relatively lower 
spatial resolution). 

This final point is probably the most crucial. 
Surgeons, hepatologists, oncologists, and interven- 
tional and diagnostic radiologists bring their unique 
and important input to the treatment planning for 
these complex cases. Treatment planning should be 
^ed after every intervention whether this is 



Table 10.7. Radiofrequencv a o la r ion .■; an alternative I. 
nique for the treatment of unresectable HCC 



>5cm <25% >75% 

As indie a fed above, resp> uise and ieau renre rates are inversely 
proportional to lesion size. Up to three separate lesions of up 
to 4 cm each can be treated with RFA, TACE, or both for that 
matter with goo,", results. Lesions larger than 5 cm or more 
than three lesions or any size should preferentially be treated 
with TACE. Percutaneous eihj.no! o: noetic joid injection has 
similar response rates to RFA and suffers from the same limi- 
tations pertaining to lesion size and number 



partial hepatectomy, TACE, systemic chemotherapy 
or percutaneous ablation therapy, as the clinical 
situation maychange for the better or worse. Amul- 
tidisciplinary team approach to treating primary or 
secondary liver cancer is a development that affords 
such patients maximum benefit from what medicine 
currently has to offer. 






l.Achenbach T e; a I. (2002 i Chemoemoolizalion for primary 
liver cancer. Eur 1 Surg Oncol 28:37-41 

2. Adam R (20C_i ) Chemotherapy and surgery: new perspec- 
tives on the treat mem of unresectable l:v T : meljsljses.Aiin 
Oncol 14 [Suppl 2]:II13-I116 

3.Camma C et al. (20C2i Trai'.sarteria! chemoembolizatiOEi 
tor unresectable hepatocellular carcinoma: meta-analysis 
of rjiiuomized controlled trials. 1-i.adiology 224:47-54 

4.E1-Serag HB, Mason AC (2000) Risk factors for the rising 
primary liver cancer m the Unite." States. Arch Intern Med 
273227-3230 

5.Feldman ED et al. (2004) Regional treatment options for 
patients with ocular melanoma metastatic to the liver. Ann 
Surg Oncol 11:290-297 

6.Georgiades CS et al. (2001) New non-surgical therapies 
in the treatment of hepatocellular carcinoma. Techn Vase 
Intervent Radiol 4:193-199 

7.HoganBAetal.i2002 ) Hep a tic metastases from an unknown 
primary (UPN'i: survival, prognostic indicators and value 
of extensive investigations. Clin Radiol 57:1073-1077 

8. Inoue ¥ et al, i I L J L J4 i Hyper vase u la i liver metastases of gas- 
tric cancer compJetely lespoitding to transcatheter oily che- 
moemoolizjiien using erirubicin hydrocliioride, mitomycin 
C and lipiodol. Gan To Kagaku Ryoho 21:1665-1667 

9.Kjwji 5 et ill. ( ] 9Q2 ) Prospective jf;6 randomized clini- 
cal trial for the treatment ol" hepaiocellular carcinoma -a 
comparison of lipioclol-transcalheler arierial embolization 
with and wiihoul adriamycin ■: firsr cooperative study!. "The 
Cooperative study Group for Liver Cancer Treatment of 
Japan. Cancer Chemother Pharmacol 31 [Suppl]:Sl-S6 



lO.Kawai S et al. (1997) Prospe 
of lipiodol-transcatheter arte: 
treatment of hepatocellular carcinoma: a comparison iA 
epirubicin and doxorubicin (second cooperative study). 
The Cooperative Study Group for Liver Cancer Treatment 
of Japan. Semin Oncol 24 [Suppl 6]:S6-38-S6-45 

ll.Llovet JM et a 1. 12002 ) A r re rial embolization or chemoem- 
bohzation versus symptomatic treatment in patients with 
unresectao.e hepatocellular carcinoma: a randomised con- 
trolled trial. Lancet 359:1734-1739 

12.Nakahashi C et al. (2003) The impact of liver metastases 
on mortality in patients initially diagnosed with locallv 
advanced or resectable pancreatic cancer. Inl I Gastromtesl 
Cancer 33:155-164 

13.Sanz-Altamira PM et al. (1997) Selective chemoemboliza- 
n on in the management of hepatic metastases in refractory 
colorectal carcinoma: a phase II trial. Dis Colon Rectum 
40:770-775 

14. SeongJ et al(1999) Combined transcatheter arterial ch emo- 
embolization and local radiotherapy .:■ I" unresectable hepaio- 
cellular carcinoma. Int ] Radiat Oncol Biol Phys 43:393-397 

15.Tanada M et al. (1996) Intrahepatic arterial infusion che- 
molherapy tor tlir colon cancer patients with liver metasta- 
ses - a comparison of arreria; embolization chemolherapv 
versus continuous annual infusion chemotherapy, Gan To 
Kagaku Ryoho 23:1440-1442 

16.Tarazov PG (2000! Arterial chemoembolization for meia- 
slatic co.orecta.l cancer in the aver. Vopr Cnlsol 4<y.:*<~. -56o 

17. Wyld Let a 1.(2003) Prognostic factors for patients with hepatic 
metastases from breast cancer. Br ] Cancer 89:284-290 

18.YuMCet al. (2000) Epidemiology of hepatocellular carci- 
noma. Can I Gastroenterol 14:703-709 



11 Radioactive Microspheres for the Treatment of HCC 



s, Riad Salem and Jean-Fra 



Introduction 141 

Clinical Considerations 142 

Patient Selection & Preparation 142 

Technique 142 

Calculation of the Total Dose To Be Delivered 142 

Calculation of the Shunt-Ratio 143 

Anatomical Considerations 143 

"'■'Y-Microsphere Embolization 144 

r'LUieiVi Recovery 145 

Follow-up 147 

Conclusion 147 

References 1 48 



11.1 

Introduction 

As in the case for transarterial chemoemboliza- 
tion (TACE, Chapter 10}, radioembolization takes 
advantage of the preferential hepatic arterial supply 
of hepatocellular carcinoma (HCC) to deliver tar- 
geted therapy to the tumor, relatively sparing the 
liver parenchyma, which is mostly supported by the 
portal venous system. Radioembolization is effected 
via intra-arterial delivery of carrier spheres onto 
which radioactive particles are attached. There are 
two types of radioactive microspheres that can be 
used in the treatment of primary (and metastatic 
for that matter) liver disease. They both contain 
Yttrium-90 ( 90 Y) as the active element but differ 
in the type of carrier particle. The first is 9fh Y glass 



C. S. Geohgtades MD, PhD 

Assistant Professor of RjJiobgy and y.ngei v. Johns Hopkins 
Medical Institutions, Blalock 545, 600 N. Wolfe Street, Balti- 
more, MD 21287, USA 
R. Salem, MD 

Assistant Professoi .if Radiology jii; Oncology, Nor duves tern 

Memorial Hospital. Department of Radiology, 676 North St. 

Claire, Chicago, IL 6061 1, USA 

J.-F. Geschwind.MD 

Associate Professo: of Racoologv, ?;::gerv and Oncology, loans 

Hopkins Medical Institutions, Blalock 545, 600 N. Wolfe Street, 

Baltimore, MD, 21287, USA 



microspheres (Theraspheres, MDS Nord ion, Ottawa, 
Ontario, Canada), which are glass spheres with 
a diameter of 25±10u.m, impregnated with 90 Y, a 
radioactive element. Following intra-arterial infu- 
sion, most Theraspheres embolize at the arteriole 
level because of their relative size. S0 Y is a pure beta 
emitter (937 KeV) that decays to Zirconium-90 with 
a half-life of 64.2 h. The emitted electrons have an 
average tissue penetration of 2.5 mm (effective max 
10 mm) [1-5]. These physical properties stimulated 
interest in the use of Theraspheres for the treatment 
of HCC as well as metastatic liver lesions. Unlike 
TACE, the effectiveness of '°Y-microsphere embo- 
lization has not been established; however it is seen 
as a viable alternative for patients whose hepatic 
neoplasm does not respond to TACE. Histological 
studies have shown that there is a disproportionate 
accumulation of J Y-mk rospheres along the vascular 
periphery of the hepatic tumor, with a relative con- 
centration of 2.4 to 50 times more than in the normal 
liver parenchyma [6,7]. Though the exact reason for 
this is not understood (perhaps the altered blood 
vessel flow and diameter that results from tumor 
angiogenesis allows preferential embosphere flow), 
this phenomenon can be used to deliver large doses 
of radiation to the tumor, while relatively sparing the 
normal liver. After lodgi iis iii ilie iisial arteriolar cir- 
culation, the microsphere radiation has a maximum 
effective tissue penetration of 10 mm, thereby spar- 
ing the normal liver parenchyma beyond this limit. 
Radiation essentially ceases 10 days after emboliza- 
tion but even before that it poses no threat to others. 
These advantages are exploited in this type of novel 
treatment and the process is described below. 

The second type of 90 Y carrier is resin-based 
microspheres with a diameter of 29-35 urn and 
are also infused via the appropriate hepatic artery 
branch to provide selective internal radiation (SIR). 
These SIR-Spheres (Sirtex, Medical Limited) have 
an average activity of 40 Bq per sphere and can be 
suspended in sterile water and contrast media to 
the desired total activity [8, 9]. Since the radioactive 
element is the same as that on glass microspheres, 



the tissue penetration and decay characteristics are prolong survival, '"Y-microsphere (either glat 
identical, as are patient selection, preparation, tech- resin-based) embolization is not considered a 
nique, and recovery. five treatment option. 



11.2 

Clinical Considerations 



11.3 

Technique 



11.2.1 

Patient Selection & Preparation 

Selection criteria are similar to those for TACE and 
are listed in section 10.2.1 ("Chemoembolization 
for Liver"). As with TACE, candidates must have 
unresectable solid, primary or secondary hepatic 
tumors. However, one crucial difference is the 
degree of shunting between the hepatic artery and 
hepatic vein. The detection of shunting during the 
pre-TACE arteriogram needs to be addressed but 
not necessarily quantified. Gelfoam embolization 
and repeat arteriogram immediately or 7-14 days 
later to document lack of visible shunting allows 
for TACE to proceed. However, in the case of S0 Y- 
microsphere embolization, shunting can result in 
life-threatening complications and must be accu- 
rately quantified. Radiation pneumonitis can result 
from 90 Y-microsphere shunting to the lungs with 
significant morbidity and possible mortality ensu- 
ing, especially if the total lung dose approaches 
30-50 Gy [3]. The method for quantifying shunting 
is described in the following section. 

Oktida stage and ECOG score should be obtained 
prior to treatment, as severe liver dysfunction is 
a contraindication to any form of hepatic artery 
embolization including °"Y- microsphere emboliza- 
tion. Laboratory values should be obtained prior 
to embolization, including: a comprehensive meta- 
bolic panel (NA, K, glucose, creatinine, BUN, total 
bilirubin, AST, ALT, alkaline phosphatase, albumin, 
total protein), hematology panel (hematocrit and/or 
hemoglobin, white blood cell count, platelet count, 
coagulation profile (INR, APTT) and tumor mark- 
ers (i.e. AFP for HCC, CEA for colon cancer). These 
values serve not only to ensure a safer procedure 
(i.e. normal coagulation) but also to allow for proper 
follow-up of hepatic and renal function and moni- 
tor response (using tumor marker levels). A baseline 
gadolinium-enhanced MRI with diffusion/perfu- 
sion sequence should be obtained as part of staging, 
for establishing response to treatment and for future 
treatment planning. Based on current evidence, the 
patients should be informed that though it may 



Once the patient has been deemed a candidate for 
M Y-microsphere embolization one has to calculate 
two important values: 
l.The total dose of radiation to be delivered which 

is related to the efficacy of the treatment 
2. The hepatic artery-to-vein shunt percentage, 

which is related to the safety of the treatment. 



11.3.1 

Calculation of the Total Dose To Be Delivered 

The total dose to be delivered depends not on the 
tumor burden but on the total volume of the liver 
(normal parenchyma plus tumor) to be embolized. 
A CT or MRI should be obtained, based on which 
the liver volume to be embolized is calculated. The 
same MRI (which should include contrast-enhanced 
and diffusion/perfusion sequences) can be used 
as baseline to quantify the response to treatment. 
Studies have shown that a total dose of 120-150 Gy 
yields better results than lower doses [1,4,8]. The 
total activity to be injected is calculated by: 

DIGylxVlLI^GLOqj^ 
50Gy/Kg.GBqx[l-F] 

where, 

• A is the total activity to be injected (in gigabec- 
querels) 

• D is the desired dose to the volume of the liver to 
be treated (in grays) 

• V is the total liver volume to be treated (in liters) 

• SGL is the specific gravity of the liver (1.03 kg/1) 

• F is the percent hepatic artery-to-vein shunting, 

• The factor 50 results from the fact that for soft 
tissue (liver) the absorbed dose is 50 Gy per GBq 
per Kg 

Therasphere vials are supplied with predeter- 
mined doses fractionated at 3, 5, 7, 10, 15 and 20 GBq, 
therefore a tailored combination should be used for 
each patient depending on the desire dose. 



e Microspheres for the Treatment of HCC 



In addition to the above dose calculation, in the ofthe liver/lungs a 

case of resin-based Yttrium-90 treatment, some is calculated by 
authors [10] calculate the tumor to normal liver 
uptake ratio (T/N) and treat only if the T/N ratio is _ ALng 

equal or greater than 2. ALng + ALi 



? obtained. The shunt percentage 



Where, 

• At/An is the ratio of activity in the tumor divided 
by the activity in nontumorous liver and can be 
calculated from the images obtained for the cal- 
culation ofthe shunt ration (see next section} 

• Mt/Mn is the ratio ofthe tumor to nontumorous 
liver volume calculated from CT or MR1 studies 



11.3.2 

Calculation ofthe Shunt-Ratio 

From the above equation the only factor that needs 
to be independently quantified is the percent hepatic 
artery-to-vein shunting. This is accomplished by 
performing a perfusion study ofthe liver with ?9 T- 
m macroaggregated albumin ( w T-m MAA) parti- 
cles, which are routinely used to perform a ventila- 
tion/perfusion scan for the diagnosis of pulmonary 
embolism. Commercially available .MAA particles 
have a diameter of 50±13 fim (i.e. more than 90% 
are within 10 and 90 u.m in diameter) [11]. Because 
a slightly larger percentage of Jf Y- microspheres are 
smaller than the diameter of the capillaries (com- 
pared to MAA particles), the use of MAA to calcu- 
late the shunt percentage tends to slightly under- 
estimate the shunt. Therefore one should be rather 
conservative in cases where the shunt percentage is 
borderline and err on the side of safety. The initial 
diagnostic hepatic arteriogram has a dual purpose: 
first, to plan which hepatic artery branch will be 
used to treat the tumor; second, to calculate the 
shunt related to that vessel. Though most commonly 
the main right or left hepatic artery is used, occa- 
sionally a secondary branch may sut lice if it supplies 
the entire tumor. Once the diagnostic catheter is 
in the branch that will be used to deliver the 90 Y- 
microspheres during the future treatment, 2-6 mCi 
(75-220 MBq} of "T-m MAA is infused. Following 
this, a perfusion study with antero-posterior planar 
images is obtained whose field of view includes 
the liver and chest. Steps 1-5 described below are 
followed. After Step 5, the patient is taken to the 
nuclear medicine department and the planar images 



• S is the shunt percentage (percent of the total 
activity that will be shunted to the lungs) 

• ALng is the activity calculated over the lung fields, 

• ALvr is the activity calculated over the liver field 

Normal lungs can tolerate a total lung dose of 
30 Gy [3], above which severe morbidity and possi- 
bly mortality may result from radiation pneumoni- 
tis. Thus, if for example, a total liver dose of 150 Gyis 
desired the shunt should be less than approximately 
20%. A safety factor can be introduced arbitrarily 
to increase the safety margin. Figures 11.1 and 
11.2 show two planar images of two patients who 
underwent such a shunt study. The first patient was 
deemed a candidate for radioembolization having 
shown negligible shunting to the lungs. The second 
patient showed significant shunting and would have 
suffered complications related to radiation pneumo- 
nitis had lie received raclioemholizafion. 



11.3.3 

Anatomical Considerations 



Radioembolization takes advantage ofthe fact that, 
while normal liver parenchyma receives most of its 




Fig. 11.1. Planar gamma camera view of the liver and chest 
following "T-m MAA infusion in the right hepatic artery of 
a patient wirJi '.inresec table HCC. Activity is seen in the liver 
(rtiTi'iri without any pulmonary activity. T lie shun: percentage 
was calculated to be 2%i thus the patient is a candidate for 
" u T-mic rosp her 




w of the liver and chest fol- 
lowing ""T-m MAA infusion in the hepatic artery of a patient 
with unresectable HOI. significant I'almonaiv activity is seen 
(ai'ivws) indicating a shunt. The liver activity is indicated by 
an ijrroiWit'iii.i The shunt percentage was prohibitive; thus the 
patient could not safely receive '"Y-microsp 



blood supply (60%-80%) from the portal venous 
system, malignant hepatic tumors, whether pri- 
mary or metastatic, are nearly exclusively supplied 
by branches of the hepatic artery. Cancer angio- 
genesis (a.k.a. neovascularization) is a process by 
which neoplastic cells recruit new blood vessels in 
order to ensure adequate local oxygen tension. Due 
to their relatively higher metabolic demands, cancer 
cells "live" in a nearly constant state of hypoxia. 
They respond by secreting chemotactic factors that 
promote the formation of new blood vessels. Arterial 
epithelial cells are much more responsive to these 
factors, vchicb explains ivhv such malignant tumors 
are supplied by the hepatic artery. Radioemboliza- 
tion then selectively targets the tumor while liver 
parenchyma is mostly (but not entirely) spared. 

Accessory and/or replaced right or left hepatic 
arteries are common (up to 20%-30%). In addition, 
common origins of the left hepatic and left gastric as 
well as right hepatic and right gastric arteries may 
be added complicating factors. Such associations are 
important insofar as they increase the risk of non- 
target embolization, mainly the stomach, proximal 
small bowel, and/or pancreas. Though self-limiting 
gastritis, duodenitis, and pancreatitis have been 
reported, no deaths have yet been documented from 
nontarget embolization (excluding pneumonitis). 
Irrespective of this, it is important to clearly delin- 
eate vascular anatomy during the initial arterio- 
gram in order to minimize morbidity. 1 f 90 Y-micro- 



sphere treatment cannot be given safely due to left or 
right gastric or gastroduodenal vascular anatomic 
relationships, one should consider coil-embolizing 

these vessels and then proceeding with '^-micro- 
sphere embolization. 



11.3.4 

^-Microsphere Embolization 

The treatment ['Ian having being decided upon, writ- 
ten informed consent is obtained and the patient's 
groin regions are prepared and draped in a sterile 
fashion on the fluoroscopy table. Informed con- 
sent should disclose the following risks: Injury to 
blood vessels and/or organs, anaphylactic reaction 
to contrast, worsening of renal function, infection, 
worsening liver function or liver failure, and pos- 
sibly death. One must document the pre-procedural 
peripheral pulses and choose the femoral artery with 
the strongest pulse for initial access. Sedation with 
i.v. versed and fentanyl is used at our institution, 
occasionally needing to be enhanced with Phener- 
gan and/or Benadryl. 

• Step 1-Obtaining vascular access. In most cases 
access using an 18-g, single-wall needle followed 
by an 0.035" guide wire is successful. In difficult 
cases a 0.018" micropuncture set can be of help, 
with or without the use of ultrasound guidance. 

• Step 2- Maintaining access. A 5 Fr short vascular 
sheath providing access in the right or left (stron- 
gest pulse) common femoral artery is used at our 
institution. A 4 Fr access set can be used in cases 
where less traumatic arterial access is needed (i.e. 
slightly abnormal coagulation profile), however 
the smaller catheters may be a bit less control- 
lable. 

• Step 3-Abdominal portogram. A flush aortogram 
via a multisideholed, pig-tailed catheter at the 
level of the celiac artery will delineate the vascular 
anatomy, tumor supply and provide a road-map 
for more selective access. For the most part this 
step can be skipped. In rare cases and after fail- 
ing to easily cannulate the SMA and celiac access 
with a selective catheter (see step 4), which may 
suggest variant anatomy, one may fall back to it. 
If performed, a 15 cc per s injection for a total of 
50 cc is adequate. 

• Step 4-Selective arteriograms. First, a 5 Fr cathe- 
ter (Simmons 1 or Cobra glide catheters, Terumo) 
is used to select the superior mesenteric artery 
(SMA) and an arteriogram is performed using an 
injection rate of 6 ccper sfor 3-4 s. Then the celiac 



ldioactive Microspheres for I lie Treat nieni of HCC 



artery is selected (again Simmon's 1 or Cobra 
glide catheters, Terumo) and a selective arterio- 
gram is performed with a similar injection rate as 
above. In most cases the celiac axis arteriogram 
will show the tumor blush to best advantage. 

• Step 5-Selecting the final catheter position. A 
glide wire 0.035" is advanced through the glide 
catheter followed by the catheter itself. Though 
one wants to be as selective as possible to avoid 
treating normal liver, being too selective will 
result in parts of the tumor not being treated. In 
general, either the right or left main hepatic artery 
is the optimal position for the treatment catheter. 
In cases where there is tumor in both lobes, the 
one showing more tumor blush on the diagnos- 
tic arteriogram should be targeted. If the 5 Fr 
glide catheter cannot be advanced to the desired 
location because of unfavorable anatomy, a 3 Fr 
mil 'roca truster (Renegade, Boston Scientific) over 
an 0.018" guidewire (i.e. Transend, Boston Scien- 
tific) can be used coaxially. 

• Step 6a-If the objective is to calculate the future 
dose to be delivered, then once the diagnostic 
catheter is in the branch that will be used to 
deliver the 90 Y-microspheres in the future, 2- 
6mCi (75-220 MBq) of 9S T-m MAA is infused. 
As described above, the patient is then taken to 
the nuclear medicine department and the shunt 
ratio is calculated (see section 11.3.2}. 

• Step 6b-If the dose has been previously calculated 
and the patient has been deemed a candidate for 
radioembolization, then the dose is delivered 
using the set-up shown in Figure 11.3. Extreme 
care regarding the set-up (tubing connections, 
stop-cock positions, etc) must be exercised as the 
volume to be injected is small (a few milliliters) 
and errors are usually irreversible. 

• Step 7-The catheter and sheath are removed and 
hemostasis is achieved with manual pressure or 
the use of a closure device. Peripheral pulses are 
rechecked and documented to make sure they 
are stable. Though exceedingly rare, significant 
changes may signify complications such as access 
artery dissection or distal thrombosis. 

The infusion set-up system is shown inFigure 11.3. 
Careful set-up and inspection of the system is cru- 
cial prior to infusion to avoid inadvertent spillage 
or misadministration of the Theraspheres. A list of 
basic materials needed is shown in Table 11.1. 

Close coordination between the Interventional 
Radiologist, Oncologist and Radiation Safety Offi- 
cer is a must for an uneventful treatment. Even then, 




Fig. 11.3. Schematic of the set-up for the treatment appara- 
tus for w Y-microsphe:e embolization, i . syringe; 2, three-way 
stop cock; 3, saline o;ig: 4. shir. Jed vi.il containing the UU Y- 
niiiTO sphere solution; 5, need.es; r.-, ■:■-■.:: 1 1 ■.:■ iv infusion catheter; 
7, excess ■"'Y-micro sphere solution collection via I. shielded 



one must abide by the ALARA (As Low As Reason- 
ably Achievable) principle to limit radiation expo- 
sure. The process must be speedy without sacrificing 
quality control, with proper shielding and radia- 
tion dosimetry and utilizing the minimum number 
of personnel possible. The infusion is given under 
continuous fluoroscopic guidance to ensure proper 
delivery. Once the total dose is given the catheter is 
flushed with a few milliliters of saline to extrude any 
particles remaining in it. The catheter and arterial 
access sheath are removed, hemostasis is achieved 
and the patient n 



11.3.5 

Patient Recovery 

90 Y- microsphere embolization for liver tumors is 
generally an outpatient procedure with minimal 
complications. Patients are recovered and observed 
for 4-6 h, essentially a post-arteriogram recovery. 
Possible complications and mitigating interventions 
are shown in Table 11.2. Mild pain and minimal 
nausea are not uncommon and if controlled with 
medication should not prevent the patient's dis- 
charge. A small minority of patients will develop 
post-embolization syndrome and require overnight 
admission to control their pain, nausea and fever. 
Even then, almost all will be discharged the next day. 
The vast majority of patients report no symptoms 
and return home the same day. A sample discharge 
order sheet is shown in Table 11.3. They present no 



ials needed for *Y-micro sphere embolizatio 



Obtaining arterial ai 



- g-single wail needle i Cook" i and 0.03a" Microp unci tire 



guide - 



(Bentson, Cook] 

vascular sheath (Cordis") 
5 Fr, pigtail- flush catheter :' Angjo J y na m - 



V .i i :v. anting arterial ao 

Pel i"o: :tt:ng abuonuna! 

a or tog ram ics B ] 

Performing selective SMA 5 Fr, hook, endhole glide catheter (Si 

& celiac arteriogram mon's 1 , Terumo-' : 

Selecting the final catheter Si m mon's 1 or Cobra glide catheter t 

position 0.035" glide wire (Terumo) 



The nticrocatheter (step a! :s used 



2-6 mCi (75-220 MBq) of w T-m MAA 

Yttrium-90 dose 

i see rig. ] i .3 for additional su pokes i 

Manual hemostasis 



4Fr, 11 cm vascular sheath (Cordis; 
4 Fr, pig tail- flush catheter i Angio cynt 



Cobra (Terumo) or Michelson (Angiody- 

3 Fr micro catheter (Renegade, Boston Sci- 
entific') coaxially through the 5 Fr or 4 Fr 
catheter above over a 0.018" wire (Transend, 
Boston Scientific) 



Closure dev 



e to unfavorable 



inly if die Simmons I glide canr.ol be advanced lo the desired location J 
it the shunt ratio through the tumor to be treated must be calculated to avoid nontarget 
:e performed during the patient's first visit followed by a plana i gamma-camera image that is used 
to calculate (his ratio. 1: deemed a candidate for embolization, lhe patient returns a: a later lime and steps 1-5 and 6b-7 are 
followed to perform me embolization. [Cook. Blooiuington : IN; Cora is. Cordis Corp., M:ami, l-L; Angio dynamics, Ciuee:tsbu:y. 
NY; Terumo, Terumo Medical Corp., Somerset, \'i Boston Scientific, Boston, MA] 



Table 11.2. Possible complications associated with J|, Y- microsphere emboliza 

aia.j mii:g.Ui:'g . in er vent ions 

Possible complications of radio-embolization Mitigating 



Nontarget embolizatio 



Turn 






; Anti-ulcer, antacid med 

Pre- and post-procedtit 
Follow creatinine 
Symptomatic support 



hydra 



Post- embolization syndrome 

(Pain, fever, nausea/vomiting! 

Racialion pneunr 'iiil:s 

Such complications from radioembolization a 

patients to have this procedure on mi outpatient basis, 1-iarely, treatmem may be complicated by 

post-embolization syndrome and symptomatic supporl may be necessary. Nontarget embolization 

can be all but eliminated by meiica.ous technique and pre-procedure planning 



Admission-supportive 
e exceedingly rare allowing the 



rity of 



Cable 11.3. Sample discharge h 

» Discontinue all i.v. lines 

» Discharge to home 

• Medications: - Ciprofloxa 



is for patients alter radioembokzatio 



250mgp.o.bidx7days 

- Oxycontin 10 mg p.o, q 12 h prn pain 

- Oxycodone 5-10 mg p.o. q 4-6 h prn breakthrough a am 

- Zofran 8 mg p.o. q 8 h prn nausea 

• Instructions: - No straining, siai: ckmbing, or driving x 48 h 

- If groin swelling or cruising, or fever, nausea/vomiting, 
or worsening abdominal pain call ion call number) 

• Diet: - As tolerated 

• Follow-up: - Contrast- en it a need \' K 1 of liver and same day clinic aaaoinl mem want 

(Interventional Radiologist! in 4-6 weeks 
Send copy of discharge summary to ! referring physician's name) 



" These are genera, guidelines. Spec 
and clinical picture 



c instructions shot 



ailored to pat 



e Microspheres for the Treatment of HCC 



radiation risk to others and thus no special precau- 
tions are required. Mild and self-limiting transami- 
nase elevation is often seen but usually of no clinical 
significance. Upon discharge, the patient is placed 
on a 7-day oral antibiotic regimen (i.e. Ciprofloxacin 
250 mgp.o. bid) and given PRN oral pain medication 
for possible abdominal pain. One must remember 
that occasionally, a patient who has been discharged 
to home without complaints may develop pain, 
nausea and/or fever within a few days afterwards. 
Therefore the above med ical ions are recommended 
for all patients regardless of symptomatology. 



11.3.6 
Follow-up 

At approximately four to six weeks, the patient is 
seen at the clinic and a contrast-enhanced, per- 
fus ion/diffusion MRI is obtained. At that time, 
one should address two issues. First, what is the 
tumor's response to treatment and second, has the 
patient's overall condition changed? The answers 
to these questions will dictate whether the patient 
still remains a candidate for embolization and if 
so, should he or she continue with '"Y-microsphere 
embolization or change the treatment plan. In estab- 
lishing response to treatment, the pertinent factor is 
the percent enhancement of the tumor in the MRI 
scan, and not as classically thought, the size of the 
tumor. Many times the tumor is killed entirely and 
replaced by an indolent cyst or necrotic/fibrotic 
tissue of similar size. Additionally, it is not uncom- 
mon to see a lack of response after the first treatment 
and a good response after the second or third treat- 
ment, and thus at least two or three embolizations 
are recommended before one decides on a change of 
venue. ECOG score and laboratory values should be 
retested to ensure that the patient remains a candi- 
date for further embolization. Finally, re-evaluation 
of the patient's clinical slams, including re-staging of 
the disease if relevant changes are observed, is nec- 
essary. Such re-evaluations may require the input of 
surgery or hepatology in case adequate down-stag- 
ing renders the patient a surgical candidate. 



11.4 
Conclusion 



have been developed to attempt to improve survival 
in these patients. Intra-arterial embolization with 
90 Y- microspheres has shownpromise in early clinical 
studies for the treatment of unresectable HCC. Such 
patients are expected to live for about 5-7 months 
with symptomatic treatment alone. Glass '^-micro- 
sphere embolization has been shown to extend the 
median survival to 12 months for Okuda stage II 
disease and 23 months for Okuda stage I [4, 12]. Sig- 
nificant turn or shrinkage has also been reported [6], 
which again requires the constant re-evaluation of 
patients in case surgical resection or transplantation 
becomes an option. Additional, combination treat- 
ments as well as treatment for colorectal metastatic 
disease are beginning to appear, i.e. 90 Y-microsphere 
embolization followed by hepatic arterial chemo- 
therapy (HAC). The 1-, 2-, 3-, and 5-year survival 
rates of patients treated with glass 90 Y-microsphere 
embolization followed by HAC are reported at 72%, 
39%, 17%, and 3.5%, versus 68%, 29%, 6.5%, and 0% 
for HAC alone [13]. S0 Y-microsphere treatment has 
the added advantage of very minimal toxicity/side 
effects and quick recovery. Additionally, glass 90 Y- 
microsphere embolization appears to yield similar 
survival benefit to those of TACE for unresectable 
HCC, with Geschwind et al. [2] reporting a 1-year 
survival of about 63% for both and significantly 
better than supportive treatment alone. Resin- 
based 90 Y- microsphere embolization has also been 
shown to provide a survival benefit, with Lau et 
al. [10] reporting a median survival of 9.4 months 
(1.8-46.4 months), with four of 71 patients becom- 
ing resectable after treatment. Lau et al. [8] in 
phase I &II clinical trials reported a median sur- 
vival of 55.9 weeks for patients who received a tumor 
dose > 120 Gy. Similarly, benefit has been shown for 
patients with colorectal metastases treated with SIR 
spheres. Gray et al. [14] and Blanchard et al. [15] 
showed that about 501" of patients have exhibited 
more than 50% shrinkage of the tumor with a con- 
comitant decrease in CEA levels. Given the large 
number of nonsurgical treatment options, patient 
selection is of paramount importance. In this con- 
text, 9fh Y- micro sphere embolization has a definite 
niche. In the end, a multidisciplinary approach that 
includes Interventional & Diagnostic Radiologists, 
Oncologists, Hepatologisis, and Surgeons is a must, 
in order to choose and tailor the optimum treat- 
ment protocol and offer the patient the best hope 
for extended survival. 



With few if any surgical optic 
i patients, many no 



; for he pat 






References 



l.Carr IB (2004) Hepatic arterial M YI1rium glass mien 

spheres liheraspherei for niiresectal-.i' ■U'p.. | :'.cllula- .;- 

cinonjii: interim safety ;i:id survival dala vr. •.:.: patienls 

Liver Transplant 10 [Supp! 1]:S107-SI 10 
2.Geschwind JF et al. (2004) Yttrium-90 microspheres for ihe 

treatment of hepatocellular carcinom.i ■. .ix:r:'L-nit:ril.-j;> 

127:S194-S205 
3. Salem R et al. (2002) Yttrium-90 microspheres: radiation 

dierapy for unresectable liver cancer. . Vase nicrv sadiul 

13:5223-S229 
4. Salem R et al. (2004) Use of yttrium--}:: glass microspheres 

: iheraspheiv; for ihe trearmem of in 

lubr carcinoma in patient 1 vt-,\\: pof.n w - ih-i:— :h:siv I 

Vase Interv Radiol 1 5:335- 34> 
5.Sarfaraz M et al. (2003) Physical aspects of Yttrium 90 

microsphere therapy for -e-resc'.ihk' "-ipst.. 1 :c*-::r' 

Med Phys 30:199-203 
i.CaoX et al.! 1000] Hepatic ndi.>e-n: - i: izaiiun wi:h y:tr:um 

a giass microspheres for . '.-ic-' >■' prmary iver 

cancer. Ch:n Med I 112:43:: *3i 
7.Campr>ell AM el al. (2000) Analysis of the distribution 

of intr; arterial microspheres in human liver lisihnvinf, 

hepatii V'lrum ■*" mitr:isphere :fie:.ipy : hys Med iiiol 

45:1023 I H33 
8.Lau WY e: al. (1991) Treatment of inoperable hepato 

cellula- carcinoma with mi rah era he arterial Yltr;jm 90 



miir:i>phe: 



I and II study. Br J Cancer 70:994- 



999 
9. Van Hazel Get al. (2004) Randomized phase 2 trial of SIR- 

■•phiio p., » 1 1 u ; :i !"-.:■ n r : i c i 1 / J c n 1 1 ''V : :■ r i n chemotherapy versus 

fiuorojra. . leucovorin oh emo therapy alone in advanced 

colorectal cancer. ] Surg Oncol 88:78-85 
1 0. 1 ju WY el si. (1 993) Selective internal radiation therapy for 

-n-rcM';:.i:-.e hepatocellular carcinoma with intraarterial 

ir:fusii:n i:: '"Yttrium microspheres. Int I R.idiai Oncol Biol 

Phys 40:583 592 
ll.HungJCel al. (2000) Evaluation of macro aggregated albu- 
sizes for use .n pr.lmonary shunt sluoies. I Am 

Pharm Assoc 40:46-51 
12. Kent; Gil, Sundram FX (2003) Radionuclide therapy 

'.■:• hepai.'.dlular ■.".'. re i noma. Ann Acad Med Singapore 

32:ilR S24 
I 3. Gray H el a:. (2001) Randomized trial of sir- spheres plus 

Lhe::u::rn" ipy vs. chemotherapy alone tor treating ra:ie:i-s 

with liver metastases rrom primary large powel cancer. Ann 

Oncol 12:17 1-1720 
1 4. Gray K el ai. (1 992) Regression of liver metastases following 

treatment with Yttrium-90 microspheres. Aust NZ J Surg 

62:1 OS 110 
I 5. Klanchard RJ et aL (1989) Treatment of liver tumors with 

Yttrium 9f: microspheres alone. Can Assoc Radiol) 40:206- 

210 



12 Yttrium-90 Radioembolization for the Treatment 
of Liver Metastases 



Intiod^ciion 149 

Pathophysiology and Therapeutic Principle 

iIliiiio-.il C oir-i e'er Li tions 150 

Anatomic and Technical Considerations 15 

Ty-.crrS-hrre kiminislraiion 153 
I ];usime!ry::ir TheraSphere 153 
'. Infusion Technique 154 

S::< Sphere* Administration 154 
. Dosimetry :'-r SIR-Spheres 154 
'. Infusion Technique 155 

Calculation of Lung Dose 155 

Results 156 

Clinical Experience, Response, 

and S.rviva 156 

S1K Spheres*; Clinical Experience, Response 

and Survival 157 

Complications 159 

Conclusion 159 

References 159 



12.1 

Introduction 

The liver is the most frequent site of metastases, pri- 
marily due to the spread of cancer cells through the 
portal circulation. Approximately 60% of patients 
diagnosed with colorectal carcinoma will eventually 
experience liver disease as the predominant site. As 
with hepatocellular carcinoma (HCC), surgical resec- 
tion of colorectal metastases otters the only chance for 
cure. However, this option is only available to a small 
percentage of patients. Many patients with other pri- 
maries such as breast, lung, and neuroendocrine will 
develop liver metastases during the course of the dis- 



R. Salem, MD, MBA 

Assist.ui; Prolvssoi .if Ha; jo logy anc neology, Nor duves tern 

Memorial Hospital. Pepniimcal of Radiology, 676 North St. 

Claire, Chicago, IL 6061 1, USA 

K.G.Thi;hston,MA 

17 Bramble Lane, West Grove, PA 19390, USA 

J.-F. Geschwind, MD 

Associate Proles?.: 1 : of kLiciologv, ?;::geiv and 0:- oology, lohiis 

Hopkins Medical Institutions, Blalock 545, 600 N. Wolfe Street, 

Baltimore, MD 21287, USA 



ease. Therefore, there is a need for novel liver-directed 
treatments for patients with unresectable metastases to 
the liver. Current therapies for the treatment of liver 
metastases parallel those for HCC and include: hepatic 
arterial infusion of chemotherapy (HAI), trans-arte- 
rial chemoembolization <TAC!-J, radiofrequency abla- 
tion (RFA), and combinations of these treatments. 
These treatments have displayed some effectiveness 
in prolonging lite lor patients with liver metastases, but 
are often associated with toxicities such as abdominal 
pain, fever, nausea, and vomiting. 

Yttrium-90 ( 90 Y> microspheres (TheraSphere, MDS 
Nordion, Ottawa, Canada and SIR-Spheres, Sirtex 
Medical, Lake Forest, Illinois) represent an intra- 
arterial therapy, infused via a catheter placed in the 
hepatic arterial system. The microspheres are selec- 
tively delivered to the tumor bed due to the hypervas- 
cularity of tumor relative to normal liver parenchyma, 
where they become entrapped in the arterioles feeding 
the tumor. Since Jf| Y emits beta radiation with a maxi- 
mum average penetration of approximately 1 cm, the 
majority of the radiation effect is directed to tumor- 
ous tissue while sparing normal liver parenchyma. 
This results in a maximum tumor icidal effect, while 
minimizing potential compromise to normal liver 
function. The therapeutic benefit derived as a result 
ot effecting tumor kill while sparing radiosensitive 
normal tissue provides a significant treatment alter- 
native for patients who have limited treatment options 
available. TheraSphere was approved by the FDA for 
unresectable hepatocellular carcinoma in December 
1999 under a Humanitarian Device Exemption, while 
SIR-Spheres was approved in March 2002 for colorec- 
tal cancer metastatic to the liver in conjunction with 
infusion of intra-hepatic floxuridine (FUDR). 



Pathophysiology and Therapeutic Principle 



The rationale for intraarterial delivery of m Y micro- 
spheres for metastatic disease to the liver involves 



anatomic and physiologic aspects of hepatic tumors 
that can be exploited for the delivery of a therapeutic 
agent. Hepatic tumors derive at least 90% of their 
blood supply from the hepatic artery, while liver 
parenchyma obtains between 70%-80% of its blood 
supply from the portal vein [1-8]. This differential 
pattern of vascular perfusion provides an intrin- 
sic advantage for hepatic arterial regional therapies 
delivered selectively to liver tumors. Additionally, 
many liver tu mors, both primary and secondary, are 
hypervascular relative to normal liver parenchyma 
as determined by contrast angiography. Thus, selec- 
tive arterial delivery, as practiced in hepatic arte- 
rial chemotherapy and chemoembolization, delivers 
therapeutic doses of radiation that are preferentially 
retained in the liver tumor, theoretically sparing the 
surrounding noi"imaliH.nanr liver tissue. It has been 
shown, using hepatic arterial injection of radiola- 
beled microspheres in experimental tumors, that 
tumor microcirculatory blood vessel density is 3-4 
times greater than that of surrounding liver paren- 
chyma [9, 10]. In particular, single photon emission 
computed tomography (SPECT) with hepatic arterial 
injection of r " Ml 'Ti;-m;KToa.!i* recited albumin ( MA. A) 
has been used to investigate the patterns of micro- 
circulation in patients with liver tumors and con- 
firm findings in experimental animal liver tumor 
models [11]. The incorporation of an appropriate 
therapeutic radioactive isotope into nondegradable 
microspheres can potentially be utilized to capital- 
ize upon the selective advantage afforded by hepatic 
arterial administration and by the increased arte- 
riolar density of malignant tissue within the liver to 
deliver a highly localized dose of radiation directly 
to the intrahepatic tumor(s). It has been known for 
some time that it is possible to inject the liver and 
other organs with doses of nondegradable (glass or 
resin) microspheres without producing overt isch- 
emic damage [1, 9, 12]. 

Radiation pneumonitis is a concern with hepatic- 
directed radiation treatment. Previous preclinical 
and clinical studies with ,0 Y microspheres demon- 
strated that up to 30 Gy to the lungs could be tol- 
erated with a single injection, and up to 50 Gy for 
multiple injections [13]. For this reason, patients 
with "Tc-MAA evidence of potential shunting to 
the lungs leading to lung doses greater than 30 Gy 
should not be treated. Similarly, any flow of m Y 
microspheres to the gastrointestinal system that 
cannot be corrected by percutaneous coil emboliza- 
tion techniques, as predicted on ' 0m Tc-MAA, is con- 
traindicated because of potential adverse gastroin- 
testinal events. 



12.3 

Clinical Considerations 

The selection process for patients undergoing radio- 
embolization is multifactorial. Patients with meta- 
static disease to the liver might have undergone one 
or several courses of systemic chemotherapy, surgi- 
cal resection, and/or radiolrecjuency ablation. Rel- 
evant clinical history having an impact on the safety 
and efficacy of radioembolization might include 
surgicallyplaced intrahepatic chemotherapy pumps 
(causing chemical vasculitis), the use of radiosensi- 
tizers (such as capecitabine or irinotecan), as well 
as treatment with groth factor inhibitors, such as 
bevacizumab. These patients have image findings 
of progressive liver-domman! meraslatie disease, 
regardless of any therapeutic benefit afforded by 
the aforementioned therapies. 

For all patients, one of the most important fac- 
tors in determining eligibility tor radioembolization 
is ECOG performance status. Patients presenting 
with clearly compromised functional status (ECOG 
2-4; see Table 12.1) are at high risk for rapid onset 
of liver failure and associated morbidity with treat- 
ment. Notwithstanding this precaution, each patient 
deserves individual consideration given the favor- 
able toxicity profile of radioembolization; some 
patients with limited ECOG performance may still 
benefit from therapy. 

Liver metastases present with relatively consis- 
tent findings on MRI, CT, or ultrasound. If a mass is 
identified, pathologic confirmation of malignancy 
metastatic to the liver is necessary. If ultrasound is 
the initial diagnostic modality, additional cross-sec- 
tional imaging should be obtained. Triple-phase CT 
is highly sensitive in detecting hepatic malignancies. 
Since the majority of liver tumors are angiographi- 
cally hypervascular, scanning in the early phases 
likelihood of detection. 



equivalent 
K.;['j]-itsky 



Table 12.1. ECOG performance st 
ECOG Characteristics 



Asymptomatic and fully active 100% 

Symptomatic; fully ambulatory; 80%-90% 

:es:iii"-ed in phy^icallv strenuous activity 

symptomatic; ambulatory; ciipaL^e of 60%-70% 

se[f-ca:e; mcie It.aii 5C?o of waking 

hours are spent out of bed 

symptomatic; limited self-care; 40%-50% 

spends more than 50% of time in bed 

Completely disabled; no self-care; 20%-30% 

:'ro:idJe;\ 



Ylrn u :•'.-'''!'■ }\ .j J ioe:v. l -1 ■:■■. i z:i i ioj j :■:■: die Treiitme:'/ .:■[' Liver !Welji-;wvt:; 



Later phase imaging is necessary to detect other less 
vascular lesions, the degree of multifocality, and to 
identify portal vein patency. MRI is also a sensitive 
modality to identity and characterize lesions, given 
specific attention to diffusion weighted imaging 
sequences. 

One of the most important clinical parameters 
that must be assessed when treating patients with 
radioembolization is the status of overall liver func- 
tion. In the absence of biliary obstruction, drug tox- 
icity (e.g., capecitabine) or metabolic abnormality 
(e.g., Gilbert's syndrome}, it is extremely unusual 
for patients with metastatic disease to the liver to 
exhibit elevated liver functions. In particular, total 
bilirubin is usually normal in this patient popula- 
tion. In cases where total bilirubin is elevated and all 
of the abovementioned factors have been excluded, 
it is likely that tumor infiltration within the hepatic 
parenchyma is the causative agent, thereby imply- 
ing a grim prognosis for the patient. The decision to 
treat such patients should be based on the thorough 
assessment of the possibility of extending survival 
or palliating pain. 

The pretreatment evaluation of a patient with 
metastatic disease to the liver should include: 
1. History, physical examination, assessment of per- 
formance status 
2. Clinical laboratory tests (complete blood count 
with differential, blood urea nitrogen, serum cre- 
atinine, serum electrolytes, liver function, albu- 
min, LDH, PT) 
3. Chest X-ray, tumor marker assay (CEA, AFP) 
4.CT/MRI scan of the abdomen and pelvis with 
assessment of portal vein patency 

Similar to hepatic artery chemoembolization, 
patients with bilobar disease should be treated in 
a lobar fashion at staged time intervals, usually 
30-60 days following the first treatment. Patients' 
eligibility for repeat radioembolization should be 
evaluated following every treatment. Patients on 
chemotherapy should have this therapy discontin- 
ued twoweeks prior to radioembolization. Che- 
motherapy may be restarted twoweeks following 
li.ni i ■ ■ e i li !_■ ■.: I i :-■ ..■ r i . .n. 



12.4 

Anatomic and Technical Considerations 



Although recent implementation of CT, MRI, and 
ultrasound with 3-D reconstruction for the iden- 



tification of first- and second-order variants have 
proven effective for the identification of large vari- 
ant mesenteric vessels, these techniques are not a 
replacement for conventional angiography. During 
the evaluation of the patient for radioemboliza- 
tion, mesenteric angiography and ""' : Tc-MAA lung 
shunting scan must be performed [14-16]. Com- 
plications as a result of inadequate assessment for 
radioembolization and nontarget embolization 
include unplanned/unexpected necrosis in unde- 
sirable arterial beds, such as the cystic artery, GI, 
cutaneous and phrenic capillary beds [17-22], 

All patients being evaluated for radioemboliza- 
tion should have the following angiographic evalu- 
ation [l-l]: 

Abdominal aoitogram-injection ot 15--20 cc/sec 
for a total of 30-40 cc. This allows for the assessment 
of aortic tortuosity, mural atherosclerotic disease, 
and facilitates proper visceral catheter selection. 

Superior mesenteric angiogram-injection of 3- 
4 cc/secfor30 cc. This allows assessment of any vari- 
ant vessels to the liver (accessory or replaced right 
hepatic), as well as visualization and identification 
of a patent portal vein. This injection rate allows for 
the opacification of the mesenteric system without 
unnecessary reflux of contrast into the aorta. 
1. Celiac angiogram- mi eel ion of 4 cc/sec for 12-15 cc. 
This allows for the assessment of normal hepatic 
branch anatomy, the presence of a replaced left 
hepatic artery, or other variant arteries without 
reflux of contrast into the aorta. 
2. Selective left hepatic arteriogram-injection of 2 cc/ 
sec for 8 cc. In cases of normal anatomy, this allows 
for the assessment of flow to segments 2, 3, 4A, and 
4B. Special attention should be paid to the falci- 
form, phrenic, right or accessory gastric arteries. 
3. Selective right hepatic arteriogram-injection of 
3 cc/sec for 12 cc. Normally, the right hepatic artery 
provides flow to segments 1 (caudate lobe may have 
other blood supply), 5, 6, 7, and 8. Particular atten- 
tion should be paid to the supraduodenal, retro- 
duodenal, retroportal and cystic arteries. 
4. Selective gastroduodenal arteriogram-injection 
of 2 cc/sec for 8-10 cc. The gastroduodenal artery 
normally provides flow to the pancreas, stomach, 
small bowel, and omentum. Attention should be 
paid to the identification of an accessory right 
hepatic artery feeding segment 6. The threshold 
lor prophylactic embolization of this vessel during 
radioembolization should be quite low. Two exam- 
ples of prophylactic embolization in preparation 
for radioembolization are demonstrated in Figures 
12.1 and 12.2. 





Fig. 1 2.1 a-c..^upi\;;Uiode:i;il,g.i si: ''duodena I. r.ghi gas:ric artery embo- 
lization. ■> Initial common hepatic arteriogram rerea.ing ;i small vessel 
artery arising kom -he rigi'.t hepatic ariery representing the supraduo- 
denal i srfijighr [iiTii! )). Relative position cc I he gas^ro duodenal artery 
is noted d'ani'il iimur). Right gastric artery .'.rises from die proper 
hepatic artery i iWj.'/V unviviicrais). t> Hollowing complete em holizji ion 
of i lie gasno-duodenal i \vli:h' iini-.;: ) and right gas- ric arteries. s : ..per- 
selective catheterization o\ the supraduodenal artery {stmiglir iii'i'ou ) 
reveals dislrihuiion to the .:. u .:■ ■■:. e tin . branches Uun-iTii blink dn L nr!. 
c Kmoolizaiion o: the supraduodenal was conipleieo {sti\uglit iin\-.'. ) 
along wi;h the CI 'A {:iinvd .ji ji- ul. as well as the right gastric artery 
allowing for optimal deliverv to the liver. The vascular s'apply to the 
liver tumors has been convened and simplified moi- two feed.ng vessels, 
the right and left hepatic arteries. The patienl underwenl safe infusion 
of Y90 microspheres. 



As described above, in order to visualize 
small vessels as well as vessels that may demon- 
strate reversal of flow (e.g., result of flow shunt, 
or sumping secondary to hypervascular tumor), 
dedicated microcatheter injection with relatively 
high rates (2-3cc/sec for 8-12cc) should be per- 
formed. Without adequate contrast bolus, many 
ancillary vessels (which have profound effect on 
hemodynamics and directed therapy) may go 
unnoticed, resulting in toxicities including post 
embolization syndrome, distal embolization, 
and/or end nontarget organ necrosis or ulcer- 
ation. Although it may be argued that high injec- 
tion rates may represent supraphysiologic flow 
dynamics, the potential changes induced as a 
result of regional therapy with i adioembolization 
(spasm, ischemia, stasis, and vessel injury) may 
result in altered physiologic states and thus reflux 
into these vessels. Every attempt must be made to 
avoid this scenario. 



Arteriovenous connections responsible for radio- 
active particle delivery from the liver to the lungs 
arise from cancers rather than from normal liver. 
Thus, in the presence of liver cancers (particularly 
hepatocellular carcinoma], n significant amount of 
W Y microspheres may shunt to the lungs. Hence, 
during the angiographic evaluation of a patient for 
M Y, 4-5 mCi of ?9l "Tc-MAA must be injected in the 
vessel of interest, followed by imaging for lung shunt 
fraction in Nuclear Medicine. Given that the likeli- 
hood of shunting is low with metastatic disease, we 
favor whole liver (i.e., proper hepatic) MAA injec- 
tion in order to assess the entire liver at one time. 
Lung shunt fraction (LSF) is defined as total lung 
counts/( total lung counts + total abdomen counts). 
The lung shunt fraction that is obtained must now 
be factored into the dosimetry portion of the treat- 
ment plan. 

The technical aspects of radioembolization are 
quite complex and should not be undertaken lightly. 



Ylrn u :•'.-'''!'■ }\ .j J ioe:v. lt ■:■■. i Z:i i ioj j :■:■: :\w Treat me:'/ .:■[' liver \:elj:;j- : ir-; 




Fig. 12.2a-c. GPA/right gastric, a Com it; on hepatLi: arteriogram demon- 
s hyper vascular tumors in ;: patient '.undergoing fjdioeiiibolizalioii. 
GDA (black arrowheads) and right gastric inline arrowhead) must be 
embolized to prevent no:;-targe: administration, b Right gastric catheieriza- 
tion (arrowhead) demonstrates lioiv aA'itg the lesser curve of the stomach. 
c GDA (bliick arroiv) and right gastric i '.viiifa iin-flir! embolization permit- 
iing safe Y a raoioembolization. 



Furthermore, the delivery of TheraSphere and SIR- 
Spheres are distinctly different. Unlike SIR-Spheres, 
the embolic effects of the 20-40 micrometer Thera- 
Sphere 90 Y particles are angiographically negligible. 
As described in this chapter, unrecognized collat- 
eral vessels with consequent infusion of radioactive 
microspheres are certain to result in clinical toxicities 
if proper angiographic techniques are not adopted 
(see package insert TheraSphere"', SIR-Spheres' fl ). 
These might include gastrointestinal ulceration, 
pancreatitis, and skin irritation as well as other 
nontarget radiation. For this reason, aggressive pro- 
phylactic embolization of vessels prior to therapy is 
highly recommended such that all hepatico-enteric 
arterial communications are completely eliminated. 
These vessels include the gastroduodenal, right gas- 
tric, esophageal, accessory phrenic, and falciform as 
well as variants arteries such as the supra/retroduo- 
denal. At our institution, where over 400 radioem- 
bolizations have been performed, we have found our 



GI toxicity rate to be well below 1%. This is due to 
our standard practice of: (a) aggressive prophylactic 
embolization of GDA/right gastric and other vari- 
ant vessels, (b) use of nonembolic TheraSpheres'* in 
a lobar and segmental fashion, and (c) use of SIR- 
Spheres' fl in a lobar, segmental, and dose-fraction- 
ated method (several small doses rather than one 
larger dose) without reaching n completely embolic 



12.4.1 

TheraSphere Administration 

12.4.1.1 

Dosimetryfor TheraSphere 

As described in the product insert, TheraSpher 
consists of insoluble glass microspheres wher 
Yttrium-90 is an integral constituent of the glas: 



The mean sphere diameter ranges from 20 to 30 pm. 
Each milligram contains between 22,000 and 73,000 
microspheres. TheraSphere is supplied in 0.05 ml 
of sterile, pyrogen-free water contained in a 0.3- 
ml vee-bottom vial secured within a 12-mm clear 
acrylic vial shield. TheraSphere is available in six 
activity sizes: 3 GBq (81 mCi), 5 GBq (135 mCi), 7 GBq 
(189 mCi), 10 GBq (270 mCi), 15 GBq (405 mCi), and 
20 GBq (540 mCi) [23]. The corresponding number 
of microspheres per vial is 1.2, 2, 2.8, 4, 6, and 8 
million, respectively. The activity per microsphere 
is approximately 2500 Bq [24]. 

Assuming TheraSphere "°Y microspheres distrib- 
ute in a uniform manner throughout the liver and 
90 Y undergoes complete decay in situ, radioactivity 
required to deliver the desired dose to the liver can 
be calculated using the following formula [15]: 
TheraSphere: A (GBq) = [D (Gy) x M (kg)] / 50 
When lung shunt fraction (LSF) is taken into 
account, the actual dose delivered to the target 
volume becomes [15]: 

D (Gy) = [A (GBq) x 50 x (1-LSF)] / M (kg) 

A is the activity delivered to the liver, D is the 
absorbed delivered dose to the target liver mass, M 
is target liver mass. Liver volume (cc) is estimated 
with CT, and then converted to mass using a conver- 
sion factor of 1.03 kg/cc. 

As an example, an authorized user wishes to treat 
a patient with the following characteristics: target 
volume 1000 cc (1.030 kg), desired dose 120 Gy, 
LSF=5%. 



guide, and a priming needle guide, all contained in 
a Lucite shield. A MONARCH™ 25 cc syringe (Merit 
Medical) is used to infuse saline containing the 
TheraSphere 90 Y microspheres through a catheter 
placed in the hepatic vasculature. 

Once the catheter is in place and the authorized 
user is ready for delivery, the catheter is connected to 
the outlet tubing. Delivery of TheraSphere is accom- 
plished by pressurizing the MONARCH syringe. For 
3 French catheter systems, the infusion pressure 
should range from 20 to 40 PSI, while for 5 French 
systems, the infusion pressure should not usually 
exceed 20 PSI. It is essential that, irrespective of 
the infusion pressure utilized, pressure and flow of 
microspheres closely mimic that observed angio- 
graphieally with gentle, hand injection of contrast. 
The authorized user should familiarize himself 
with the actual flow dynamics of the vessel being 
infused and use a correspondingly lowered infusion 
pressure where necessary, such as might be seen in 
patients with decreased cardiac output. Given the 
small volume of microspheres contained in a given 
activity of TheraSphere (typically 27-90 mg}, a low 
volume of saline is required to infuse a vial of Thera- 
Sphere; the majority of the microspheres are infused 
once a few milliliters of saline are injected. Further- 
more, given the low number of microspheres infused 
with TheraSphere (typically 1.2-4.0 million), the 
entire vascular bed is never saturated. Hence, live 
fluoroscopic guidance while the infusion is occur- 
ring is not necessary. A complete infusion usually 
requires 20-40 cc. 



Activity required = (120 x 1.030} / 50 = 2.47 GBq 12.4.2 

SIR-Spheres Administration 
The patient receives an infusion of2.47GBqto the 
target volume. Given a 5% LSF, the actual delivered 12.4.2.1 
dose was: Dosimetry for SIR-Spheres 



D (Gy) = [2.47 x 50 x (1-0.05)] / 1.030 = 114 Gy 



D (Gy) = 50 x (2.47 x 0.05} = 6 Gy 



12.4.1.2 

Infusion Technique 



The TheraSphere Administration Set consists of one 
inlet set, one outlet set, one empty vial, and two 
interlocking units consisting of a positioning needle 



As described in the product insert, SIR-Spheres* 
consist of biocompatible resin-based microspheres 
containing ,0 Y with a size between 20 and 40 microns 
in diameter. SIR-Spheres' is a permanent implant 
and is provided in a vial with water for injection. 
Each vial contains 3 GBq of yttrium-90 (at the time 
of calibration) in a total of 5 cc water for injection. 
Each vial contains 40-80 million microspheres [25]. 
The corresponding activity per microsphere for SIR- 
Spheres is much lower than that of TheraSphere (50 
Bqvs. 2500 Bq respectively) [24]. 

Just as with TheraSphere, assuming SIR-Spheres 
W Y microspheres distribute in a uniform r 



Ylir.i: :•>'"' K .. k1 i '.■riv.L'v.iz.i; io;"; :"■:■■: die Trer.tme:'/ .:■[' liver !\ ! ^ l,i ;i.i - : t ; 



throughout Che liver and undergo complete decay in Table 12.2. Radioactivity delivered to the liver with SIR- 



situ, radioactivity delivered to the li 
lated using one of two available methods: 

The first method incorporates body surface area 
and estimate of tumor burden as follows: 

A (GBq} = BSA (m 2 ) - 0.2 + (% tumor involvement/ 100) 
[26] 



leu- Sphere;, based c- 



>50% 
<25 % 



3.0 '.".Bo 
2.5 GBq 
2.0 GBq 



where BSA is body surface area. 

The second method is based on a broad estimate 
of tumor burden as described in Table 12.2 [25]: 

For either SIR-Spheres© dosimetry models, A 
(GBq) is decreased depending on the extent of LSF 
(<10% LSF no reduction, 1096-15% LSF-20% reduc- 
tion, 15%-20% LSF-40% reduction, >20% LSF no 
treatment). 

As an example, an authorized user wishes to treat 
a patient with the following characteristics: total 
weight 91 kg, height 1.83 meters (6 ft}, target volume 
1000 cc, tumor volume 300 cc, LSF=S%. 

Using the first method, the formula for BSA as 
described by Dubois and Dubois [27] is: 

BSA = 0.20247 x height 0725 (m) x weight 0425 (kg) 

Therefore, 

A (GBq) = 2.13 - 0.2 + (30/100) = 2.23 GBq 

would be required using the BSA formula. 

Alternatively, given the tumor burden of 25%- 
50%, the patient could be prescribed 2. 5 GBq in activ- 
ity based on Table 12.2 [25]. Given the 5% LSF, no 
reduction is activity would be required. It should be 
noted that for SIR-Spheres, the dosimetry described 
in the product insert is based on whole liver infu- 
sion. If a lobar infusion is intended, the infused 
activity should be calculated assuming whole liver 
volume, and then "corrected" to the proportional 
volume of the target lobe. For example, if the right 
lobe is the target and represents 70% of the entire 
liver volume, the calculated activity to be delivered 
should be multiplied by 0.7. 



12.4.1.2 

infusion Technique 

The SIR-Spheres administration set c 
Perspex shield, the dose vial, inlet and o 
with needles. Standard 10- or 20-c 
syringes preloaded with sterile water s 



nsists of a 
itlet tubing 
: injection 



to infuse the microspheres into the delivery cath- 

Given the larger number of microspheres (40-80 
million) and lower activity of SIR-Spheres (50 Bqper 
microsphere) compared to TheraSphere, the delivery 
of SIR-Spheres is distinctly different than that for 
TheraSphere. Once the catheter is in place and the 
authorized user is ready for delivery, the catheter is 
connected to the outlet tubing. Given the very large 
number of SIR-Spheres microspheres required to 
deliver the intended dose, it is not uncommon for the 
entire vascular bed to become saturated with micro- 
spheres and an embolic state to be reached. For this 
reason, fluoroscopic guidance is essential during 
the infusion. The technique of SIR-Spheres infusion 
involves the alternating infusion of sterile water and 
contrast, never allowing direct SIR-Spheres contact 
with contrast. This allows the authorized user to 
adequately monitor the injection and ensure that 
vascular saturation has not been reached. In cases 
where unrecognized vascular saturation occurs and 
microsphere infusion continues, reflux of micro- 
spheres and nontarget radiation become a distinct 
possibility. The infusion is complete if either (1) 
the entire intended dose has been infused without 
reaching stasis, or (2) stasis has been reached and 
only a portion of the dose has been infused. Given 
the risk of reflux and nontarget radiation once stasis 
has been reached, the continued infusion of SIR- 
Spheres is not recommended. 



12.4.3 

Calculation of Lung Dose 



Radiation pneumonitis is a concern with hepatic- 
directed radiation treatment. Previous preclinical 
and clinical studies with °°Y microspheres dem- 
onstrated that up to 30 Gy to the lungs could be 
tolerated with a single injection, and up to 50 Gy 
for multiple injections [13]. For this reason, patients 
with 9?m Tc-MAA evidence of potential pulmonary 
shunting resulting in lung doses greater than 30 Gy 
should not be treated. 



The absorbed lung radiation dose is the total 
cumulative dose of all treatments [28]): 

Cumulative absorbed lung radiation dose = 

50 x lungmassf A L *LSF L 

where A; = activity infused, LSFi = lung shunt frac- 
tion during infusion, n = number of infusions, 
approximate vascular lung mass (for both lungs, 
including blood) = 1 kg [29]. 

This dose should not exceed the limit of 30 Gy per 
single infusion and 50 Gy cumulatively. In patients 
who require more than two treatments to achieve 
tumor coverage or in patients being retreated in the 
same target volume after progression, repeat 99ln Tc- 
MAA LSF should be performed before each treat- 
ment and calculation of cumulative absorbed lung 
radiation dose included from all previous treat- 



12.5 
Results 

12.5.1 

TheraSphere: Clinical Experience, Response, 

and Survival 

Andrews et al. [12] presented data on 24 patients 
including 17 with colorectal metastases to the liver, 
six with metastatic neuroendocrine tumors, and one 
HCC patient. Imaging at week 16 indicated a partial 
response in five patients, minimal response in four, 
stable disease in seven, and progressive disease in 
the remaining eight patients. Other than mild gas- 
trointestinal symptoms in four patients (unrelated 
to TheraSphere), no hematologic, hepatic, or pul- 
monary toxicities were observed. The authors con- 
sidered the hepatic tolerance to radiation delivered 
by 90 Y to be excellent at doses of up to 150 Gyused in 
the study. Herba and Thirlwell [30] performed a 
prospective dose-escalation study with TheraSphere 
starting at 50 Gy and escalating in 25 Gy increments 
to 150 Gy. There were 37 patients with liver metas- 
tases, 33 of whom had colorectal metastases to the 
liver. The authors observed no major hematological 
or pulmonary complications but did observe some 
gastroduodenal ulceration, which occurred early in 
their clinical experience with TheraSphere, due to 
inadvertent deposition of spheres in the GI tract. 
There was a beneficial response observed by CT 



i where t 



uld be resolved. Stabil: 



22/30 patients (73%). Due to the small sample size 
of the study, no statistically significant relationship 
between dose and clinical or radiological beneficial 
effects was observed. However, the authors con- 
cluded that TheraSphere treatment was a feasible 
and safe technique with beneficial effects. Wong 
et al. [31] presented data on TheraSphere treatment 
of eight patients with unresectable colorectal liver 
metastases. Tumor response was evaluated using 
imaging (CT/MRI) and metabolic evaluation via 
l8 F-FDG-PET and serum CEA. Five of the eight 
patients had an improvement in their tumor activ- 
ity, as assessed by a decrease in 13 F-FDG-PET meta- 
bolic activity and confirmed by parallel changes in 
serum CEA. However, as observed in other stud- 
ies, the use of imaging by CT/MRI illustrated that 
only some of the tumors that responded by meta- 
bolic criteria revealed a corresponding decrease in 
size. This study suggested that using tumor size 
as an indication of treatment response would lead 
to an underestimate of the effect of TheraSphere. 
The authors concluded that there was a significant 
metabolic response to TheraSphere treatment in 
patients with unresectable colorectal liver metas- 
tases. This treatment appealed to provide signifi- 
cant palliation for patients with otherwise incur- 
able disease. In a subsequent study, Wong et al. 
[32] presented data on TheraSphere treatment of 
27 patients with metastatic colorectal cancer to the 
liver. Tumor response was evaluated via la F-FDG- 
PET and serum CEA. The study evaluated the use 
of l8 F-FDG-PET to quantify the metabolic response 
to treatment comparing visual estimates to stan- 
dardized hepatic uptake values. Visual estimates 
were graded as: progression, no change, mild, 
moderate or dramatic improvement. Visual esti- 
mates indicated 20 patients responded to treatment 
while seven patients experienced progression or no 
change in their disease. There was a significant cor- 
relation (r=0.75, p<0.0001) between the response 
group identified through visual estimation and 
as determined by hepatic standardized uptake 
values. There was no statistically significant cor- 
relation observed with CEA values (p=0.13), which 
was attributed to the effect of extrahepatic lesions. 
The authors concluded that treatment significantly 
reduced hepatic tumor metabolism and appeared 
to be palliative in patients with unresectable liver 
metastases. 

Goin etal. [33] perloi med a dose-escalation study 
withTheraSpherein43colorectalmetastasespatients. 
The study assessed dose-related effects on survival. 



.'.-'''0 Ajilioc'r.^TO.izLiiioij :■:■: ~.\w Treiitme; 1 ^ of Liv, 



i 27 patients 



tumor response and toxicity. There we 
threatening or fatal toxicities. The medi; 
was 408 (95% CI=316-565) days. Tumor response 
was evaluated by decrease in tumor size assessed 
by CT imaging. By these criteria, two patients had a 
complete response, eight had a partial response, and 
35 (81%) were at least stable. Higher doses were asso- 
ciated with greater tumor response and increased 
survival (p=0.05). In addition, tumor hypervascu- 
larity (p=0.01), higher performance status [base- 
line] (p=0.002) and less liver involvement(p=0.004) 
were associated with enhanced response or survival. 
Clinical toxicities included duodenal/gastric ulcers 
in six patients (14%) that resolved with medical 
management. These were most likely due to inad- 
vertent deposition of microspheres into the GI tract 
via unappreciated collateral vessels. Other related 
complications included single 
fever and fatigue. There was no do 
toxicities observed in the study. 

Salem et al. [34] presented da 
with colorectal metastases treated with Thera- 
Sphere. Patients who had life-threatening colorec- 
tal metastases to the liver for whom other therapies 
were judged to be inappropriate or had failed were 
treated. The majority of patients entering the study 
had extrahepatic lesions (85%), 89% of patients had 
undergone prior systemic chemotherapy, 52% had 
bilobar disease, and 22% had >25% of their liver 
replaced by tumor. Patients underwent baseline CT 
and 18 F-FDG-PET imaging and follow-up imaging 
for determination of efficacy. Metabolic response 
was also evaluated by CEA levels. Greater than 
80% of patients displayed response to treatment 
assessed by 18 F-FDG-PET. The response observed 
via CT imaging was less dramatic but paralleled the 
l8 F-FDG-PET results. Almost all (96%) of patients 
showed stabilization or response by one of the two 
imaging methods. There was an increase in survival 
tor patients with <25% tumor repla 
[95% CI=250-481] days) versus tl 
(162 [CI=153-237], p=0.0001). The overall median 
survival was 286 [CI=218-406] days, likely as a 
result oi the prevalence of extra hepatic lesions in tile 
majority of the patients (23/27). However, patients 
with an ECOG of (n=17) had a median survival of 
406 [CI=250-490] days. Treatments were well toler- 
ated with most events being transient (mild fatigue 
[n=13], nausea [n=4], and abdominal pain [n=5]) 
and resolving without medical intervention. Six 
patients (22%) experienced non-treatment-related 
ascites/pleural effusion or laboratory toxicities as a 
consequence of liver failure in advanced-stage, met- 



t (339 
: with >25% 



astatic disease. The response rate compared favor- 
ably to hepatic arterial chemotherapy and fewer 
complications were anticipated due to the relatively 
simple procedure required and the minimal toxicity 
associated with TheraSphere treatment. The authors 
concluded that TheraSphere appeared to provide 
therapeutic benefit with minimal toxicity inpatients 
with progressive metastases following failure on 
systemic chemotherapy. In an ongoing study at our 
institution, a cohort of 65 patients with metastatic 
disease to the liver troin diverse primaries including 
colorectal, pancreatic, melanoma, lymphoma, blad- 
der, breast, and neuroendocrine were treated with 
TheraSphere. All patients were treated on an outpa- 
tient basis. Tumor response rate using RECIST cri- 
teria was approximately 35%, while the 18 FDG-PET 
response rate was significantly higher. Response 
rates were accentuated in those patients who under- 
went systemic chemotherapy following a full course 
of liver-directed therapy with TheraSphere. 



12.5.2 

SIR-Spheres ': Clinical Experience, Response, 

and Survival 

Gray et al. [35] published a phase III randomized 
clinical trial of 74 patients conducted to assess 
whether a single injection of 90 Y in combination 
with intrahepatic FUDR could increase the tumor 
response rate, time to disease progression in the 
liver, and survival compared to FUDR alone. Treat- 
ment-related toxicities or change in quality of life 
were also examined. All patients had undergone 
complete surgical resection of a primary adeno- 
of the large bowel, and only those with 
:table metastases limited to the liver and 
lymph nodes in the porta hepatis were included in 
the study. In addition, patients were required to 
have a WHO performance status of 0-2, adequate 
hematological and hepatic function, and not have 
evidence of cirrhosis or ascites. Both treatment 
arms received 12-day cycles of continuous infusion 
floxuridine (FUDR) at 0.3 mg/kg of body wt/day 
that were repeated at four weekly intervals, and 
continued for 18 cycles (or until evidence of tumor 
progression, extrahepatic metastases requiring a 
systemic chemotherapy change, unacceptable tox- 
icity, port failure, or at the patient's request). The 
SIR-Spheres treatment arm also received a predeter- 
mined quantity of 90 Y that varied (2 GBq, 2.5 GBq, or 
3 GBq) depending on the size of the tumor. Yttrium- 
90 microspheres were administered one time only, 



within four weeks of insertion of the hepatic artery 
access port. The mean S0 Y dose administered was 
2.156 +/- 0.32 GBq. There was no difference between 
the 90 Y arm and control arm in the mean chemo- 
therapy dose (1,863 +/- 1,735 mg FUDR vs. 1,822 
+/- 1,323 FUDR per patient) or the mean number 
of cycles of chemotherapy (8.7 +/- 5.6 vs. 8.0 +/- 5.0 
cycles per patient). Six of 34 patients (18%) in the 
hepatic artery chemotherapy (H AC) arm had at least 
a PR, while 16/36 patients (44%) in the HAC + SIRT 
arm had at least a PR. (p = 0.01). 

Stubbs et al. [36] published a clinical trial of 50 
patients with extensive colorectal liver metastases 
not suitable for either resection or cryotherapy. The 
study compared experience with S0 Y alone (n = 7) 
and in combination (n = 43) with fluorouracil (5-FU). 
For all patients, 90 Y microspheres were administered 
as a single treatment within 10 days of hepatic artery 
port placement. The dose was titrated to the esti- 
mated extent of d isease (< 25% liver replacement: 2 
GBq, 25-50% liver replacement: 2.5 GBq, and > 50% 
liver replacement: 3 GBq) and given over 10 minutes, 
a few minutes after 50 meg angiotensin II. Forty- 
three of the 50 enrolled patients also received 5-FU 
given at the time of ,0 Y continuously over 4 days 
(1 gm/day), every 4 weeks. Prior to administration 
of ?0 Y, a Wm technetium-labeled macroaggregated 
albumin ( ?9m Tc-MAA) test was conducted to discern 
the percentage of lung shunting and assess the risk 
of radiation pneumonitis. Acute pain and/or nausea 
was experienced in 14 patients (28%) at the time of 
administration of 90 Y, and was managed with nar- 
cotics and antiemetics. Six patients (12%) developed 
an acute duodenal ulcer within 2 months after m Y 
therapy and the initial cycle of 5-FU that was due 
to misperfusion of the duodenum by either 9ft Y, 5- 
FU, or both. Antitumor effect was assessed by tumor 
marker (CEA) and CT response. Median CEA levels 
were reduced to 25% of baseline values atone month 
post-treatment with 90 Y, and remained < 30% of 
baseline when followed for 6 months. Median sur- 
vival for all liver metastases patients from the time 
of diagnosis was 14.5 months (range 1.9 to 91.4) and 
from the time of treatment was 9.8 months (range 
1.0 to 30.3). 

Stubbs et al. [37] published on 38 patients with 
extensive colorectal liver metastases who received 
SIR-Spheres. Liver involvement was < 25% in 19 
patients, 25%-50% in 9 and > 50% in 10. Patients 
received 90 Y in the hepatic artery via an arterial port 
and subsequent 4-weekly cycles of hepatic artery 
chemotherapy with 5-fluorouracil. The treatments 
were well tolerated, and no treatment-related mor- 



tality was observed. Response to SIR-Spheres ther- 
apy, as indicated by decreasing tumor markers and 
serial 3-monthly CT scans were seen in over 90% of 
patients. Estimated survival at 6, 12, and 18 months 
was 70%, 46% and 46%, respectively, and was prin- 
cipally determined by the development of extrahe- 
patic metastases. The authors concluded that SIR- 
Spheres was well tolerated in patients with extensive 
colorectal liver metastases and achieved encour- 
aging liver tumor responses, which are well main- 
tained by hepatic artery chemotherapy. 

Van Hazel et al. [26] published a randomized 
clinical trial of 21 patients with untreated advanced 
colorectal liver metastases (with or without extrahe- 
patic metastases) that compared the response rate, 
time to progressive disease, and toxicity of systemic 
5-fluorouracil/leucovorin chemotherapy versus 
5-fluorouracil/leucovorin plus a single adminis- 
tration of °°Y. Systemic chemotherapy consisted of 
425 mg/m 2 fluorouracil + 20 mg/m 2 leucovorin IV 
bolus for 5 days, and repeated in 4 weekly cycles. 
Mean SIR-Spheres activity infused was 2.25 GBq. 
There were five cases of grade 3/4 toxicity follow- 
ing FU/LV and 13 cases following combined SIRT + 
FU/LV, which were primarily elevated liver function 
tests. Tumor response rates using RECIST criteria 
were 10/11 (91%) and 0/10 (0%) for the combined 
chemotherapy/ S0 Y and chemotherapy only arms 
respectively (p<0.001). The time to progression was 
significantly greater in those receiving combination 
therapy compared to the chemotherapy arm (18.6 
vs. 3.6 months, p<0.0005). Survival was signifi- 
cantly improved in the combination arm compared 
to chemotherapy (29.4 vs. 12.8 months,p=0.02). The 
authors concluded that a single administration of 
,{I Y significantly increased both tumor response and 
time to progressive disease when added to systemic 
FU/LV chemotherapy, with acceptable toxicity. 
Studies are now underway to assess the efficacy of 
combining SIR-Spheres with oxaliplatin, irinotecan 
and bevacizumab based therapies. 

Rubin et al. [38] presented a case report of a 
patient with metastatic breast cancer to the liver 
treated with SIR-Spheres. The authors concluded 
that using an integrative approach to cancer treat- 
ment including SIR-Spheres was successful in pal- 
liating a patient with metastatic breast cancer. Boan 
et al. [39] presented data on a nine-patient cohort 
with primary and metastatic disease to the liver 
treated with SIR-Spheres. Although no response, 
toxicity or long-term data was presented, the authors 
concluded that SIR-Spheres was well-tolerated by all 
patients and that further evaluation was underway. 



.'.-'''0 Aadioeraae.izaiioij :■:■: die Treatment of Liv, 



Coldwell et al. [40] presented an abstract 
describing 84 patients receiving 127 infusions for 
colorectal metastases to the liver. The target dose was 
90 Gy to the tumor and 30 Gy to the normal paren- 
chyma. Objective response rates were 35% by CT, 
70% by CEA, and 90% by l8 FDG-PET. Mean follow- 
up time was 12 months, with median survival not 
having been reached at the time of presentation. No 
life-threatening toxicities were noted. All patients 
who exhibited fluoroscopic cessation of blood flow 
(stasis) during 90 Y infusion experienced post-embo- 
lization syndrome. The authors concluded that 
radioembolization provides encouraging response 
rates with an improvement in overall survival with 
acceptable toxicity in the group of patients treated. 



control the progression of radiation hepatitis. With 
radioembolization, care must be taken to ensure 
that the normal parenchyma is not receiving exces- 
sive radiation to a level where radiation induced 
liver disease might occur. Finally, it must be stated 
that the true mechanism of radiation hepatitis from 
radioembolization is nut currently understood. Any 
evidenci ■' ■' ii ■■■ ■ . otitis (e.g. anicteric ascites, 
elevated alkaline phosphatase, elevated transami- 
nases [42] represents an extension of the knowledge 
gained from external beam radiation. 



12.7 

Conclusion 



12.6 

Complications 



The most common complications of radioemboli- 
zation include nontarget radiation (GI ulceration, 
pancreatitis), radiation pneumonitis, and radiation 
induced liver disease (radiation hepatitis). The inci- 
dence of nontarget radiation should be minimized 
it the above-described technical principles are fol- 
lowed, including aggressive embolization of collat- 
eral vessels and the use of fluoroscopic guidance. 
The risk of radiation pneumonitis is mitigated if 
dosimetry planning incorporates the 30-Gy lung 
limit. The last possible complication of radioem- 
bolization is radiation hepatitis. This mechanism 
involves the irradiation of normal parenchyma 
beyond that which is tolerated. Ingold et al. [41] 
published a landmark series on radiation hepatitis 
in a cohort of patients treated with whole abdomen 
external beam radiation for gynecologic malignan- 
cies. The classical findings of anicteric ascites, ele- 
vated alkaline phosphatase, thrombocytopenia, and 
veno-occlusive disease occurred in those patients 
receiving greater than 30 Gy to the liver. Although 
the mechanism of selective "internal" microsphere 
radioembolization is distinctly different than exter- 
nal beam radiation, liver failure in this unique form 
of radiation hepatitis is a possibility. When radio- 
embolization with microspheres is undertaken, the 
objective is to administer the microspheres to the 
tumor without affecting the normal parenchyma. 
However, if the normal parenchyma receives a 
threshold dose above a yet undetermined amount for 
this mode of therapy, irreversible liver failure will 
invariably ensue. Systemic steroid treatment may 



There is a significant body of evidence supporting 
the safety and effectiveness of radioembolization in 
the treatment of metastases to the liver. The afore- 
mentioned studies representing the collective clini- 
cal experience supporting the safety and therapeutic 
benefit of TheraSphere and SIR-Spheres in patients 
with metastatic disease to the liver suggest further 
investigation for additional applications. Disease 
states where further work should be initiated include 
HCC (primary therapy, randomization against 
TACE, bridge to transplantation, tumor downstag- 
ing), colorectal (combination with radiosensitizers 
[capecitabine, CPT-11] or newer agents [oxaliplatin, 
bevacizumab, celuximab]). and metastatic neuroen- 
docrine cancer to the liver. Furthermore, this tech- 
nology appears to be ideal for extrahepatic applica- 
tions, such as the treatment of renal cell carcinomas, 
meningiomas, as well as other malignancies that 
are readily accessible am;ioj;raphically. There is also 
much to be gained from a more rigorous approach 
to investigating patient selection criteria, presenta- 
tion of disease, and optimal dosimetry to obtain the 
desired therapeutic effect given these factors. 



References 

1. Breed is C, Young G (1954) The blood supply of neoplasms 
in the liver. Am I Pathol 30:969-985 

2.SchenkWG,McDonaldJR,McDonaldK,DrapanasT(1962) 
Direct measurement of hepatic blood l"ew in *urpical 
patients. Ann Surg 156:463 

3. Lin G, Junderquist A, Hagerstrand I, Boijsen K (1984) 
Post-iv.eitem examination of die blood ■■i;."; , y a::d vas- 
cular pattern of small liver metastases -.- —.an. Surgery 
96:517-526 



4-Grindky |H, Her rick IF, Mann FC (1941] Measurement of 
the blood flow of the liver. Am J Physiol 132:489-496 

5.TyastrupN,WinlklerK,MellemgaardK,etal.(1962)Deter- 
m j nation of the hepatic arterial blood flow ami oxygen 
supply in man by clamping ihe hepatic artery during sur- 
gery. ] Clin Invesl 41:447-454 

:-. Almersio O. riengmark '•. iingevik L e: al. !19oo! Hepatic 
artery ligaiion as pretreatmenl for aver resection of meta- 
static cancer. Rev Surg 23:377-380 

7.BiermanHR, Byron RL, Kelley KH et al. (1951] Studies on 
die blood supply o: tumor in man ill. Vascular patterns 
of the liver by hepatic arteriography in vivo. JNCI 12:107- 
131 

S.Healy JE (1965 /Vascular pal terns in human metastatic hver 
tumors. Surg Gynecol Obstet 120:1187-1193 

9.Blanchard RJ, Grotenhuis I, Lafaye JW et al. (1965) Blood 
supply io hepatic VI carcinoma imp-ants as measured by 
radioactive microspheres. Proc Soc Exp Biol Med 118:465- 
463 

10. SundqviuslK.HafstromL.PerssonB (1978) Measurements 
of total and regional tumor [Mood flow and organ blood 
flow using Tc99m label. e; ; . microspheres. Fur 1 Surg Res 
10:433-443 

11. Gyves ], Ziessman HA, Ensminger WD et al. (1934) Defini- 
tion of hepatic tumor microcirculation by single photon 
emission compulei .;\-a lomegi ;v: ay : <]■■■£ C7 ;. | Nucl Med 
25:972-977 

12. Andrews JC, Walker SC, Ackerman RJ, Cotton LA, Ens- 
minger WD, Shapiro h (1967) Hepatic ladieembolization 
with yt-.ir.n-i-^O containing y.a ,; s rccio-spheres: Prelimi- 
nary results and clinical follow-up. i Nucl Med 35:1 o37— 
1644 

13.Leung TWT, Lau WY, Ho SKW et al. (1995) Radiation 
pnettmon.ilis after selective internal radiation treatment 
with intra -arterial 90 -ytirium- micro spheres for inoperable 
hepatic tumors. Int I Radiation Biol Phys 33:919-924 

U.Liu D, Salem R, Bui. JT, Courtney A, Barakat O, Sergie Z, 
Atassi B, Barrett K, Lewandowski RJ, Wong CO, Gates VL, 
Thurston KG Angiographic considerations in patients 
undergoing .irer-directed Iherapy with yl'rijm 90 micm 
spheres and chemoenibol;/i'.:'ii i ...imtrehctisivL review. 
J Vase Interv Radiol 2005, n press 

15. Salem R, Thurston KG, Carr Kl, ;lo;n |K, Gesi:hwi::d Jh' 
(2002) Yttrium-90 microspheres-. Kadiahrn therapy for 
ttnreseclable liver cancer. I V.isc Inte-v h;d:ii| 13: 22JS 
229S 

16. Salem R, Lewandowski RJ, Roberts C, Cicun J K, Thurston 
KG, Abouljoud M, Courtney A (2004) Use of Yllnum 90 
Glass Microspheres (TheraSphere) for the Treatment of 
Unresectable !-!epaiocelluls* t^r.iroma in Patients with 
Portal Vein Thrombosis. J Vase Interv Kadiol 1 1:33 5 345 

17. Carr BI (2002) Hepatic iriery chLmue—.fi: ti?aliori for 
advanced Siage HCC: exp. -it*.,e <\ $•'■'. p£r:i-nK Itt'pitio 
gastroenterology 43:7 t; -?:- 

18.ChunHJ,Byun JY.Yoo SS. C'i:; KG (200.1) Ajde^ benefit 
of thoracic aortography after :rar.vi:-cria. enir>ivi?alion ir. 
parents ivi:h hemoptysis. - iv»i , :::(!i , ::.»l Irf' 1 ■'.<//' 

1531 

19. Chung ]W, Park JH, Han JK, Choi BI, Han MC, Lee HS et aL 
; ] '"■'>o i Hepatic minors: arecisposing factors lb: complica- 
tions of Iranscatliete: oiiy chenieerabolization. Radii:- logy 
198:33-40 

20.1naba Y.Arai Y.Matsueda K, Takeuchi Y.Aramaki T (2001) 



Righl gasinc artery embolization :o- prevent acute gastric 
mucosal lesions in pal lents undergoing ivaeat hepatic ane- 
ria! infusion chemotherapy. I Vase interv Kadiol 12:957- 
963 

21. Ueno K, Miyazono N, Inoue H, Miyake S.Nishida H, Nakajo 
M :]''"''5j FmPo.ization o: Ihe hepatic falciform artery :o 
prevent supraumbilical skin rash during trail scatheter 
arterial chemoen'.bojzaiion for hepatocellular carcinoma. 
Cardiovasc Intervent Radiol 18:183-185 

22.Arora R, Soulen MC, Haskal ZJ. Cutaneous complications 
of hepatic chemoembolizaiion via extrahepatic collaterals 
(1999) J Vase Interv Radiol 10:1351-1356 

23. Yttrium-90 microspheres (TheraSphere) Package Insert, 
MDS Nordion.Kanata, Canada 2004 

24. Kennedy AS, Nutting C, Coldwell D, Gaiser J, Drachenberg 
C (2004) Pathologic response and microdosimetry of U "Y 
microspheres in man: Review of four explnated whole 
livers. Int. J Radiation Oncology Biol Phys 60:1 552-1 563 

25. Yttrium-90 microspheres (SIR-Spheres) Package Insert, 
Sirtex Medical, Lake Forest, IL 2002 

26. Van Hazel G, o.ackwel! A. Anderson 1, l-rice D, Moroz P, 
Bower G, Cardaci G, Gray B (2004) Randomised phase 2 
trial of SIR-ipheres clus fluorouracil/leucovorin chemo- 
therapy verses flu orouraci l/leu covor in chemotherapy alone 
in advanced colorectal cancer. | Surg Oncol 88:78-85 

27. Medical College -:A Wisconsin. htlp://wYvw.Jnimed.mcv.-. 
edu/clinical/body.html 2005 

28.Berger, M.J (1971) Distribution of absorbed dose around 
point sources of electrons and beta particles in water and 
other media, fournal of Nuclear Medicine Suppl 5: 5-23 

29.Synder WS, Ford MR, Warner GC, Waston SB. Absorbed 
Jose per unit cumulate J activity ror selected radionuclides 
.)n.:. organs., in Ml HP Pamphlet Number : 1 . New York, NY: 
Society of Nuclear Medicine, 1975-1976 

30.Herba MJ, Thirlwell MP (2002) Radioemlu: lira lion for 
hepatic metastases. Semin Oncol 29:152-15" 

31. Wong CYO, Salem R, Roman S et al. (20H2) Kvalualtng 
■"'Y-glass m.c iii-spa ere treatmenl response t i .;irc%e.la'ik 
colorectal liver metastases by [ 10 F] FDG PET: n comparison 
with CT or MRI.Eur J Nucl Med 29:815 

32. Wong C-YO, Salem R, Qing F, Wong KT, Barker D, Gates V 
ei a I. ( J 004) Metabolic response after int :a arterial L 'Y-Glass 
microsphere treatment for colorectal liver metaslases: A 
comparison of quantitative and visual analyses by '-F-FPG 
PET. J Nucl Med 45:1 392-97 

33. Goin JE, Dancey JE, Hermann GA, Sickles CJ, Roberts CA, 
Macdonald JS (2003) Treatment of unresectable metastatic 
colorectal carcinoma to the live: with intrahepatic Y-90 
microspheres: Jose-rangaig study. World j of N'uc Med 
2:216-225 

34. Salem R, Thurston KG, Goin JE, Wong CO, Lewandowski 
RJ, Gates VL et al. (2005) TheraSphere for unresectable 
metastatic carcinoma io the aver: treatment response al 
targeted doses of 135-150 Gy as measured by FDG-PET and 
CT imaging, in press 

35. Gray E, Van Hazrl G. Ho-ae M, huiton M. Moroz P.Anderson 
J, Gebski V (2001) Randomized trial of SIR-Spheres plus 
chemotherapy vs chemotherapy alone for i real in g patients 
with live: metastases from primarv large bowel. Annals of 
Oncology 12:1711-1120 

36. Stubbs RS, Cannan RJ, Mitchell AW (2001 ) Selective Internal 
Radiation therapy with Yltiiuni -90 microspheres lor exlm- 
sive colorectal metastases. J Gastrointest Surg 5:294-302 



Ylr n u iV. -'''!'■ jv.idioer.too.izaiion for the Treatment of liver \:ela:ta- : ir-; 



37.StubbsRS,Cannan RJ, Mitchell AW (2001) Selective inter- 
im' radiation therapy (S1RT) with 90Yttrium microspheres 
for extensive colorectal live: metastases. Hepatogastroen- 
terology 48:333-337 

38. Rubin D, Nutting C, [ones B (2004) Metastatic Breast 
Cancer in a 54 year old woman: [ulcerative Treatment with 
Yttrium-90 Radioembolization. Integrative Cancer Thera- 
pies 3: 262-267 

39. Boan JF, Marti-Climont JM, Martinez A, Sangro B, Rodri- 
guez I, Penuelas I, Richter JA (2004) Abstract P954, Euro- 
pean lotirn.'.! of Nil o lea: Medicine and Molecular Imaging 
Vol31,Suppl.2 



40.Coldwell D, Nutting C, Kennedy A (2004) Initial clinical 
results in the treatment of unresectable hepatic minors 
with resin-o-ar.ed yttriii:i:-90 radioetnbolization. Presented 
at the Cardiovascular .:w\.:. Interventional Radiological Soci- 
ety of Europe (CIRSE) conference. September 25-29 

41.Ingold JA,Reed GB, Kaplan HS, Bagsjaw MA (1965) Radia- 
tion hepatitis. Am ] Roentgen 93:200-208 

42. Cheng JC.Wu JK, Huang CM.HuangDY, Cheng SH, Lin YM, 
Jian J], Yang PS, Chuang VP, Huang AT (2002) Radiation- 
. :'d. i ce'1 liver c:se.:se after radiotherapy for hepatocellular 
clinical manifestation and dosimetric descrip- 
l. Radiother Oncol Apr;63(l):41 -45 



13 Portal Vein Embolization 



Alain J. Roche and Dominique Elia 



Introduction 163 

Phytopathology 163 

Portal Blood and Liver Regeneration 163 

Portal Vein Emboliz.it i on and Liver 

Regeneration 164 
1 Mechanisms Inducing Regr:'.era;ion After PVE 164 
1 is Winnie E j u j" e l; s e Cone!a:ed to ¥:.w. iional 

Increase? 1 64 

IndicaliiT. 1 :. ' Portal Vein Embolization 164 

Calculation . i FRL.'Totrd Etiitcriomu Liver Volume 

Kami ifii 

Hepatocollular Carcinoma 165 

;:'::: jr'i; -..■.'linoma 155 

l.iver Metastasis 155 

utuel jiuIj nations 156 

Anatomy 165 
Technique 156 

Personal Technique 165 
Other Techniques 16S 



.1 I Usui Embolization or Proximal Ligation? 16S 
.3 Embolic Agents 169 

Complications 169 

Tolerance 169 

Complications 170 

Does PVE Accelerate Growth Rate of Liver 

Metarnasesr 170 

Results 171 

Liver Hemodynamic Effects of Portal Vein 

Embolization 171 

Induced Hypertrophy, Delay to Surgery 171 

Hvlv: ; ropery :n Paiienis w:t:'. LLodei -ying Liver 

Disease 171 

Is Hypertrophy Predictable? 172 

Is Liver Failure After Surgery Predictable? 172 

Long- term Rrsuks jnd Survival 172 

Future Developments and Research 172 

Conclusion 173 

References 173 



13.1 

Introduction 

One of the prerequisites tor partial hepatic resection 
is the presence of enough remaining functional liver 
parenchyma to avoid life-threatening postoperative 
liver failure. Therefore, the possibilities of curative 
resection of liver tumors are strongly dependent on 
the volume of the future remnant liver (FRL). In clini- 
cal practice, these possibilities are frequently limited 
when an extended right hepatectomy is mandatory 
since the lef t lobe is small, or when major liver surgery 
is indicated in patients with impaired liver function, 
whatever the cause (cirrhosis, cholestasis, noncir- 
rhotic fibrous disease, or severe fatty steatosis). The 
aim of portal vein embolization (PVE) is to selectively 
induce hypertrophy of the FRL during the preopera- 
tive period. This is achieved by embolization of the 
intrahepatic portal branches of the future resected 
liver, therefore leadinj', to distribution of the entire 
portal blood flow, containing hepatotrophic factors, 
exclusively towards the FRL. Lhis technique was first 
applied for patients with hepatocellular carcinoma 
[29] with the double goal to induce hypertrophy of the 
FRL and to prevent retrograde intraportal tumor dis- 
semination from the tumor lobe. Fewyears later, PVE 
was proposed in patients with hilar cholanpo.arvi- 
noma carcinoma who frequently require major hepa- 
tectomy in relation to their tumor location [39], and 
in patients with liver metastasis where PVE extends 
the indications for curative surgery [■!?]. 



13.2 
Physiopathology 



13.2.1 



A. J. Roche, MD 

Head of I nter ven 1 1 om; I Radiology Sec 

C-u stave Rotissy, 39 Rue Cunulle Pesr 

Cedex, France 

_ _ .,_ It nas been c 

D. Elias, MD 

Head of Digestive Surgery Section, Institut Gustave Roussy, 39 branch ligat 

Rue Camiile Pesmoclins, 94800 Villejuif Cedex, France sponding lobe and hypertrophy of the contralateral 



d for a long time that portal 
shrinkage of the corre- 



A. I. Roche and D.Elia: 



one. Moreover, it is well known in clinical prac- 
tice Chat liver trophicity closely depends on hepatic 
portal blood perfusion. Liver atrophy after surgi- 
cal or spontaneous portocaval shunting, hypertro- 
phy of the remnant liver alter partial hepatectomy, 
Spiegel lobe hypertrophy in Budd-Chiari disease 
where the caudate lobe remains the only one to still 
have hepatopetal portal blood flow, occlusion of 
portal veins from cholangiocarcinoma giving rise 
to hypertrophy of the unaffected lobe, are some 
examples illustrating this very close relationship. 
It was established along the seventies that portal 
venous blood flow promoted hepatic cell regenera- 
tion [8] and that blood arising from duodenopancre- 
atic area had strong hepatotrophic properties [50]. 
Insulin and glucagon were then soon recognized 
as growth -regulatory factors which, when infused 
concomitantly, synergistically stimulated hepatic 
regeneration. More recently, hepatocyte growth 
factor (HGF) could be isolated in different labora- 
tories and described to rise after partial hepatec- 
tomy. Multiple other factors such as cytokines or 
transforming growth factor-alpha (TGF-a), have 
also been demonstrated to play a role in hepatic 
regeneration. 



13.2.2 

Portal Vein Embolization and Liver 

Regeneration 

13.2.2.1 

Mechanisms Inducing Regeneration After PVE 

(Table 13.1) 

Occlusion of portal branches of the liver paren- 
chyma to be resected redistributes the totality 
of its portal blood flow, and consequently all its 
hepatotrophic contents, towards the FRL. This is 
the basic rationale of the method that triggers off 
regenerative activity of the nonembolized portion of 
the liver. Moreover, PVE dilates the portal branches 
in the FRL, exposing liver vasculature to stretch 
stress which act as a trigger for IL-6 release from 
endothelial cells and contribute to the activation 
of regenerative cascade in the FRL [26]. Induction 
of heat shock protein in the nonembolized lobe is 
supposed to have similar effects [40]. PVE also acts 
through two potentially complementary pathways 
specifically related to embolization: ischemia and 
inflammation. With most of the embolic agents, 
PVE induces a mild ischemia: apoptosis or necrosis 
of some hepatocytes, and intercellular disjunction. 



These lesions lead liver cells of the embolized liver 
to produce regenerating factors [35]. Moreover, the 
injection of embolic material induces a foreign body 
reaction and a cascade of inflammatory phenomena 
with production of cytokines and liver growth fac- 
tors by Kup tier 1 cells and yiamilocytes. This pathway 
may be more or less predominant, depending on 
the intensity of the inflammatory reaction induced 
by the embolic agent used for PVE. Consequently, 
embolic agents inducing a strong inflammatory 

:h as cyanoacrylate or ethanol, should 

hypertrophy than others. 



13.2.2.2 
fs Volume 
Increase? 



Correlated to Functional 



Even if volume increase of the FRL after PVE was 
demonstrated by all the first publications, it had 
to be proved that it corresponded to a functional 
increase. Nowadays it has been clearly demon- 
strated by the mean of many different techniques 
that volume increase did parallel function increase. 
PVE produces a significant increase in bile volume 
and biliary indocyanine green concentration in the 
FRL [53]. Histology examination of FRL and assess- 
ment of volumetric, cell kinetic and morphometric 
parameters attributed to PVE a gain of functional 
hepatocyte mass and early induction of hepatocyte 
proliferation following hepatectomy [19]. Levels of 
erythrocyte polyamine, that is known to be related 
to liver regeneration, increase along the seven days 
following PVE [52]. Hepatic energy charge levels in 
the FRL remain comparable to that of normal liver 
[9], hepatic plasma clearance of sorbitol and antipy- 
rine were stable after PVE [48], while the percent- 
age of FRL to total liver volume increased, thereby 
demonstrating that the functional reserve of FRL 
increased. Functional improvement after PVE has 
also been demonstrated through evaluation of the 
first-pass lidocaine extraction [51] and (99m)Tc- 
galactosyl serum albumin scintigraphy [34]. 



13.3 

Indications for Portal Vein Embolization 

Limits for hepatic resections, and consequently for 
PVE, depend on multiple factors. The age of the 
patient is one of these: the younger the patient is, 
the larger a resection is tolerated. Liver function 



> : ::'iT;i. Veii: t.iuLx'lizniii.'--:''. 



Table 13.1. Mechanisms involved in i'.v^ertiopiiy Hirer PVii. Cv:o/:nes .111.:. grown h f.iciors produced by the 
embolized liver need recira.l.'.Uon to be eriecUve upon FRL. 



PORTALVEIH EMBOLIZATION 







is obviously a predominant factor for de 
ing the limit for a safe resection. Bilirubin 
indocyanine green clearance and presence or not 
of a cirrhosis are widely used as parameters for 
evaluating liver function before hepatic resection. 
Liver function is considered as normal if there is 
no jaundice and indocyanine green clearance at 
15min is <10%. Presence of jaundice or ICG clear- 
ance 10%-20% indicates mild liver dysfunction.ICG 
clearance >20% indicates more severe compromised 
liver, also meaning that PVE should not be followed 
by dramatic increase in FRL volume. At last, it is the 
FRL/Total functional liver ratio for a given patient 
(with or without compromised liver tunction) that 
determines possibilities for a safe and complete 
resection. Depending on the authors, PVE is consid- 
ered when this ratio is expected to be <25%-40% in 
patients with normal liver function and <40%-50% 
in patients with liver dysfunction [3, 12, 32]. 



13.3.1 

Calculation of FRL/Total Functional Liver 

Volume Ratio 

Many studies have demonstrated that CT estima- 
tions of liver volumes in vivo were correctly corre- 
lated to real volumes despite partial volume effect, 
respiratory phase, or inter- observer variations. It is 
widely admitted that tumors do not contain func- 
tional hepatocytes. Consequently, tumor volume 



must be subtracted from that of liver. Peropera- 
tive radio frequency ablation (RFA) has been widely 
developed along the recent years. When RFA is 
planified for treating a tumor located in the FRL 
simultaneously to the hepatectomy, one should pay 
attention to subtract the supposed volume of the 
future RFA lesion (tumor volume + safety margin) 
from the total FRL volume. 



13.3.2 

Hepatocellular Carcinoma 

PVE was initially proposed in HCC at least for con- 
trolling retrograde tumor thrombus invasion in the 
portal vein [29], but today there is little evidence 
to support this supposed interest of the technique. 
On the contrary, some authors pointed out that the 
compensatory increase in arterial flow in the embo- 
lized lobe might accelerate the tumor growth [54]. 
Most of hepatocellular carcinomas occur in patients 
with compromised liver. Thereby this dramatically 
increases the risk of severe postoperative compli- 
cations and limits the possibilities for major cura- 
tive liver resections. Consequently, PVE is actually 
proposed in selected cases to extend indications for 
curative surgery and increase its safety. Apart from 
increasing the FRL volume and tunction, minimiz- 
ing the sudden increase in portal pressure at resec- 
tion in these cirrhotic patients may also be an advan- 
tage of preoperative PVE. 



A. [. fvodie.iiid li. t.i;v; 



13.3.3 
Cholangiocarcinoma 



Radical surgery of hilar cholangio 
ally necessitates major liver resection, mainly an 
extensive right hepatectomy. Such a major interven- 
tion is frequently impeded by a too small left lobe, 
which consequently indicates preoperative PVE. 
Moreover, the patient frequently develops obstruc- 
tive jatindice that may severely injured functionality 
of the FRL. Preoperative PVE is performed in these 
patients after biliary drainage of the FRL. Ideally 
patients should undergo PVE only when decrease in 
the serum bilirubin level is obtained, which facili- 
tate hypertrophy of the nonembolized sector. On 
the contrary, biliary drainage of the future resected 
parenchyma should not be performed in the preop- 
erative period to allow a more important shrinkage 
of the embolized territory. 



13.3.4 

Liver Metastasis 

Curative resection of liver metastases is mainly 
performed in patients presenting with colorec- 
tal primary cancer. Liver metastases are found in 
40%-70% of patients with a colorectal cancer. In 
about one third of cases, the liver is shown to be the 
only site of cancer spread, even at autopsy. There 
is no spontaneous long-term survival in untreated 
patients, whose median survival time range from 
6 to 18 months. Furthermore, liver involvement is 
the most important factor associated with decreased 
patient survival. However, at time of diagnosis, the 
majority of patients present unresectable tumors, 
and resection can be performed in <20% of all 
patients with colorectal liver metastases. The main 
limitation for resectability is the impossibility to be 
curative while leaving a sufficient residual amount 
of functional liver parenchyma. Consequently, pre- 
operative PVE may dramatically improve the pos- 
sibilities for a curative (RO) resection of liver metas- 
tases by increasing the volume and the function of 
the future remnant liver. 



huge benign adenoma (personal data), or even for 
allowing resection in primary sclerosing cholangitis 
[43]. 



13.3.5 

Other Indications 

PVE has been reported prior to resection of mul- 
tiple benign adenomas which dissemination in the 
liver parenchyma impeded curative surgery [11], 



13.4 

Anatomy 

A thorough knowledge of hepatic segmentation and 
portal venous anatomy is essential before perform- 
ing PVE. Variations frequently occur and must 
be well known. One may remember that almost 
all variations concern right branches, making left 
branches remarkably constant in their disposition. 
The most frequent variation is slipping from right 
to let! ol segment V anil VIM branches, separalelv 
or together. Therefore two main variations are fre- 
quently encountered: (i) trifurcation of the portal 
vein in left branch, segments V + V1II branch and 
segments VI + VII branch, when the slipping is lim- 
ited; (ii) bifurcation of the portal vein in a right vein 
limited to segments VI + VII and leftvein giving also 
rise to segments V+V1II branch, when the slipping 
is complete. Segmental branches of the right liver 
mayalso slip separately, but the most frequent varia- 
tions concern segment VIII branch that may slip to 
the left as well as to the right. 



13.5 

Technique 



13.5.1 

Personal Technique 



The procedure maybe performed undei 
sedation and analgesia but most of the teams prefer 
general anesthesia that provides more comfort for 
the patient as for the operator. When the goal of PVE 
is occlusion of right branches, the preferred access 
of the portal vein is mostly the anterior subxiphoid 
left route that allows antegrade catheterization of 
all right branches to be occluded and free flow 
embolization, thereby providing safer maneuvers. 
The puncture is achieved under sonographic guid- 
ance with a 5F-needle catheter (Table 13.2). When 
branches for segment IV have not to be occluded, the 
entry point in portal veins may be the Reix recess. 
If segment IV is concern by PVE, it is recommended 
to enter the segment III branch, upstream from the 
recess, to facilitate catheterization of segment IV 



Portal Vein Embolizi 



Table 13.2. Personal cookbook 

!-.[' ,; i I'.'.Ci'r 

Puncture: 5F-needle catheter, 27 cm (Cook) 
Catheterization: 

5F catheter from the needle catheter 

Guide wire: Kayak 0.035 J glide wire (Boston Scientific) 
Embolic agent: 

Cyanoaci yli'.te: Hisioacryi, 0.5 ml vials (B. Brjun) 

Lipiodol Ultrafluide, 10 ml (Guerbet) 
Embolization technique: 

Three-way stopcock resistant to Lipiodol (Cook) 

isotonic glucose solution. 

Two 1-ml syringes (Terumo) 

20 ml syringe 



Echotip needle, 18G,20cm (Allege 



iirae catheter with on demand shaping of the tip 
0.035 shapeable glide wire (Terumo) 



Embogold (700-900 urn) (Biosphere Medica"' 



branches. Retrograde catheterization of the portal 
vein forperforming a portography is the first step of 
the procedure in order to identify individual intra- 
hepatic branches and anatomical variations. In all 
patients with a known or suspected compromised 
liver, the portal pressure must be measured prior to 
embolization as it represents a prognostic param- 
eter. Catheterization of every branch to be embo- 
lized is then performed with the 5F catheter of the 
needle catheter device, with the help of a J shaped 
0.035 glide wire. In case of left approach and very 
tightened portal bifurcation, catheterization of the 
right portal vein may necessitate to extemporane- 
ously shape the catheter tip. Depending on indi- 
vidual anatomy, a 1-2 cm length and 30°-90° angu- 
lated tip is then shaped under steam to make further 
maneuvers easier. Every main trunk to be occluded 
is selectively catheterized for performing a distal 
and free flow embolization. The degree of selectivity 
(sectorial, segmental, or subsegmental) before each 
embolization depends on individual anatomy and 
local hemodynamic. It is chosen for each vein to 
ensure a stable selective positioning of the catheter, 
providing best conditions for free flow embolization 
and preventing from inadvertent reflux of embolic 
material. Massive reflux of embolic agent in the FRL 
would annihilate its hypertrophy, or induce almost 
total portal occlusion and thereby fatal portal hyper- 
tension when the rest of the portal vasculature has 
already been totally embolized. Consequently, right 
branches originating close to the portal bifurcation 
should be hyperselectively catheterized. Caution 
should also be exercised to avoid reflux into left lobe 
veins when occluding veins in segment IV. Due to 
this potential risk, segment IV portal veins should be 
occluded first for added safety, and sometimes even 
with particulate embolic agent instead of cyanoacry- 
late. As many authors, we mostly perform emboliza- 



tion with a mixture of cyanoacrylate and Lipiodol. 
Safe use of this embolic agent necessitates following 
a very strict technique but, to our point of view, 
presents multiple advantages. It permits to achieve 
complete and durable occlusion. Its radio-opacity 
increases safety at time of embolization. Histoacryl 
and Lipiodol are mixed in a ratio of 1 part of Histo- 
acryl for 1 to 3 parts of Lipiodol, the more Lipiodol 
in the mixture, the longer the polymerization time 
of the glue. Consequently, it allows distal emboliza- 
tion in every case since the polymerization time can 
be adapted to individual and instant hemodynamic 
variations. Furthermore, the cyanoacrylate induces 
a very strong inflammatory reaction, involving ves- 
sels as well as bile ducts, that is thought to increase 
production of hepatotrophic factors. The mixture 
(Histoacryl/Lipiodol ratio close to 1/2) is prepared 
in an insulin syringe, immediately prior to the first 
embolization. If needed during the procedure, it is 
possible to increase the dilution by adding Lipiodol. 
The mixture is pushed with isotonic glucose, follow- 
ing the "sandwich technique": the volume of every 
injection of mixture being lower than the catheter 
content. This is repeated as many times as necessary 
for obtaining a distal and complete occlusion. The 
total dose of Histoacryl will be 1-3 cc, administered 
in 4-6 successive injections of mixture. Catheter 
occlusion along repetitive injections of glue is a 
risk of this technique. Pushing the 0.035 glide wire 
through the catheter still in position, immediately 
after each injection of glue minimizes it. This cleans 
the inner wall of the catheter from residual glue/Lip- 
iodol mixture, and gently push it out in the embo- 
lized vein under fluoroscopic control. The three- 
way stopcock also needs to be cleared from residual 
glue after each injection. Anyway, both three-way 
stopcock and catheter must be exchanged in time 
before occlusion, at least every 3-5 injections. A 



A. [. Roche mid li. t.i;v; 



control portography is performed at the end of the 
procedure and postembolization portal pressure is 
registered. In our experience, the transhepatic tract 
does not need to be embolized. 



13.5.2 

Other Techniques 



been successfully used in cases of impossibility to 
perform the conventional transhepatic technique, 
due to tumor interposition or severely impaired 
hemostasis [46). The surgical transileocolic approach 
needs laparotomy but for some authors allows tumor 
extension to be better assessed preoperatively [21]. 
Nevertheless most of the surgeons actually prefer the 
transhepatic approaches. 



13.5.2.1 
Approaches 



13.5.2.2 

Distal Embolization c 



Prior to complex hepatectomiesPVE may c 
and right portal branches. A right transhepatic access 
may be then chosen, giving preference to entering a 
vein not to be occluded (Fig. 13.1). Some authors advo- 
cate a right transhepatic approach in all cases, using 
double- or triple-lumen balloon catheters for embo- 
lization [42], Recently, the transjugular approach has 



1 Proximal Ligation? 



Distal embolization is achieved with particulate 
agents, cyanoacrylate or other liquid agents. Proxi- 
mal ligation is surgically performed or may be 
done percutaneously with steel coils or detachable 
balloons. Considering that the intrahepatic portal 
vasculature was classically considered as terminal 



Fig.1J.1a d. PVE under right tr 

Mich la) for inducing 
<if!f;t!Vf hypertrophy of the posterior 
:::!.mr, of (he liver prior to hepa- 

Si'tMmy for metastases aiming to 
--esi-vt' ;:n.( segments VI and VII 
. icrylrite+Lipiod. 'I cast 

L'i emoonzed branches is 
pl'.in h.m :!>) :;:'d CT conlrcl prio 
surgery (ar 




Portal Vein Embolizi 



type, interest of performing a distal embolization 
rather than a proximal surgical ligation has been 
contested. Therefore, nowadays there are strong 
arguments for preferring a distal occlusion. The 
efficacy of PVE vs. right portal vein ligation before 
extended right hepatectomy has been recently dem- 
onstrated [6]. The increase in FRL volume was sig- 
nificantly higher with PVE (188 +/- 81 ml vs. 123 
+/- 58 ml; £=0.012). A strictly proximal occlusion 
allows distal reentry through the intra -parenchyma- 
tous vascular shunts opening [57]. After having con- 
trolled patients with a previous proximal ligation we 
also encountered antegrade porto-portal collateral 
circulation bypassing the ligation and supposed to 
be parabiliary veins homologue to the parabiliary 
arteries well known to play a predominant role in 
the development of intrahepatic arterial collateral 
circulation's. Lastly, in rare cases we encountered 
macroscopic disial intra hepatic porlo-pornil anas- 
tomosis (Fig. 13.2} that could also minimize the effi- 
cacy of a too proximal occlusion in some patients. 



13.5.2.3 
Embolic Agents 

Many embolic agents have been used and compared, 
either in animal experiments or clinical use. There is 
no definitive argument in the literature infavor of one 
specific type of embolus, excepted that the embolic 
agent must provide a distal occlusion (Table 13.3). 
Among particulate and liquid embolus, the actual 
tendency is to favor agents inducing complete, distal 
and durable occlusion as well as a strong inflamma- 
tory reaction. Most of teams actually seem to prefer 
Histoacryl/Lipiodol mixture. Absolute ethanol has 
also been experimentally assessed and clinically 
used by several teams [49, 57, 58]. Fifteen to 65 ml 
of ethanol had to be injected to induce an adequate 
occlusion, either in right portal vein or at a segmen- 
tal or subsegmental level. Technical easiness of its 
use and efficacy to inducing hypertrophy are clear 
advantages of ethanol. On the opposite, its clinical 
and hepatic biological tolerance appeared tobemuch 
poorer than that of cyanoacrylate. Consequently, 
one should be very careful in using this embolic 
agent, especially in patients with compromised liver. 
Recently, new agent have been studied in attempt to 
benefit from ethanol advantages while limiting its 
adverse effects [30] or using sclerosing agents com- 
bined with gelatin sponge [25]. Coils do not induce 
distal occlusion and are generally used combined 
with a distal embolus [38] or in rare cases where 




Fig. 13.2. ?-ponuinec-'.;s ii-iirLdiepjuc pixie-porta] 
in the right liver \iii rv n-sj, demon si :Li:ed d : .j mg a direct tran- 
shepatic po: togi jpj'.y under let': approach 

Table 13.3. Com; 1 .; red ertk.'.cy of uifrereat : .,s : .:.'.l embojc jgems 





Embolic agents 


k'.d'.Kvd 
.'.VLVl'MYohv 


nE BAEREet al. [10] 


Gelatin sponge 


53% 


i hv-.-ei iri.i-.--hv m 4 weeks) 


Coils 


44% 




cvunoacrvlate 


68% 


Kaneko etal. [25] 


ijehitin sponge 


21.9% 


(hvpertrophv ai 1 weeks) 


Fibrin glue 


13.5% 




cynnoa cry late 


25.0% 



an efficient and safe occlusion is not feasible with 
cyanoacrylate or particulate embolus. Particulate 
calibrated agents may also be used: polyvinyl alcohol 
particles [7] or Embospheres (personal data). 



13.6 
Complications 

13.6.1 
Tolerance 

Clinical tolerance is generally excellent with only 
mild abdominal pain or discomfort and slight fever 
which disappear in less than 3 days. A flush syn- 
drome may occur in the post embolization period in 

patients with carcinoid primary, and should be sys- 
tematically prevented. During the post PVE period, 



A. J. Roche and ['. Hihs 



prothrombin time remains above 70% of the base- 
line value. Serum aspartate transaminase and ala- 
nine transaminase slightly increase and may reach 
a maximum value of threefold the normal value on 
the first day post PVE, excepted when using ethanol 
that induces a greater cytolysis. Alterations in the 
total bilirubin level are insignificant. Normally, the 
total duration of hospitalization for the procedure 
does not exceed 3 days. 

At histopathological study, there is no macro- 
scopic difference between embolized and nonembo- 
lized liver. Inpatients embolized with gelatin sponge, 
small vessels remain patent even though the main 
portal branches are obstructed. In patients emboli- 
zed with Histoacryl (Fig. 13.3), portal vein walls are 
damaged with their lumen filled with embolic mate- 
rial and macrophage cells and periportal inflam- 
matory reaction and fibrosis are constantly associ- 
ated. A massive peribiliary fibrosis, as encountered 
in sclerosing cholangitis, is also observed in most 
of the cases treated with ivanoacrvlate. Specimens 
contain very rare and small foci of necrotic tissue, 
excepted for ethanol embolization where there are 
more important. 



13.6.2 
Complications 

In the literature from experienced teams, complica- 
tions rate rarely exceed 1.5%, without any reported 
mortality. Main reported complications include 
pneumothorax, subcapsular hematoma, arterial 
pseudoaneurysm a Her inadvertent arterial puncture 
and portal vein thrombosis [31]. The risks are higher 
in patients with portal hypertension and/or blood 




Fig. 1 3.3. I.iver hi^top.'di'.OA'gy of [lie reseaed paieiiii'.vma 
4 iveeks after postal embolization with Histoacryl. Portal 
branch (P) filled with cyanoacrylate (C). Bile duct (B) pre- 
senting a mosfivc pe:idik"ial lioiosis {arrows) 



coagulation disorders. In patients who underwent a 
duodenopancreatectomy and who chronically pres- 
ent an infected biliary tree, the PVE can be done 
without risk of hepatic abscess, contrarily to that 
occurs after intra-arterial chemoembolization or 
after radiofrequency ablation. 



13.6.3 

Does PVE Accelerate Growth Rate of Liver 

Metastases? 

It is well known that partial hepatectomy acceler- 
ates local tumor growth [45]. Some of the cytokines 
and growth factors involved in liver regeneration or 
hypertrophy may also be involved in tumor burden. 
For example, transforming growth factor- a is a 
strong stimulator of liver regeneration but has also 
been demonstrated to have stimulatory effects on 
tumor growth in vitro [36]. Consequently, one could 
suppose PVE to accelerate tumor growth of lesions 
located in the FRL. Our group studied volumetry of 
the FRL and that of liver metastases located in the 
FRL in five cases [13]. Volumes measurement prior 
to PVE and one month later showed that increase of 
the normal liver varied to 59%-127%, compared with 
60%-970% for the metastases. The ratio between the 
growth rate of the left lobe and the liver metastases 
varied from 1.0 to 15.6. However, the spontaneous 
growth rate of metastases prior to PVE, that should 
be subtracted to the total tumor growth to define the 
exact potential enhancement by PVE, was unknown 
in this study. More recently, Kokudo et al. demon- 
strated an increase in proliferative activity of intrahe- 
patic colorectal metastases inpatients who underwent 
PVE compared with a control non-PVE group [33]. 
The Ki-67 labeling index of metastatic lesions was 
significantly higher in the PVE group. Long-term sur- 
vival was similar in the two groups but disease-free 
survival was significantly poorer in the PVE group. 
To overpass this potential adverse effect, some have 
proposed a two-stage hepatectomy, PVE being per- 
formed after a primary resection of metastases that 
are present in the FRL [22], Radiofrequency ablation 
of lesions located in the FRL, prior or simultaneously 
to PVE, is also an alternative option (Fig. 13.4). To 
conclude, complementary studies are still needed but 
it is logical to consider that PVE accelerates tumor 
growth in some patients and some tumors. However, 
clinical experience demonstrates that this rare and 
probably limited adverse effect remains negligible 
compared to advantages of PVE in widely extending 
indications for curative surgery. 



Porro' Vein H 1 1 l _"> .l.;:i:i'- 




Fig. 1 j.4a,b. PVE ui'.dcj lei": ironsliep.Ui,: rippii-ridi prior to iigli: l"iepotee:o:v.y extend e.". to segment IV for over metasioses from 
ponoreiit!.: coremoid luiiior (iP.<fiT, : </ij. Perai to neons i :id:ol"requen.."Y o Not ion of o let": lobe single lesio:: (iinr:- 1 . ) is pel formed 
along the sonic session. Control portogiophy lb) nflei PVH. the triple eooled KV needle oeing in plo:e {crossed arrow ) 



13.7 
Results 

13.7.1 

Liver Hemodynamic Effects of Portal Vein 

Embolization 

When performed on a normal liver, right portal vein 
embolization induces an immediate mild increase in 
portal pressure, from 2 to 5 cm of saline. However, 
some minutes or even seconds after embolization, 
the portal pressure comes back to its initial value. 
In patients with abnormal liver parenchyma, portal 
pressure increase tends to be greater and more dura- 
ble. Interestingly, in both cases, the portal pressure 
registered immediately after PVE is similar to levels 
registered after hepatectomy, and subsequently may 
have a diagnostic interest in predicting the hemody- 
namic surgical outcome [55]. 



13.7.2 

Induced Hypertrophy, Delay to Surgery 

Along the last 10-year period we have performed 
PVE in 108 patients, before right hepatectomy in 43 
cases, right extended hepatectomy in 58 cases and 
more complex hepatectomy in 7 cases (6.4%). We 
have reported a mean increase of 70% in the size of 
the FRL and a ratio of FRL/Total liver that increased 
by 12.4% and measured 32% four weeks after PVE 
[12]. Main series report increases in volume of 30 
to 42 % and FRL/Total liver ratio increases from 
19%-36% to 31%-59%. Disparities maybe explained 



by different delays before surgery (2-4 weeks), use 
of diverse embolic agents and different proportion 
of patients with compromised liver in the respective 
studies [1, 21, 27, 28, 37, 58]. In animals, complete 
hypertrophy of the contralateral hepatic lobe occurs 
3-6 months after portal branch ligation. Hepatic 
regeneration follows an exponential curve, with a 
250% increase in the remnant volume during the 
first week after resection of 80% of the liver. Delay 
between PVE and surgery should be as short as pos- 
sible to preclude any tumor growth. In our expe- 
rience, all patients reached the critical FRL/Total 
functional liver volume ratio of 25% after 4-5 weeks. 
Consequently, as several other teams, we consider a 
4-5 weeks delay before surgery to be a good com- 
promise between hepatic hypertrophy and tumor 
dissemination. Other teams consider the hypertro- 
phy gain negligible after 2-3 weeks and prefer to 
perform the resection earlier. Interest of PVE prior 
to large resections was evaluated through retrospec- 
tive [2, 16, 20] or prospective [15] comparisons with 
non-PVE series. In all retrospective studies, FRL 
were significantly smaller at presentation in PVE 
group and thank to PVE were similar at surgery. 



13.7.3 

Hypertrophy in Patients with Underlying Liver 

Disease 

Almost all patients with normal liver experience 
hypertrophy after PVE since only 86% develops 
hypertrophy in patients with chronic liver disease 
[15]. Furthermore, in most of the studies, hyper- 



A. [. fvodie.iiid li. t.i;-< 



trophy is milder and slower (35%-40% i 
volume of the FRL at 1 month) than in normal liver. 
In damaged liver due to viral hepatitis, increase in 
volume of the FRL was 25% at 2 weeks, compared 
with a 34% value in normal liver [56]. In HCC with 
injured liver, Ji et al. recently reported FRL/Total 
liver ratio increasing from 36% to 40.8% at 1 week 
and 47.3% at 3 week [23]. More and more frequently, 
major hepatectomy for metastases has to be done in 
patients with liver fibrosis following long-term sys- 
temic chemotherapy or intraarterial infusion. We 
have reported that hypertrophy can also be achieved 
in such cases of liver injury [12]. 



13.7.4 

Is Hypertrophy Predictable? 

Patients with diabetes mellitus have lower hyper- 
trophy [41]. Imamura et al. showed that diabetes 
mellitus, a high total bilirubin level at time of PVE 
and being male each reduced the extent of hyper- 
trophy in FRL and that cholestasis accelerated the 
process of atrophy in the embolized lobe [21]. The 
hypertrophy rate has a significant correlation with 
the absolute value of portal blood flow velocity on 
day 1 post PVE [18] and Doppler evaluation of left 
portal branch velocity along the post PVE period 
seems to easily predict the hypertrophy rate of the 
nonembolized left lobe [17]. The parenchymal volu- 
metric rate of the FRL before PVE in patients with 
damaged liver parenchyma due to viral hepatitis, 
and both this volumetric rate and prothrombin time 
have been showed as independent parameters pre- 
dicting the hypertrophic ratio of FRL after PVE [56]. 
Indocyanine green retention rate at 15min is an 
adverse predicting factor for all patients and platelet 
count is significantly correlated with the hypertro- 
phic ratio in hepatocellular patients [24]. 



13.7.5 

Is Liver Failure after Surgery Predictable? 



Patient selection criteria for hepatectomy following 
PVE are still under investigation. For Wakabayashi 
et al. postoperative liver failure appears to be more 
severe in patients having high portal pressure and 
hypertrophy of the FRL lower than 20% [55]. In 
another study, the same team founded that portal 
pressure and serum level of hyaluronate measured 
before and after PVE were the most useful param- 
eters in prediction of the outcome of the following 



hepatectomy [56]. The cut-off points of significance 
for serum hyaluronate were 130 ng/ml and 160 ng/ 
ml before and after PVE respectively. Cut-off for 
portal pressures was 16 cm and 25 cm of saline, 
measured before and immediately after PVE respec- 
tively. Consequently, lush imtialportalpressureand 
important elevation after PVE both indicate limited 
resection, and an initially elevated pressure should 
be considered as a poor indication for PVE. Preop- 
erative 99mTc-galactosyl serum albumin has been 
reported as a useful tool for predicting residual liver 
function before hepatectomy [44]. In patients who 
needed major hepatectomy for biliary tract malig- 
nancy and presenting with jaundice, skeletonization 
of the hepatoduodenal ligament was significant Iv 
correlated to postoperative hyperbilirubinemia, 
even after PVE [16]. Anyway, in high risk group 
of patients, PVE suppresses rise in total bilirubin 
and thrombin-antithrombin complex, decreases 
the incidence of postoperative complications and 
reduces intensive care unit or total hospital stay after 
hepatectomy [15, 16]. 



13.7.6 

Long-term Results and Survival 

In patients presenting hepatobiliary malignan- 
cies without chronic liver disease, Abdalla et al. 
reported equivalent median survival durations 
when they underwent PVE or not prior to extended 
hepatectomy (> or = 5 segments), respectively 40 
and 52 months [2]. We have reported 5-year sur- 
vival and 5-year disease-free survival of 34% and 
24% respectively in t>0 patients who underwent PVE 
for liver metastases, that was comparable with the 
survival rates obtained after resection without PVE 
[14]. Compared long-term results from the litera- 
ture, in specific groups of patients presenting with 
liver metastases from colorectal primary or with 
hepatocellular carcinoma, are summarized in Table 
13 4. All these studies demonstrated equivalence in 
5-year survival and 5-year disease-free survival 
between groups of patients preoperatively treated 
by PVE or not (Table 13.4). 



Future Developments and Research 



Cyanoacrylate, the main efficient embolic agents for 
PVE, necessitates sophisticated manipulations and 



^orm' Vein Embolization 



e 13.4. Long-term survival of pauenls vcith liver metastases from colorectal cancer or hepatocellular car> 
.valence in long-ierm st.rvival rates between groups -A patients who underwent preoperative PVE ; PV'Ej 



: ~i\\\ isncal 
(NPVE) 



Metastases from colorectal c, 



Number of % of patients 
resected resected after 
patients PVE 



Overall actuarial si 
yfvalrate(%) 





:LAYetal.[4] PVE 19 


61 


40 


AZOULAY 


et al [5] 


PVE 


89 


67 


44 




NPVE 88 




38 






NPVE 


60 


S3 


53 


Hlia 


i et al. [14] PVE 41 

NPVE 357 


87 


37.3 
38.1 


Wakabay 


s.shi et al 


. [56] PVE 

NPVE 


72,2 


72.2 
80.2 


39.9 
44.1 



very specific training for its safe use. Ethanol is also 
efficient in inducing hypertrophy but carries out 
poorer tolerance that limit its use at least in com- 
promised liver. Therefore, researches for improving 
embolic' agents for PVE are certainly still required. 
Accessibility to a combined Angio-CT suite allows 
realtime precise intrahepatic portal mapping during 
the procedure and increases possibilities for on- 
demand atypical and hyperselective PVE, adapted 
to the most complex types of hepatectomies. A 
FRL/Total functional liver ratio of more than 30% is 
widely admitted as a requested condition for allow- 
ing a safe hepatectomy in patients with normal liver. 
However, curative resection is frequently discussed 
inpatients presenting with an injured liver: liver cir- 
rhosis and any type of liver fibrosis, previous intra- 
arterial (or even systemic) long-term chemotherapy, 
severe fatty steatosis, chronic hepatitis, or previous 
hepatic resection. The cut-off of liver volume to be 
preserved remains still unclear in these patients with 
compromised liver and should be clarified through 
prospective studies. Computer simulation of liver 
resection or nnliotrequeikv ablution, for preopera- 
tive assessment of volumes and function (possibly 
based on the indocyanine green clearance) should 
also be developed. 



13.9 
Conclusion 

PVE is a safe technique allowing efficient preopera- 
tive hypertrophy of the FRL in selected cases. It is 
recommended to perform distal embolization rather 
than proximal occlusion or ligation. Embolic agents 
producing high local inflammatory reaction induce 
greater hypertrophy than others do. 

PVE neither improves nor worsens long-term prog- 
nosis but it allows curative resection in patients that 
otherwise are considered as unresectable. Nowadays, 



it may be frequently combined with multiple other 
efficient therapeutic modalities (systemic or intraar- 
terial chemotherapy, chemoembolization, radiofre- 
quency ablation) in order to lead more patients with 
malignant liver tumor to radical surgery. 



l.Abdalla EK, Hicks ME, Vauthey JN (2001) Portal vein 
embolization: rationale, technicaie and future prospects. 
Br] Surg 88:165-175 

2.Abdalla EK, Barnett CC, Doherty D et al. (2002) Extended 
heoatectomy at pal. ems will: hepatobiliary mal.siiianciirs 
with and without preoperative portal vein embolization. 
Arch Surg 137:675-680 

3. Adam R, Lucidi V, Bismuth H (2004) Hepatic colorectal 
metastases: methods o: improving resectaraihty. Surg Clin 
North Am 84:659-671 

4.Azoulay D, Castaing D, Smail A et al. (2000a) Resection of 
iionresectable liver metastases from colorectal cancer after 
percutaneous portal vein em raolizat ion. Ann Sn rg cal :4S0- 
486 

S.Azoulay D, Castaing D, Krissat J et al. (2000b) Percutane- 
ous portal vein embolization increases the feasibility and 
s.'.fely o: major aver resection for hep;', to cellular carcinoma 
in injured liver. Ann Surg 232:665-672 

6.Broering DC, Hillert C, Krupski G et al. (2002) Portal 
vein em I" oiiz.it ;o n vs. port.'.l vein iigation for induction of 
iiypr: tropin" of th r future liver remnant. I Castro in lest Sn re 
6:905-913 

7. Brown KT, Brody LA, Decorato DR et al. (2001) Portal vein 
embolization with use of polyvinyl alcohol particles. I Vase 
Interv Radiol 12:882-886 

S.Bucher NLR, Swaffield MN (1975) Regulation of hepatic 
n rais by synergistic action of insulin and 
• Natl Acad Sci USA 72:1157-1160 

9.Chijiiwa K, Saiki S, Noshiro H et al. (2000) Effect of pre- 
operative portal win eiiioo.izaiion on liver volume ;tnd 
hepatic energy status o: the nonemoo.ized liver lobe in 
humans. Eur Surg Res 32:94-99 
10.De Baere T, Roche A, Elias D et al. (1996) Preoperative 
portal vein embolization for extension of hep.iteciomv 
indications. Hepatology 24:1386-1391 
ll.Denys AL, Abehsera M, Leloutre B et al. (20001 Intrahe- 
L'.i I ic r. em't dynamic chanties lolkw.i'.ti ■;■■.:■■: I.;, vein emho- 



A. [. Roche and I'. Elias 



1707 



■eDoppler study. Eur Radiol 10:1703- 29.Kinosh: 



12. Elias D, de Baere T, Roche A et al. (1998) Preoperative selec- 
tive portal vein embolizations .'.re ^n effective means of 
extending I he indications of major hep a tec iomy in normal 
and injuried liver. Hepatogastroetirerology 45:170-177 

13. EliasD.de Baere T, Roche A etal.( 1999) During liver regen- 
eration follow. nt : i u _-_ r port.;, embolization the growll: 
rate of liver metastases is more rapid than that of the liver 
parenchyma. Br ] Surg 86:784-788 

14. Elias D, Ouellet JF, de Baere T et al. (2002) Preoperative 
seleaive portal vein embolizalion oelore hepateciomy for 
■ iver metosioses: .ong-term results and impocl on survival. 
Surgery 131:294-299 

15. Farges O, Belghiti ], Kianmanesh R et al. (2003) Portal vein 
embokz.itioii herore rigkt krpateciom.v: prospective clini- 
cal trial. Ann Surg 237:208-217 

16. Fujii Y, Shimada H, Endo I et al (2003) Effects of portal vein 
embokzatioii before ma:or hepateciomy i-iep.-t-.jg.i si :"':en- 
terology 50:438-442 

17. Gerunda GE, Bolognesi M, Neri D et al. (2002) Preoperative 
selective port.il ve.n emno.izaiion '^SPVii; oefore niaior 
kepatic resection. Lfrecliveness o: Poppler estimation of 
kepatic blood flow to predict the hyp e r I top it y :ate of non- 
embokzed live: segmenls. Hrpatogitslr uenterol 43:1405- 
1411 

18. Goto Y, NaginoM,NimuraY(1998) D op pier estimation of 
portal blood flow after percutaneous transhepatic port.il 
vein embolization. Ann Surg 228:209-213 

19.Harada H.Imamura H, Miyagawa S et aL (1997) Fate of the 
Itumon live: .ifter kemikrpoiic portal vein emfokzoiion: eel. 
kinetic and morphometric study. Hepatology 2b:i 162-1 1 70 

20. Hemming AW, Reed Al, Howard RJ et al. (2003) Preopera- 
tive portal vein embolization fo: extended kepairctomy 
Ann Surg 237:686-691 

21.1mamura H, Shimada R, Kubota M et al. (1999) Preopera- 
tive porta! vein embolization: .m; audit or sU paiients. Hepa- 
tology 29:1099-1105 

22. Jaeck D, Bachellier P, Nakano H et al. (2003) One or two- 
stage liroateciomv combined with portal vein emboliza- 
tion for initially nonresectable colorectal liver metastases. 
Am J Surg 185:221-229 

23. Ji W, Liu WH, Ma KS et al. (2003) Preoperative selective 
portal vein embolization in Iwo-step keratectomy for 
Itep.itocellular carcinoma in miured livers: a preliminary 
report. Hepatobiliary Pancre.it I Us Int 2:216-220 

24. Kaido T, Arii S, Shimada Y et al. (2003) Portal embolization 
in various tvpes of liver: novel variables lo predict liyper- 
tropky. Hepaioeasiroenterologv 50:140-145 

2?.Kaneko T, Nakao A, Takagi H (2002) Clinical studies of 

new material tor portal vein embolizonon: corvpansi::: of 

embolic erfeci wiln different agents. Hrpatoga-'rietilLTol 

ogy 49:472-477 
26.Kawai M, Naruse K, Komatsu S et al. 12002) Ycchanical 

stress- den en cent secret ion of interletikm £ by t tuk-he.ia. 

cells after portal vein embolization: clinical :.*:.l iiper: 

mental studies. J Hepatol 37:240-246 
27. Kawasaki S, Makuuchi M, Kahazu T et aL (1994a) Kesecltun 

tor multiple metastauc liver tumors: after port: 

tion. Surgery 115:674-677 
1ft. Kawasaki S, Mai. audi. .VI, Miyagawa S el al. (19E4;>; K..d .:i 

operation after ponal enioo.ization tor tumor 'it Itilar bi.e 

duct. J Am Coll Surg 178:480-486 



H, Sakai K, Hirohashi K et al. (1986) Preopera- 
tive portal vein embolization tor hepatocellular carcinoma. 
World J Surg 10:803-808 

KoGY, Sung KB, YoonHKet al. (2003) Preoperative portal 
win embolization witk a new liqtucl embolic agent, kadiol- 
ogy 227:407-413 

Kodama Y, Shimizu T, Endo H et al. (2002) Complications 
of perctiianeotis transhepatic portal vein embolization. I 
Vase Interv Radiol 13:1233-1237 

Kokudo N, Makuuchi M (2004) Current role of portal vein 
em folizat ion/It epatic artery cl'.emoembolizal ion. L : urg till in 
North Am 84:643-657 

Kokudo N.Tada K, Seki M et al. (2001 ) Proliferative activity of 
intrahepatic colored;;! meiastases atter preoperative k.emilte- 
pat.c portol veal emb' ■..toi.oii. Hepaioiogv .'4:2:: 7-172 
Kubo S, Shiomi S, Tanaka H et al. (2002) Evaluation of 
the etYecl of portal vein embolization on liver function 
bv(99m)tc-gak;ctosyl h union serani albumin sc:ntigraniiv. 
(Surg Res 107:113-118 

Kusaka K, Imamura H.Tomiya T et al. (2004) Factors affect- 
ing kvrr regeneralion after rigkt poriol ve.n emoo.izauon. 
Hepa ^'gastroenterology 51:532-535 
Lee GE, Merlino C. kausto N ; : 392 i Pevelopmenl of liver 
tumors in transforming growth factor alpha transgenic 
mice. Cancer Res 52:5162-5170 

Lee KG, Kinoshita H, Hirohashi K et al. (1993) Extension of 
j.i it i.: a. im.'ic: Lous for :n- p . L ■: ■ . " e I . ,i I . ■ : care. noma by portal 
vein embolization. World [ Surg 17:109-115 
Madoff PC. Hicks ME, Abdalla EK et al. (2003) Portal vein 
tmlvi|:/ai ;i with polyvinyl alcohol particles and coils 
■ ■ "ii for maior liver resection for hepaiobniary 
s.ifeiy and et'reciiveness-sUic.y :n 2:- oatients. 
Hadiology 227:251-260 

Mak-uci-.iM.ThaiBL.TakayasuKetal. (1990) Preoperative 
;■ ::'.i. i iif. lizalion to increase safety of maior hep .'.tee- 
ter::)' tur h.lar bile duct carcinoma: a preliminary report. 
Surgery 107:521-527 

Miyake H, Fujii M, Sasaki K et al. (2003) Heat shock protein 
70 inci'jction in kepatocytes atter rigkt portal vein emboli- 
zation. Hepatogastroenterology 53:2084-2087 
Nagino M. Ximura Y, Komiya ] ei ok !i°9?i Changes in 
hepatic lobe volume in biliary tract cancer palients atter 
right portal vein embolizotton. Hepatology 21:434-439 
Nagino M, Nimtiro Y. Kamiya ) et a I. 1 : 99i?i Se lee live per- 
cutaneous :r;:n ,; k.epot:c embokzritioti ■:.( the portol vein in 
preporaiktn for extensive liver resection: tke ipsi.ateral 
approach. Radiology 200:559-563 

Nagino M, Kamiya J, Kanai M et al. (2000) Right triseg- 
mem portal vein embolization for biliary tract carcinoma: 
technique and clinical utility. Surgery 127:155-160 
N:>ihiyamj Y, Yamamoto Y, Hino I et al. (2003) 99mTc 
j.,a.:ii'':vyi human serum albumin liver dynamic 5PE7 for 
■e assessment of hrpatectom.y in relation to 
pe-cuiar-t. us transhepatic portal embolization. Xucl Med 
Oimmun 1 1:809-817 

I'anis V, K:heiro J, Chretien F et al. (1992) Dormant liver 
meuslase- an experimental study. Br ] Surg 79:221-223 
l'LTti:::a- IV.Daradkeh S, Johann M et al. (2003) Transjug- 
jlar [irtiy.-i *ative porta, emoo.izaiion ;TIPE ) a p. lot si tidy. 
Ilepaldj^iiroenterology 50:610-613 
Roche A, SuyerP, Elias D et al. (1991) Preoperative portal 
win enrbo kz.it i:t n tor kepatic metastases. I Inter vent 1-i.adiol 
6:63-66 



! J orra' Vein Embolization 



-.-(-. S it i mad a ji. imam lira !-!, N ; I k .1 y ; ; : :: : ■ A e i ill. :. ii'OJ i Cij.inge-5 
in mood flow iind function of the liver .ifier right ponal 
vein embolization. Arch Surg 137:1384-1388 

49.Shimamura T.Nakajima Y.Une ¥ et al. (1997) Efficacy and 
safety of preoperative percutaneous transhepatic portal 
embolization will - , absolute ethanol: a cliiiic.il study. Sur- 
gery 121:135-141 

50. Starzl TE, Francavilla A, Halgrirason CG (1973) The origin, 
hormonal nature and action of hepatotrophic substances 
in portal venous blood. Surg Gynecol Obst 137:1 79-1 99 

Sl.Tominaga M, Ku Y, Iwasaki T et al. (2002) Effect of portal 
vein embolization on function of the nonemoolized lobes 
of the .ire r: eva hi a I ion by firslpass hepatic lid oca ineexti ac- 
tion in dogs. Surgery 132:424-430 

52.TsukamotoT,KinoshitaH,Hirohashi Ketal. (1999) Human 
erythrocyte polya.ir.me .rrels after porta' rein emboliza- 
tion. Hep a to gastroenterology 46:31 78-3183 

53. Uesaka k.N.mv.ia V.Mat in ;Hn ?9t>\ '."hanges in hepatic lob.'.r 
function after nghi penal vein eiv.bo.izanon. An appraisal bv 
biliary indocyan.ne given excretion. Ann Surg 223:77-83 



54. Wakabayashi H, Ishimura K, Okano K et al. (2001) Is pre- 
operative ponal vein embolization effective in improv- 
ing pro a ii:: si- ;i:ie: maio-: hepatic resection in p;:tients 
iv i :h advanced--! age hepa;Oce;L;]..tr caicinoma: Cancer 
92:2384-2390 

55. Wakabayashi H, Yachida S, Maeba T et al. (2002a) Evalu- 
ation of live: function for the application of preoperative 
ponal vein embolization on maior hepatic reseciion. Hepa- 
to gastroenterology 49:1048-1052 

56. Wakabayashi H, Ishimura K.Okano Ketal. (2002b) Applica- 
tion ■.■if n:e opera i.ve j. "■-.:■ i " I - - 1 vein em I" ■.. ■ . i ; . L . ■. ■ 1 1 oefi 'ie : i ". .- 1 ■ .■ :" 
henatic resect ion :n patiems with normal or aonointa! live: 
parenchyma. Surgery 131:26-33 

57. Yamakado K, Takeda K.Nishide Y et aL (1995) Portal vein 
embolization vein ilrel coils ,;nd absolute ethano!: a com- 
parative experiment;'.' study with canine .irer. !-!epatoh::gv 
22:1812-1818 

58. Yamakado K, Takeda K, Matsumura Ketal (1997] Regen- 
eration of the un-embol.zed .irer parencliynia to I. owing 
portal vein embolization. 1 Hepatol 27:871-880 



14 Embolotherapy for 

Neuroendocrine Tumor Hepatic Metastases 



Kong Teng Tan and John R. Kachura 



,.1 Introduction 177 

-2 Pathophysiology a:o.l Clinical Considerali 

,.3 Technique 179 

,.3.1 Patient Selection 179 

t.3.2 Emboliz.it ion Protocols ISO 

7.3.3 Pre-Erabolization Preparation ISO 

,.3.4 Embolization Technique (Table 1 4.2) J 8. 

t.3.5 Post-Emboiization and Follow-up JS J 

7.4 Results 184 

7.5 Complications (Table 14.3) 1S4 
7.5.1 Angiographic Complications 184 

7.5.1.1 Nontarget Embolization 1S4 

7.5.1.2 Embolization complications 1S5 

7.6 Pitfalls 185 

7.7 Conclusion 186 
References 1S6 



14.1 
Introduction 

Neuroendocrine tumors comprise an interesting 
group of rare neoplasms, which are derived from 
neuroendocrine cells interspersed within the gas- 
trointestinal system and throughout the body. In 
the gastrointestinal tract, they mainly manifest as 
pancreatic islet-cell tumors and carcinoid tumors 
which are essentially serotonin-secreting tumors 
that originate from enterochromaffin cells. Due to 
its rarity, the epidemiology of malignant 



K. T. Tan, MB, BCh, FRCS, FRCR 

Division of Vasai In: and Iroerveniional Radiology, 

Pepailmen: of Medio:; I imaging, To.viiio General Hospital. 

585 University Avenue, NCSB 1 C-563, Toronto, Ontario, 

Canada M5G 2N2 

J. R. Kachuha, MD, FRCPC 

Division of Vasai I:;: and li^erveniional Radiology. 

Deparlnien; of Medio a I imaging, Toronto General Hospital. 

200 Elizabeth Street, Eaton South l-454d, Toronto, Ontario, 

Canada M5G 2C4 



docrine tumors was not well established until rela- 
tively recently. A large population-based registry in 
France documented incidences of 7.6 and 5.0 cases 
per million population for men and women, respec- 
tively, with increasing incidence over time for both 
sexes [1]. All together, these tumors account for 1% 
of all gastrointestinal tumors. The small bowel is the 
most common primary site (40%), followed by the 
large bowel (30%) and the pancreas (20%) [1], 

Although these tumors are well known for pro- 
ducing various hormonal syndromes, approxi- 
mately half are nonfunctioning or do not produce 
hormones ot clinical significance. In addition, these 
tumors are often indolent, described by Moertel 
[2] as "cancers in slow motion", with which patients 
may live for many years. Indeed at the time of diag- 
nosis, the duration ot symptoms attributable to the 
tumor frequently exceeds several years. Neverthe- 
less, patients with carcinoid tumor metastatic to the 
liver have a median survival of only 3 years, with 
at most 30% of them still alive 5 years after diag- 
nosis [2,3]. Therefore, the index of suspicion must 
be high in order to diagnose these tumors prior to 
the development of metastases in a timely fashion. 
When these tumors present clinically, the symptoms 
are caused by either hormonal excess, local tumor 
growth, or metastatic spread. Although surgical 
extirpation remains the only curative modality, it 
is feasible merely in patients presenting with local 
disease, which accounts for <30% of all cases. On 
;ral or lymph node metastases 
l >70% of patients at first clini- 
cal presentation, with liver metastases accounting 
for 60% of all secondaries [1,4,5]. 

The main prognostic factors of these tumors are 
the stage of disease at presentation, anatomical loca- 
tion of tumor, histological subtype, and adequacy 
of surgical resection [6-8]. Five-year survival of 
patients with and without metastatic carcinoid 
tumors are 0%-40% and 75%-99%, respectively 
[2,3,9,10]. The corresponding figures for the second 
most common gastrointestinal neuroendocrine 
tumor, i.e. gastrinoma, are 20% and 65% [11]. 



the other hand, i 
tlready are present ii 



K. T. Tan and J.R.Kachura 



14.2 

Pathophysiology and Clinical 

Considerations 

Gastrointestinal neuroendocrine tumors often 
develop extensive hepatic metastases that remain 
clinically silent until the liver's metabolic capacity 
is overtaken by the increasing serotonin and neu- 
ropeptides produced by these tumors [12]. When 
tumor burden overtakes hepatic metabolic func- 
tion, the excess hormones are released into the sys- 
temic circulation producing a variety of syndromes, 
lest of which is the carcinoid syndrome, 
:ed by flushing, diarrhea, cardiac val- 
vular damage, broncho con strict ion, and pellagra. 
Although these symptoms of are usually well-con- 
trolled using somatostatin analogues (octreotide), 
tolerance generally occurs which eventually leads to 
a decrease and finally absence of therapeutic effec- 
tiveness [13,14]. Furthermore, the use of octreotide 
remains solelyas a palliative measure, as this therapy 
has no significant effect on the neuropeptide pro- 
duction nor reduces metastatic load [15,16]. Hence, 
since the introduction of somatostatin analogues, 
the commonest cause of death in these patients is 
liver failure from tumor progression [17]. 

Liver metastases from neuroendocrine tumors 
may be found synchronously with the primary 
tumor, may occur following resection of a primary 
tumor, or may occur in the absence of a detectable 
primary tumor. Imaging modalities available for 
the localization of both primary neuroendocrine 
tumors and their metastases include ultrasonog- 
raphy (US), computed tomogiaphv (CT), magnetic 
resonance imaging (MRI), endoscopic ultrasonog- 
raphy (EUS), scintigraphy, portal venous sampling, 
and arteriography. Conventional cross sectional 
imaging studies (US, CT, MRI) individually have 
poor accuracy in locating the primary tumor, with 
reported sensitivities of less than 30% [18-20]. When 
combined with somatostatin receptor scintigraphy, 
however, the detection rate improves significantly 
to 70% [19]. For primary islet cell tumor detection, 
EUS is the method of choice with reported sensitivi- 
ties of 80-90% [21,22]. Although reported to have 
good sensitivity, both portal venous sampling and 
selective arteriography are invasive diagnostic tests. 
As with the detection of primary tumors, the diag- 
nosis of neuroendocrine tumor hepatic metastases 
improves when somatostatin receptor scintigraphy 
is added to US, CT or MRI, with the resultant sen- 
sitivity exceeding 90% in comparison to 40%-50% 
with cross sectional imaging alone [18-21]. In addi- 



tion, scintigraphy is also the best screening test for 
extrahepatic metastases, the presence of which has 
significant impact on the treatment planning and 
outcome [23]. 

Surgical management of metastatic neuroendo- 
crine tumors remains a significant challenge. A few 
authors have advocated hepatic resection for resect- 
able hepatic metastases, with results supporting 
palliation and prolonged survival in these patients 
[2 I -33], Hov.'ever, most patients ( = 90 ! !.->) with meta- 
static disease are unresectable at diagnosis; other 
treatment options such as aggressive chemotherapy, 
cytoreductive procedures, interferon, and symp- 
tomatic control with conventional pharmacological 
methods, all have disappointing results and cure is 
rare [34-40]. 

Although neuroendocrine tumors vary in behav- 
ior, these tumors share the clinical tendency to 
metastasize diffusely to the liver and to be extremely 
vascular [41]. Therefore, it seems logical to target 
the liver with regional treatment when metastatic 
neuroendocrine cancers have a substantial hepatic 
component. The unique dual blood supply of the 
liver allows interruption of the arterial supply with- 
out major risk of hepatic infarction, as normal liver 
parenchyma derives approximate!;-' T5% of its blood 
supply and one-third of its oxygen supply from 
the portal venous system. Tumors, however, derive 
their blood supply entirely from the arterial system. 
Some authors recommend bland liver embolization 
whereas others suggest chemoembolization for neu- 
roendocrine tumor hepatic metastases. In chemo- 
embolization, an intra-arterial chemotherapeutic 
agent is an added component to the embolization 
procedure, with the premise that such an agent may 
achieve a drug concentration greater than is pos- 
sible using an intravenous route, with a long expo- 
sure of the tumor to the drug [42] while attaining 
low systemic toxicity. Furthermore, ischemia from 
the embolization may render the tumor cells more 
sensitive to the chemotherapeutic agent [43], 

Detailed clinical management of patients with 
liver metastases from neuroendocrine tumors is 
beyond the scope of this chapter. A suggested treat- 
ment algorithm is shown in Figure 14.1 [44]. Briefly, 
candidates for treatment can be divided into three 
groups: (1) those with surgically resectable liver 
metastases with no extrahepatic disease, (2) those 
with unresectable liver metastases with no extrahe- 
patic disease, and (3) those with liver metastases as 
well as extrahepatic disease [44]. Group 1 patients 
could be offered surgicalmetastasectomy as this pro- 
vides the potential for svmplonta tic improvement, as 



EmbolothcTiipy for Ne^ro-endo-ciine Tuir.o: Hepatic Metastases 






i \ 



.14.1. Treatment algorithm for patients ■ 



n 5 year survival from 20%-30% 
without surgical excision to 50%-80% with resec- 
tion [45,46]. In those patients with large liver metas- 
tases, pre-resection hepatic artery embolization can 
be performed to reduce tumor bulk and hence may 
facilitate complete resection [47,48]. In addition, 
pre-resection portal vein embolization of the lobe/ 
segments to be resected can be used to induce com- 
pensatory hypertrophy and increase function of the 
future liver remnant in patients with questionable 
hepatic reserve. For group 2 and 3 patients, conven- 
tional anti-hormonal pharmacological therapies are 
still considered first line treatment although this is 
being challenged by newer treatment alternatives. 
Systemic chemotherapy such as streptozocin has 
been used on its own or in combination with other 
agents. The results are variable and often not long 
lasting, however, with significant systemic toxicity 
[39, 49]. Although early results with the therapeutic 
use of 1131 metaiodobenzylguanidine (MIBG) are 
encouraging, as a 5-year survival of 85% and symp- 
tomatic relief in a majority of patients with meta- 
static neuroendocrine malignancy can be achieved, 
only approximated 511*0 ot patients are receptorpos- 
itive for MIBG [50]. Moreover, a complete response 
is rare and the long-term outcome and side effects 
of MIBG are currently unknown [51-54]. Similarly, 



intra- arterial infusion of Indiumlll pentetreotide 
has been used in a small number of patients with 
encouraging result [55]. Orthotopic liver transplant 
for hepatic metastases of neuroendocrine tumor is 
controversial. Early transplant aiming for cure has 
been suggested for group 2 patient'. [44]. With the 
perpetual donor organ shortage and variable results, 
however, this approach requires further evaluation 
[56-60]. 

Hepatic artery embolotherapy (with or without 
chemotherapeutic agent) has been established as an 
effective method in long-term control of hormonal 
symptoms and pain, and in the reduction of tumor 
growth in patients who are not suitable for surgi- 
cal excision and refractory to medical therapy [48, 
61-65]. Recently, Roche and coworkers proposed 
the use of chemoembolization as the primary line 
of treatment inpatients with unresectable neuroen- 
docrine liver 1 metastases [<.>■!]. The authors reported 
a 90% symptomatic and a 75% hormonal response 
in comparison to the 30% -75% response rate with 
somatostatin analogues, 43% objective tumor 
reduction in comparison to 5%-15% with soma- 
tostatin analogues, and a 5- and 10-year survival of 
83% and 56%, respectively, which are considerably 
better than the previously reported survival rates of 
0%-40% inpatients who received only conventional 
medical treatment [2,3,9,10]. 



14.3 

Technique 

14.3.1 

Patient Selection 

Eligibility requirements for hepatic artery embolo- 
therapy for neuroendocrine tumor metastases are 
summarized in Table 14.1. They include adequate 
hepatic and renal function, acceptable coagulation 
parameters, and hepatopedal portal venous flow. 
While what constitutes an adequate amount of resid- 
ual uninvolved liver is not clear, in most published 
series 50% to 60% tumor replacement is considered 
as the acceptable upper limit. It is well recognized 
that over 75% tumor replacement is associated with 
higher incidence of hepatic failure post emboliza- 
tion [48]. In those patients with borderline liver 
and/or renal function, superselective embolization 
using smaller amounts of embolic agent and iodin- 
ated contrast could be considered. 



K.T. Tan and [. R. Kachui 



14.3.2 

Embolization Protocols 

Unfortunately, due to the rarity of neuroendocrine 
tumors, there is no consensus on the best embolo- 
therapy protocol. A variety of methods have been 
used, and no prospective comparative studies have 
been conducted: (1) particles (bland) emboliza- 
tion including polyvinyl alcohol (PVA}, tris-acryl 
gelatin microspheres, or pledgets of absorbable 
gelatin sponge, (2) chemoembolization using a 
cytotoxic agent (such as doxorubicin, mitomycin 
C, or cisplatin) and Lipiodol (ethiodized oil) with 
or without a bland embolic agent, (3) cyanoacry- 
late embolization, and, more recently, (4) selective 
intra-arterial Indiumlll pentetreotide infusion 
[27, 30, 42, 43, 47, 48, 55, 61-66]. Stokes and col- 
leagues in 1993 suggested that chemoembolization 
is associated with a shorter recovery period (pain 
and liver enzyme elevations) in comparison to par- 
ticle occlusion while producing similar efficacy 
or response rate [67]. They compared 20 patients 
treated with hepatic arterial chemoembolization 
to their prior experience using gelatin sponge 
powder alone for embolization ot metastatic endo- 
crine tumors. Gelatin sponge powder, however, has 
fallen out of favor as an embolic agent as it pen- 
etrates distally and causes tissue necrosis. Also, 
Brown and colleagues remind us that no study 
has demonstrated superiority of adding either che- 
motherapeutic agents or Lipiodol for the treatment 
of neuroendocrine tumor hepatic metastases, and 
that these agents may increase the risk of severe 
complications [48]. Most published series reported 
good symptomatic palliation, with response rates 
ranging from 75% to 100%, following all of the 
above embolization protocols. At our center, we 
use PVA or tris-acryl gelatin microspheres for 
embolotherapy of patients with neuroendocrine 
tumor hepatic metastases. 

For chemoembolization, the most widely used 
agent is doxorubicin (50 ing/nr body surface area} 
mixed with iodinated contrast agent and Lipiodol. 
The amounts of contrast and Lipiodol used have 
varied, ranging from 0-10 ml and 10-20 ml, respec- 
tively. Following intra-arterial chemotherapeutic 
injection, many routinely occlude the feeding artery 
with particulate agents such as PVA or gelatin 
sponge. In our experience, we have not been able to 
predict the volume of Lipiodol and/or embolic mate- 
rial that a right or left hepatic arterial system can 
accommodate, based on tumor size, number, and 



Inclusion criteria 

Unresectable tumor 
Patent portal venous system 
Satisfactory live: function 
Satisfactory renal function 
Normal coagulation 

Exclusion criteria 

Occluded [lortal venous system 

Hepatofugal portal venous flow 

> 7?°s of hepatic parenci'.vma :eplaced by tumor 

Hepatic encephalopathy 

Poor liver function 

Biliary obstruction 

Renal failure 

Uncorrectable coagulopathy 
Life expectancy < 3 months 



14.3.3 

Pre-Embolization Preparation 

All patients undergo analysis of baseline liver func- 
tion, electrolytes, renal function, complete blood 
count, and coagulation studies within 1-4 weeks 
prior to the procedure. CT or MRI is performed 
prior to treatment primarily for assessment of portal 
vein patency and to document lesion size and dis- 
tribution. 

In order to decrease the risk of carcinoid crisis in 
those patients with symptomatica!!)" active tumors, 
150-500 micrograms of somatostatin may be given 
the day before embolotherapy and continued for 3 to 
5 days post-treatment [35]. Despite pre-medication, 
however, carcinoid crisis can occur during the pro- 
cedure, often manifested by hemodynamic instabil- 
ity. For noncarcinoid or islet cell tumors, treatment 
of underlying endocrinopathy must be initiated 
before referral for embolotherapy. In our institution, 
prophylactic intravenous antibiotic coverage with 
cefazolin 1 g qSh and metronidazole 500 mg q8h is 
used routinely during the inpatient stay. In some 
centers, allopurinol and lactulose are also admin- 
istered to prevent urate-induced renal failure and 
post-embolization hepatic encephalopathy. 



14.3.4 

Embolization Technique (Table 14.2} 



The patient is usually admitted the day before or on 
the morning of the procedure after overnight fast- 



Embolotherapy for Neuroendocrine Tumo: Hepatic Metastases 



ing. Blood pressure, pulse, and oxygen saturation 
are continuously monitored throughout the proce- 
dure, and intravenous sedation (midazolam) and 
analgesia (fentanyl) are administered as required. 
Access to the common femoral artery is achieved 
using Seldinger technique following the infiltration 
of local anesthesia. A 4 or 5 French vascular sheath 
is then inserted into the artery. Selective arteriogra- 
phy of the celiac and superior mesenteric arteries is 
performed to evaluate the anatomy of these vessels, 
to assess portal venous flow, and to identify the 
distribution of the metastases (Fig. 14.2). Once the 
arterial anatomy is clearly understood, a catheter 
is advanced selectively into the right or left hepatic 
artery, depending upon which lobe has the great- 
est tumor burden. If both lobes of the liver have 
a similar metastatic load, then the hepatic artery 
that is the easier to catheterize is embolized. A 
4 French hydrophilic cobra catheter used with an 
hydrophilic guidewire usually suffices. Embolic 
and/or chemotherapeutic material is injected into 
the selected hepatic artery. It is imperative that the 
tip of the catheter is beyond any visceral branches, 
such as gastric arteries, the cystic artery, and the 
gastroduodenal artery, in order to minimize the 
risk of nontarget embolization. Small vessels and 
branches unable to be accessed with a standard 
angiographic catheter may be selected with a 
microcatheters (3 French or smaller}. To minimize 
the risk of post-embolization hepatic failure, only 
one lobe of the liver is treated on each occasion. 
We use PVA particles or tris-acryl gelatin micro- 
spheres > 100 u.m diameter, switching to larger par- 
ticles (e.g. 300-500 |im) when arteriographic tumor 
blush decreases, and terminating embolization 
when there is flow stasis throughout the targeted 
artery distribution. Although manufacturers have 



Options for celi.'.c Littery I'.itd superior mesenteric artery 
catheterization, in preferential order: 

• 4 French Cobr.i 1 hydrophilic catheter and angled 
hydrophilic guidewire 

• 4 or 5 French Sos Omni catheter to engage the origin, 
then exchange 'or :. 4 Hrencli Oc'fra 1 hydrophilic cath- 
eter over a 03." straight Rentson wire or hydrophilic 

• 4 or 5 French Simmons 1 or 2 catheter 

• 4 French Simmon; 1 hycionhilic catheter 

• Sos Omni or Simmons catheter to engage the celiac ori- 
gin, and use microcalheter for st:pe:s elective catheter- 



specific instructions for preparation of the particle 
suspension, we find a 20-ml mixture of 50% full 
strength nonionic contrast and 50% normal saline 
satisfactory for any standard vial of embolic agent. 
A 10-ml syringe is filled with this particle suspen- 
sion, and is attached to an empty 10-ml syringe 
and to the catheter by means of a three-way stop- 
cock. By injecting the suspension from one syringe 
to the other intermittently between embolization 
injections into the catheter, a uniform suspension 
of particles can be nicely maintained. Some advo- 
cate the administration of lidocaine (10 mg bolus) 
directly into the treated artery during emboliza- 
tion, in an effort to reduce the pain experienced 
during or after the procedure [68]. 



14.3.5 

Post-Embolization and Follow-up 

Following the embolo therapy, most patients will 
have some degree of right upper quadrant pain, 
nausea, vomiting, and low-grade pyrexia due to 
post-embolization syndrome. The pain, nausea, and 
vomiting usually resolve within 24 hours. Complete 
blood counts, coagulation status, electrolytes, liver 
and renal function tests are performed on daily 
basis until the patient is discharged. Liver enzymes 
typically become elevated immediately, peaking in 
24-36 hours, and then decrease to pre- embolization 
levels in less than one week. In those patients with 
hormonally active tumors, production of 5-hyroxyin- 
doleacetic acid, gastrin or glucagon diminishes con- 
siderably (by 90%) within two weeks of treatment, 
along with associated symptomatic relief [69]. 

Triphasic contrast-enhanced CT scans of the liver 
areobtainedl day,l month, 3 months, 6 months, and 
yearly after embolization, or as required, depend- 
ing on the clinical status of the patient (Fig. 14.3). 
The CT scans reveal changes in tumor morphology, 
tumor size, overall liver size, and the development of 
new lesions or metastases. Appropriate biochemical 
tumor markers such as 5-hydroxymdoleacetic acid 
for carcinoid tumor may be measured before and 
after embolization at the similar intervals. 

Most patients with neuroendocrine tumor liver 
metastasis have extensive bilobar involvement. As 
only one lobe is treated during each embolization 
session, most patients require a minimum of two 
treatment sessions in order to treat the entire liver, 
depend ing on the hepatic arterial anatomy. An inter- 
val of 4 to 6 weeks is recommended between treat- 
How for liver function recovery. 



K.T. Tan and [. R. Kachui 




Fig.14.2a-h. Sixly-fou:-yeai-old female vcidi raa.dly progressing carcinoid liver metastases. The gastroduoaena! arteiy was 
sargically ligaiea at I he nine of resection o'i ilie or unary tamo: at tbe gasno-dnodenal : unction 1 vear:; befoie. Arterial- phase 
la} aa.d venoas-.'aase lb) CT anages :T iiver shove greater iv.eta slal. ■." barden in die len lobe. ^elecnve early (c,d) and delayed 
(e,f) ane: iographic imaees of I - 1 ; heaanc arleiy :.I.HA:, berore (i.e.) and afier (d,f| eaacioazauon using [^ vials of 100-300 |jm 
t:.s-aarvl £ea^:i:\ aa.crospaeres via a 4 r re nail '"ob:a II hvoroaiiuc catneter. Arrows de:'o:e stagnant coaarast coin inn i:; proximal 
a spec I of eaaoonzed i.HA. Mos- of I lie ai.eaed contrast is see:' relieving int.:- ihe i iehl hepatio artery in (d J. Celiac a rtei iographic 
images before (g| and after (hi LHA embolization 



Emboli: 1 therapy tor Neuroendocrine Tumor Hepatic Metastases 





Fig. l-!.3a--.l. !-if;v-si\-year-old fema.e with nonfunctioning rectal carcinoid luir.or me:asratic to live:, compl;'.:ned of upper 
I'.bdonun.U pa :n and h. ::■ l" m ■. -::..i I symptoms, (a) l-'iv-embokz;'.! ion venous -phase '.IT .ma tie of the .iver shows ihe largest m eta si a sis 
in the left lobe (arrow), (b) CT image with';;:: intii'.venous contrast injection, oblaineo immediately aftet i.HA emboliza:ion 
: as:ng 4.5 viiils of tr:s-acryl gelatin microspheres, shows retained arteni'.llv :n:ected contrast within, the larges: me:as:asis (iin-i'ii'i. 
(c) Venous-phase CT :mage 3 cavs pos:-emboliza:io:i shows decreased attenuation a:id a small amount of gas wiilun the large': 
metastasis, consisteni w.th necrosis, id) Vrnoiis phase CT i::'.:'.ge a months posi-rmo-o-.izaii'.'n shows a sienir.cant decrease in 
size of the largest metastasis, but growth -:A~ a new m curst, '.sis in Ihe caudate lobe (arrow) 



K. T. Tan and J. R. Kachura 



14.4 
Results 

At present, there is no conclusive data regarding 
the effectiveness of embolotherapy for liver metas- 
tasis from neuroendocrine tumor in comparison 
to other methods of treatment due to various rea- 
sons. It is an uncommon condition with a wide 
spectrum of clinical presentations and severity in 
addition to multiple treatment options. Hence, it 
is highly unlikely that randomized trials will ever 
be conducted to investigate this matter. All results 
for embolotherapy published so far are case series 
involving small numbers of subjects ranging from 
14 to 41 patients with a wide range of disease sever- 
ity. Despite this, allowing for different stages of dis- 
ease and method of treatments, these series show 
a good clinical response rate (reduction of symp- 
toms, somatostatin requirement, tumor size) of 
between 50 to 100%, with symptom-free intervals 
of 5-10 months in 90%-100% of patients [43, 47, 48, 
61-75]. Interestingly, the reported 5-year survival 
of 53% to 83% post-embolotherapy is superior in 
comparison to historical controls receiving medical 
treatment alone (0%-40%) [2, 3, 9, 10]. However, 
there is no definitive evidence that any particular 
embolic agent is more superior to others. 

Our center recently prospectively evaluated 
11 patients with metastatic neuroendocrine tumors 
treated with 20 hepatic artery embolizations using 
tris-acryl gelatin microspheres for pain, hormonal 
symptoms, and/or rapid tumor growth. On follow- 
up CT scanning, 90% of lesions showed reduction in 
vascularity suggesting tumor necrosis. All patients 
had amelioration ol original pain and/or hormonal 
symptoms (mean 31% decrease in pain score) lasting 
3-15 months. There was one major complication: 
encephalopathy for which no etiology was found and 
which resulted in a prolonged hospital stay [70], 



14.5.1 

Angiographic Complications 

Access site complications are unusual as a large- 
diameter common femoral artery access is rarely 
required for the embolization. Iatrogenic arterial 
dissection occasionally occurs at the celiac artery 
origin during difficult catheterization. In patients 
with borderline hepatic or renal function, the risk 
of contrast induced nephropathy and hepatorenal 
syndrome can usually be prevented with optimal 
hydration and by limiting the volume of liver embo- 
lized. 



14.5.1.1 

Nontarget Embolization 

Inadvertent extrahepatic deposition of embolic 
material is relatively common. Although any non- 
target embolization is undesirable and may cause 
complications, the frequency of significant clini- 
cal sequela is low [72]. Asymptomatic deposition of 
embolic material may be seen in the lung, stomach, 
pancreas, duodenum, gallbladder, diaphragm, and 



Table 14.3. Complications of embolothenvv for neuroendo- 



Angiographic complications 
Access-related 

Hisseclion / thrombo:;^ 

Distal embolization 

Hemorrhage / hematoma ,' false aneui 

Nerve damage 

- Contrast media 

- .Allergic / anaphylactoid 

- Nephrotoxicity 

- Catheter and general complications 

- Air embolism .' :hromboembolism 
Hraeturc <<f guide wire nl catheter 



14.5 

Complications (Table 14.3) 



S'lXilurget emholi/aliun 

(!hi:lt:cysliii> 

I'an.Tc^lili 1 

Splenic infarct 

(iaslric or howo! :r!a:c:i 

Serious complications following embolization of Pulmonary embolism 

metastatic neuroendocrine tumor are uncommon 

with careful patient selection, thorough pre-pro- 

cedure preparation, and meticulous angiographic 

technique [72]. Reported major complication rates 

and death rates after embolotherapy for neuroen- 
docrine tumor hepatic metastases range from 1% to 

10%, and 0% to 3%, respectively. 



Embolization compli< 

Severe/prolonged post- 
Carcinoid crisis 

Anemi.'., net 
:c!Xic Jgeiji ) 

- Liver abscess 

- Liver failure 



'botization syndrome 
trc'penia, and throir.bocyropeaia ic 



Embolotherapy for Neuroendoi 



■r Hepatic Metastases 



spleen. Some patients may develop significant com- 
plications, however, from such deposition of embolic 
material. Acalculous cholecystitis, gangrenous cho- 
lecystitis, acute pancreatitis, and splenic infarction 
have all been reported. Fortunately, most of these 
complications can be treated conservatively and 
they rarely require surgical interventions. 



14.5.1.2 
Embolizatioi 



(implication: 



Although the main objective of embolotherapy is to 
cause tumor cell death, this by itself may have unde- 
sirable effects. Post-embolization syndrome (pain, 
nausea, vomiting, pyrexia, and malaise) practically 
occur in all patients and can last for 1 to 2 weeks. 
Anemia, neutropenia, and thrombocytopenia are 
invariable if a cytotoxic agent is used. Secondary 
infection of the necrotic liver/tumor can occur and 
may lead to the formation of liver abscess (Fig. 14.4), 
which may require percutaneous drainage. Previous 
biliary tract surgery such as enterobiliary anasto- 
mosis or sphincterotomy is associated with a greater 
incidence of liver abscess, and this is likely due to 
the colonization of the biliary tree by gut organisms 
[76-78]. There is some evidence to suggest that bowel 
preparation combined with tazobactam/piperacillin 
given intravenously for 3 days reduces the incidence 
of this complication in high risk patients [79]. 

Until the introduction of somatostatin analogues, 
exacerbation of the symptoms of carcinoid or frank 
isis was common with embolization of 



carcinoid metastases. Now, with somatostatin ana- 
logues used routinely for hormonally active tumors, 
both during and after embolization, carcinoid crisis 
occurs in only a small percentage of patients. Treat- 
ment of these patients, who often have severe disease 
of the pulmonary and tricuspid valves, is difficult 
because there is little room for error when reducing 
preload to treat pulmonary edema. Right atrial pres- 
sure must be maintained to ensure adequate flow 
through the diseased tricuspid valve. Maintenance 
of right atrial pressure requires close collaboration 
with an experienced anesthesiologist or cardiolo- 
gist. 



14.6 
Pitfalls 



There are many potential variations of the anat- 
omy of the arterial supply to the liver. Scrupulous 
attention to hepatic arterial anatomy, variants, and 
nontarget branches is necessary to avoid potentially 
devastating complications. In addition, hypervascu- 
lar metastases can create a sump effect, reversing 
the flow in the gastroduodenal artery (GDA). Thus 
a superior mesenteric artery injection may fill the 
hepatic artery via an enlarged GDA, which could be 
mistaken for a replaced right hepatic. Furthermore, 
a celiac injection in such a case may fail to opacify 
the GDA because of the flow reversal and may give 
the impression of an occluded GDA. After emboliza- 
tion of the tumor, the flow will revert to antegrade 




Fig. 14.4a,b. Fil'iy-fonr-year-old male wiri; me:as:auc neuroendocrine tumor of unknown r/riniary. (a) r':e- embolization venons- 
phase CT image of die liver shows the largest me:as:asis in the left lobe i.-invirj. (IV) Venous- phase ;_T image of die liver 
obtained 7 wks posi-e:rioojza:ior. of trie I..HA using r.- vials o: :ris-acry! gelatin. :v..-.":ospr.eres shows Ih.d ;h.e laities: meiasias.s 
has decreased in size ,uv\ enhancement. bin contains a moderaie anion m o:' gas. The pa tie:'.: complained ...f :ever and r.oniii.ng, 
which resolved with, an.iibiotic therapv. Tee -esion was not aspirated or drained, ttv sr.gges: perciiianeoi.s drainage only if a 
palient becomes septic or if dieie is no response :o antibioses. Atrsivhc,;-.!; denote inleiva! growth of a right lobe i 



K. T. Tan and ]. R. Kachura 



in the GDA. Thus it is important to have the catheter 
tip distal to the origin of the GDA to avoid nontarget 
embolization. 

Some advanced or superficial tumors mavpniasil- 
ize arterial supply from thearteries of adjacent organs, 
especially after multiple prior embolization proce- 
dures. Such pniasitization may require embolization 
of branches arising from such arteries as the right 
renal, colonic, gastric, phrenic, internal thoracic, and 
intercostal arteries. It is important to recognize that 
not all such parasitized vessels can be safely treated 
without risk to other important organs. 

A patent portal venous system with hepatopedal 
flow is essential to minimize the risk of possible 
liver necrosis/failure post-embolization. In patients 
with occluded or reversed flow (hepatofugal) portal 
venous systems, embolization can still be performed 
using a modified technique of superselective injec- 
tion and reduced amounts of embolic material [71]. 



14.7 

Conclusion 

Current management strategies for neuroendocrine 
tumor hepatic metastases have relied on data from 
anecdotal evidence and retrospective studies involv- 
ing small numbers of patients. It is unlikely that 
this situation will change in the near future, since 
prospective studies are difficult to perform in such 
a relatively rare and biologically heterogeneous dis- 
ease. Embolotherapy is a widely accepted method 
of treatment for nonresectable hepatic metastases 
from neuroendocrine tumors. Long-term palliation 
of pain and hormonal symptoms is possible using 
repeated treatment. Both bland hepatic artery embo- 
lization and chemoembolization have been used, 
and there is no conclusive data to indicate which 
embolization method and which embolic agents are 
most efficacious. 



i. Lepage C, Bon vier AM, Pheap l\!,et a 1. (2004! Incidence and 
man a en:t enl .:■[' m.U.gnaiil digestive enaocrine tumours in 
.'. well defined French population. Gut 53:549-553 

2.Moertel CG (1987) An odyssey in the land of small tumors. 
I Clin Oncol 5:1503-1522 

3.Soreide O, Berstad T , Bakka A (1 992) Surgical treatment as 
i pr.nciple in palients wilh a ova need ao.toii: a la. c.acinoio 
minors. Surgery ] I 1 :4S 



4. Hemminki K. Li X iJOCUj Incidence trends and risk fac- 
tors of carcinoid tumors: a nationwide epidemiologic study 
from Sweden. Cancer 92:2204-1 

5. Quaedvlieg PF, Visser O, Lamers CB, et al. (2001) Epidemi- 
ology an.: stir viva, in patiems with carcinoid disease in The 
Net 'inlands. An ep.d em i '•■logic.! sttidy with 233] patients. 
Ann Oncol 12:1295-300 

S.Hochwald SN, Zee S, Conlon KG, et al. (2002) Prognostic 
factors in pancreatic endocrine neoplasms: an analysis <yi 
13d cases with a proposa' for iow-grade and inlei mediate- 
grade groups. I Clin Oncol 20:2633-42 

7. Shebani KO, Souba WW, Finkelstein DM, et al. (1999) Prog- 
nosis and survival in patients with gastrointestinal tract 
carcinoid tumors. Ann Surg 229:815-21 

8. Madeira I, Terris B, Voss M, et al. (1998) Prognostic factors 
in patients with encocrine tarn oars of the daodencpancre- 
atic area. Gut 43:422-7 

9. Godwin IP [I (1975) Carcinoid tumors: An analysis of 
2837 cases. Cancer 36:560-569 

lO.Zeitels J, Naunheim K.Kaplan EL, et al. (1982) Carcinoid 
tumors: A 37 year experience. Arch Surg 1 17:732-737 

ll.Mignon M, Ruszniewski P, Haffar S, et aL (1986) Cur- 
rent approach a:-- the management of tumoral process in 
patients with gastrinoma. World I Surg 10:703-710 

12.Creutzfeldt W (1996) Carcinoid tumors: development of 
our knowledge. World I Surg 20:126-131 

13. Arnold R, Frank M, Kajdan U (1994) Management of ga s- 
tioeineio pancreatic endocrine minors: The place of soma- 
tostatin analogues. Digestion 55 (Suppl3):107-113 

14. Oberg K, Erik son B (1989) Medical treatment of neuroendo- 
crine gut and pancreatic minors. Acta Oncol 28:425-431 

15. Ruszniewski P, Ducreux M, Chayvialle JA, et al. (1996) 
Treatment of the carcinoid syndrome wit.: ti'.e long acting 
somatostatin aii.Uo-e.ne lame. -tide: .; prospective study in 
39 patients. Gut 39:279-283 

16. Arnold R (1 9 t; r.-: Medical ireaimenl .:■!" in etas- a sizing carci- 
noid tumors. World I Surg 20:203-207 

17.Eriksson B, Skogseid B, Lundqvist G, et al. (1990) Medical 
treatment and iong-terin survival m a prospective study 
■oj" 84 patients with enaocrine pancreatic tumors. Cancer 
65:1883-1890 

18. Chiti A, Fanti S, Savelli G, et al. (1998) Comparison of soma- 
[ostal.n receph ■: .mag my, coinpulrJ r ■: ■ j ~t ■ giapnv and ahr.:- 
souna in I he canic.U management ot neuroendocrine gastro- 
entero-pancreatic tumours. Eur I Nucl Med 25:1396-1403 

19. Gibril F, Reynolds JC, Doppman JL, et al. (1996) Somatosta- 
tin receptor scintigraphy: iis sensitivity compared with 
that of other imaging methods in delecting primary and 
metastatic gastrinomas. A prospective study. Ann Intern 
Med 125:26-34 

20.FruchtH, Doppman ]L, Norton JA.etal. (1989) Gastrino- 
mas: comparison of MhL imaging with ■■.T. ang.ogiaphv. and 
US. Radiology 171:713-717 

21.Zimmer T, Ziegler K, Bader M, et al. (1994) Localization 
of neuroendocrine tumours of Uie upper ga s t ro i nl es una I 
tract. Gut 35:471-475 

22.Zimmer T, Stolzel U, Bader M, et al. (1996) Endoscopic 
intra sonography and somaiostatin receptor scintigraphy 
in the preoperative looil.zatinii ot insulinomas and gastri- 
nomas. Gut 39:562-568 

23.Anthuber M, fauch KW, Briegel J, et al. (1996) Results of 
liver transplantation for gaslioenteicpaiicreatic tumor 
World I Surg 20:73-76 



Emboli: 1 therapy for Neuroendocrine Tumor Hepatic Metastases 



; 137:88-93 

etaJ. (1999] Hepatic arte- 

e management of advanced 

il J Gastroenterol Hepoitol 



Ahlman H, Westberg G, Wangberg B, et al. (199<i) Trent- 
meiii of liver metastases of c.rcinoid tumors. World I Surg 
20:196-202 

Chamberlain RS, Canes D, Brown KT, et al. (2000] Hepatic 
neuroendocrine met.'.st.ises: I 'oes intervention alter out- 
comes? I Am Coll Surg 190:432-445 
Chen H, Hardacre JM, Uzar A, et al. (1993) Isolated liver 
metastases from neuroendocrine tumors: doe;; resection 
prolong survival; J Am Coll Sui 
'. 'ominguez i. Penys A, Menu Y. e 
rial chemoembolization i: 
digestive endocrine tumo 
31(Suppl2):213-215 
McEntee GP, Nagorney DM, Kvols LK, et al. (1990) Cytore- 
ductive hepntic surgery for neuroendocrine tumors. Sur- 
gery 108:1091-1096 

Soreide O, Berstad T, Bakka A, et al. (1992) Surgical treat- 
ment as a principle in patients with adv.mceo .ibdominal 
carcinoid 1 ii mors. Surgery 1 11:48-54 
Yao KK, Talamonti MS, Nemcek A, et al. (2001) Indica- 
tions and results of liver resection .in..: hepntic chemoem- 
bohzation for metoslotic giistro.mes.iin;'.! neuroendocrine 
tumors. Surgery 1 30:677-685 

Benevento A, Boni L, Frediani L, et al. (2000) Result of liver 
resection as treotmeni for metastases from noncolorectal 
cancer. J Surg Oncol 74:24-29 

Grazi GL, Cescon M, Pierangeli F, et al. (2000) Highly 
.liZsies-ive pokey oi hepntic resections for neuroendocrine 
liver metastases. Hepoiogasrroenterology 47:481-486 
Que FG, Nagorney DM, Batts KP, et al. (1995) Hepatic resec- 
tion for metosiotic neuroendocrine cure mom as. Am | Surg 
169:36-42 

Akerstrom G (1996) Management of carcinoid tumors 
of the stomach, duodenum, and pancreas. World J Surg 
20:173-182 

Faiss S, Scherubl H.Riecken EO, et al. (1996) Drug therapy 
in m ei a sialic neuroendocrine tumors of die g.'.stroentero- 
pancre.nic system. Recent Re suits C. nicer Rrs 1 -ii:l 93-1107 
di Bartolomeo M, Bajetta E, Buzzoni R, et al. (1 996) Clinical 
efficacy of octreotide in the treatment of metastatic neuro- 
endocrine tumors. A study by the Ituj.m ."rial;; in Meiiic.il 
Oncology Group. Cancer 77:402-408 
Moertel CG, Hanley |A (1979! Combination chemotherapy 
[nrds in metastatic carcinoid tumor .md the mokgnom c.ir- 
cinoid syndrome. Cancer Clin Trials 2:327-334 
Oberg K, Norheim I, Lundqvist G, et al. (1987] Cytotoxic 
treatment in patients with malignant carcinoid tumors. 
Response to slreptozocin — alone or in combination with 
5 FU. Acta Oncol 26:429-432 

Moertel CG, Lefkopoulo M, Lipsitz S, et al. (1992] Strep- 
[ozocm-doxoruoicin. strepiozocin-fiuorourac.il, or ohloro- 
zotocin in the treatment of advanced islet-cell c; 
N Engl J Med 326:519-523 
Rivera E, Ajani JA (1998) Doxorubicin, streptoz 
5- flu orouraoi I chemotherapy for patients with r 
islet-cell carcinoma. Am J Clin Oncol 21:36-38 
Gray RK, Rosch J, Grollman ]H (1970] Arteriography in the 
diagnosis of islet-cell tumors. Rod iology P7:o9 — 14 
Andersson M, Aronsen F, Balch C, et al. (1989) Pharma- 
cokinetics of mtrii-arteri.il mitomycin-." with or without 
degradable starch microspheres (DSM) i. 
of non- resect able liver cancer. Acta Oncol 28:219-222 
Ruszniewski P, Malka D (2000) Hepatic arterial chemoe 



boliz.'.tion in the m.magemeiit of novo need oigestive endo- 
crine tumors. Digestion 62(suppl 1):79-S3 

4. Sutcliffe R. .Via p ire I ', Romoitr '. rt .;.. . i ■ : 4i Man.: item mi 
of neuroendocrine liver metosloses Am i Surg 187:39-46 

5. Norton )A, Warren RS, Kelly MG, et al. (2003) Aggressive 
surgen" for meloslotic kver neuroendocrine tumors. Sur- 
gery 134:1057-1063 

6. Sarmiento JM, Heywood G, Rubin J, et al. (2003) Surgical 
treatment of neuroendocrine metostases to lire liver: a pleo 
for resection to increase survival. J Am Coll Surg 197:29- 
37 

7. Eriksson BK, Larsson EG, Skogseid BM, et al. (1998) Liver 
embolizations of patients with malign.mt neuroendocrine 
gastrointestinal tumors. Cancer 83:2293-2301 

8. Brown KT, Koh BY, Brody LA, et al. (1999) Particle embo- 
lization :yi henotic metastases for control of pain and hor- 
monal symptoms. ) Vase Interv Radiol 10:397-403 

9. Rivera E, Ajani JA (1998] Doxorubicin, streptozocin, and 
5-fluorourocil chemotheropy for putients with metastatic 
islet-cell carcinoma. Am J Clin Oncol 21:36-38 

0. Faiss S, Pape UF, Bohmig M, et al. (2003) Prospective, ran- 
dom izeo, mull ice nter trio J on the a miproli ter.it ive effect of 
lonreotide, interferon rofa, .md their combinotion for ther- 
apy of metasnitio neuroendocrine gasiroenteroponcrearic 
tumors — The intern. it ion.'. I Lonreotide and Interferon Alfo 
Study Group. J Clin Oncol 21:2689-2696 

1. De Jong M, Breeman WA, Bernard HF, et al. (1999) Therapy 
of neuroendocrine tumors with radiolabeled somatostatm- 
analogues. Q I Xucl Med 43:3.~ii'-Ii!i 

2. MukherjeeIJ,KaltsasGA,IslamN,etal. (2001) Treatment of 
metastatic carcinoid tumours, phaeochromocytoma, paro- 
gongiioma and medullory c.rcinomo ':■• the thyroid with 
(131)I-meta-iodobenzylguanidine [(131)I-mIBG]. Clin 
Endocrinol 55:47-60 

3.Castellani MR, Chiti A, Seregni E, e 
131Imetaiodobenzylguanidine (MIBGI i 
of neuroendocrine tumours. Exper 
Cancer Institute of Milan. Q J Nucl Med 4 

4.Chatal JF, Le Bodic MF, Kraeber-Bodere F, 
Nucleoi medicine opplic.it ions for neu roe rid oc 
World J Surg 24:1285-9 

it. Chatziio.mnou A, Ladopoulos C, Limouris GS, et ill. (2004! 
Selective intra. uteri a! injection of ' '■ In-pentelreotioe in 
the treatment of somatostatin positive (receptors II, SSTR,) 
neuroendocrine metastatic o.sntse in the kver. ■'. I K-1-. _ : ri 4, 
Barcelona. Poster 150 

6.LeTreutYP, Delpero JR. Dousset B, et al. (1997) Results of 
liver tiYiiispl.Li'troio;'. in ike treatment of metastatic neuro- 
endocrine tumors. A 31-case French multicentric report. 
Ann Surg 225:355-364 

7.Dousset B, HoussinD, SoubraneO, et al. (1995) Metastatic 
endocrine it; mors: is there o place for kver transplantation: 
Liver Transpl Surg 1:111-117 

S.Knechtle SJ, Kalayoglu M, D'Alessandro AM, et al (1997) 
Proceed with caution: liver Iransplont.'.tion for metastatic 
neuroendocrine tumors. Ann '-urg cc?:?4i ; -34fi 

9. Lang H, Oldhafer KJ, Weimann A, et al. (1997) Liver trans- 
pkmtation for metostotic neuroendocrine tumors. Ann 
Surg 225:347-354 

O.Coppa J, Pulvirenti A, Schiavo M, et al. (2001) Resection 
versus transpl. mtat ion for liver metosloses from neuroen- 
docrine tumors. Transplant Proc 33:1537-1539 

1. Perry LJ, Stuart K, Stokes KR, et al. (1994) Hepatic arte- 



1. (2000) Role of 
r the treatment 
e of the National 

4:77-87 

it al. (2000) 
ine tumors. 



K. T. Tan and J.R.Kachura 



rial chemoenibolization for metastatic neuroendocrine 
tumors. Surgery 116:1111-1116 

62.Drougas fG, Antony LB, Blair TK, et al. (1998) Hepatic 
artery di em o embolization for management of patients 
with advanced mei.ist.iiic carcinoid tumors. Am ] Surg 
175:408-412 

63.Kim YH, Ajani [A, Carrasco CH, et al. (1999) Selective 
hepatic arieri.il chemoemholization for liver i 
in patients with carcinoid tumor or islet cell i 
Cancer Invest 17:474-478 

64. Roche A, Girish BV, de Baere T, et aL (2003) Tram cath 
eter arterial chemoembotizalion as first-line tr. riniitii lor 
hepatic metastases from endocrine tumors, l-u- aadiol 
13:136-140 

65.Schell SR, Camp RE, Caridi JG, et al. (2002) Hepatic 
Arlery iimholization for Control -:A symptoms .icirciHtde 
ji.ee. a i re mem s, and Tumor Progression in Met..stalic liar 
cinoid Tumors. ] Gastrointest Surg 6:664-670 

66.WmkelbauerFW,Nieder!eB,Piet5chriiannF(lSv ■: Ik' .n, 
artery em bolo therapy of hepatic metastases Inm carci 
noid tumors: value of using a mixture of cy 
and ethiodized oil. Am j Roentgenol 1(15:323-32/' 

67. Stokes KR, Stuart K.CIouse ME (1993) Hepatic arterial che- 
moenibolization for melasiatic endocrine tumors. 1 Vase 
Interv Radiol 4:341-345 

68.Hartnell GG, Gates J, Stuart K, et al. (1999) Hepatic che- 
moenibolization: et'fecl of intraarterial .idocaine on pain 
and postprocedure recovery. Cardiovasc interven: Radio. 
22:293-297 

69.Clouse ME, Perry L, Stuart K, et al. (1994) Hepatic arte- 
rial chemoenibolization for metastatic neuroendocrine 
tumors. Digestion 55(suppl 3):92-97 

70. Lem SL, Asch M, Kachura IR, et al. (2004) Toronto hepatic 
artery embolization study I Vase Interv Radiol 15:52-}? 
[Poster No. 297 presented at 29 th Annual Scientific Meet- 



ing of the Society of Inlervenl iona. Radiology. Phoenix. 
Arizona, March 25-30, 2004] 

71. Pentecost M|,DanielsJR,TeitelbaumGP,etal. (1993) Hepatic 
chemoenibolization: safety with portal vein thrombosis. 1 
Vase Intervent Radiol 4:347-351 

72. Gates J, Hartnell GG, Stuart KE, et al. (1999) Chemoem- 
bohzation of hepatic neoplasms: safety, complications, and 
when to worry. Radiographics 19:399-414 

73. Carrasco CH, Charnsangavej C, Ajani J, et al. (1986) The 
carcinoid syndrome: palliation ov hepatic artery emboliza- 
tion. A™ T Roer'penol 147-1 49 154 

74.Mar!i:ik Kli, l.okich J), Rollins JK, et al. (1990) Hepatic 
arterial embolization fur mctastah; horrrainc stcret- 
.">; 'inrnr'- Itihr^.jCL. tltei tive-fs. ar.d tump lica lions. 
Dancer SS.2227 2232 

75. Ila|ari*adeh II, Ivancer K, .Vljeiler CR, el al. (1992) Xffec- 
:w |m.i.ihv trtaimeni <\ — eia\:jr.i nriii.>.d tumors 
with mlra arterial chcrrM-rRrapv/ihi ir .at :•: :■.!./. ition 
romhined w:lh i:clrei:l:de acelale. Am | S_r^ '63 479- 

7ft. Ishikawa II, Kanai I, On.i T, e: al. (1994) Analysis :if cases 

wiin liver aosctiis iollcwing iraiiscameier arterial oliemo- 
emoc-jzation iTAE! fit--:" malignant hepatic tumors ■: lapa- 
nese). Gan To Kagaku Ryoho 21:2233-2236 

77. Song SY, Chung IW, Han JK, et al. (2001 ) Liver abscess after 
transcatheter oily chemoembohzation for hepatic tumors: 
incidence, predisposing factors, and clinical outcome. J 
Vase Interv Radiol 12:313-320 

78. Kim W, Clark TW, Baum RA, et al. (2001 ) Risk factors for 
Ijver abscess formation after hepanc chemoemoolizaiion ■ 
Vase Interv Radiol 12:965-968 

79. Geschwind IF, Kaushik S.Ramsey DE,et al. (2002) Influence 
of a new prophylactic antibiotic therapv :.'n I he incidence of 
liver abscesses after chemoembohzation trealment o: liver 
tumors. I Vase Interv Radiol 13:1163-1166 



15 Bone Metastases from Renal Cell Carcinoma: 
Preoperative Embolization 



Introduction 1S9 

Clinical Features and Physiopathology 19L 

Angiography and Embolization Technique 

Ejid:-;:iNons 190 

Preprocedural Preparation 191 

Angiographic Technique 191 

Embolization Technique 191 

Results 193 

Obliteration of Tumor Blush / Estimated 

Blood Loss 193 

O^eon;- Healing and Survival 196 

Complications 196 

Conclusion 198 

References 199 



15.1 
Introduction 

The American Cancer Society estimates that there 
will be approximately 36,160 new cases of kidney and 
other urinary carcinomas in the U.S. in 2005. Renal 
a (RCC) is the most common type of 
:er in adults, accounting for 70% of the 
, approximately 25,312 new cases. Fur- 
ince the 1970s, incidence rates for RCC 
/erage of 2% per year, but 
e appears to be leveling off [1, 
e-tlurd of p.iiiei if s with KCC hnve metas- 
tases at presentation [3], Eighty percent of patients 
with RCC eventually have metastases, and nearly 
one-half of these patients have metastases to the 
bone [4-6]. However, more recent reports indicated 
that with the significant improvement of imaging 
techniques, such as ultrasonography, computerized 
tomography (CT), magnetic resonance imaging 
(MRI), and positron emission tomography (PET), 



kidney ca 
tumors, i 
thermore : 

recently th 
2]. Up 



S. Sun, MD 

Associate Professor o:' Radiology, 1 ;ep-.n iintnt of Radiology. 
University of Iowa, College of Medicine, 200 Hawkins Dr., 3955 
JPP, Iowa City, IA 52242, USA 



asymptomatic RCC is more frequently detected and 
detected at lower stages ot disease, when tumors may 
be resected with curative intent or when a metastatic 
lesion is in the early stage [7, 8]. As a result, fewer 
than 20% of patients with RCC have overt metastasis 
at the initial presentation [7]. 

Treatments for RCC includes radiation therapy, 
chemotherapy, and surgical intervention. RCC is 
relatively radioresistant. The response rate to radia- 
tion therapy is 50% at best, and symptomatic recur- 
rences are common [4-9]. Despite extensive evalu- 
ation of many different treatment modalities, RCC 
remains highly resistance to systemic therapy, and 
the median survival of RCC patients is ~8 months. 
About 10% to 20% of patients exhibit complete or 
partial response to interferon and/or interleukin-2 
with/without chemotherapy or each therapy alone, 
with a durable response of >2 years in 5% or less [3]. 
In patients with localized disease, surgical resec- 
tion of the primary tumor remains the mainstay of 
therapy [7, 8]. 

Although metastasis to the bone represents an 
advanced stage of underlying disease, it may be asso- 
ciated with relatively prolonged survival. However, 
reports of survival conflict in a range of less than 
15% to more than 50% [4, 10]. Therefore, indications 
for surgical intervention and the appropriate extent 
of surgery remain controversial. Wide excision of 
metastatic lesions was advocated because of the unre- 
sponsiveness of RCC to noninvasive measures, such 
as chemotherapy and radiation therapy, and the pos- 
sibility that survival may be relatively prolonged in 
treated patients [10]. Existing reports on the surgical 
management of metastatic RCC of bone have limited 
value since they are based on only a small number of 
patients or failed to mention the surgical technique 
and its effect on local tumor control [10]. 

Most metastases from RCC are hypervascular 
and tend to bleed massively during surgery [11]. 
Biopsies, open reductions, internal fixations, and 
resections have been associated with an average 
intraoperative blood loss of 1.5-3 L [12]. A blood loss 
of 2000-18,500 ml (mean 6800 ml) was reported in 



20 patients operated on during the pre- emboliza- 
tion era [13]. Preoperative embolization of these 
lesions appears to be a safe and effective technique 
to decrease intraoperative blood loss and the dura- 
tion of surgery, and thus reduce the surgical morbid- 
ity/mortality:! nd hospital stay [11-20]. Preoperative 
embolization has become a standard measure for 
this type of patient before surgical intervention. 



Clinical Features and Physic-pathology 



Systemic symptoms may also occur, such ashypocal- 
caemia. Occasionally, patients may have hyperten- 
sion from the tumor affecting the rennin-angioten- 
sin pathway. RCC metastases most commonly affect 
the spine, proximal long bone, pelvis, ribs, sternum, 
and skull [3, 22, 23]. Since the kidney is comprised 
of mostly blood vessels, RCC is normally a hyper- 
vascular tumor. RCC metastases usually mimic the 
primary tumor in vascularity, being hypervascular 
in 65%-75% of patients, and bleeding extensively 
(even audibly) after a simple biopsy [3, 8]. 



Due to its visceral location and the existence of a 
second functional kidney, RCC is characterized by a 
lackof early warning signs, resulting in a high propor- 
tion of patients with either locally advanced disease 
or metastases already present at the time of diagnosis 
[3, 21]. RCC metastases take place via the lymphatic 
or venous routes. The lung parenchyma, bone, liver, 
and brain are the most common sites of metastases 
[3, 21, 22]. Although metastasis to the bone from RCC 
ranks as approximately the sixth most common site, 
compared to other tumors, this tumor has several 
unique features that increase its significance: (1) the 
metastases may occur many years (up to 10 years} 
after the primary tumor has been treated surgically; 
(2) the metastases may occur as a solitary lesion and 
as such an en bloc surgical resection may render the 
patient free of cancer and offer hope for a cure; (3) 
even though the incidence of RCC is proportionally 
small, the tumor has a high avidity for the skeletal 
system and thus produces a relatively large number 
of bone lesions; and (4) the bone lesions can be large, 
with an average size of 7 cm in diameter, and have an 
aggressive appearance [3, 19, 21, 22]. 

Pain is the most common presenting symptom. 
Pathological fracture rarely occurs without a history 
of a few weeks or months of increasingly severe pain. 



Angiography and Embolization Technique 

15.3.1 
Indications 

Due to the fact that most embolization procedures 
were performed preoperatively, surgical indications 
dictate the number and extent of the procedures. 
Indications for surgery are divided into two groups 
according to signs of mechanical skeletal failure 
(Table 15.1). For patients who required amputa- 
tion or wide radical resection due to massive tumor 
extension to the soft tissue with invasion of the major 
neurovascular bundle of the extremity, preoperative 
embolization may not be indicated [10]. Similarly, for 
patients with a small, easily accessed lesion or with an 
angiography-proven very hypovascular tumor, pre- 
operative embolization may not be necessary [18]. 
Otherwise, all other patients should undergo preop- 
erative embolization with the intent to obliterate all 
tumor feeders and decrease intraoperative blood loss. 
General indications tor 1 Ira nscatheter embolization of 
bone metastasis are listed in Table 15.2. Even though 
listed, practically, the applications of this technique 
in the indications other than preoperative emboliza- 



Table 15.1. Indicator* for ■angeiy 

Without mechanic;!' bone failure • Solitary bor 



With mechanical bone failure 



• Intractable pain 

• Impending or pathologic: 



Table 15.2. Indications for embolizatio 
Preoperative embolization (72%) 
Control of hemorrhage 
Inhibition of tumor growth 
Reducing viable minor volume to facil 



.e Metastases from Keiul Cell Ciiiiaoau: Preoperative Emboliza 



tion have been limited, with only sporadic reports ii 
the literature [11-20]. 



15.3.2 

Preprocedural Preparation 

Due to the preoperative nature of the procedure, the 
orthopedic surgeon should do a complete work-up 
of the patient to rule out all potential major medical 
contraindications for surgery. Interventional radiol- 
ogists should review every document available and 
focus on issues related to the embolization proce- 
dure. One should perform and document findings 
of physical examination in terms of the patient's 
general condition, the status of affected limb, and 
circulation of the extremities in order to set up a 
baseline and compare with postembolization find- 
ings later. For patients who have a history of contrast 
allergy, premedication should be given according to 
the institutional protocol. Routine preprocedure lab 
tests include CBC, coagulation tests, and renal func- 
tion tests. Patients may have impaired renal func- 
tion due to previous nephrectomy for RCC. These 
patients need to be sufficiently hyd rated before the 
embolization procedure. The amount of contrast 
media given during the procedure should also be 
limited if possible. For patients who have significant 
coagulopathy, correction is necessary with Vitamin 
K or transfusion of fresh frozen plasma or platelets. 
Placinga Foley catheter may be helpful, as the embo- 
lization procedure can be lengthy if multiple tumor 
feeders need to be embolized, and since the patient 
may have a pathological fracture, ambulation could 
be difficult. Furthermore, for patients with a pend- 
ing pathological fracture, early ambulation postem- 
bolization mav complicate The pathological fracture 
[19]. A review of available imaging studies is impera- 
tive for choosing a treatment plan. Consents for the 
procedure and conscious sedation are obtained rou- 
tinely. Whether or not one should give prophylactic 
antibiotics routinely before the procedure is contro- 
versial. Most authors do not prescribe antibiotics for 
the purpose of prophylaxis [18-20]. 



15.3.3 

Angiographic Technique 

Most preoperative embolizations can be performed 
under intravenous conscious sedation via a trans- 
femoral approach. Lesions in the proximal femur are 
treated from the contralateral approach. Lesions in 



the distal femur may be accessed in an ipsilateral 
antegrade fashion to facilitate catheter manipula- 
tion [19]. For lesions located in the pelvis and upper 
extremities, a right femoral approach is routinely 
used. A5-F sheath is used for most procedures. A 5-F 
Davis catheter, JB-1 or Cobra visceral catheter is com- 
monly used for selective catheterization. Microcath- 
eters such as the 2.9-F Renegate Super-flow (Boston 
Scientific) through a coaxial system are used for 
superselective catheterization that allows a safe and 
effective embolization of the tumor feeders. 

The pre-embolization arteriogram should be 
started at the main territory artery in order to cover 
the entire area and identify all tumor feeders. For 
example, a distal abdominal aortogram or common 
iliac arteriogram should be performed when a 
tumor is located in the proximal femur, because 
the tumor may derive blood from superior/inferior 
gluteal arteries, medial/lateral circumflex femoral 
arteries, and descending muscular branches from 
the profunda femoris artery. In rare situations, the 
obturator and internal pudendal arteries from the 
anterior group of the internal iliac artery may also 
provide blood supply to the tumor. The postem- 
bolization arteriogram should include all possible 
collateral pathways, because untreated collater- 
als may become more prominent after occlusion 
of other feeders (Fig. 15.1a-c). The amount of con- 
trast medium given during an arteriogram must be 
sufficient to make all possible tumor feeders well- 
opacified. Therefore, in addition to an appropriate 
injection rate, the period of injection should also be 
long enough (lasting more than 3 sec). The acqui- 
sition should be long enough to cover all phases 
of the arteriogram (arterial, capillary, and venous 
phase) to document tumor vascularity and arterial 
to venous shunting. Frequently, multiple selective 
arteriograms in different projections maybe neces- 
sary in order to show the origin of a tumor feeder 
in profile and facilitate superselective catheteriza- 
tion. The fluoro-fade, or road mapping, technique is 
available for use with most modern digital arterio- 
graphic equipment. This is very helpful for superse- 
lective catheterization, especially when a tumor is 
located in the extremities (reduced artifacts caused 
by movements of breathing and bowel peristalses). 



15.3.4 

Embolization Technique 



Thus far, there has been no single embolic ager 
that is ideal for preoperative embolization of bon 




. Collateral supply of rumor vessels, a Pre-embohzation arte- 
riogram. Tumor supply from Li ranches of :be i i t: [it Literal femoral circum- 
flex [tinge arrow), profunda isin-.iii iin-n ui, .ind the superior gluteal (<UTpir 
/i['i7i/t) arteries, b Supei se.e.tjve catheterization of the lateral femoral cir- 
cumflex artery. Note th lU the desceuc.ing branches do not supply the tumor 

■).c Arteriogram after em bo. izat ion of the tumor with PVA.Note sub- 
traction artifact from coil [i-.n^c mi ■■.<«) thai was p.accfr into the descending 
branch of lateral femoral cncumf.ex artery before l-'VA embolization. Also 
note that gluteal s.ipp.y is more obvious after embolization of the main 

feeders ismall at 



metastases from RCC. An ideal embolic agent should 
be easily delivered through a microcatheter, should 
reach and permanently occlude small blood ves- 
sels deep within the tumor, and should be nontoxic 
and easy to prepare and control during delivery. 
Different embolic agents have been used for pre- 
operative embolization of bone metastases from 
RCC, including absolute alcohol, tissue adhesives, 
coils, gelatin sponge, Embospheres, and polyvinyl 
alcohol particles (PVA) [11-20, 24-31]. The use of 
liquid embolic agents (e.g., ethanol, tissue adhesive) 
was not advocated for preoperative embolization of 
bone metastasis since they may be associated with 
a high rate of complications (even in experienced 
hands) such as tissue ischemia, skin necrosis, and 
neurologic impairment when used for spinal metas- 
tasis [13]. Coils are a permanent embolic agent, but 
they induce relatively proximal occlusion of target 
vessels, and thus are less effective for decreasing 



estimated blood loss (EBL), because bone metastasis 
from RCC shows angiomatous vascularization that 
may reconstitute collaterals within hours. Practi- 
cally, it is not uncommon to find new tumor feeders 
that were not evident on thepreembolization angio- 
gram, immediately after the major feeders were 
occluded. Coil embolization made no significant 
difference in EBL compared with a control group 
in hypervascular spinal lesions [13, 19]. The same 
studies also suggested that the additional use of coils 
after PVA particles provides no further benefit. 

Gel loa in particles have been used in the early stage 
of preoperative embolization of bone metastasis. Due 
to the fact that it is biologically de tradable and not 
definite in size, Gelfoam pledge only creates proxi- 
mal and temporary occlusion of target vessels. Early 
re canalization and revascularization of embolized 
vessels have been observed, which resulted inunfavor- 
able outcomes regarding EBL. Therefore, compared 



.e Metastasis from Renal Cell Car 



'.: Preoperative Embolization 



with PVA particles, Gelfoam pledge is less reliable in 
controlling intraoperative bleeding [10, 13, 19], and 
it should be avoided in preoperative embolization for 
bone metastasis from KCC, espet in II;' when surgery is 
not to be performed within 48 h [19]. 

PVA particles are considered a permanent periph- 
eral embolic agent, and have been used successfully 
for the treatment of hemorrhage, vascular malforma- 
tions, and tumors throughout the body [31, 32]. PVA 
particles in sizes between 250 and 1,000 u.m were 
used for preoperative embolization in the major- 
ity of published series [14, 19-21, 32]. The technical 
requirements are more demanding for PVA than for 
larger embolization materials that can be deployed 
more proximally, e.g. coils or Gelfoam pledges. Selec- 
tive and superselective catheterization can provide 
a safer environment for deploying PVA and reduce 
the possibility of errant embolization, but caution 
must be taken. Large anastomoses, such as those 
between the inferior gluteal or the obturator arteries 
and the femoral artery, may provide an escape route 
for particles into nontarget territories [33]. The size 
of the embolic material needs to be adjusted to the 
size of these potential collateral vessels and the size 
of existing A-V shunts, which are often present in 
hypervascular metastases (Fig. 15.2a-g). 

However, smaller particles (-200 fim in size) 
should be used when a catheter or microcatheter can 
be superselectively positioned in a small, peripheral 
feeder that supplies the tumor only. Smaller PVA 
particles can also be used when branches from a 
tumor feeder that supply surrounding normal tissue 
are protectively embolized with coils (Fig. 15.1a,c). 
Occasionally, a main tumor feeder gives many 
branches that supply most of the tumor, but bifur- 
cates from a major branch supplying normal tissue. 
In order to make the embolization safer, superselec- 
tive embolization of each of tumor feeders will be 
required, but is very time-consuming. Under such 
a situation, coil embolization of the major normal 
branch will make the procedure much easier and 
safer. Embolization with smaller PVA particles by 
means of a coaxial microcatheter system is more 
effective, and complete obliteration of all tumor 
blushes can be expected [13, 19]. This is partially 
due to the fact that smaller particles make imme- 
diate revascularization from vessels distal to the 
embolized pedicles through existing collaterals less 
likely. Immediate revascularization is frequently 
seen when larger particles or proximal emboliza- 
tion was conducted, which may give one the impres- 
sion that complete obliteration of the tumor blush 
can never be achieved. It is also frustrating for the 



operator because the new tumor feeders are usually 
too small to be catheterized, not to mention the time 
one has to spend. 

Nonetheless, when embolizing an artery that 
may supply important structures with a smaller size 
of PVA, great care must be taken to avoid nontar- 
get embolization. For example, internal iliac artery 
branches (mostly inferior and superior gluteal 
artery) and deep femoral artery branches may supply 
the sciatic nerve, buttock, and leg muscles. Emboli- 
zation of these arteries may result in an inadvertent 
event [33]. The embolization should begin with the 
major pedicles and then proceed to the accessory 
ones to avoid embolization through backflowof any 
neighboring area. It is not uncommon to see that 
the accessory feeders have already partially or com- 
pletely occluded through the communication with 
the major feeder when the major feeder has been 
embolized (Fig. 15.3a-e). PVA is mixed with con- 
trast medium for better fluoroscopic delectability 
and more controlled PVA particle delivery. To keep 
the PVA particles in suspension during the delivery, 
many methods have been used. The most commonly 
used technique is to perform, as frequently as possi- 
ble, mechanical exchanges of the solution containing 
PVA particles between two syringes on a three-way 
stopcock. Ex vivo experiments showed that the best 
PVA particle suspension could be achieved when the 
ratio of contrast medium and normal saline was 6:4, 
i.e., 60% contrast and 40% saline [28] . For better flu- 
oroscopic visualization during the particles' deliv- 
ery, full-strength contrast was also used with the 
delivery syringe held upwards to keep the particles 
floating on the top of the syringe [19]. The mixture 
of PVA is manually delivered under fluoroscopic 
observation. When the flow slows down or stagna- 
tion is observed, the delivery is halted and residual 
PVA is slowly flushed forward with normal saline. 
Saline clearly defines the contrast material interface 
and emptying of the catheter from particles [19]. 



15.4 
Results 

15.4.1 

Obliteration of Tumor Blush /Estimated Blood 

Loss 

Intra- operative blood loss is the major criterion in 
evaluating the el flaw vol preoperative embolization. 
Significant intra-operative blood loss was defined as 





Fig 15r>a-g. Fifty- iiine-ve.M-0:d female with RC'Z metastasis of left proxi- 
mal hunter lis ana i:r.pe:'.dHig fracf.ue. i-'VA i.i?i)-L;ni i enfnolizatic-n with 
obliteration of t timer stain by more than l ">0 c 'v ai'.d intra-opeiative blood 
loss of 350 ml. li Arteriogram showed hypervascul.M- uimor in the left 
proximal humerus supplied oy lateral circumflex aj terv. b Venous phase 
showec s i gniric, i m iLimcr ~= Ili z=Jj .mi," A-V shuntii'.g : : r f " .■ L "■■ s). c PosteMbQ- 
l:7,i : ion arleric-gi urn with catheter tip at the major feeder shows complete 
occlusion of the feede: with no tumor oltish seen : :;ii::n).d Arteriogram 
with catheter pulling b-aciv shows ■,-, minor tumor feeder supplying part of 
lite tumor (anoirsi. e $uhselective pc-steinbohzation arte! iogfjm shows 
tout! occlusion of the minor feeder fan-on.'), f Arteriogram after repo- 
sitioned catheter to the r.istal subclavian artery shows iMiollte: minor 
feeder with tumor h-iush, winch was less evident before the other two 
tumor feerlers were embolized i^ncuij. g Late-phase arteriogram .Lite; 
the thud feeder embolized will! citlieter positioned at proximal subcla- 
vian artery shows complete occlusion of all tumor feeders 



.e Metastases from Renal Cell Carcinoma: Pre 




Fig 15.3a-c. Left glenoid solitary RCC metastasis, s/p pre-operation 
embolization with PVA (350-500 |im), estimated blond loss of 75- 
100m!. a P re-em bolization arteriogram showed the communication 
oetween I wo- mm or feedeis iiirw.: sj. b 5,'p embolization of the maior 
feeder, selective ane::og:am shows pa ;' : i a I ly C'Ccmded miner feeder 
wiih shiag.sh flow liiiioir;. c Comaleie.v embol.zed feeders with no 
".iimor stain identified 



a loss of more than 600 ml of blood from a lesion 
of extremities or 1,200 ml from a pelvic lesion [10]. 
The amount of blood loss has been positively cor- 
related with the percentage of obliteration of tumor 
blush I OTB) piisiemholizalion. The more tumor 
feeders embolized, the better EBL results that could 
be achieved. Different categories for rating OTB that 
resulted in a significant difference in EBL have been 
used [13, 15, 19, 20]. Achieving greater than 70% or 
75% of OTB was recommended in order to effec- 
tively reduce intra- operative blood loss [15, 19]. For 
peripheral lesions, patients with OTB >70% had an 
average of 550 ml EBL, which was significantly less 
thanpatients with OTB of <70% [15, 19]. With proper 
embolization technique, sufficient OTB could be 
obtained in more than 75% of patients who under- 
went preoperative ejnboliz.it ion [19]. Some authors 
used the criterion of complete/incomplete OTB to 
indicate the success of the procedure [13, 20]. An 
average of 535 ml EBL (ranging from 200-1600 ml) 
was observed in patients who had complete OTB. 
However, complete OTB could be obtained only in 
36% of all patients [20]. EBL did not significantly 
correlate with tumor size or vascularity prior to 
embolization [19]. The effect of embolotherapy on 
OTB was based on a planimetric comparison of pre- 



and fostemloolizahoii arteriograms [19], ("mises [in- 
patients with <70% of OTB were multifold: (1) severe 
tortuosity and irregularity of vessels secondary to 
remarkable atherosclerotic disease precluded super- 
selective catheterization; (2) tumor feeding vessels 
that arose directly from a limb- supplying artery 
were so small that a microcatheter could not be 
seated well enough for embolization without risk of 
nontarget ischemia (Fig. 15.4a,b); and (3) proximal 
embolization with coils or Gelfoam pledge [19]. 

It is generally recommended that surgery be per- 
formed within 24 h after embolization, since flow 
re constitution through collaterals increases with 
time [14]. For patients who were embolized with 
coils only and who were operated more than 48 h 
after embolization, significant intraoperative blood 
loss was encountered during the surgery. This high 
blood loss was observed although more than 70% 
obliteration of tumor blush had been achieved in 
some patients [19]. The small number of patients 
makes it difficult to assess whether this result is 
related to the type of embolization material or the 
timing of surgery. In patients who were embolized 
with smaller-sized PVA, the timing of surgery may 
not be critical; there was no significant difference 
in EBL between the patients who were operated on 




g 15.4a,b. Obliteration of ;nmor si.- in oy 00%, 700 ml of estimated blood loss. survival j months, a Pre -embolization arte- 
riogram showing a hype: vascular lesion in the rig lit r.islal ft in ur. The branches supplied a tumor arising directly from the 
poolitea! a:\ery; many of ihem vvere too small for s.ile embolization Mi tousi. b f'os'.emb' 'kzation ,11 lei :■ 'gum vci;h residua! 



within 24 h and those who had surgery 36 to 120 h 
after embolization. Use ot'PVA in cases of an antici- 
pated delay in surgery is therefore recommended 
[19]. 



15.4.2 

Osseous Healing and Survival 

Bone healing can be affected adversely by many 
variables, such as presence of tumor cells, poor 
local blood supply, systemic disease, malnutrition, 
corticosteroid therapy, and iatrogenic inference [34, 
35]. Preoperative embolization has the potential to 
induce ischemic changes in the tissues adjacent 
to the tumor and may theoretically interfere with 
osseous union. However, there has been no evi- 
dence of delayed postoperative healing or nonheal- 
ing reported in the literature [10, 19, 34, 35]. In all 
reported patients, bone healing ensued as evidenced 
by radiography, progressive callus formed at 3 and 
6 months, and hardware was in place without com- 
plication (Fig. 15.5a-e). When bone healing was evi- 
dent, functional recovery largely depended on the 
type of surgery the patients received. 

The oncological objective of excising metastatic 
RCC of bone is the achievement of local tumor con- 
trol. Patient survival is determined by metastatic 



disease at other sites, inherent biological behavior 
of the tumor, and tumor response to adjuvant treat- 
ment modalities. Therefore, the rate of local recur- 
rence is the most appropriate criterion by which 
to evaluate the oncological adequacy of resection 
margin [10]. 

The appropriate goal of pre -operative emboliza- 
tion is to decrease intra-operative blood loss, fol- 
lowed by reduction of the amount of blood transfu- 
sion and surgical morbidity/mortality. As a result, 
shortened duration of surgery and hospital stay can 
be expected [10, 19, 25]. A median hospital stay of 
8 days was reported with minimal surgical compli- 
cations in a relative large surgical series [10]. Res- 
toration of musculoskeletal function and release of 
pain therefore are paramount for the quality of life 
of these patients [19]. 



15.4.3 
Complications 



Postembolization syndrome (PES) it 
occurrence after pre-operative embolization of bone 
metastases. It was reported that PES is not neces- 
sarily regarded as a complication [36]. Marked PES 
may indicate a successful embolization and has been 
associated with a good clinical response to the embo- 



.e Metastases from Renal Cell Car 



'.: Preoperative Embolization 




Fig k"."a--d. Obi iter.; I ion o:' iumor stain by mo:e tha:'. t !?'o. Iniracpei alive bloo; loss of 100 mi. survival 0-1/2 yvars wi:h norma! 
healing jn; ljmiup.ared function of her left uppe: extremr.y. a,b Pre-^nbolizaiion; c,d poslenfLicazalion. a Plain ladiograofi 
vmh la:ge ■. -^:e- 'lv:io .esi..:: of ih e let": humerus aiv.i paihol igie iraouire : -.a ■ ei:>:. b E.irly a: :ei iai oaase showing hvpervasoulai 
c Arteriogram ,i:'ter "■■.aerse;eo:iv- embolization o) the let": ."ire 1 .: mil ex humeral ai;e:ies with 500-|.im PVA particles 
i'j). d Radiograph -■:■ months after ein notation and surgioal repair shows [lorm.ii ocne heaang w-i:h hardware in place 



{"' 



") 



lization [36]. Common symptoms are fever (usually 
below 38°C), malaise, and pain. PES usually subsides 
within 3-5 days without treatment. However, severe 
PES niav deter ri surgeon from im mediate interven- 
tion. When PVA particle is used, such a delay should 
not have adverse effects on intraoperative blood loss. 
Reported complications include non target emboli- 
zation, skin necrosis, and ischemic ulcer [36]. One 
spontaneous pathologic fracture was reported 12 h 
after the embolization with PVA and additional 



coils. More than 95% of obliteration of tumor blush 
had been achieved by superselective embolization in 
this patient (Fig. 15.6a-e). The patient reported no 
apparent external forces on his leg after the emboli- 
zation. It is conceivable that acute ischemic changes 
following embolization in a hypervascular tumor 
may cause volume changes eliciting a spontaneous 
cortical collapse. Although patients with pending 
fractures are already handled carefully, extra cau- 
tion during patient transport and immobilization of 




Fig 15.6a-e. Patient complicated 
with pathologic fracture 12 h after 
the embolization, a Plain radiograph 
shows a large osteolytic lesion in the 
proximal one-third of right femur 
with impending pathologic fracture 
(arrows), b A hypervascular lesion 
supplied by branches of the 
femoris artery (am; its), c Complete 
embolization of tumor feeders. d,e 
Plain radiographs show pathologic 
fracture and surgical internal fixation 



the affected extremity is recommended after embo- 
lization [19]. 

While complications can be reduced through 
abiding by proper technique, risks are higher in cer- 
tain anatomic locations. During pelvic emboliza- 
tion, occlusion of blood vessels supplying the sciatic 15.5 
nerve may cause ischemic neuropathy, which can be Conclusion 
avoided by sparing the inferior gluteal artery during 
embolization. Also, muscle branches of the supe- 
rior gluteal artery or deep femoral artery shi 
be spared to prevent 
emholixationshould 1 



of tumor feeders with si 
vertent embolization. 



irticles to avoid 



Retrospective studies are of limited value : 

mining the exact effect of a certain form 

Coil ment. Intra- operative blood loss depends 

formed before emboliza- on the type of surgery, but the individual 



skills of 



.e Metastases from Renal Cell Car 



'.: Preoperative fimholiza 



surgeon as well. Theoretically, a prospective and 
randomized study would be desirable for precisely 
evaluating the effects of pre-operative emboliza- 
tion. However, according to the nature of clinical 
studies, the necessity and possibility of conducting 
such a study are in question. Practically, pre-opera- 
tive embolization ol hypervnsailar bone metastases 
from renal cell carcinoma is a safe and minimally 
invasive procedure in experienced hands. Best 
results can be achieved with the use of PVA particles 
and when >70% of tumor blush is obliterated. This 
has become a standard pre-operative procedure for 
patients with hypervascular metastatic disease of 
bone from RCC. 



Parker SL, Tong T, Bolden S and Wingo PA [1005) Cancer 
;, 2005. CA Cancer J Clin 65:5-27 
;a SS, Silverman DT, McLaughlin JK et al. (1990) Com- 
»n of the descriptive epidemic-. ogy of urinary tract 
r. Cancer Causes and Control 1:133-141 
i,P (2000) Renal eel! card noma: Presentation, staging 
and surgical treatment. Eenun Oncol 27:160-176 
Swanson DA, Orovan Wl, Johnson DE, Giacco G (1981] 
Osseous metastases secondary to renal cell carcinoma. 
Urology 18:556-561 

Selli O, Hinshaw WM, Woodard BH, Paulson DF (1983) 
Gratification of risi* factors in renal ceil carcinoma. Cancer 
52:899-903 

King GJ, Kostuik JP, McBroom RJ, Richardson W (1991) 
Miigica! management of metastatic renal carcinoma of the 
spine. Spine 16:265-271 

Kessler O, Mukamel E, Hadar H et al (1994) Effect of 
improved diagnosis oi renal cell carcinoma on the course 
of the disease. J Surg Oncol 57:201 

Maldazys |D and deKernio IB (1986) Prognostic factors in 
metastatic renal carcinoma. ; Urol 136:376 
Kollender Y, Bickels J, Price WM. et al. (2000) Metastatic 
renal cell carcinoma ':•': Pone: Indications .md technique of 
surgical intervention. J Urol 164:1505-1508 
Bowers TA, Murray ]A, Charnssngayej C, Soo CS, Chuang 
VP VVal.ace i : : ?BJ : hone metastases from renal carcinoma: 
The pre-operative use of transcatheter arterial occlusion. 1 
Bone Joint Surg (Am) 64-A:749-754 
11. Carpenter PR, Ewing JW.CookAI.Kuster AH (1977) Angio- 
graphic assessment and control of potential operative heni- 
orrnage with pathologic fracttires secondary lo metastasis. 
Clin Orthop 123:6-8 

Manke C, Bretschneider T, Lenhart M et al. (2001 ) Spinal 
metastases from rena. cell carcinoma: effect of preopera- 
tive pari icle embolization on intra ope rat ive blood loss. Am 
I Neuroradiol 22:997-1003 

Carrasco CH (1991 ) Embolotherapy of bone and soft-tissue 
tumors. In: Kadir S ieo I Current practice of interventional 
radiology. Decker, Philadelphia 
Gellad FE, Sadato N, Numaguchi Y, Levine AM (1990] Vas- 



n of the spine: preoperative emboliza- 
tion. Radiology 176: 683-686 

s.Keller FS, Roach I, Bird CB (1983) Percutaneous emboliza- 
iion or bony pelvic neoplasms with tissue adhesive. Radiol- 
ogy 147^1-27 

S.Rowe DM, Becker GJ, Rabe FE et al. (1984] Osseous metas- 
tases from rena, cell carcinoma: embolization anc. surgery 
for restoration of function. Radiology 150:673-676 

7. Barton PP, Waneck RE, Karnel FJ et al. (1996) Embolization 
of bone metastases. J Vase Interv Radiol 7:81 -88 

3. Sun S, lang EV (1998) Bone metastases from renal cell 
carcinoma: Preoperative embol.zauon. I Vase Interv Radiol 
9:263-269 

?. Chatziioannou AN, lohnson \!E, Pneitmaticos SG et al. 
(2(100) Preoperative embolization of bone metastases from 
renal cell carcinoma. Eur Radiol 1 0:593-596 

}. Lavrenkov K, Meller I, Cohen Y (2002] Solitary bone metas- 
tasis of renal cell carcinoma treated with I into -spa ring sur- 
gery followed by radiotherapy. Isr Med Assoc 4:385-386 

I. Motzer RJ, Bander NH, Nanus, DM (1996) Renal-cell carci- 
noma. N Engl J Med 335:865 

'.Tuncay IC, Condrad EU (1993) Metastatic bone disease. J 
Arthroplasty Arthrosc Surg 7:80-821 8 

i. Rossi C, Ricci S, Boriani Set al (1990] Percutaneous trans- 
catheter arterial embolization o: bone and soft tissue 
tumors. Skeletal Radiol 19:555-560 

l.Berkefeld J, Scale D.Kirchner Jet al. (1999) Hypervascular 
spinal tumors: .nfhintce of litr embolization technique 
on perioperative it e in or rh age. Am I Neuroradiol 20:757- 
763 

5. Olerud C, Johnson H Jr. Lofberg AM et al. (1993) Emboliza- 
tion of spinal metastases reduces perioperative blood loss. 
Acta Orthop Scand 64:9-12 

i. Smith TP, Gray L, Weinstein JN et al. (1995) Preoperative 
traittsrtriia.-rntb'j/at.C'it :f spina: commit neoplasms. 
Vase Interv Radiol 6:863-869 

7. Siskin GP, Englander M, Stainken BF et al. (2000) Embolic 
agems used lor uterine tibroid embolization. Am j Roent- 
genol 175:767-773 

s. Kunstiinger F.Brunelle F.Chaumont P.Doyon D (1981 ) Vas- 
cular occlusive agents. Ant I Roentgenol 136:151-156 

). White RI, Stranberg JV, Gross G, Barth K (1977) Therapeu- 
tic embolization with long term occ. tiding a gems and their 
effects on embolized tissues. Radiology 125:677-687 

IHortonJA, Marano GD, Kerber CW et al. (1983) Polyvinyl 
alcohol foam -gel foam for therapeutic embolization: A syn- 
ergistic mixture. Am | Neuroradiol 4:143-147 

I. Pisco JM, Martins JM, Correia MG (1989] Internal iliac 
artery: Embolization to control hemorrhage from pelvic 
neoplasms. Radiology 172:337-339 

'.Merland J-J, Charas J (1981) Arteriography of the pelvis. 
Djiigiicsf!;" aid TViiTi incline P'0\.\'jtircs. pp. 3-4. Springer 
Verlag, Berlin 

S. Buckwalter JA, Cruess RL (1991) Healing of the musculosk- 
eletal tissues. In: Rockwood CA Jr, Green DP, Bucholz RW, 
eds. hoikii-ooti tiiiii 'jtveu's fiitiiitiis .» intuits. 3rd ed., pp 
181, Lippincott, Philadelphia 

l.Hulth A (1989) Current concepts of fracture healing. Clin 
Orthop 249:265-284 

s. Hemingway AP and Allison I'll ;'955l Complications 
ot embolization: analysis of 410 procedures. Radiology 
166:609-672 



16 Embolotherapy for Organ Ablation 



David C. Ma 



6.1 Eraboloiii e:\ipv m M judgement or Keiul 

Tumors 201 
6.1.1 Introduction 201 

6 I .'. A 'xi: mil and Angiographic Consideralior 
6 .1 I'.vl:*' .„■■.. Considerations 203 
6.!.; Results 207 
6.1 .4.'. Krabolottlerapy for Renal Cell Car 

6.1 .4.2 Embolotherapy for Angiomyolipor 
6.1.5 Future Directions 211 

6.2 Embolotherapy in Management 
of Hypersplenism 211 

6.2.1 Introduction 211 

6.2.2 Anatomic Considerations 2J2 

6.2.3 Technical Considerations 212 

6.2.4 Results 214 

6.2.5 Future Development and Research 

6.3 Conclusion 215 
References 217 



16.1 

Embolotherapy in Management of Renal 
Tumors 

16.1.1 

Introduction 

Malignant tumors of the kidney and renal pelvis 
accotint for -3% of all adult malignancies. There 
were -31,200 new cases in 2000 contributing to 
-11,900 deaths in the United States [1]. Renal cell 
carcinoma is the most common primary malignant 
tumor of the kidney and occurs in both sporadic and 
hered itary forms [2]. Most cases are sporadic, but at 



D. C. Madoff, MD; R. Verua, MD; K. Ahrar, MD 
Section of Interventional kiidiology, Division of Diagnos- 
tic Imaging, The "University of Texas, \'I> Anderson Cancer 
Center, 1515 Holcombe Boulevard, Unit 325, Houston, TX 
77030-4009, USA 



least four forms of hereditary renal carcinoma have 
been identified. The most studied form of heredi- 
tary renal cancer is von Hippel-Lindau syndrome, in 
which affected individuals have a predisposition to 
develop tumors in various organs, including the kid- 
neys. In the hereditary syndromes, kidney cancer 
is often bilateral and tends to occur in younger 
patients. Renal cell carcinoma may remain clinically 
occult for most of its course; the classic presentation 
of pain, hematuria, and palpable mass occurs in 
only 9% of patients. Approximately 30% of patients 
with renal cell carcinoma present with metastatic 
disease, 25% with locally advanced tumor, and 45% 
with localized disease. Widespread use of routine 
abdominal imaging has led to increasing detection 
of smaller lower stage renal tumors [3]. Therefore, 
the natural history of renal cell carcinoma may be 
changing [3]. Historically, radical nephrectomy was 
considered the only effective treatment for patients 
with renal cell carcinoma. More recently, nephron- 
sparing surgery has been accepted as an alternative 
for smaller tumors (less than 4 cm) localized to the 
kidney [3]. Although promising for the management 
of smaller tumors, energy ablat ive therapies, includ- 
ing cryotherapy and radiofrequency ablation, are 
investigational and are reserved for patients who 
are not good surgical candidates, patients with only 
one kidney, and those with multiple bilateral tumors 
[4, 5]. Metastatic renal cell carcinoma is unrespon- 
sive to conventional chemotherapy used to treat 
other metastatic solid tumors, but treatment with 
immunotherapy has resulted in durable responses 
in selected patients [3]. Before the era of immuno- 
therapy, the course of metastatic renal cancer was 
not affected by debulking nephrectomy, and surgery 
was reserved only for palliation ot svmptoms. Recent 
trials of nephrectomy and immunotherapy versus 
immunotherapy alone have demonstrated statisti- 
cally significant improvement in overall survival in 
patients with metastatic renal cell carcinoma [6]. In 
light of these findings, nephrectomy has become 
the initial component of multimodality therapy for 
liietii.itatic renal cancer. 



P. C. Madoffel 



Surgical resection of larger i 
is challenging because both the kidney and the 
tumor are extremely vascular and dissection of 
the tumor may lead to clinically significant blood 
loss, necessitating replacement of a large volume 
of blood. In addition, renal hilar lymph nodes may 
interfere with visualization and access to the vas- 
cular pedicle [7]. Early ligation ot T lie renal .me it 
is necessary to avoid substantial bleeding, but the 
renal vein lies anterior to the artery and may be 
encountered before the renal artery. For these rea- 
sons, starting in the 1970s, the technique of renal 
artery embolization was popularized as an adjunct 
to surgery. Since that time, embolization has been 
used before resection of larger tumors and tumors 
invading the renal vein or inferior vena cava [8, 
9]. When used appropriately and in the right set- 
ting, the technique purportedly decreases intra- 
operative blood loss and creates a plane of edema 
that facilitates the dissection of the tumor [8]. 
The same technique has been used for palliation 
of patients who may not be surgical candidates. 
Renal artery embolization has been used to allevi- 
ate tumor-related symptoms like hematuria, flank 
pain, paraneoplastic symptoms such as hypercal- 
cemia or polycythemia, congestive heart failure, 
and hypotension [10, 11]. Other potential, but as of 
yet unproven, benefits of renal artery embolization 
include inhibition of tumor growth and improved 
rarvival [12]. 

Angiomyolipoma is generally a benign tumor 
of the kidney, although a more aggressive sub- 
type (epithelioid angiomvolipoma) has also been 
described [13]. The tumor is composed of mature 
fat cells, vascular tissue, and smooth muscle in 
various proportions. Clinically, angiomy.'lipi: 



eoft 



Table 16.1. Clinical 
contemporary surgical 



> distinct settings. It may be 



Tuberous Sporadic p-vatue 
sclerosis- angiomyo- (chi-squar 
associated lipoma test) 

angiomyo- 
Upon.. 



Mean age 30.3 

Tumor diameter fern) 8.9 
% Multiple tumors 97 
% A- presentation: 

- Symptomatic 64 

- A.rute hemorrhage 44 



l: Nelson and Sanda [13] -(printed with pi 



associated with the tuberous sclerosis complex, 
which is an autosomal dominant disease with 
incomplete penetrance. The triad of seizures, 
mental retardation, and adenoma sebaceum char- 
acterizes tuberous sclerosis in its classic clinical 
presentation. More commonly, angiomyolipoma is 
encountered sporadically in individuals who oth- 
erwise have no clinical features of tuberous sclero- 
sis. Angiomyolipoma is also associated with pul- 
monary lymphangioleiomyomatosis. The clinical 
presentation of angiomyolipoma maybe similar to 
that of renal cell carcinoma. Patients may present 
with flank pain, a palpable tender mass, or gross 
hematuria, and possibly nausea, fever, hyperten- 
sion, or anemia. In patients with tuberous sclerosis 
and multiple a ninomvohpomas, renal insult ideiR'v 
or failure may be the initial sign of the tumor. The 
most dramatic and life-threatening presentation of 
angiomyolipoma is spontaneous retroperitoneal 
hemorrhage. Angiomyolipomas associated with 
the tuberous sclerosis complex are often multiple, 
bilateral, and larger at presentation. These tumors 
are more prone to spontaneous hemorrhage than 
their sporadic counterparts (Table 16.1). 

Before 1976, 93% of reported angiomyolipomas 
not associated with tuberous sclerosis were treated 
with total nephrectomy, whereas in 1984, only 50% 
of all angiomyolipomas reported in the literature 
were treated with total nephrectomy [14]. Because 
of advances in embolization and partial nephrec- 
tomy techniques, most angiomyolipomas are now 
treated conservatively where the goal is control of 
symptoms, prevention of life-threatening hemor- 
rhage, and preservation of renal function. Other 
than patients who present with spontaneous bleed- 
ing and require urgent treatment, the timing of 
intervention for ajigiomyollpoma remains a sub- 
ject of debate. In one study, 90% of symptomatic 
tumors were 4 cm or larger and 64% of asymptom- 
atic tumors were smaller than 4 cm [14]. In another 
study, small (less than 4 cm), medium (4-8 cm), 
and large (greater than 8 cm) tumors were symp- 
tomatic in 10%, 54%, and 100% of cases, respec- 
tively [15]. Primary indications for intervention 
include pain, retroperitoneal hemorrhage, and 
hematuria regardless of tumor size, but prophy- 
lactic intervention is reserved for larger tumors in 
high-risk patients, in females of childbearing age, 
and for those patients for whom follow-up or access 
to emergency care may be inadequate. Regardless 
of the indication, preserving renal tissue and func- 
tion is a significant concern when treatment is con- 
templated. 



Embolotherapy for Organ Ablat 



16.1.2 

Anatomic and Angiographic Considerations 

A thorough understanding of the renal vascular 
anatomy is a prerequisite for successful emboliza- 
tion of renal tumors and helps minimize inadvertent 
nontarget embolization. Most commonly, a single 
renal artery to each kidney arises from the abdomi- 
nal aorta at the level of L1-L2 disc space. Multiple 
renal arteries to one or both kidneys may be pres- 
ent in 12%-32% of individuals [16]. The main renal 
artery frequently divides into anterior and posterior 
branches. Because of the location of the kidney in 
the retroperitoneum, the ventral or anterior divi- 
sion of the renal artery supplies the lateral border 
of the kidney in an anteroposterior angiogram, 
whereas the dorsal or posterior division provides 
blood supply to the medial border of the kidney. Fur- 
thermore, the anterior division appears as a direct 
continuation of the main renal artery, whereas the 
posterior division is smaller in caliber and appears 
as a branch of the main renal artery [17]. Branching 
of the renal arteries continues to form segmental, 
lobar, interlobar, and arcuate arteries. The arcuate 
arteries are end arteries and do not anastomose with 
one another; instead, they subdivide into interlobu- 
lar arteries, which in turn give rise to afferent glo- 
merular arterioles [18]. 

Perforating arteries, an important collateral path- 
way to the kidney, arise from the intrapareikhyma! 
branches of the renal artery and exit from the kidney 
to anastomose with various retroperitoneal arteries 
[18]. In addition to the main renal artery and per- 
forating arteries, the superior, middle, and inferior 
capsular arteries should be considered as well. The 
superior capsular artery may arise from the infe- 
rior adrenal artery, main renal artery, or aorta. The 
middle capsular artery, which may consist of one or 
more branches, arises from the main renal artery. 
The inferior capsular artery may originate from the 
gonadal artery, an accessory or aberrant lower pole, 
or even the main renal artery. These vessels form a 
rich capsular network that anastomoses freely with 
perforating arteries and other retroperitoneal (espe- 
cially lumbar) arteries and also with internal iliac, 
intercostal, and mesenteric arteries [18]. 

Angiography is no longer performed in the diag- 
nostic workup of renal masses and is reserved for 
therapeutic interventions only. The study should 
start with an abdominal aortogram to determine the 
number and location ot arteries supplying the kidney 
and the tumor. Selective catheterization and angiog- 
raphy of each feeding artery is performed to assess 



the extent of neovascularity, degree of arteriovenous 
shunting, and risk of nontarget embolization. 

Most renal cell carcinomas appear hypervascular 
on angiography (Fig. 16.1} and demonstrate tumor 
vessels that are irregular, tortuous, randomly distrib- 
uted, variable in size, and unpredictable in branching 
[19]. Other findings may include pooling of contrast 
in dilated vessels, arteriovenous shunting, staining of 
the tumor during the capillary phase, mid renal vein 
invasion or occlusion by the tumor. 

The angiographic appearance ot a iii:i.i myoli- 
poma is that of a hypervascular mass with a large 
feeding artery [19]. The vascular components of the 
tumor are thick-walled arteries characterized by 
the absence of the internal elastic membrane and 
a disordered adventitial cuff of smooth muscle, 
which results in the numerous saccular aneurysms 
frequently seen on arteriograms. Tortuous vessels 
may be circumferential!;' arranged in the arterial 
phase. Overall, a stinburst or whorled appearance 
in the nephrography and venous phases is sugges- 
tive of these tumors. Arteriovenous shunting is not 
a prominent feature ot angiomyolipomas. 



16.1.3 

Technical Considerations 

The size and extent of the tumor, the need to pre- 
serve any of the renal parenchyma, and the overall 
goal of embolization should be considered in plan- 
ning embolotherapy of renal tumors. Historically, 
the choice of embolic agents depended on the expe- 
rience and preference of the operator. Although 
more than twenty embolic agents have been used 
(Table 16.2), it is most worthwhile to consider three 
classes of embolic agents currently used for emboli- 
zations of renal tumors: liquid agents (the prototype 
of which is absolute ellianol), particulate materials 
(the prototype of which is polyvinyl alcohol foam}, 
and metallic coils. 

A recent review of the literature demonstrated a 
trend toward the use of ethanol for embolization of 
renal tumors [20]. Ethanol is a nonviscous liquid, 
and after it is injected into the main renal artery, it 
diffuses into the distal vascular bed of the tumor, 
potentially causing tumor necrosis rather than 
simple occlusion of the embolized artery [21]. Trans- 
catheter administration of ethanol is technically 
easy, and theoretically, reflux of a small amount of 
ethanol may not be as toxic as other embolic agents 
to other organs of the body because alcohol dilutes 
rapidly in a large volume of blood [22]. In reality, 



D. C. Madoff et al. 




Fig.16.la-c. A 42 -year-old man with heirralaria was referred 

for preoperative embolization o\ a large renal tumor. Axial 

CT image of die abdomen ia; shows a large enhancing renal 

with tumor thrombus extending lo the inferior vena 

(IVC). Selective right :c:'.a! arteriogram (b) shows neo- 

larity of the tumor. Arteriogram after embolization 

lc) shows complete occlusion of renal artery branches and 

complete devascikanzation of the tumor. At surgery, the 

right l-.idniry, adrenal gland, and [VC tumor thrombus were 

resected. Patient repaired trail -fa -ion of :wo units of packed 

red blood cells 



however, reflux of ethanol can lead to nontarget 
embolization of the inferior mesenteric artery and 
the lumbar arteries [11, 23-26]. 

Ellman et al. [21] first described ablation of renal 
tumors with absolute ethanol. On the basis of their 
experience with an animal model and six patients, 
they recommended selective injection of ethanol at 
s many tumor arteries as pos- 
ted that injection of ethanol at 
iults in tissue toxicity, thereby 
the perivascular areas, '\ltuig- 
l small arteries and glomeruli, 
tall arteries; subsequently, the 
ged and sloughs over several 
hours, resulting in complete occlusion of damaged 
vessels. Ellman et al. further elaborated that slow 
injection of ethanol (0.1 ml/s) results in little direct 
tissue toxicity and that instead, small clumps of 



i rate of 2 ml/s i 
sible. They hypothes 
a rate of 1 to 5 ml/s n 
leading to necrosis ir 
ing" of erythrocytes 
and spasm of these s 
i-aclc'thelium is dam 



damaged erythrocytes and denatured proteins lead 
to proximal occlusion of the arteries. The desired 
angiographic end points were described as occlu- 
sion of all arteries smaller than major segmental 
branches, stagnation of flow in patent major arteries, 
and extravasation of contrast material into the 
parenchyma. According to their report, injection ol 

1 ml of ethanol per4 lb (1.8 kg) of body weight results 
in a systemic blood ethanol level of about 50 mg/dl, 
which is half the intoxicating level of 100 mg/dl and 
far below the toxic level of 500 mg/dl. 

In a modification of the technique of Ellman 
et al., Rabe et al. [22] used an occlusion balloon 
catheter in eight patients. In this technique, the 
occlusion balloon is inflated in the main renal 
artery (Fig. 16.2), and ethanol is injected at a rate 
of 1 to 5 ml/s. The balloon is kept inflated for 10 s to 

2 min, and after 10 min, the catheter is aspirated to 



!-_ it.iL>-- "■ I. Mil e:.ipv :'.:■[■ Organ Ablation 



Table 16 J. Agents li-ed for diempeulic embolization of renal 
n alphabetic. I order 
:e ethanol 
Autologous muscle particles 
Avitene (microfibrillar collagen hemostat) 
Coils (nretaU steel / mini / Gianturco / GAW) 
Collagen 

Detachable balloons 
Dura particles 

Ethibloc (oily contrast- labeled amino acid) 
Fibro spurn 

■ 7- e ' M : i j j foam / . 7- ■=■ L lo ;i j j i 
Gelatin sponge / GeLtspon 
Gelfoam prepared with BCG 
Histoacryl 

Ivalon (polyvinyl alcohol] 
I C RA ( isobu ty 1- 2 - cy a no a cry late ] 
Lyodura 

MMC (microencaps'jlated or nonencapsdjied mitomycin C) 
P.i.iiCi"':. i m ethyl n; ethacrylate) 
Sp on go st an 
Tachotyp flocculi 
Thrombin 

Vilan 

From: Kal.man and Y'akenhorst [10] (primed with permis- 



e residual alcohol from it. After the catheter 
is flushed, a test injection is performed with con- 
trast material. This sequence is repeated until total 
occlusion of the main renal artery is achieved. The 
advantages of using an occlusion balloon catheter 
are several: the balloon interrupts renal blood flow, 
markedly prolonging the contact time of ethanol 
with the endothelium and thereby reducing the 
volume of ethanol needed for complete ablation of 
the target tissue; the balloon inhibits reflux into 
the aorta; and with the use of a balloon catheter, the 
main renal artery can be injected with ethanol and 
selective catheterization of segmental branches is 
not necessary. Rare et al. also suggested that injec- 
tion of ethanol in the main renal artery without an 
occlusion balloon is ineffective because ethanol 
dilutes rapidly in a large volume of blood. Instead, 
they endorsed the technique of Eliman et al. with 
selective catheterization and embolization of seg- 
mental arteries when a balloon occlusion catheter 
could not be used. 

Bakal et al. [7] described a variation of the bal- 
loon occlusion technique. They performed all renal 
embolizations during selective placement of an 
occlusion balloon catheter in the distal main renal 
artery. Estimates of ethanol volume needed for 
embolization were made by test injecting contrast 
material during balloon occlusion. After infusion of 
the ethanol, the balloon was left inflated for 5 min. 
As the balloon was deflating, gentle suction through 
the distal endhole of the catheter prevented reflux of 



residual ethanol or thrombus into the aorta. Unlike 
the study by Rare et al., no attempts were made 
to selectively catheterize or embolize other tumor 
vessels; embolization of the kidney rather than the 
tumor was the intended effect. The retrospective 
analysis of Bakal et al. demonstrated that complete 
ablation of large, hypervascular tumors was asso- 
ciated with a significantly reduced blood transfu- 
sion requirement at nephrectomy, whereas tumors 
that were partially ablated (accessory renal arteries, 
polar branches, or lumbar arteries were not embo- 
lized) required larger amount of blood transfusion 
than the control group did. 

Absolute ethanol is readily available, inexpensive, 
and easy to handle, but its major drawback is that 
it is not radiopaque. Early experiments in mixing 
ethanol with radiographic contrast resulted in pre- 
cipitation of the mixture [22]. However, ethanol may 
be mixed with iodized oil (Lipiodol or Ethiodol) in 
a 1:3 mixture for successful embolization of angio- 
myolipomas [27]. Konya et al. [28] suggested that a 
1:1 mixture of ethanol and iodized oil followed by 
absolute ethanol may be an effective embolic agent 
for ablation of renal tissue. 

Particulate materials, such as absorbable gela- 
tin sponge (Gelfoam) and polyvinyl alcohol foam 
(Ivalon), have been used alone and with metallic 
coils [29]. Newer embolic agents such asEmboGold 
Microspheres (BioSphere Medical, Rockland, MA) 
and Contour SE (Boston Scientific; Natick, MA) 
are also used for embolization of renal masses, 
but no reports of their effectiveness or lack thereof 
are available in the literature; theoretically, these 
newer agents should not be any less effective than 
Gelfoam or Ivalon. There is no scientific evidence 
pointing to a particular particle size for emboliza- 
tion of renal masses. Despite the lack of informa- 
tion, some recommendations can be made here. 
In general, distal embolization of the tumor bed 
rather than proximal occlusion is desirable; there- 
fore, embolization should start with particles small 
enough to avoid proximal occlusion. Hyper 
lar renal tumors may have considerable a 
nous shunting, and nontarget embolization of the 
pulmonary bed is a concern. In tumors with rapid 
arteriovenous shunting, the use of a sclerosing 
agent such as ethanol or larger particles may be 
prudent. The use of coils for palliation of tumors 
is discouraged because renal tumors in advanced 
stages often have an extensive collateral blood 
supply, rendering proximal occlusion of the feed- 
ing arteries ineffective. Furthermore, many tumors 
require additional interventions, so arterial access 



D. C. Madoff et aL 




.a and right flank pain was referred for pallia- 
tive embolization. Selective digital s.c. refaction angiogram of the right main renal artery ;aj shows early [".furcation of the 
renal aneiv, n.eovascularily. tumor soain, jno scosiamial aneriovenoi.s shinning. The mferio: vsi^a cava is opacified in the 
background. After place men I cc" .in occlusion balloon catheter il>), ti'.e anterior branch -:A the renal arterv was embolized wiih 
I ml of dehydrated alcohol ! absolute ei ha noli. Selective angiogram of Ir.e posteiior branch ot" the righ: renal arterv ;c; snows 
blooo supply to ihe reminder of the tumor. Embolization, of mis branch was completed using EmboGo.d Microspheres. Selec- 
live angiogram o: ti'.e main rena. artery id! shows complete occlusion of bolh anterior and posterior branches. The tumor is 
completely devascularized 



to the neoplasm must remain intact [30]. If coils are 
used for preoperative embolization, they should be 
sized and placed appropriately to minimize the 
risk of dislodgment at the time of surgery and to 
avoid difficulties with renal artery clamping or 
ligation [29]. 

Although firm scientific evidence is lacking, 
complete embolization of the renal tumors may be 
a desirable end point both for preoperative and pal- 



liative purposes [7, 11, 27, 31]. Because most renal 
cell carcinomas are hypervascular and often recruit 
collateral blood supply from other sources as well, 
many operators use a combination of techniques 
and embolic agents to achieve complete emboliza- 
tion (Fig. 16.2). Therefore, it seems appropriate for 
the operator to be familiar with various techniques 
and have access to a full complement of different 
embolic agents. 



iimbo I. vrli e:.ipv :or Orpin Ablution 



16.1.4 
Results 

16.1.4.1 

Embolotherapyfor Renal Cell Carcinoma 

Renal artery embolization was popularized by Alm- 
gard in the early 1970s. Over the next two decades, 
investigators debated the usefulness of embolother- 
apy in the management ol renal cell carcinoma, but 
the decreasing number of reports in the literature 
since then indicates a loss of interest in this tech- 
nique over time, particularly in the last decade. Most 
of the published reports are now over 20 years old. 
Angiography, catheterization, and embolization 



techniques have evolved over the last three decades. 
A review of this older literature, which may not be 
reflective of the current practice of embolotherapy, 
is nevertheless warranted to elucidate the evolution 
of the technique, point out some of the shortcom- 
ings of previous studies, and highlight questions 
that remain to be answered. 

In 1999, Kalman and Varenhorst [20] pub- 
lished a survey of the pertinent literature which 
appeared during 1973-1997 on renal artery embo- 
lization for management of renal cell carcinoma 
(Table 16.3). Each one of the selected articles pre- 
sented a minimum of 20 embolization cases, and 
each study aimed to investigate the clinical effect of 
embolization. All reports were scrutinized for study 



Table 16.3. Case sei 


:e^ : ■.'. biis:: ed in i:ie - :■«.!■■:: .ke:,i:iu e. i 


n order of year c 


fpublicatior 


■., agents used for the 


pen tic embolizatio 


n, number of 


patients, and indication for embolization 






First Author 


Year of 


Method 


Number of 


Pre- 


Palliative Not 




publication 




L\iiie:iis 


oper.iiive 


specific 


AlmgArd [45] 


1977 


Muscle particles 


38 


29 


9 


Fhasson [49] 


1978 


Gelfojim 




35 


10 


ScBttLMAN [141] 


1980 


Coils 

Geltornu 
Gelfoam + coils 
ICBA 


3 
10 
9 


26 


2 


Frasson [50] 


1981 


Coils 

Gelfoam 
Gelfoam + coils 
ICBA 
Muscle particles 


3 
2 


241 


- 


Gnn.iANAl53] 


1981 


Coils 


1 










Gehl.n fo;ini 




40 








ICBA 


25 






Kato [142] 


1981 


Encaps MMC + gel 
Sponge 


33 


23 


10 


Mobilio 148] 


1981 


Geif '.iir. 
ICBA 


f 


1 




Wallace 129] 


1981 


Gelfoam + coils 


100 




26 


Le Glillol- [143] 


1982 


No: specified 


247 


203 


44 


Teasdale [144] 


1982 


Coils 


3 










Gelfoam 


22 


26 


2 






Gelfoam + coils 


3 






Bono [39] 


1983 


Avitene 


47 


47 








Geltoum 


48 


48 








ICBA 






4 


Nakano [36] 


1983 


Gelatin sponge 
ICBA 


21 


12 


9 


Ekelund [40] 


1984 


Et!:.inol 


20 






Kafsary [35] 


1984 


Coils 

Gelatin sponge 














55 


49 








Thrombin 












C omnium ions of the ,ibo 








KUHTH [32] 


1984 


Coils 

Etlianol 












Gel to;; in 


25 


25 








ICBA 









Table 16.3. Continued 








First Author 


Year of 


Mel::od 


Number of Pre- 




pilNkMi-. 




p :-.i. . e i u s 


opera ti 


Mcivor [33] 


1984 


Coils 










Dura particles 


29 


29 






Geli"' ;;i:"r. 










Thrombin 






Mebust [52] 


1984 


Coils + gelatin sponge 
Elhanol 


5 


4D 


Christens en [42] 


1985 


Coils 


36 


36 


GOTTESMAN [41] 


1985 


Coil + Gelfoam 


30 


30 


Klimbehg [8] 


1985 


Elhanol 


25 


21 


Lavimer [24] 


1985 


E-.h,ni::i| 


7 










85 


81 






[v.: Ion 


25 




Leinonen [38] 


1985 


Edionol 


12 


10 


Wejgel [34] 


1985 


H:hoi]:nl 


22 


22 


Chudacek [145] 


1986 


r:.hil- ,:-: 
Vilan 


30 




Karwowski [146] 


1987 


Gelfoam 


81 





Palliative Not 



[•AT- [40] 

UwnUH [4: 
Bakal [7] 



E7I1..1110I 

ICBA 

Ethanol 

Ethanol + coils 

Gelfoam 

Gelfoam + coils 

ICBA 

ICBA + coils 

ICBA + coils + Gelfoam 

Detachable balloons 

Hi' : k'.K:y. 

Ivalon 

Coils 

i .jclj"i;;"i:r. 

Ivalon 

Gelfoam + BCG 

Encaps MMC 

K j l ^ Li y-^ MY.'Z - Gclto.im 

Elhanol 

Ethanol 

Ethanol + coils 

Ethanol + Gelfoam 

Ethanol 

1st [20]-( with modification,] 



design, control-comparison group, recruitment, 
patient selection, tumor stage, method of emboli- 
zation, delay between embolization and operation, 
measurement of intended outcome, adverse out- 
come of treatment, and assessment of the extent to had historical control 
which therapeutic embolization provided a solution spective randi 
to the clinical problem. 



Of the 51 publications, only 7 were prospective 
studies [8, 32-37]. The remainder were retrospec- 
tive studies, or the study design was not specified. 
Most studies were from observation, and only four 
i. As of 2004, no pro- 
trial of renal artery 
embolization has been reported. The methodologic 



!-_ it.iL>-- "■ I. Mil e:.ipv :'.:■[■ Orenn Ab hit ion 



weaknesses of the published studies make it diffi- 
cult to compare the reports and draw firm conclu- 
sions; therefore, evidence regarding renal artery 
embolization in the literature is weak and must be 
interpreted with caution. 

Recruitment of patients to the studies on preop- 
erative embolization varied widely. Some investiga- 
tors included patients with "large primary tumors", 
and others recruited patients with tumors larger 
than a specified diameter [34, 38, 39]; one study did 
not consider tumor size as a selection criterion [24]. 
Eligibility criteria for palliative embolization were 
more commonly reported lis palliation ot pain, hema- 
turia, or life-threatening endocrine tumor activity 
[11, 24, 37, 38, 40, 41]. In one study, palliation was 
not defined with respect to any symptoms but as an 
alternative therapy for patients with advanced dis- 
ease who were not surgical candidates [24]. Several 
of the 51 papers reported on patients who underwent 
either preoperative or palliative embolization. Over- 
all, the studies varied greatly in their patient selec- 
tion criteria, and details regard iiij; patient condition 
and tumor stage were often lacking. 

Over the span of three decades, more than 20 
embolic agents have been used. Few authors used 
one agent only for embolization in their study [8,40, 
42-45]; most introduced combinations of agents or 
used different agents for embolization in their trials. 
Earlier studies used absorbable gelatin sponge and 
coils, which are still used in preoperative devascu- 
larization of renal tumors. For palliation, more per- 
manent agents such as Ivalon and ethanol are pre- 
ferred. More recently, absolute alcohol has become 
the recommended agent for embolization of renal 
tumors for all indications [7,43,44]. 

The time between embolization and nephrectomy 
was reported in 34 of the 45 studies on preopera- 
tive embolization. This interval varied trom 8 hours 
[8] to 183 days [46]. In one study, a planned delayed 
nephrectomy was thought to result in development of 
edema and facilitate dissection [9], whereas another 
study reported a more difficult dissection 3 days or 
longer after embolization because of increasing col- 
lateral circulation [47]. On the basis of the results of 
the reviewed articles, Kalm an and Varenhorst [20] 
concluded that the optimal delay between emboliza- 
tion and operation was less than 48 hours. 

Several variables have been considered in mea- 
surement of the intended outcome, including intra- 
operative blood loss. Twelve studies evaluated intra- 
operative blood loss after renal artery embolization. 
In three studies, blood loss was based on subjec- 
tive estimation only [48-50]. One study compared 



blood loss after embolization using ethanol and 
Gelfoamand found no significant difference [38]. In 
three studies, the difference in the amount of blood 
loss in embolized and nonembolized patients was 
not statistically significant [42, 43, 51]. One study 
reported greater blood loss for embolized patients 
compared with historical data, but an appropriately 
matched control group was not selected [52]. Bakal 
et al. [7] demonstrated that estimated blood loss is 
an unreliable measurement and used instead the 
transfused volume as a measure of successful pre- 
operative embolization. They demonstrated that the 
transfusion volume was statistically significantly 
lower after embolization for patients with large, 
hypervascular renal cell carcinoma (volume greater 
than 250 ml, diameter larger than 7.8 cm) than for 
smaller or hypovascular tumors. Another group 
recorded a statistically significantly lower volume 
of blood transfused in T3 and T4 tumors than in a 
historical control group [53]. Some groups believe 
that resection of renal cell carcinoma is facilitated 
by preoperative embolization [8, 47]. Reportedly, a 
plane of edema develops after embolization, making 
dissection of the tumor easier. In tumors with exten- 
sion into inferior vena cava, embolization causes the 
tumor to shrink and facilitates its resection from the 
vena cava [9]. In addition, in larger tumors extend- 
ing to the renal hilum and perihilar lymph nodes, 
preoperative embolization allows for ligation of the 
renal vein before the renal artery, alleviating some 
of the technical difficulties with resection of these 
bulky tumors [8]. 

Only four studies reported duration of surgery 
as an objective measure of intended outcome. One 
study found a shorter operating time compared with 
nonembolized patients in other published reports 
[8], and three trials found no benefit from preopera- 
tive embolization [42, 51, 52]. 

Immunologic response was also evaluated as a 
measurement of intended outcome. Wallace et 
al. [47] suggested that angioinfarction of the tumor 
may result in release of tumor antigens, resulting in 
enhanced immunity against the tumor, and also that 
embolization prolonged survival in a selected group 
of patients. Although several reports showed dis- 
torted immunologic homeostasis after renal artery 
embolization, there was no conclusive evidence that 
embolization provided immunotherapeutic benefits 
in the management of advanced renal cell carcinoma 
[36, 40, 46]. In one study, patients with metastatic 
renal cell carcinoma who were treated with emboli- 
zation and nephrectomy had a longer survival than 
patients who underwent embolization only [35]. In 



a more recent study not included in the review by 
Kai.vian and Varenhorst, preoperative emboliza- 
tion resulted in improved survival after nephrec- 
tomy: the overall 5- and 10-year survival rates were 
62% and 47%, respectively, for 118 patients emboli- 
zed before nephrectomy and 35% and 23%, respec- 
tively, for a matched group of 116 patients treated 
with surgery alone (p = 0.01) [12]. But this optimism 
is not shared by the experience of other researchers 
[11,41,42]. 

Palliation of symptoms in nonoperable patients 
was also included in some outcome analyses. The 
details of symptoms and therapeutic effects of renal 
artery embolization are generally lacking, but stud ies 
generally reported that hematuria, pain, and paraneo- 
plastic symptoms were alleviated. In one study, severe 
hematuria resolved in 11 of 14 patients, and incom- 
plete embolization of the tumor blood supply from 
parasitized lumbar arteries resulted in persistent 
hematuria in 3 of 14 patients [11]. In another study, 
malignant hypercalcemia resolved after emboliza- 
tion [10]. Kalman and Varenhorst concluded that 
a small group of patients with specific, tumor-related 
symptoms may benefit from embolization. However, 
the palliative effect of embolization cannot be evalu- 
ated from the available data, and the effectiveness of 
the procedure awaits validation. 

The most common side effect of embolotherapy 
is the post-embolization syndrome, which consists 
of low-grade fever, pain, nausea, and vomiting that 
start shortly after embolization and may last sev- 
eral days [32, 39, 47, 49]. These symptoms are often 
self-limiting and require only supportive therapy. 
Although solid evidence was not provided, some 
studies suggested that ablation of tumors with etha- 
nol may result in a lower frequency of nausea and 
vomiting and an overall milder post-embolization 
syndrome than ablation of tumors with other par- 
ticulate embolic agents will [8, 21, 38]. More serious 
complications have been reported as a result of non- 
target embolization of the large bowel, spinal cord, 
contralateral kidney, or gonadal artery [23-26]. 
Contrast-induced nephropathy, renal abscess, and 
hypertension have also been reported [47]. In their 
initial description of the technique, Rabe et al. [22] 
recommended the use of broad-spectrum antibiotics 
before and after embolization to prevent superinfec- 
tion of necrotic tumor. Many reports lack specifics, 
but the need for prophvlactic antibiotics does not 
seem to be universally accepted. In 1985, Lammer 
et al. [24] reported a 9.9% overall complication rate 
in 121 renal tumor embolizations and a mortality 
rate of 3.3%. The most common complications in 



this series were renal failure and nontarget emboli- 
zation. The complication rate was four times higher 
in the palliative embolization procedures than in 
the preoperative embolizations. The authors attrib- 
uted this discrepancy to the larger embolized tumor 
mass and the severely impaired health of patients 
who underwent palliative embolization [24]. A 
decade later, Bakal et al. [7] reported a puncture 
site hematoma as the sole complication in a group of 
24 patients who underwent preoperative emboliza- 
tion. It appears that with advancements in technol- 
ogy and experience in embolotherapy, complication 
rates have decreased substantially. 

In a recent review of our database, we identified 
30 consecutive patients (20 males, 10 females) with a 
mean age of 65 years (range, 42-85). who underwent 
embolization and surgical resection of their renal 

gery, tumor thrombus was removed from IVC in 25 
patients (83%) and from the right atrium in 3 patients 
(10%). The surgeons reported substantial bleeding 
from venous collaterals in 8 patient'; (27%), marked 
inflammatory response around kidney in 6 patients 
(20%), and ascites in 1 patient (3%). Median blood 
transfusion requirement was 2 units of packed red 
blood cells. Mean tumor size was reduced tol0.5 cm 
after resection (ji-value, 0.003). In 3 patients (10%) 
a significant reduction in size of IVC thrombus was 
noted during surgery. 



16.1.4.2 

Embolotherapy for Angiomyolipoma 

Nelson and Sanda [13] summarized contempo- 
rary advances relevant to the clinical management 
of renal angiomyolipoma. Among other factors, 
the authors cited refinement of embolization and 
partial nephrectomy techniques as major develop- 
ments in the management of these benign tumors. 
Their review of the literature identified 24 reports 
[22, 27, 31, 54-74] of a total of 76 patients regard- 
ing therapeutic embolization for angiomyolipoma 
(Table 16.4). The most common indication for embo- 
lization was acute hemorrhage. Other indications 
were symptomatic tumors inpatients who were poor 
surgical candidates or had limited renal reserve 
and asymptomatic tumors for which prophylactic 
treatment was deemed appropriate. Post-emboliza- 
tion syndrome was recorded for 85% of patients, 
other complications were reported in 10% of cases 
including renal abscess (5%) and pleural effusion 
(3%). Over a median follow-up of 23 months (range, 



Embolotherapy for Organ Ablat 



0-204 months), 17% of patie 
Corns or hemorrhage that rei 
Cion or surgery. 



16.1.5 

Future Directions 



recurrent symp- 
'epeat emboliza- 



Preoperative and palliative embolization of renal cell 
a may benefit a large number of patients, 
city of scientific evidt 



However, be 
the techn 

although embolization 
for acute hemorrhage : 
lipoma, it is not widely i 



underutilized. Similarly, 
s an accepted treatment 
patients with angiomyo- 
treat asymptomatic 



patients with this condition. Patient selection, tech- 
nical details, expected complications, and outcomes 
all remain unclear at this time. Prospective clinical 
trials and randomized clinical trials are necessary 
to evaluate the true value of embolotherapy in the 
management of renal tumors. 



Embolotherapy in Management 
of Hypersplenism 



16.2.1 
Introduction 



Surgical removal or transcatheter ablation of splenic 
parenchyma is often performed for the management 
of hypersplenism, a pathologic condition in which 
there is increased pooling or destruction of the 
corpuscular elements of the blood by the spleen, 
resulting in reticuloendothelial hyperplasia [75]. 
Hypersplenism may be seen in many disorders, 



including cirrhosis with portal hypertension [76, 
77]; hematologic abnormalities such as idiopathic 
thrombocytopenic purpura, thalassemia major, 
and hereditary spherocytosis [78-82]; and diffuse 
splenic infiltration from primary malignancies 
such as leukemia and lymphoma [83-86]. Signs of 
hypersplenism include splenomegaly, thrombocyto- 
penia, leukopenia, and anemia, and symptoms may 
include abdominal discomfort, pain, respiratory 
distress, and early satiety [87, 88]. Removal of func- 
tional splenic tissue may also improve hematologic 
abnormalities related to bone marrow suppression 
from systemic chemotherapeutic and immunosup- 
pressive agents so that optimal doses of such medi- 
cations can be maintained [88-90]. 

Total splenectomy may be an effective treatment 
for hypersplenism, but it impairs the body's ability 
to produce antibodies against encapsulated microor- 
ganisms and predisposes patient to sepsis [91]. Par- 
tial splenectomy has been proposed as a way to avoid 
this life-threatening complication [92, 93]. Despite 
surgery (total or partial splenectomy), the patient's 
condition for which the operation is performed may 
relapse, resulting in the need for a second opera- 
tion or additional transfusions [81, 92-96]. Other 
patients may not be considered surgical candidates 
because of their underlying medical condition and 

During the past three decades, splenic arterial 
embolization has been advocated in the nonopera- 
tive treatment of patients with these difficult clini- 
cal scenarios by intentionally infarcting splenic 
tissue and reducing its consumptive activity. In 
1973, Maddison [97] reported the initial clinical 
experience of splenic artery embolization, but severe 
complications that resulted from complete splenic 
infarction prevented acceptance of the technique as 
a viable treatment option. Since this initial descrip- 



Table 16.4. Results o: embdixiiei.ipv :or iiiigioiv.yoliponia 
Reference No. of % Complici 



% Recurrent % Repeat 


%Sn 


rj>ery 


Median, fol 


svmpioms or emboliZLidc 
hemorrhage 






(mos.) frai 


60 40 
D 

13 13 


20 





IS (0-29) 

24 (12-4?) 
24 (3-96) 


29 


20 





15 (1-204) 
28 (8-72) 

20 \1-S4i 


8 4 


27 




5 (0-60) 



Kess 



US [54] 
R[S5] 



Lee [27] 

Hamlin [56] 

Has [57] 14 

Soulem [58] 5 

Others [22,31.59-74] 25 

(few 



Totals 

Fioir.: N:-:i.:; 



a [i 3] : punted wirh pe: 



tion, many authors have advocated incomplete or 
partial splenic embolization (PSE), a method by 
which a portion of the splenic parenchyma is left 
viable, thus preserving the spleen's immunologic 
function and limiting complications while still 
managing the underlying condition [78-82, 87-90, 
98, 99]. PSE has also been advocated as a preopera- 
tive tool to improve the safety of and reduce intra- 
operative blood loss during open or laparoscopic 
splenectomy [100-103]. 



16.2.2 

Anatomic Considerations 

The splenic artery supplies the spleen 
substantial portions of the stomach and pa 
[104]. The splenic artery courses superior and ante- 
rior to the splenic vein along the superior edge of 
the pancreas. Near the splenic hilum, the artery 
usually divides into superior and inferior terminal 
branches, and each branch further divides into four 
to six segmental intrasplenic branches. The supe- 
rior terminal branches are usually longer than the 
inferior terminal branches and provide the major 
splenic arterial supply. A superior polar artery usu- 
ally arises from the distal splenic artery near the 
hilum, but it may originate from the superior ter- 
minal artery. The inferior polar artery usually pro- 
vides the left gastroepiploic artery, but it may arise 
from the distal splenic or inferior terminal artery. 
The left gastroepiploic artery then runs along the 
greater curvature of the stomach. Numerous short 
gastric branches arise from the terminal splenic or 
left gastroepiploic artery to supply the gastric cardia 
and fundus. In addition, the splenic artery has many 
branches to the body and tail of the pancreas; the 
largest of these are the pancreatic magna and dorsal 
pancreatic artery. When PSE is considered, knowl- 
edge and visualization o\ these arteries is essential 
to reduce the risk for nontarget embolization. 



16.2.3 

Technical Considerations 

PSE refers to partial obliteration of the peripheral 
intrasplenic vascular bed by injection of embolic 
material through the angiographic catheter placed 
within the splenic artery. This technique evolved 
as initial attempts to treat hypersplenism by proxi- 
mal splenic arterial occlusion proved unsuccessful. 
Response failure was attributed to the abundant 



collateral circulation from the short gastric and 
gastroepiploic arteries that reestablish the blood 
supply to the spleen around the occluded segment 
of the splenic artery [105-107]. Proximal arterial 
occlusion, although ineffective for management 
of hypersplenism, is a useful technique for reduc- 
ing intraoperative blood loss in thrombocytopenic 
patients undergoing splenectomy [108, 109]. 

Because of the need for more enduring and effec- 
tive hemodynamic and hematologic responses, total 
infarction of the spleen was performed with autolo- 
gous clots or particles in single or staged procedures 
[83, 97, 110, 111]. However, the widespread use of 
this technique was limited because of the severity 
and high incidence of complications such as splenic 
abscess, splenic rupture, septicemia, splenic vein 
thrombosis, and unremitting bronchopneumonia 
[83, 110-115]. Several mechanisms may cause com- 
plications after complete infarction of the spleen: 
induced immunocompromised state of the patient, 
predisposition of anaerobic bacterial growth within 
the hypoxic ischemic tissues, introduction of exog- 
enous bacteria through the angiographic catheter 
or embolic agents, and contamination of the devas- 
cularized splenic tissue with organisms originating 
within the intestines by retrograde portal blood flow 
[83,106,112, 116, 117]. 

Hemodynamic and hematologic responses and the 
severity of complications correlate with the amount 
of splenic tissue that is infarcted after embolization 
[110]. Therefore, PSE has been advocated to reduce 
complication rates after splenic infarction. Abla- 
tion of more than 80% of the splenic mass has been 
reported, but most authors have attempted to embo- 
lize60%-70% of the parenchyma [115, 118-120]. This 
amount of embolization allows for reduced seques- 
tration and destruction of the blood elements while 
maintaining the spleen's immunologic function and 
preserving antegrade flow within the splenic vein. 

Spigos et al. [112] adopted a strict protocol that 
resulted in a remarkably low number of complica- 
tions. The protocol included broad- spectrum anti- 
biotics started 8-12 hours before the procedure and 
continued for 1-2 weeks, local antibiotics (such 
as gentamicin) suspended in the solution used to 
deliver the particulate embolic agents and admin- 
istered through the angiographic catheter, strict 
attention to sterility (whole-body povidone-iodine 
bath or wide surgical scrub at the site of catheter 
insertion), selective catheterization with the cath- 
eter tip beyond pancreatic branches, effective pain 
control with narcotics or epidural anesthesia for 
48 hours (which prevents the splinting that may 



!-_ it.iL>-- "■ I. Mil e:apv :'.:■[■ Organ Ablution 



lead to pulmonary co mp lie at ions), and avoidance of 
overembolization. A Pneumovax vaccine was also 
administered before the procedure to help prevent 
pneumococcal infection. 

More recently, Harked et al. [99] evaluated the 
effect of PSE where only 30%-40% of the splenic mass 
was ablated and found that the reduced infarction 
volume still led to increased platelet counts, albeit 
the increase was less impressive in those patients 
than in patients in whom 70%-80% of the spleen 
was ablated. However, their technique also resulted 
in significantly lower morbidity. Therefore, a more 
conservative embolization procedure with lower 
complication rates may be prudent, and a second 
embolization can be performed if necessary. 

PSE can be performed by two methods. In one 
approach, a few distal branches of the splenic 
artery are selectively catheterized and embolized 
to complete stasis, and several other branches are 



left untreated (Fig. 16.3). Parenchymal phase of 
angiogram may be used to estimate the volume 
of remaining viable splenic tissue. Additional 
branches can be catheterized and embolized 
until the desired effect is achieved. In the second 
method, the working catheter tip is placed more 
proximally in the main splenic artery but beyond 
the origin of major pancreatic branches. Embolic 
particles are injected until the parenchymal blush 
is reduced nngiographicully to the desired amount. 
Contrast-enhanced CT scan may be used for follow 
up purposes (Fig. 16.4). 

In patients undergoing splenectomy, distal or 
proximal splenic arteries can be completely occluded 
to reduce the risk of intraoperative hemorrhage. 
After the embolization, if patients have thrombocy- 
topenia, a femoral arterial closure device may help 
promote hemostasis at the puncture site. Patients 
commonly experience fever, leukocytosis, and ele- 





Fig. 16.3a-c. A 47-year-old woman with locally advanced 
pancreatic carcinoma and thrombocytopenia, precluding 
fuithe: c:".cniC'th.er.iL , y, was interred for p;'.:u.;l splenic embo- 
lization. Pre-embokzittion I'.rtenogr.tm iaj shows normal 
splenic artery anatomy. The superior splenic artery (b) 
was selectively o.\ the ten zed and embolized using particles 
i range. .!00-?00 microns i. Posi-emboiizalion arteriogram :. c: 
shows complete occlusion of the s : ape:ior splenic artery. One 
month after emholizauon, her platelel count was greater than 
400,000/mm 3 



D. C. Madoff et aL 




Fig. 16.4a,b. A 4S-vei'.r-old man with pancreatic cancer pre- 
sented with persistent neutropenia and thrombocytopenia. 
Patient was referred for pai;ial splenic embolization (PSE i in 
nil ir.lempi io increase white b I ::■.:■ .". cell ami clatelei coiinis 
prior to additional chemotherapy. Pre-embolization axial 
'".'.' image or I :". e- ahdomr:'. iai - : .'.ows splenomegaly. i."T sea:; 
performed 4mon:hs arler PSE !l>j shows massive necrosis of 
splenic parenchyma. VVi:hin _ week:, of partial splenic embo- 
I. ;:.:i..: i"i. i.'.e pkie.ei c- '.'.;":; ;:■ a maa/.ed i.v l ' , , l 'i , '»''/iaia : : 



vated serum amylase levels, but these complications 
are transient. 

The embolic agents most commonly used for 
splenic ablation are Gelfoam pledgets and poly- 
vinyl alcohol particles. Yoshioka et al. [105] also 
showed excellent increases in platelet counts when 
coils were placed within the intrasplenic branches, 
and Hickman et al. [108] successfully used Gel- 
foam, polyvinyl alcohol particles, and coils alone 
or in combination for preoperative embolization. In 
animal studies, additional materials that have been 
used for this purpose are absolute ethanol [121, 122], 
microfibrillar collagen [123], tissue adhesives [124], 
balloon catheters [125], silicone particles [126], and 



others [127], but none of these agents have been 
widely accepted for use in humans. 

The techniques developed and used at different 
centers vary greatly, but some general recommen- 
dations can be made here. Pre-procedure antibi- 
otics are highly recommended. Povidone-iodine 
baths are not commonly used, but careful attention 
to sterile preparation is warranted. Embolization 
can be performed through the diagnostic catheter 
placed in the splenic artery only if the catheter tip 
can be advanced beyond the origin of major pan- 
creatic branches; a coaxial system with the use of a 
micro catheter often facilitates catheterization of the 
appropriate branches. Adequate pain control after 
the procedure with the use of patient-controlled 
analgesic pumps is warranted to minimize the sever- 
ity and possibly duration of the post-embolization 
syndrome and to decrease the risk of pulmonary 
complications (such as atelectases and pneumonia). 

Similar to complete splenic arterial emboliza- 
tion, PSE is prone to complications and adverse 
effects, but PSE is much better tolerated than com- 
plete splenic ablation. In addition to those men- 
tioned above, patients might develop pancreatitis 
(likely a result of nontarget embolization of dorsal 
pancreatic and pancreatic magna arteries), pleural 
effusions requiring thoracentesis, paralytic ileus, or 
the post-embolization syndrome consisting of fever, 
leukocytosis, and abdominal pain [1 1 (| ], 

After PSE, the spleen retains its ability to regener- 
ate. Kumpe et al. [120] found substantial regeneration 
of splenic tissue in 9 of 11 patients despite 70%-80% 
embolization, as visualized on Tm-99m sulfur colloid 
liver-spleen scans performed 4 to 16 months later. In 
contrast, Watanabe et al. [128] reported that PSE of 
80% or more resulted in initial increases in spleen 
size followed by substantial and stable reductions by 
4 months. Despite these conflicting results, a repeat 
PSE procedure can be performed with similar effec- 
s should symptoms recur [98]. 



16.2.4 
Results 

Few studies have compared splenectomy with PSE 
in a randomized, prospective fashion. In a series 
by Mozes et al. [118], 53 patients who were to later 
undergo renal transplant with aznthioprine immu- 
nosuppressive therapy (which causes leukopenia 
and thrombocytopenia) were randomly assigned to 
a splenectomy ai'onp (25 patients) or a PSE group (28 
patients). For patients in the PSE group, a mean of 



Embolotherapy for Organ Ablat 



65.4% ± 16.6% of the splenic mass was ablated. The 
early postoperative morbidity rate and the duration 
of hospital stay were similar in the two study groups. 
Abscess and splenic rupture were not encountered. 
The two cases of severe pancreatitis resulting in 
death occurred in the splenectomy group, and one 
death from pneumococcal pneumonia occurred 
3 months after PSE. Equivalent numbers of renal 
transplantation were carried out in both groups 
and resulted in similar long-term (2.5-4.0 years) 
graft survival (60% vs. 66%) and long-term patient 
mortality. However, splenic regeneration occurred 
in most patients after PSE, and doubling of splenic 
parenchyma was seen in 40% of them. As demon- 
strated in this study, PSE may be an effective alterna- 
tive to splenectomy, especially for patients who are 
not suitable surgical candidates. 

Numerous retrospective studies have demon- 
strated that PSE is an effective short-term therapeu- 
tic alternative to splenectomy for a wide spectrum 
of patients with hypersplenism [77,78,80,82,88-90]. 
Kimura et al. [80] reported the results of initial and 
repeated PSE in patients with chronic idiopathic 
thrombocytopenic purpura. Thirty-nine patients 
underwent initial PSE, and 12 underwent a repeat 
PSE. The therapeutic effects of initial and repeat PSE 
were classified as complete response if the patient's 
platelet count rose to more than 100,000/mm 3 with- 
out steroids 1 year after the initial or repeat embo- 
lization, as partial response if the platelet count 
increased by 50,000-100,000 under similar circum- 
stances, or as no response. Twenty patients (51%) 
responded to the initial PSE (11 complete response, 
9 partial response) with signilieantlv higher peak 
platelet response (p = 0.029). Differences between 
responders and nonresponders in terms of age, sex, 
lowest platelet count, and steroid response before 
PSE were nonsignificant. Of the 11 patients with 
complete response (median follow-up, 58 months; 
range 21-156 months), one relapsed after 32 months 
and underwent repeat PSE. Of the 9 patients with 
partial response, four maintained a platelet count 
of more than 50,000/mm 3 without relapse during a 
median follow-up period of 73 months (range, 14 to 
42 months), and five relapsed after a median follow- 
up period of 34 months (range, 15-123 months). 
Repeat PSE resulted in four partial responses and 
one no response. However, in 6 of 19 patients who 
had no response to the initial PSE (median follow-up, 
8 months; range, 3- 22 months), repeat PSE elicited 
only one partial response. The 51% remission rate 
was maintained by means of repeat PSE for a median 
follow-up of 76 months (range, 14-147 months) after 



the initial PSE. Kimura et al. concluded that PSE 
combined with repeat embolization may be an 
effective alternative to splenectomy in patients with 
chronic idiopathic thrombocytopenic purpura. 

Palsson et al. [129] reviewed 26 severely ill 
patients (median age, 63.5 years) who were treated 
with PSE a total of 52 times, mainly because of 
bleeding esophageal varices and thrombocytope- 
nia. The mean hemoglobin values, leukocyte counts, 
and platelet counts increased significantly after 
PSE, and the frequency of bleeding episodes from 
esophageal varices was statistically significantly 
reduced. As defined by the authors, the integrated 
PSE effect was judged as improved in 19 patients, 
unchanged in five, and worse in two. Median sur- 
vival time was 50.5 months (range, 0.5-272 months). 
Complications consisted mainly of fever, atelectasis, 
and abdominal pain, although two patients died of 
PSE-related complications. Thus, a standardized 
and graded PSE is reasonably safe even in patients 
with advanced disease in whom it is hazardous to 
perform splenectomy. Palsson et al. concluded that 
PSE maintains long-term effects on hematologic 
parameters, esophageal variceal hemorrhage, and 
may substantially improve patients' diniatl statu',. 

Nio et al. [130] published a retrospective study 
in which 41 PSE procedures were performed in 36 
children with liver disease and thrombocytopenia 
resulting from hypersplenism. The average volume 
embolized was 70.1%, and the mean follow-up was 
71 months (range, 20 days-182 months). Eleven 
patients (30.6%) had recurring thrombocytopenia 
(defined as fewer than 100,000/mm 3 ). There were no 
significant differences in the volume embolized or 
platelet count before PSE between the patients with 
and without recurring thrombocytopenia. However, 
the peak platelet counts after PSE was significantly 
lower in the patients with recurring thrombocy- 
topenia (p = 0.0091). In 17 of 24 survivors who did 
not undergo liver transplantation, platelet counts 
remained normal throughout the follow-up period. 
Hematologic indices improved in all 36 patients 
after PSE, and its long-term efficacy was shown in 
70% of the: 



16.2.5 

Future Development and Research 

External beam radiation therapy is an alternative 
treatment for patients with splenomegaly [131-135]. 
Similarly, transcatheter arterial brachytherapy 
using yttrium-90 microspheres has been reported 



D. C. Madoff et al. 



[136, 137]. At the time of this writing, the cost asso- 
ciated with intraarterial brachytherapy and the 
complexity of the delivery system discourage its 
widespread use. When intraarterial bra chy therapy 
becomes more widely available, further investiga- 
tion of its usefulness in patients with hypersplen- 
ism may be warranted. Thermal ablation therapy, 
which has received considerable attention over the 
last decade, has been used to treat patients with 
hepatic malignancies, small renal cell carcinoma, 
and lung tumors [138, 139]. More recently, radiofre- 
quency ablation has been proposed as an alternative 
treatment for hypersplenism [140]; the safety and 
usefulness of this approach remains to be carefully 
evaluated in clinical setting. 



16.3 
Conclusion 

Embolotherapy for organ ablation, as highlighted 
in this chapter for treatment of patients with renal 
tumors and hypersplenism, was developed in the 1970s 
and enjoyed a period of intense activity over the next 
two decades. During that period, numerous articles 
describing single-center experiences with small series 
of patients appeared in the medical literature. Retro- 
spective studies of small series of cases demonstrated 
the safety and potential benefits of embolotherapy for 
organ ablation in selected patients. During the same 
period, technological advancements have elevated the 
fields of angiography, vascular catheterization, and 



Table 16.5. Catheters and cmbokc agents used for organ 
Renal Artery Embolization 

Abdominal aortogram 

Pigtail catheter, 5 Fr Cook, Bloomingtc 

Selective renal arteriogram 

C2 Cobra Visceral, 5 Fr Cook, Bloomingtc 

Sos Omni Selective (2), 5 Fr Angiodyn amies, C 

Occlusion balloon catheter, 5 Fr 
Subselective renal arteriogram 

Renegade HI-FLO, 3 Fr 
Embolic agents 

Dehydrated alcohol 

EmboGold microspheres 

Contour.SE Microspheres 

Absorbable Gelatin Sponge 



Splenic artery embolization 

Selective splenic arteriogram 
C2 Cobra Visceral, 5 Fr 
Sos Omni Selective (2], 5 Fr 

Subselective splenic arteriogram 

Renegade HI-FLO, 3 Fr 
Embolic agents 

Absorbable Gelatin Sponge 
EmboGold Microspheres 
Contour.SE Microspheres 



Boston Scientific Medi-Tech, Natick, MA 

American Regent, Shirley, NY 
Biosphere Medical, Rockland, MA 
Boston Scientifrc, Natick, MA 
Ethicon, Somerville, NJ 



Cool., R!oomiag:on. ;i^ 
Angiodyn amies, Queensbury, NY 

Boston Scientific Medi-Tech, Natick, MA 

Ethicon, Somerville, NJ 
Biosphere Medical, Rockland, MA 

Boston Scientific, Natick, MA 



Fig. 16.5. A simp.e and r:se:iil algo- 
rithm for planning embolization of 
renal tumors. Interventional radiolo- 
gist should be familiar with various 
embokc .:genls including eikriio. and 
solid particles and different methods 
for delivery of these agents. Neverthe- 
less, die algorithm may be modified to 
suite individual expertise and available 
embolic agents 



Embolotherapy for Organ Ablat 



embolotherapy to new heights. Table 16.5 provides a 
short list of supplies currently used for embolotherapy 
at our institution. A simple algorithm for embolization 
of renal tumors is also suggested (Fig. 16.5). In this era 
of evidence-based medicine, advanced embolotherapy 
techniques warrant re evaluation and validation in the 
management of renal tumors and hypersplenism. 



Greenlee RT, Murray T, Bolder) S.VY'ingo PA (2000) Cancer 
Statistics, 2000. CA Cancer ] Clin 50:7-33 
Linehan WM.Zbar B, Bates SE, Zelefsky M), Yang |C (2001 ) 
Cancer of the kidney and ureter. In: DeVita VTJ, Hellman 
S, Bosenoerg SA ieols! Oincer principles and practice of 
oncology, l.ippincol: Williams and VVi!l-:iiis, Philadelphia, 
PA, Dp 1362-1396 

Pantuck AJ, Zisman A, Belldegrun AS (2001 ) The changing 
n.iv.i :.! . .- : :'. ry of :enal eel! carcinoma. I Urol ::■<:' :1 ol 1 -!.• 
Lowry PS, Nakada SY (2003) Renal cryotherapy: 2003 
clinical status. Curr Opin Urol 13:193-197 
Gervais DA, McGovern FJ, Arellano RS, McDougal WS, 
Mueller PR (2003) Renal cell carcinoma: clinical experi- 
ence and technical success with, radio-frequency iibl.iti-.jn 
of 42 tumors. Radiology 226:417-424 
FlaniganRC,MickischG,SylvesterR,TangenC,vanPoppel 
H, Crawford ED (2004) Cytoreductive nephrectomy in 
pai.enis wiili meta static renal cancer: a combine;", analy- 
sis. J Urol 171:1071-1076 

Bakal CW, Cynamon J, Lakritz PS, Sprayregen S (1993) 
Value •:■: preoperative renal .irtery embolization in reduc- 
ing o.ood transfusion, requirement daring iiepiiic.rloniy 
for renal cell carcinoma. J Vase Interv Radiol 4:727-731 
Klimberg I, Hunter P, Hawkins IF, Drylie DM, Wajsman Z 
i I ^5: Preoperative angioinfarotion o: .ocalizecl renal cell 
carcinoma using absolute ethanol. ] Urol 133:21-24 
Craven WM, Redmond PL, Kumpe DA, Durham JD, Wet- 
tlauret INN ;,i, l .: VI a lined ;i clave.: nephrectomy .ifie: elha- 
nol embolization of renal carcinoma. J Urol 146:704-703 
Jacobs JA, Ring El, Wein AJ (1981) New indications for 
renal infarction. I L'i'o] 125:243-24? 
Nurmi M, Satokari K, Puntala P (1987) Renal artery embo- 
lization in die palliative treatmem o: renal aoenocarci- 
noma. Scand I Urol Nephrol 21:93-96 
Zielinski H, Szmigielski S, Petrovich Z (2000) Comparison 
■ ■:' pi eop era live emoo.izauon followed by radical nepbrec- 
lomv willi radical nephrectomy alone for renal cell carci- 
noma. Am J Clin Oncol 23:6-12 

Nek. in Cl-i Sand a MG '2002 : Comemporarv diagnosis and 
m a :".a Yemeni of rer.al a iigiomvo lipoma. I Urol 165:: 31 5- 
1325 

Oesterling IE, Fishman EK, Goldman SM, Marshall FF 
i I : ' : S'.<) The m a r.a gnne nl of renal angionivo.ipoma. I Urol 
135:1121-1124 

Dickinson M, Ruckle H, Beaghler M, Hadley HR (1998) 
Rer.al angiomvo.iponia: opiimal Ireatnieiit based on size 
and symptoms. CHn Nephrol 49:281 -286 
Kladir S : i ;, S:- : I ! ia g no: si .c angiography [ si ed::. Saunders. 
Philadelphia, PA, pp 445-495 



Boiisen E (1997) Renal agniography. In: Baum S (ed) 

A or:", ill's angiography, -"ill r.." :i. Little Brown. New York. NY, 
pp 1101-1131 

Kadir S ! : 991 ) Atlas o: normal and variant anci. ■cr.iraic 
anatomy, I si ecu. Saunders, Philadelphia, PA, pp 3S7-- f2 ; 
Abrams HL, Grassi CJ (1997) Renal tumo versus renal 
cyst. In: Baum 5. I ed ) Abram's angiography. 4tli edn. Little 
Brown, New York, NY, pp 1 1 32-1 1 77 
Kalman D.Varenhorst E (1999) The role of arterial embo- 
lizaiion in renal cell carcinoma. Scand 1 Urol Nephrol 
33:162-170 

Ellman BA, Parkhill BJ, Curry TS 3rd, Marcus PB, Peters 
PC (1981 1 Ablation of renal minors with, absolute ethanol: 
a new technique. Radiology 141:619-626 
Rabe FE, Yune HY, Richmond BD, Klatte EC (1982) Renal 
minor infarction wall ao-solute edianol. Alk Am ! Boem- 
genol 139:1139-1144 

Cox GG, Lee KR, Price HI, Gunter K, Noble MJ, Mebust WK 
:. 1 9B2 ) Colonic infarction fol. owing etlianol enioolizaiion 
of renal-cell carcinoma. Radiology l 45:343-345 
Lammer J, Justich E, Schreyer H, Pettek R (1985) Compli- 
cations of renal minor embolization. Card i ova sc Imerveni 
Radiol 8:31-35 

Mulligan BD, Espinosa GA (1983) Bowel infarction: com- 
plication of ethanol ablation of a renal mm or. Card i ova so 
Intervent Radiol 6:55-57 

Teertstra H], Winter WA, Frensdorf EL (1984) Ethanol 
embolization of a renal tumor, complicated by colonic 
infarction. Diagn Imaging Clin Med 53:250-254 
Lee W, Kim TS, Chung )W, Han JK, Kim SH, Park JH (1998] 
Rnia] angionivolipoma: e mb o loth era pv with a mixture of 
alcohol and iodized oil. I Vase Interv Radiol 9:255-261 
Konya A, Van Pelt CS, Wright KC (2004) Ethiodized oil- 
el hail':'! capillary embolization .11 rabbi: kidneys: temporal 
histopathologic findings. Radiology 232:147-153 
Wallace S, Charnsangavej C, Carrasco CH, Richli WR, 
Swanson li M333: Therapeuiic angiographic tec.miqaes, 
renal tumors: clinical resuhs. In: lionclelinger R1-. Rossi 
P, Kurdziel ]C, Wallace S ieos! Interventional radiology. 
Thieme Medical, New York, NY, pp 468-477 

30. Ray CE, Waltman AC (2832 i General principles of emboli- 
za:ion and chemoembo;iza:ion. In: Bahal CW, Siloerzv. : -iL T 
IB, Cynamon I, Sprayregen S : eds'i Vascular and imerven- 
iional radiology principles and practice. Trneiue. New 
York, NY, pp 39-100 

31. Zerhouni EA, Schellhammer P, Schaefer ]C, Drucker )R, 
laife AH, Gonzales IB el a I. ! : t; B-*'i Man age mem ^ bleed - 
nig renal aiigioiuvolioomas by t ra n so a tiieter embolization 
following CT diagnosis. Urol Radiol 6:205-209 

32. Kurth KH, Cinqualbre J, Oliver RT, Schulman CC (1984) 
1- mb-.ilizaiion and s::osequer.: neniireciomy in metas:a:ic 
renal cell carcinoma. Prog Clin Biol Res 153:423-436 

33. Mclvor J, KaisaryAV, Williams G, Grant RW (1984) Tumour 
imaiction af:er pre-ooerative embolisalion of renal carci- 
noma. Clin Radiol 35:59-64 

34. WeigelJW,Mebu5tWK,PoretJD,NobleMI,VotapkaT,Krish- 
nanECetal. (1985) Treatment of renal cell carcinoma with 
renal infarction, delayed nephrectomy, medroxvoiogirsiei- 
one, and xenogeneic immune BNA. Urology" 25:133-135 

35. Kaisary AV, Williams G, Riddle PR (1984) The role of 
preoperative embolization in renal cell carcinoma, j Urol 
131:641-646 

36. Nakano H, Nihira H, Toge T (1933) Treatment of renal 



i.'.ncef paiienls ny lianscaiheter rinboliz.il ion and iis efrecls 
□11 lymphocyte prokferaiive it spouses. [ Urol 130:24-27 
Kurth KH, Debruyne FM, Hall RR, Denis L, Verbaes A, 
Bollack C et al. (1987) Embolization and postinfarction 
nephrotomy in patients with primary metastatic ren.u 
adenocarcinoma. Eur Urol 13:251-255 
Lei none n A :. ] ,: 8?i Embolization of renal carcinoma. Com- 
parison between :iir early results of Gel foam jii i ab solute 
ethanol embolization. Ann Clin Res 17:299-305 
Bono AV, Caresano A (1983) The role of embolization in 
the treatment of kidney carcinoma. Eur Urol 9:334-337 
Ekelund L, Ek A, Forsberg L, Haukaas 5, Heniakson H. 
KallandT et al. (1984) Occlusion of renal arterial tumor 
snpplv '■villi .ih ,; ok;> ririanol. experience ividi 20 cases. 
Acta Radiol Diagn (Stockh) 25:195-201 
Gottesman JE, Crawford ED, Grossman HB, Scardino P, 
McCracken I!- (I98sj hifaiciioii-nephreciomy for meta- 
static :e:i.il carcinoma, l-ouihwes: oncology group study. 
Urology 25:248-250 

':liris[eiisenK,DyreborgU,Andersen)F,NissenHM(1985) 
"Hie viiitie of iransvascinar einbclizanon in I lie tie;'. I merit 
of renal carcinoma. J Urol 133*3* 193 
LaniganD, JurriaansE, H;--:inds|C, WelLi ir.Ch.ia RG 
; ! ™2 ; The current statui <'. emh:]|i/alii;n ir. renal cell 
ca:cinoma-a survey of I'C.il ;\:\ n^lnnal practice i'.'.ta 
Radiol 46:176-178 

Park JH, Kim SH, Han ;K, Chung |W, Man MC (1994) 
Transc.itheter .naerial enf)i>h/aii'N <.:' i;rroi'..'.il\t: ror.al 
cell carcinoma with a module wl eLiunoi and il-uiZcU uil. 
Cardiovasc Intervent Radiol 17:323-327 
Almgard LE, Slezak P (1977) Treatment of renal adenocar- 
cinoma by emboli zillion: ;: follow- up o: 38 oases. Eur Urol 
3:279-281 

Kato T, Sato K, Abe R, Moriyama M (1989) The role of 
emholizaLion/chemeembohzaiion :n the treatment of 
rena. cell carcinoma. In: A. an K led ) Telinipeutio progress 
in urological cancers. Liss, New York, pp 697-705 
Wallace S, Chuang VP, Swanson D, Bracken B, Hersh EM, 
Ayala A et al (1981) Embolization of renal carcinoma. 
Radiology 138:563-570 

Mobilio G, Cavalli A, Bianchi G (1981) Preoperative arte- 
rial occlusion in renal minors: a years experience. In; Urol 
Nephrol 13:25-33 

r'rasson K Kigazzola L, Bianchi G. i-ranzohn N. Caresano 
Ajdel Faveio C el a I. i 1 97S! Selective arie: ial embolization 
in renal tumors. Radiol Clin (Basel) 47:239-251 
Frasson F, Roversi RA, Simonetti G, Ziviello M (1981) 
embolization of rena! tumors. A survey of die [Lilian 
experience : 282 patients. Ann Radiol (Paris) 24:396-399 
Fischedick AR, Peters PE, Kleinhans G, Pfeifer E (1987) 
f'leopeialive renal mm or embolization. A userul proce- 
dure? Acta Radiol 28:303-306 

Mebust WK, Weigel IW, Lee KR.Cox GG, Jewel] WR, Krish- 
nan EC (1984) Renal cell carcinoma-angioinfarction. J 
Urol 131:231-235 

Giuliani L.Carmienani G. he-grnno E, Puppo f 1 . Quaitrini 5 
l ! a S! ; usefulness of preoperative liansciiiheie: emboliza- 
tion in kidney minors. Urology 17:431-434 
Mourikis ','-. ChatziiOLinnou A, Antoniou A, Kehagias '.-. 
Cikas D, Vlahos 1 ■: ! 339: Selective arterial emno.izauon in 
ihe m an age mem of sympiomatio renal aiig.omyol.pomas. 
Eur] Radiol 32:153-159 
Kessier ul, Gillon G. Neuman M, Engelsiein '.'-. Winkler 



H. km; el 1 i 1 998) Management or rena. angiomyonpoina: 
analysis of 15 cases. Eur Urol 33:572-575 

6. Hamlin JA, Smith DC, Taylor EC, McKinney JM, Ruckle 
HC, Hadley HR ( 1 997) Renal angiomyolipomas: long-term 
fo. low-up of embolization for acme hemorrhage. Can 
Assoc Radiol J 48:191-198 

7. Han YM, Kim JK, Roh BS, Song HY, Lee JM, Lee YH et al. 
i I 997) Renal angiomyolipoma: selective arieiaal embol.za- 
non-effecl iveness una changes in a neio myogenic compo- 
nents in long-term follow-up. Radiology 204:65-70 
Soulen MC, Faykus MH Jr, Shlansky-Goldberg RD, Wein 
Al, Cope C i I 994! Elective embo.izalion for prevention -.A 
hemorrhage f:o:i: renal angiemyclipomas. I Vase iiuerv 
Radiol 5:587-591 

Ciancio S[, Vira M, Simon MA, Lerner SP, Schulam PG 
(2001) Giant bilateral renal aiigiemyolipomas associated 
with tuberous sclerosis. Urology 57:554 
kiibalius C-A, Siablis P. Uatsikos EN, Ya: men it is 8, Karn- 
abatidis P. Dimopoulos 1 (1998) Renal angiomyolipoma 
with hueme-i rhuge treated by urgent embol.zatinn. Scniid 
J Urol Nephrol 32:54-55 

Henslee D, Ross G Jr, Beale G (1991) Bilateral angiomyo- 
lipomas: ::ir benefits and limitation of embolization for 
renal salvage. A case report. Mo Med 88:292-294 
Jonsson E, Sueoka BL, Spiegel PK, Richardson JR Jr, Heaney 
]A i"39! ; Angiographic ir.anagensenl of relropei itnneal 
hemorrhage- i'ja. -Hi ivna. iingiomyoliaoiiia in polycystic 
kidney disease.] Urol 145:1248-1250 
Ou YC,Wu HC, Yang CR, Chang CL, Hwang TI, Chang CH 
i I 991 ) Rena! angiomyolipoma: experience of 2? patients. 
Zhonghua Yi Xue Za Zhi (Taipei) 48:217-223 
Edelman MA, Mitty HA, Dan SJ, Birns DR (1990) Angio- 
myol. plana: posiem.bojzatiei: liquefaction and percuta- 
neous drainage. Urol Radiol 12:145-147 
Van Baal JG, Lips P, Luth W, Bakker P, Davis G, Karthaus 
P et al. (1990) Percutaneous transc.itheter embolization 
of symptomatic renal angiomyolipomas: a report of four 
cases. Nether J Surg 42:72-77 

Uchino A, Itoh K.Egashira K.Ohno M (1987) Therapeutic 
embolization foi renal angiomvoupoina: case report and 
review of the literature. Radial Med 5:191-194 
Earthraan WJ, Mazer MJ, Winfield AC (1986) Angiomyoli- 
pomas in tuberous sclerosis: s an selective en: no- oilier a pv 
ivith alcohol, with long-ierm follow-up study. Kaoiohigv 
160:437-441 

Sanchez FW.Vujic I, Ayres RI, Curry NS, Gobien RP (1985) 
Hna.o: rhagic renal angio in yo lipoma: superseded :ve renal 
aiieiial embolization for pieseiv.ition o: renal function. 
Cardiovasc interveni Radio! 8:39-42 
Aii ler I, '..".rewelcmge: ", i.iiz.^v G ( l ''84! "Macro" aneurysm 
in renal angiomyolipoma: two cases, with iheiape.nic 
embolization in one patient. Urol Radiol 6:201-203 
Rosen R), Schlossberg P, Roven SJ, Romberg M (1984) 
■Vlaiiaaemem of symptomatic renal angiomyolipomas bv 
embolization. Urol Radiol 6:196-200 
Lingeman JE, Donohue JP, Madura JA, Seiko F (' 1 982 ) Angi- 
omyolipoma: emeiging ooncepis in man a gem em. Urolegv 
20:566-570 

Hagley[i,AppellR,PingoudE,McGuireEJ (1980) Renal angi- 
omyolipoma: diagnosis a::.-, manaeement. Urology 15:1-5 
Moorhead if, Fritzsche P, Hadley HL [1977! Managemenl 
o: hemorrhage secondary to renal angiemvolipoma wiih 
selective arterial embolization. J Urol 117:122-123 



!-_ it.iL>-- "■ I. Mil erapv :'.:■[■ Organ Ablution 



, EasonAA,C:itt-:MicaEV,\k.L-r.!tbTW! 1979) Massive renal 
angiomvolinoma: preoperative infarction by baboon cath- 
eter.J Urol 121:360-361 

i. Peck-Radosavljevic M (2001) Hypersplenism. Eur T Gas- 
troenterol Hepatol 13:317-323 

-. Sangro h. Bilbao i. Herrero i. Core-]. 1 . C. Longo I, Reloqiji 
et a I. i 1993) Parti a! sp.en.ic embolization tor the i real men I 
of Hypersplenism in cirrhosis. Hepatology 1 8:309-31 4 

'. Romano \-l, Gioielli A, Capuano G, Pompom P, Salvatore 
M i2ilil4! Partial splenic embolization in patients wiih 
idiopathic portal hypertension. Eur I Radiol 49:268-273 

:. Stanley P, Shen TC (1995) Partial embolization of the 
spleen in patients with lhalassemia. I Vase Inieiv Radiol 
6:137-142 

i. Kimura F, Ito H, Shimizu H, Togawa A, Otsuka M, 
Yoshidome H et al. (2003) Partial splenic embolization 
for the treatment of hereditary spherocytosis. AJR Am J 
Roentgenol 181:1021-1024 

i. Kimura F, Itoh H, Ambiru S, Shimizu H, Togawa A, 
Yosliioieme H et al. i2f)i)2 I Long-term results o: initial and 
repeated parti.;, splenic embolization for the treatment of 
chronic idiopathic iliroiiibocyiopemc purpura. AIR At. 1 
Roentgenol 179:1323-1326 

. Kumar S, Diehn FE, Gertz MA, Teiferi A (2002) Splenec- 
tomy for immune Lhrombocytopen.ic purpara: long-term 
results .md trealmenl of pestsplenectomy relapses. Ann 
Hematol 81:312-319 

. Miyazaki M, Itoh H.Kaiho T.Ohtawa S, Ambiru S, Hayashi 
S et al. (1994) Partial splenic embolization for the treat- 
ment of chronic idiopathic ihrombocytopenic purpura. 
AJR Am I Roentgenol 163:123-126 

-. Wholey MH, Chamorro HA, Rao G, Chapman W (1978) 
Spleii.c i iita r. "i..:- ii ju.: spontaneous rupture ot die ,; plreii 
after therapeutic embolizalion. Cardiovaso Radiol ". :249- 
253 

, Athale UH, Kaste SC, Bodner SM, Ribeiro RC (2000) 
Splenic rupture in children with hematologic malignan- 
cies. Cancer 88:480-490 

:. Gardner RV, Warrier RP, Loe W, Ward K, Haymon M, 
Graver R i2.fi O: Splenic artery embolization as emergen, "v 
treiitment of splenic rupture in a child with T-cell acute 
lyinpiiocytic .ei:..e:i\.a having t'S:;14: iians.ec.Uioii. Med 
Pediatr Oncol 41:492-493 

Bird MA, Amiadi D, Behrns KE (2002) Primary splenic 
lymphoma complicated by hematemesis and gastric ero- 
sion. South Med J 95:941 -942 

Pringle KC, Spigos DG, Tan WS, Politis C, Pang EJ, Reyez 
HM et al. (1982) Partial splenic embolization in the man- 
agement of thalassemia, maior. [ Pediatr Surg 17:884-891 
Jonasson O, Spigos DG, Mozes MF (1985) Partial splenic 
embolization: exper.eiace .n la:- patients. World I Surg 
9:461-467 

Lokich [. Costelle- V i 1 S2j Splenic emboli?.-. I. on io- pi event 
dose limitation of cancer chemotherapy. AiR Am | Roent- 
genol 140:159-161 

Gerlock AJ Jr, MacDonell RC Jr. Muhletaler CA, Parris WC, 
Johnson HK, Tallent MB et al. (1982) Partial splenic embo- 
l.i'.-i. ii i.-r hypei sp.eo.ism in renal tian-plai'.tation. AIK 
Am J Roentgenol 133:451-456 

ZarrabiMH, Rosner F (1984) Serious infections in adults 
following spleneciomy for trauma. Arch Intern Med 
144:1421-1424 
Rice HE, Oldham KT, Hillery CA, Skinner MA, O'Hara 



SM, Ware KE io003; Clinical and hem .'.to logic benefils of 
panial spleneciomy for congenital hemolytic .'.nemias in 
children. Ann Surg 237:281-288 

93. De Buys Roessingh AS, de Lagausie P, Rohrlich P. Berrebi 
1-. Aigrain Y (2002) rol.c-w-up oi partial splenectomy in 
children with hereditary spherocytosis. 1 Peelialr Surg 
37:1459-1463 

94. Schwartz J, Leber MD, Gillis S, Giunta A, Eldor A, Bussel JB 
iLOC-.i ) Long term fol.ow-up a tier sp.enectomv performeo 
for immune thrombocytopenic purpura {ITP|. Am ] 
Hematol 72:94-98 

95. Kahn MJ, McCrae KR (2004) Splenectomy in immune 
th r-.j m bocy lopen ic purpura: recent controversies :ind 
long-term outcomes. Curr Hematol Rep 3:317-323 

96. McMillan R, Durette C (2004) Long-term outcomes in 
adults with chronic ITP .'.fier splenectomy fiilure. Blood 
104:956-960 

97. Maddison FE (1973) Embolic therapy of hypersplenism 
(abstract). Invest Radiol 8:280 

98. Brandt CT, Rothbarth LJ, Kumpe D, Karrer FM, Lilly JR 
!1989i Splenic emnelizatioi 1 . in children: leng-ierm effi- 
cacy. J Pediatr Surg 24:642-644; discussion 644-645 

99. Harned RK 2nd, Thompson HR, Kumpe DA, Narkewicz 
MR, Sokol RJ (1998) Partial splenic embolization in five 
children vo.lr. hypersplenism: effect-; -of reduced -volume 
em bo! i ; :tl ion on efficacy and morbidity. l\adio!ogv 
209:803-806 

lOO.Hiatt IR, Gomes AS, Machleder HI (1990) Massive sple- 
nomegiily. Superior results wiih a com fined ei;dovas..-- L ilai- 
;'.nd operative approach. Arch Surg 125:1363-1367 

lOl.Poulin EC, Mamazza I, Schlachta CM (1998) Splenic artery 
emb' ■! 1 7 ::l. -on before .aparoscop.c splenectomy. An update. 
SurgEndosc 12:870-875 

102.Totte E, Van Hee R, Kloeck I, Hendrickx L, Zachee P.Bracke 
P et al. (1998) Lap arose o-p;c splenectomy after arterial 
emboli sat ion. Hepaiogastroenlerology 45:773-776 

103.IwaseK,HigakiJ,MikataS, Tanaka Y, Yoshikawa M.Hori S 
et al. ! 1 '''99! Laparoscopic ally assisted spleneciomv to I low- 
in s; preoperative splenic arlerv embolization using con- 
tour emboli for myelofibrosis wiih massive spienoniega.v. 
Surg Laparosc Endosc Percutan Tech 9:197-202 

104.Kadir S, Lundell C, Saeed M (1991) Celiac, superior and 
inferior mesenteric arteries. In: Kadir S (ed) Atalas of 
normal and variant angiographic anatomy. Saunders, 
Philadelphia, PA, pp 204-237 

105.Yoshioka H, Kuroda C, Hori S, Tokunaga K, Tanaka T, 
Nak rim lira H et a I. ! 19S?) Splenic emPo.ization for hyper- 
splenism using sieel coils. AIR Am I Roentgenol i 44:1269- 
1274 

106.Anderson JH, VuBan A, Wallace S, Hester JP, Burke JS 
! 1 L1 77 i Transcatheter splenic arterial occlusion: an experi- 
mental study in dogs. Radiology 125:95-102 

107.Thanopoulos BD, Frimas CA (1982) Partial splenic embo- 
lization in the mrinagemenl ot hypersplenism secondary 
to Gaucher disease. J Pediatr 101:740-743 

108. Hickman MP, Lucas D, Novak Z, Rao B, Gold RE, Parvey 
L et al. (1992! Preoperative embolization of lite spleen in 
children with hypersplenism. J Vase Interv Radiol 3:647- 
652 

109.Levy JM, Wasserman P, Pitha N 
transcatheter occlusion of the sp 
114:198-199 

llO.Castaneda-Zuniga WfR, Har 



1979) Presplenectomy 

enic artery. Arch Surg 



schmidt DE, Sanchez R, 



Amplatz K (1977) Nonsurgical splenectomy. A]R Am ] 
Roentgenol 129:805-811 

lll.Vujic I, Lauver JW (198". ; Severe complications from par- 
tial splenic embolizauon in patients with liver failure. Br I 
Radiol 54:492-495 

112.Spigos DG, JonassonO, MozesM, CapekV (1979) Partial 
splenic embolization :n :he ireaimenl -.:■]" hypersplenism. 
AJR Am ] Roentgenol 1 32:777-782 

m.Witte CL, Ovitt TW, van Wyck DB, Witte MH, O'Mara RE, 
Woo [fend en IM (1976) Ischemic therapy in thrombocyto- 
pen.a from hypersplenism. Arch Sr.rg I I I :l : : ?-! : _l 

1 14.Ahvnii.irk A. fieiigmaik S, Gal 1st rand l- : . loelsson B, Lunder- 
q u i:;i A. 'I'wman T i I 98 1 1 Eva 111 a lion, of splenic emboliza- 
tion i:i paiiems ivi:li porL.il hyperiensien and hypersplen- 
ism.Ann Surg 196:518-524 

115.0wmanT,LunderquistA,AlwmarkA,BoriessonB(1979) 
embolization of I he sp.een for irealment of sp.en.cmegaly 
anal hypersplenism in paiiems will: port.'.l hypenensien. 
Invest Radiol 14:457-464 

I 1 !■. Sola fain 8| U9S1 I The role of angiographic he mo stasis in 
salvage of the i mil real spier::. Radiology 141:645-650 

I IJ.Tsapogas Ml, Karmedv AM. Peaoody RA. Ch'antrasal-;ul C 
(1971) The fate of the spleen after partial or total dearte- 
rialization. Br J Surg 58:866-867 

118.MozesMF,SpigosDG,PollakR,AbejoR, Pavel DG, Tan WS 
ei a I. i I 984) Paioial splenic e iitb ::■ 1 1 zn i i >: :"■_. :w\ a lie: native lo 
splenec:em.y-resuhs of a prospec:ive, randomized study. 
Surgery 96:694-702 

119.Spigos DG, Tan WS, Mozes MF, Pringle K, Iossifides I 
( i 950) Splemc embolization. Cardiovasc Intervent Radiol 
3:282-287 

120.Kumpe DA, Rumack CM, Pretorius DH, Stoecker T], Stellin 
GV (I985J Panial splenic embolization in children with 
Hypersplenism. Radiology 155:357-362 

121.Latshaw RF.Pearlman RL, Schaitkin BM, Griffith JW,Wei- 
dner YVA (1985) Intraarterial eihanol as a long- term occlu- 
sive a gem ;:'. ren.n, hepal :.:. anil gastrosp.en.ic ai aeries of 
pigs. Cardiovasc Intervent Radiol 8:24-30 

122.Mineau DE, Miller F] Jr, Lee RG, Nakashima EN, Nelson 
IA :. I 9SJ. i Experimental Iranscatheier splenectomy as.ng 
abseluie eihaii::'l. lia die logy : 42:355-359 

123.Kaufman SL, Strandberg JD, Barth KH, White RI Jr (1978) 
TranscaLheiet emoo.izaiion. w.th microfibrillar a e. In gen in 
swine. Invest Radiol 13:200-204 

124.Goldman ML, Skorapa V ]r, Galambos JT, Oen KT, Jove DF, 
iiloe: ii'.an .VI i I 978; I mra -arterial [ issue aahesives for med- 
ical splenectomy in dogs. Am 1 Gastroenterol 70:489-495 

125.Mazer M, Smith CW, Martin VN (1985) Distal splenic 
artery emboliza:iei: with a f.C'W-d i rected galloon catheter. 
Radiology 154:245 

126.Yamauchi T, Furui S, Irie T, Kusano S (1994) Partial splenic 
embolization ivi:h V-siiaped silicone pa: nicies. Acra Radio. 
35:335-339 

127. Wright KC, Anderson JH, Gianturco C, Wallace S, Chuang 
VP !!9Sa! Partial splenic embolizauon using polyvinyl 
a lac ho! foam, d extra n. poly sly re ne, or silicone. An experi- 
mental study in dogs. Radiology I 42:351-354 

128.WatanabeY,TodaniT,NodaT (1996) Changes in splenic 
volume afier aan.al splen.c embolization in children. I 
Pediatr Surg 31:241-244 

129.Palsson B, Hallen M, Forsberg AM, Alwmark A (2003) 
Pama. sp.eme embolization: .ong-lerm ci.itcome. Langen- 
becksArch Surg 387:421-426 



lO.Nio M, Hayashi Y, Sano N, Ishii T, Sasaki H, Ohi R (2003) 
Long-term efficacy of partial splenic embolizauon in chil- 
dren. J Pediatr Surg 38:1760-1762 

ll.Galbraith PR (1967) The mechanism of action of splenic 
irradiation in chroma myelogenous leukemia. Can Med 
Assoc J 96:1 636-1 641 

!2.Abrams RA, Liu PJ, Ambinder RF, Haulk TL, Korman LT, 
Herman MG et al. (1997) Hodgkin and non-Hodgkin 
ivmchoma: .oca, -regional radiation therapy after oone 
marrow transplantation. Radiology 203:865-870 

!3.EUiott MA, Chen MG, Silverstein MN, Tefferi A (1998) 
splenic irradiation for symptomaiic splenomegaly asso- 
ciated with myelofibrosis with myeloid metaplasia. Rr I 
Haematol 103:505-511 

!4.Schratter-Sehn AU, Cerveny M, Simmel H, Schlogl E, Sch- 
latter A ia33.i :■ Short-time splenic irradiaiion for spleno- 
megaly. Onkologie 26:21-24 

!5.McFarland JT, Kuzma C, Millard FE, Johnstone PA (2003) 
Palliative irradiation of the spleen. Am J Clin Oncol 
26:178-183 

!6.Ariel IM, Padula G (1973) Irradiation of the spleen by the 
imra-artetia. admimstralion of a i) yttrium microspheres 
in patients veidi malignant lymphoma. A prel.nunarv 
report. Cancer 31:90-96 

!7.BeckerCD,RosIerH,BiasiuttiFD,BaerHU(1995)Conges- 
uve hypers.plenis.m: treatment by means of raaioemboli- 
zation of the spleen with Y-90. Radiology 195:183-186 

IS.Nahum Goldberg S, Dupuy DE (2001) Image-guided 
fadioiVeqiiencv tumor ablauon: challenges anal opponu- 
nities.part I. J Vase Interv Radiol 12:1021-1032 

J9.Dupuy DE, Goldberg SN (2001) Image-guided radiofre- 
quency tumor a b la i ion: c::;i.letigcs anal opportunities, pari 
II. J Vase Interv Radiol 12:1135-1148 

10 .Liu QD, Ma KS, He ZP, Ding J, Huang XQ, Dong JH (2003) 
Lxpe: niiri'.i;]! study en. the lV.o : ih:l::y and safety of radio - 
frequency ablalion for secondary splenomagely ana 
hyp ersplenism. World J Gastroenterol 9:813-817 

il.Schulman CC, Struyven 1, Giannakopoulos K. Mathieu 1 
(I98l'i) Preoperanve embolization o: renal rumors-Com- 
parison of different methods. Eur Urol 6:154-157 

!2.Kato T, Nemoto R, Mori H, Takahashi M, Tamakawa Y 
i I 981 j Transcalheie: arterial c he mo embolization of renal 
cell carcinoma ivith microencapsiilaied mitomycin C. i 
Urol 125:19-24 

!3.LeGuillou M, Mer land JJ (1982) The indications of emboli- 
sation in renal aimor: whal remains :o be srna; Piog Clin 
Biol Res 100:603-607 

!4.Teasdale C, Kirk D, Jeans WD, Penry JB, Tribe CT, Slade N 
i I °8aJ i Arterial embolisaiion iij renal carcinoma: a useful 
procedure? Br J Urol 54:616-619 

IS.Chudacek Z, Zavaza. V ( I 98? 'i r'alliaiive embolization <A~ 
renal tamers. I. Immunologic reaciii'-n o:- the emboliza- 
tion. Radiol Diagn (Bed) 27:211-213 

!6.Karwowski A, Wojtowicz J (1987) Long-term results of 
embolization in renal tumors. Radiol Diagn (Bed) 28:533- 
535 

i7.L : toesslein F, "c liner. l-.e A. Muensirr W (]C88i Percutane- 
i.' ■'.'.■• iransluminal emboljza:i':;'. IV -j" impi'oVrJ :■ ■£:.-.■■■.■■ ■ 
renal cell aa re. n.:aii a -dependence '.■:"; tumor slages. Cardio- 
vasc Intervent Radiol 11:91-96 

i8. Swansea"! ["'A. Wallace S ( I 'W* Margery '.■]" metastatic renal 
cell carcinoma and use of renal infarciion. Semin iurg 
Oncol 4:124-128 



17 Research and Future Directions 
in Oncology Embolotherapy 



Introduction 221 

Advances in Chemotherapeutic Agents 21 
.1 Convention Li I Chtiviccherapeulic Agents 1 
2 Novel Drug Regimens 222 

Update on Current Use and Development 

of Embolic Agents 223 
.1 Ethiodol 223 
1 Gelfoam 224 

.3 Polyvinyl Alcohol (PVA) Particles 225 
A N-Butyl-2-Cyanoacrylate 225 
.5 Spherical Eincojc Agents 22" 
.5.1 Plain Spherical Agents 226 
.5.2 Drug-Loaded Embolic Agents 22S 

Advance m^:'.:? in E:y.:iojza:io:' Technique 

Conclusion 229 

References 230 



17.1 
Introduction 

Oncology embolotherapy consists of a catheter- 
based group of treatments where embolic agents are 
intravascularly directed to target cancerous lesions. 
This group of therapies is mainly represented by 
transcatheter arterial chemoembolization (TACE) 
for primary and metastatic hepatic lesions; how- 
ever, oncology embolotherapy has been applied 
beyond liver for palliative treatment of bone, pul- 
monary, renal, oral cavity or anterior oropharynx 
lesions [11, 29, 30, 71]. The underlying principle for 



E.Liapi.MD 

The Russell K. Moi ga:'. I 'epai^meni '.■:' Radiology and Radio- 
iogicul Sciences, Johns Hopkins Medical l:vs:iluiions, Balti- 
more MD, USA 
J.-F. H. Geschwind, MD 

Associate Professor ■ 'I" Had [.:■;■ >gv. S.ngeiv j.i'.d Oncology, Divi- 
sion of Vascular and Intervention.;! Radiology, 600 N Wolfe 
Street, Blalock 545 Baltimore, MD, 21287, USA 



the procedure is selective and precise delivery of 
high doses of chemotherapeutic agents to malig- 
nant hepatic lesions, so as to achieve local tumor 
control, while minimizing systemic toxicity. While 
exploiting the well-known arterial blood supply of 
malignant hepatic lesions, oncology embolother- 
apy involves the infusion of chemotherapeutic and 
embolic agents. 

In practice, despite its promising concept of 
design, TACE has not shown yet to be as effective 
and potent as in theory. Several challenging obsta- 
cles that have not been exceeded yet include chemo- 
therapeutic dose-limiting toxicity, development of 
mechanisms for drug resistance and tumor revas- 
cularization. Moreover, the techniques and agents 
used for intra-arterial treatment of primary and 
metastatic liver cancer are very heterogeneous. This 
hinders a more systematic approach and interpreta- 
tion of the results of clinical trials as well as imple- 
mentation of meta-analyses. 

Despite its heterogeneity and limitations, trans- 
catheter arterial chemoembolization has gained wide 
acceptance over the past 20 years and is currently 
considered as the mainstay therapy for unresectable 
primary and metastatic liver cancer [64, 67]. Recent 
advances in infusible drug regimens, embolic agents 
and embolization design, as wellas the positive results 
of two recent randomized controlled trials and a 
meta-analysis, have boosted investigators' optimism 
to further develop this method of local tumor control 
[7, 38, 39]. The extent of tumor necrosis after chemo- 
embolization has been reported to range from 60% 
to 100% [58]. However, these data need to be further 
supported by conducting prospective randomized 
clinical trials, with use of standardized interventional 
oncologic protocols. Moreover, adequate knowledge 
on the molecular basis of hepatic tumorigenesis may 
optimal treatment of primary 
ancer. New developments and 
i hepatic oncology embolo- 
imalgamation of its evolution- 
with future trends in chemotherapeutic 
agents, embolic agents and embolization techniques. 



and metastatic liver c 
research activities o: 
therapy represent a 



E. Liapi and J.-F. H. Geschwind 



Following, we present a comprehensive approach to 
recent advances on hepatic embolotherapy. Advances 
on radioactive microspheres embolization are not 
included, as they are thoroughly discussed inaprevi- 



Advances in Chemotherapeutic Agents 

17.2.1 

Conventional Chemotherapeutic Agents 

Currently, there is no good evidence for the best 
chemotherapeutic agent. A number of chemothera- 
peutic cocktails have been used for chemoemboli- 
zation in the past decade, and controversy persists 
regarding the selection of the most potent of these 
drugs. A large variety of drugs including mitomy- 
cin C, doxorubicin, epinibicin, cisplatin have been 
used with TACE. The most common chemothera- 
peutic drug used as a sole agent is doxorubicin, 
whereas the combination of cisplatin, doxorubicin, 
and mitomycin C is the most common combination 
drug regimen for treatment of HCC [6]. All of these 
drugs exhibit preferential extraction when deliv- 
ered intra hepatic ally and they can achieve favor- 
able tumor drug concentration with concurrent low 
systematic drug load. Despite their favorable and 
high intratumoral concentrations, most random- 
ized controlled trials have failed to demonstrate 
advantage of one agent over another [51]. In one 
study, cisplatin was shown to be more effective than 
doxorubicin as a single agent against HCC; however, 
this improved effectiveness could not be correlated 
with improved survival [9]. Recent data suggest that 
injectable volumes of chemotherapy and long-term 
arterial patency were improved by embolizing the 
tumor-feeding vessel(s) only after the entire dose 
of chemotherapy had been delivered [20]. In our 
institution, a standardized regimen of chemothera- 
peutic agents, based on the Hospital of the Univer- 
sity of Pennsylvania protocol, regardless of tumor 
type, is currently used. This regimen comprises of 
cisplatin 100 mg (Bristol Myers Squibb, Princeton, 
NJ), doxorubicin 50 mg (Adriamycin; Pharmacia- 
Upjohn, Kalamazoo, MI) and mitomycin C 10 mg 
(Bedford Laboratories, Bedford, OH) mixed in 10 
ml of water-soluble contrast medium (Omnipaque; 
Winthrop Pharmaceuticals, New York, NY). This 
cocktail is then mixed with an equivalent volume 
oflipiodol. 



17.2.2 

Novel Drug Regimens 

Advances in the knowledge of the molecular patho- 
genesis of HCC and hepatic tumor igenesis have led to 
the testing of some novel cytostatic agents that may 
interact upon some disrupted pathways. Among their 
significant properties is their ability to overcome drug 
resistance, inhibit angiogenesis and limit chemother- 
apeutic dose-related toxicity. Phase I/II/II1 studies are 
currently being conducted to explore whether anti- 
angiogenesis agents, inhibitors of growth- factor- sig- 
naling and cell cycle enzymes, nonspecific growth 
inhibitory agents, specific ;.i unionists of HCC tumor 
markers, and a nt i- inflammatory agents, may have a 
role in the treatment of liver cancer [8]. 

Among those agents, bevacizumab (Avast in, 
Genentech Inc., San Francisco, CA), a humanized 
monoclonal antibody that binds vascular endothe- 
lial growth factor (VEGF) and prevents its interac- 
tion to receptors on the surface of endothelial cells, 
has been recently added to the triple chemoemboli- 
zation cocktail for patients with primary and meta- 
static liver cancer. A recent pilot study, in selected 
HCC patients undergoing TACE who additionally 
received intravenous bevacizumab, showed encour- 
aging results with good drug tolerance and pro- 
longed disease control [4]. Currently, there are two 
phase II trials evaluating the safety and efficacy of 
bevacizumab in patients with primary and meta- 
static unresectable liver cancer [47, 48]. The com- 
bination of bevacizumab with this TACE regimen 
seems challenging, as the formation of new blood 
vessels may effectively be reduced while the targeted 
tumor may maintain high cytotoxic chemothera- 

3-Bromopyruvate (3-BrPa) is another example 
of a drug disrupting a metabolic pathway, which 
has been recently tested via transcatheter infusion. 
3-BrPa is a hexokinase II specific inhibitor, which 
potently abolishes cell ATP production via the inhi- 
bition of glycolysis [19]. Preliminary experiments on 
the rabbit VX-2 tumor model for liver cancer with 
direct intraarterial infusion of 3-BrPa showed very 
specific necrosis of the implanted lesions [19, 21]. 
Additionally, intraarterial injection did not affect 
the viability of surrounding normal liver tissues, nor 
damaged the animals' major tissues during systemic 
infusion [19]. The mechanism of innate resistance of 
normal cells against 3-BrPa treatment has not yet 
been clarified, though it might be related to the dif- 
ference of hexokinase II expression levels between 
normal and malignant cells [17]. 



Rr? ■?;'.: ch .iiid Future Din 



u Oncology Embdcdier.ipv 



Recombinant adenoviral vectors, such as those 
expressing recombinant b-galactosidase or human 
hepatocyte growth factor, soaked in gelatin sponge 
pledgets, have been recently tested for transcatheter 
delivery in canines [53]. The intratumoral injec- 
tion of oncolytic adenovirus seems to be a promis- 
ing approach for the treatment of tumors resistant 
to other modalities, but its intra-arterial infusion 
warrants additional research. To date, the pub- 
lished clinical experience with these agents has been 
almost exclusively limited to intratumoral injection 
on Phase I and II studies [62, 63]. 

The ideal chemotherapeutic agent for transcathe- 
ter treatment of primary and metastatic liver cancer 
is yet to be developed. This agent should combine 
effectiveness against the tumor and reduced toxicity 
to the normal or cirrhotic liver. In the near future, 
some of the above novel drugs are expected to 
enter the clinical arena and be prospectively tested, 
whereas some of the currently used chemotherapeu- 
tic agents such as doxorubicin, cisplatin and mito- 
mycin C are expected to be tested in a more system- 
atic and prospective way. 



Update on Current Use and Development 
of Embolic Agents 

Several embolic agents have been employed over the 
past two decades in order to enhance the effects of 
transcatheter intraarterial drug delivery for hepatic 
malignancies. These agents may produce different 
effects on vasculature, resulting in permanent or 
transient obstruction, by acting at different levels in 
the arterial system. Usually, the injection of embolic 
particles follows the injection of the chemotherapeutic 
mixture, yet, some centers favor mixing the particles 
in slurry with the chemotherapeutic drugs and oil 
[20]. Embolization without chemotherapy (transarte- 
rial embolization) has often been categorized as a form 
ot chenioemboli^i.ition [?$]. Tins torn) ot emboloth cr- 
apy has been occasionally described in the literature. 
Arterial embolization alone has been investigated in 
two randomized trials from Barcelona, which failed 
to demonstrate a survival benelii [5, oS]. 

Despite the extensive research that is available on 
hepatic embolization, the precise effects of emboli- 
zation on tumor cells remain largely unknown. In 
fact, recent data suggest that hypoxia, generated by 
arterial embolization, may activate several genes, 
including those for vascular endothelial growth 



factor (VEGF) and hexokinase II, leading therefore 
to compensatory angiogenesis and tumor growth 
[24]. A direct link among the degree of emboliza- 
tion, tumor hypoxia, and the stimulation of new 
blood vessels has been suggested in a number of 
recent studies. Kobayashi et al. found that blood 
levels of VEGF were markedly increased in patients 
who had been treated with embolization [27]. Poon 
et al. reported that serum VEGF level was signifi- 
cantly elevated in patients with HCC, and signifi- 
cantly high serum VEGF levels were associated with 
the absence of tumor capsule, the presence ofvenous 
invasion and microsatellite modules, and advanced 
TNM stage [56]. More recently, high serum level of 
VEGF was found to be a predictor of poor outcome 
after resection of HCC [57]. VEGF may also help in 
predicting treatment response and monitoring dis- 
ease course after chemoembolization [35]. 

From a different standpoint, similar simple obser- 
vations have already confirmed the angiogenesis 
theory by the formation of early revascularization 
after proximal and temporary embolization induced 
with gelfoam [20]. Moreover, it has been demon- 
strated that proximal embolization of tumor-feeding 
arteries in hepatic metastases with large particles or 
coils may lead to immediate peripheral hepatic cir- 
culation re constitution through collateral vessels. 
It seems that the earlier the revascularization of the 
tumor occurs, the more incomplete the necrosis will 
be. An occlusion of more peripheral vessels gener- 
ates a nearly complete tumor necrosis and current 
trends in oncology embolotherapy seem to favor 
distal occlusion. Gelatin sponge powder and pled- 
gets and polyvinyl alcohol are the most commonly 
used agents for chemoembolization [20]. Combina- 
tions of ethiodol with other embolic agents have also 
been reported for transcatheter use. Autologous clots, 
coils and microcoils, or collagen have also been used 
for oncology embolotherapy, but few reports have 
assessed their efficacy [37]. An overview of charac- 
teristics, current use and future prospective of some 
commonly used embolic agents is next presented, as 
well as an introduction to some novel embolic agents 
(Table 17.1). The latter new category of new embolic 
agents may lead to more accurate tumor targeting 
and ultimately, improve patients' survival. 



17.3.1 
Ethiodol 

This oily medium is a key ingredient to the chem 
embolization procedure due to its unique combir 



: 17.1. Size of emir oik ,i£e:v.~ irv.:. ::.■<: oncology em bolo therapy 



Embolic Agents 



P;i-:k-:ir Siz. 



Polyvinyl alcohol foam 

Gelfo-.im igc-kicii] sponge particles) 

Tris-acryl gel micro spheiei iH::".'o.:go!ii and Embospher 

Polyvinyl alcohol microspheres iContourSE) 

Polyvinyl alcohol hydoigel :v..-.":o<pheres (Bead Block) 



150-500 Li m 

Microspheres) 40-1200 urn 
45-1180 u.m 
100-1200 u.m 



E. Liapi and J.-F. H. Geschwind 



tion of properties as a drug-carrier, tumor-seeking 
and embolic agent. Since the observation that this 
poppy seed oil accumulates preferentially in hepato- 
cellular carcinoma (HCC) and other hepatic malig- 
nancies, ethiodized oil or lipiodol (Ethiodol; Savage 
Laboratories, Melville, NY) has been successfully 
used as a suspension medium for chemotherapeutic 
agents [13, 28, 45](), as a radiolabeling means by 
labeling part of its iodine with 131 I to deliver targeted 
radiotherapy, or as a plain embolic agent [2, 28, 50, 
60]. In most cases, ethiodol is used along with a 
chemotherapeutic cocktail and constitutes the basis 
of most TACE procedures. 

Although ethiodol has been used for more than 
20years for chemoembolization of HCC and hepatic 
metastatic lesions, the mechanism of its uptake by 
cancer cells is not clearly understood. A biochemical 
pump in the tumor cell wall recognizes ethiodol and 
transfers the emulsion in the intracellular space, 
and this pump is subsequently disabled by hypoxia, 
thus trapping the ethiodol emulsion within the cell. 
Ethiodol localizes in hepatic tumors when adminis- 
tered via the hepatic artery, and is retained by HCC 
for many weeks, even up to 1 year, while it is cleared 
from normal or cirrhotic liver within 4 weeks. 
When injected into the hepatic artery, it traverses 
the peribiliary plexus to the portal veins resulting 
in a dual embolization [69]. The amount of lipiodol 
emulsion has been shown to proportionally depend 
on the tumor size. However, hepatic parenchymal 
damage or bile duct ischemia have been reported 
by use of large amounts of lipiodol [12]. Individual- 
ized adjustment of lipiodol dose, according to blood 
supply pattern and tumor diameter as measured on 
CT, has been recently suggested in a controlled ran- 
domized trial [10]. The degree of lipiodol accumula- 
tion at computed tomography has been shown to be 
an independent indicator of better prognosis [44]. 

Labelingpart of lipiodol's iodine with 131 I attracted 
researchers' interest early in the course of investigat- 
ing lipiodol's potential for hepatic embolotherapy 
[75].Intra-arterial 131 I-lipiodol has been successfully 
used to treat inoperable hepatocellular carcinoma 
and is well tolerated and effective in small tumors 
(<5 cm) [42, 76]. Randomized studies have shown 



that treatment with 131 I-lipiodol is at least as effec- 
tive as but less toxic than chemoembolization and 
that there is a significant increase in the survival of 
patients presenting with an HCC with portal throm- 
bosis [61]. Moreover, it may lead to a 50% reduction 
in the recurrence rate 3 years after a resection [31]. 
Nevertheless, the administration of 131 I-lipiodol has 
limitations related to radiation protection issues. 
Following treatment with i3l I-lipiodol, patients are 
not eligible for surgery for up to 4 weeks, due to the 
relatively long physical half-life of m I (8 days) [31]. 
Another drawback is the required hospitalization of 
up to 7 days, due to the high energy of the gamma- 
ray emission of l3l I (365 keV) and its physical half- 
life. 

There have been various attempts to label lipi- 
odol with other isotopes, in particular 90 Y, l88 Re 
and recently g?m Jc [31, 77, 78] .Currently, the best 
approach seems to be the labeling of a lipophilic 
chelating agent with 188 Re, which is then put into 
solution secondarily with the Lipiodol [77]. 188 Re is a 
radionuclide with a higher energy of the beta-emis- 
sion (2120 keV and 1960 keV for i88 Re versus 606 
keV for 131 I) and its use, with its physical half-life 
of 17 h, avoids most of the radioprotect ion problems 
and allows for a shorter hospitalization, as well as 
further dose escalations. Following 188 Re-lipiodol 
treatment, patients are eligible for transplantation 
after an interval of only 1 week. However, certain 
problems remain, such as the relatively weak label- 
ing efficiency, and further studies to determine the 
maximum tolerated dose and potential hepatotoxic- 
ity should be assessed. 



17.3.2 
Gel foam 

Gelatin bioabsorbable embolic agents, in the form 
of gelfoam sponge or powder, have been exten- 
sively used as an intravascular embolization agent 
for TACE. Gelatin sponge, which blocks circulation 
transiently and is absorbed within 48-72 h, is cur- 
rently the most commoiilv employed material. Gel- 
atin sponge causes temporary vascular occlusion. 



Reseitich .iiid Future Diri 



n Oncology Entbolodierupy 



with recanalization occurring in approximately 
2 weeks [20]. When compared to powder, gelfoam 
sponge provides a more proximal obstruction to 
blood supply. However, proximal obstruction may 
enhance the development of revascularization of 
treated lesions through aberrant collaterals and 
limit tilt 1 eftieacv ol Inrtlter embolization. On tile 
other hand, gelfoam powder can induce ischemic 
bile duct necrosis. Improved overall patient survival 
rates with the addition of geltoam sponge to the lipi- 
odol emulsion were initially described by Nakao et 
al. in a retrospective study on 343 patients [46]. In 
this study, no dose-response relationship was found 
and based on these data, [he combination of lipiodol 
and gelfoam sponge was further suggested for trans- 
catheter treatment of hepatic malignancies. The use 
of gelatin-sponge allows repetitive chemoemboliza- 
tion procedures, which in some centers is desirable 
[20]. Today, gelfoam is still widely used and new 
applications of its use, such as its soaking in recom- 
binant adenoviral vectors may shed new light in the 
use of this traditional embolic agent [53]. 



17.3.3 

Polyvinyl Alcohol (PVA) Particles 

Polyvinyl alcohol (PVA) particles have been success- 
fully used since 1974 as an intravascular embolic 
agent and for many years, it has been considered 
the standard embolic agent for TACE [68]. The earli- 
est method of its preparation for use as an embolic 
agent first involved its conversion into foam that 
can absorb water and become readily compressible. 
According to this preliminary method, PVA par- 
ticles were prepared by scraping a compressed block 
or by cutting off pieces, which were then filtered 
through different sizes of sieves. Their irregular 
shape, however, did not ensure uniformity of the 
final compound, as large particles were likely to pass 
through small holes depending on their orientation 
during filtration. This problem created variability 
in size and shape; however, later on, changes were 
made to ensure improved uniformity. 

Polyvinyl alcohol is considered to be a permanent 
embolic agent because it is not biodegradable. The 
histologic effects of PVA particles embolization have 
been well documented, varying from inflammatory 
and foreign body reactions to focal angionecrosis 
of the vessel wall [36]. The duration of the vascular 
occlusion induced by PVA is variable. Occlusions 
may last for several months as a result of organiza- 
tion of the thrombus, with recanalization attributed 




Fig. 17.1. Polyvinyl alcohol piirdcles. Their irregulur slu;p 
does not gu-.iriii'.iee precise wssel occlusion, l Courtesy of Bio 
sphere Medical Inc.) 



to thrombus resorption and angioneogenesis [36]. 
Unexpected and non-targeted embolization has 
been reported, creating some uncertainty over their 
use. In a study testing the effects of embolization 
protocol on injectable volume of chemotherapy and 
subsequent arterial patency, the type of chemoem- 
bolization protocol rather than the type of embolic 
material had a significant impact on the rate of 
arterial recanalization or arterial patency [20]. Sur- 
prisingly, the assumption that PVA particles result 
in deeper penetration, when compared to gelfoam 
pledgets, was not confirmed in this study [20]. 



17.3.4 
N-Butyl-2-CyanoacryIate 

This glue, first used for bleeding control, has been 
successfully utilized for preoperative portal vein 
embolization [15]. This adhesive, that polymerizes 
on contact with blood and endothelium, does not 
affect the peribiliary arterial plexus, and this prop- 
erty led to the assumption that it can be used for 
transcatheter arterial embolization [55]. Mixed with 
radiopaque lipiodol, the polymerization time can be 
prolonged to 10-15 s, depending on dilution. There- 
fore, a more peripheral and permanent embolization 



In an initial short report, patients with c; 
hepatic metastases treated with a mixture of W- 
butyl-2-cyanoacrylate/ethiodol, showed complete 
and long-lasting relief of symptoms, with signifi- 
cant decrease or normalization of levels of 5-HIAA 
in the urine, and a reduction of metastatic tumor in 
the liver [72]. Another study demonstrated prom- 
ising results in the treatment of metastatic hepatic 



E. Liapi and J.-F. H. Geschwind 



insulinomas [73]. Other retrospective studies have 
shown that TACE with use of cyanoacrylate and 
lipiodol for unresectable HCC and neuroendocrine 
hepatic metastases is feasible [41, 42]. Cyanoacrylate 
was recently used in a retrospective study of patients 
treated for unresectable HCC, combined with other 
embolic agents without concurrent addition of che- 
motherapy [59]. Still, this type of embolic material 
is used for bland embolization and assessment of its 
effectiveness compared to the use of other embolo- 
therapy regimens is rather complicated. 



17.3.5 

Spherical Embolic Agents 



tithe: 



echm 



sofa. 



!S Of Ct 



Investigati 

mercially available embolic agents demonstrated 
that novel embolic materials should possess a com- 
bination of certain mechanical and physicochemi- 
cal properties in order to maximize their efficiency 
and minimize potential side effects. First, they 
must be spherical in shape to allow for accurate 
calibration, optimal and complete geometric vessel 
occlusion. Spherical particles have a single dimen- 
sion; moreover, depending on their size, they may 
block tightly and gradually seal the vascular lumen, 
whereas non-spherical particles produce a more 
incomplete occlusion. Second, the particles must 
not aggregate, which may lead to catheter obstruc- 
tion and proximal arterial occlusion. Other desir- 
able properties include unproblematic fabrication, 
controlled deformability, good biocompatibility and 
in vivo stability when mixed with chemotherapeutic 
agents. Moreover, potential drug-loading combined 
with sustained drug release seem to be very attrac- 
tive properties that may lead to the establishment of 
a new category of embolic agents and transform the 
profile of oncology embolotherapy. 

With the recent bloom in na no technology, sev- 
eral types of microspheres have been developed [3]. 
Some of these new types of spherical agents, such 
as polyvinyl alcohol and tris-acryl microsphi 
are non-degradable, resulting in more permanent 
occlusion. Others, such as gelatin, albumin, poly- 
saccharides (dextran), starch, ethyl cellulose, and 
poly-(D,L lact id e/glycos id e)- copolymer include a 
resorbable component, and are not suitable for per- 
manent vascular occlusion [18]. The optimal size of 
these embolic agents tor chemoembolizationhasyet 
to be established. Spherical embolic agents may be 
categorized into plain (unloaded) and drug-loaded 
(or drug-eluting) spheres. 



17.3.5.1 

Plain Spherical Agents 

17.3.5.1.1 

Tris-acryl Gelatin Microspheres 

Tris-acryl gel microspheres were the first spherical 
embolic agents to be commercially available [32]. 
Tris-acryl is an entirely synthetic, hydrophilic, and 
nonresorbable material. It has been demonstrated 
that this material produces non-toxic tissue reac- 
tion, thus allowing absorption and cellular adhe- 
sion [32]. Colored and non-colored tris-acryl gelatin 
microspheres (Embogold and Embosphere Micro- 
spheres; Biosphere Medical, Rockland, MA} are cur- 
rently commercially available. 

These microspheres are precisely calibrated, 
spherical, hydrophilic, microporous beads made 
of tris-acryl co-polymer coated with gelatin. They 
come in defined range of sizes, ranging from 40 to 
1200 |im in diameter. Their smooth hydrophilic sur- 
face, deformability and minimal aggregation ten- 
dency have been shown to result in a lower rate of 
catheter occlusion and more distal penetration into 
the small vessels [32]. Their efficacy has been evalu- 
ated in several conditions, and when compared to the 
standard polyvinyl alcohol particles (PVA) particles, 
a deeper penetration and embolization of smaller 
and more peripheral vessels may be achieved. This 
distal embolization may limit the development of 
any collateral blood supply. Also, in a study where 
PVA particles and tris-acryl microspheres of similar 
size were compared, the level of vascular occlusion 
with calibrated tris-acryl microspheres precisely 
correlated with particle size whereas the level of vas- 
cular occlusion with PVA particles did not. Another 
study has demonstrated that in embolized tumors, 




7<ChrO 



Fig.17.2. Embosphere microspheres. These tris-acryl gelatin 
micro spheres are precisely c-.ihoi LUeJ. spheres!, hydrophilic. 
microporous beads, i Oout tesy of Andy Lewis, biocompatibles, 
UK, Ltd, Surrey, UK) 



Research and Futu 



n Oncology Embolorheiiipy 



a great majority of occluded vessels were located 
within the tumoral tissue, and vessels located out- 
side the tumor were rarely occluded [33]. Interest- 
ingly, the median diameter of the occluded vessels 
located within the tumors was 210 u.m. However, the 
authors avoided suggesting this number as a thresh- 
old for the penetration, due to tumor vessel variabil- 
ity and histological processing piikilK 

Tris-acryl gelatin microspheres have be 
to be stable in a standard chemoembolization solu- 
tion and some centers have adopted their use [1]. 
Tris-acryl gelatin microspheres have been also 
tested for compatibility with several chemothera- 
peutic agents and can be mixed with carboplatin, 
mitomycin C, 5-fluorouracil, or pirarubicin for 
chemoembolization without any risk of harmfully 
altering their morphology, dimensions, or geomet- 
ric characteristics [70]. 

17.3.5. 1.2 

Polyvinyl Alcohol Microspheres 

A long track record of surety and efficacy of polyvi- 
nyl alcohol led to the design of spherical PVA. PVA 
(Contour SE Microspheres; Boston Scientific/Medi- 
tech, Natick, MA) and PVA hydrogel (Bead Block, 
Terumo Medical Corp., Somerset, NJ) are currently 
available for transcatheter use. 

Interestingly, PVA microspheres have been 
observed to result in a very mild inflammatory 
response [65]. This finding seems rather counterin- 
tuitive and unexpected, considering the aggressive 
inflammatory reactions that have been described 
with PVA particles. Further studies, however, should 
be conducted to assess whether this reduced intra- 
vascular and perivascular inflammation may have 
significant favorable clinical implications [65]. 

17.3.5.1.3 

Other Types of Plain Microspheres 

Poly-L -lactic acid (PLLA) microspheres have also 
been tested for transcatheter delivery, but they are 
biodegradable and tend to clog microcatheters, like 
PVA particles. The diameter of these particles 
can be changed according to the polymeriza- 
tion time and therefore, various sizes of PLLA 
microspheres can be prepared [74]. Loading of 
these particles with neutron- activated radioactive 
holmium 166 ( lt6 Ho) has also been tested for trans- 
catheter administration in an animal HCC model 
[49]. A recent study demonstrated that the Ho on 
the surface of neutron-irradiated Ho-PLLA-micro- 




Fig.17.3. Contour SE microspheres. PVA microspheres with 

smooth, isydraphikc su: tace and sLuidnrdizc-d caliber of size. 
(Courtesy of Andv Lewis. Riccom\-\i;iblcs. UK, Ltd, Surrey, 




Fig.17.4. Bead Block (Tei 
!-'VA hvdiogel microsphere 
patibles UK, Ltd, Surrey, I 



lino Mcdicil Coip.. ?i':-:eiset, Nl). 
^ (Co : .'ircsy of A iid y i.eivjs, Biocom- 



spheres is probably the reason for their poor sus- 
pending ability in saline and that a pharmaceuti- 
cally acceptable solvent (1% pluronic F68 or F127 
in 10% ethanol) can be used for the formulation of 
a homogeneous suspension, making these systems 
feasible for further clinical evaluation [79]. 

Radiopaque microbeads made of polyaciyloni- 
trile (PAN) hydrogel have also been evaluated in 
vitro and invivo for transcatheter embolization on a 
swine model showing potential for future use [23]. 

Superabsorbent polymer microspheres is another 
example of nontoxic and non-biodegradable solid 
particle with a spherical shape that has been recently 
tested in Japan [52]. The particle size is precisely 
calibrated in 50-um increments ranging from 53 to 
350 um and initial experience suggests that emboli- 
zation with use of these particles leads to extensive 
tumor necrosis of large nodular HCC, sparing use of 
chemotherapeutic agents [52]. 



E. Liapi and J.-F. H. Geschwind 




a Stable solution of Doxorubicin-elm^ng lie ads iPKHumON He.Ldsi m ;.„.iiie. Note thiit red-jolored doxorubicin 
e the bead, b Confer I last-:' nii^i'ostOj.iy i::..-,^. ?::.:iwiijg d> 'Xo: Lilian di-n it-;:: !■:■:'. in (he bend. Progressive 
sei/ii'.'iiiiig slji.'ws iiKi: ".lit; ■r::-^. :s co:'.iViiL[\i:ed iii the o..".e: [•■'.■r.ioi's of riie be;id. i C ■.■'..: r.esy ci A:'dy Lewis. R i i. .j ■. > j j ] y ;■ . i lt ^ e s . 
UK, Ltd, Surrey, UK] 



17.3.5.2 

Drug-Loaded Embolic Agents 

The concept of drug-loaded microspheres for che- 
moembolization is not new, and several attempts 
have been made over the past decade to design 
microspheres loaded with chemotherapeutic agents 
suitable for intraarterial delivery. Cisplatin-loaded 
poly(d,l-lactide) microspheres for chemoemboliza- 
tion [66], doxorubicin incorporated into albumin 
microspheres [26], poly (benzyl l-glutamate) (PBLG) 
microspheres containing cisplatin [34], 5-fluoro- 
uracil-loaded chitosan microspheres [16] are some 
examples of initial attempts to create vehicles of che- 
motherapeutic agents for precise tumor targeting. 
Today, the development of nanotechnology com- 
bined with the robust research activity in tumor 
pathophysiology has boosted up the production of 
noveldrug carriers for systematic or local tumor tar- 
geting. A promising example of this new category of 
embolic materials is the doxorubicin-eluting beads, 
which will be further analyzed. 

17. 3. 5.2. J 

Polyvinyl Alcohol Hydrogel Drug-EIuting Beads 

PVA hydrogels are non-biodegradable and relatively 
biocompatible and can be easily fabricated since they 
possess high stability under a range of temperature 
and pH conditions [43]. Their structural mechanical 
properties are similar to those of human soft tissues 
and provoke a limited inflammatory response [54]. 



Because of the amphophilic nature and microporous 
architecture of hydrogels, drugs can be incorporated 
in hydrogel coatings and can be typically released 
within hours to weeks [54]. The release of the loaded 
drugs in acidic environment represents another 
important property of these polymers. 

Recent development of a new PVA hydrogel bead 
has enabled accurate doxorubicin loadings to be 
achieved, with subsequent slow first order release. 
This novel drug-delivery system has been recently 
evaluated for intraarterial treatment of hepatic 
lesions [22]. Doxorubicin-eluting beads (DC Bead 
for loading by the physician and PRECISION Bead 
preloaded with doxorubicin, Biocompatibles UK 
Ltd, Surrey, UK), are designed for intraarterial infu- 
sion and selective tumor targeting [14]. 

The significant advance of these beads is the dem- 
onstration of an effective process for consistent drug 
release that controls local tissue response. Recent 
animal studies on a model of liver cancer have con- 
firmed the in vivo slow release of doxorubicin over- 
time within the tumor, with maintenance of a high 
concentration of drug for up to 14 days. Pharmacoki- 
netic data of this study showed near complete absence 
of doxorubicin within plasma, while intratumoral 
doxorubicin concentrations remained high [22]. 

Clinical studies that are necessary to support this 
initial report of efficacy are currently in progress in 
Europe and Hong Kong. These beads are not cur- 
rently available in the US market. Irinotecan-elut- 
ing beads for metastatic colon cancer are also under 
development. 



iieSe;l:ch .illu lr 11 T LI : c I"' if. 



:t Oncology Embolorlieiapy 



D mo rub I tin Ptiarmaco kinetics 





Fig. !7.'"'. 1- 1 i.ii in -.!■."■ 'ki nc lie sluciy results of cioxoi ubicin-eliiiiiig beads I ! : zBl 1:1 i; VX-J i-.ibbi: model of liver Initio: show lug:' 
g concentration wiih mini in ill systemic toxicity. Noie riie sigiiilicinUy lugije: levels of doxorubicin found ir 
or, peaking 3 o.iys -.me: ueaiineivi. ^usljined levels of ioxoi'ubici:'. iiie-.isnied in the tumor 14days i 



A future approach on this category of embolic 
agents would include a sophisticated regimen of 
timed release drugs appropriate to the pathologic 
stage of cancer, followed by a treatment that sup- 
presses angiogenesis (such as bevacizumab) or dis- 
rupts a neoplastic metabolic pathway (like 3-BrPa). 



Advancements in Embolization Technique 

Several variations in the techniques of embolo- 
therapy have been reported. Some of the common 
steps of the procedure include the patient's over- 
night fasting and volume loading, administration 
of conscious sedation and a visceral angiogram 
to identify the arterial anatomy of the liver, the 
size and locations of the tumors, and their feed- 
ing vessels and portal venous potency. A catheter 
is advanced beyond the gastroduodenal artery (to 
avoid extrahepatic embolization), and depending 
on tumor location and institution's protocol to 
lobar, segmental, or subsegmental branches feed- 
ing the target lesion(s). 

A common objective of every interventional 
oncologist is to preserve as much functional liver 
tissue as possible. Currently, there is no consensus 
on how selective (lobar vs. segmental) chemoem- 
bolization should be. Many centers prefer to focus 
on one lobe of the liver at each treatment session, 
regardless of the extent or number of tumors, so as 
to avoid a prolonged postembolization syndrome or 
postinterventional liver failure. Others may choose 
an even more selective approach, by engaging a seg- 
mental or subsegmental feeding artery. Longer sur- 
vival seems to be related to multiple TACE sessions, 



which require, however, long-term maintenance of 
arterial patency. Further research though is required 
to assess the effectiveness of long-term arterial 
patency [25]. Controversy also exists over the best 
treatment scheme for repetitive treatments and over 
the timing for repetition of the treatment session. 
Some centers prefer to treat patients at fixed timing, 
whereas others upon disease progression after the 
initial response. In our institution, the decision to 
re-treat is made on the basis of imaging and clini- 
cal assessment of tumor response. Appointments for 
imaging and clinical evaluation though are sched- 
uled within a fixed interval of time after treatment. 
In the future, a better knowledge of genetics and 
causes of hepatic malignancies may result insignifi- 
cant modifications of hepatic artery embolization 
techniques. 



17.5 
Conclusion 



Several questions remain unanswered, such as which 
is the best embolization agent, which is the best 
chemotherapeutic drug, or how can we increase the 
intratumoral concentration of the drug. The lack of 
large prospective randomized trials and the current 
difficulty in conducting meta-analyses on hepatic 
oncology embolotherapy, along with the absence of 
effective systemic therapy for unresectable primary 
and metastatic liver disease, urge intense efforts and 
the continuation of research on oncology embo- 
lotherapy. New developments in drug regimens, 
embolization materials and new variations in the 
embolization technique are rapidly changing the 
image of oncology embolotherapy and hopefully, 



1J.-F.H. Geschwind 



will positively influence the outcome oft: 
and patient survival. 

The interventional radiologist should also bear 
in mind that successful embolotherapy depends on 
factors beyond the embolic agent selection, or the 
choice chemotherapeutic cocktail. Technical skills, 
experience, familiarity with the underlying patho- 
logic processes, and appreciation of the importance 
for constructive collaboration with other special- 
ties, are also essential for every successful oncology 
embolotherapy treatment. 



l.Ball DS, Heckman R, et al. (2003) In vitro stability of tris- 
aci'yl ge.aiin microspheres :n a in a it ;p har mac eu ti,".i I che- 
moerabolization solution. I Vase Inierv Radiol 14:83-88 

2.Bretagne JF, Raoul JL, et al. (1988) Hepatic artery injec- 
tion of [-131 -iaiaeled lipiodol. Pari ]]. Preliminary results of 
: hej.ipe.i I .-." use in p.itients will: hepatocellular carcinoma 
and liver metastases. Radiology 168:547-550 

3.Brigger I, Dubernet C, et al. (2002) Nanoparticles in cancer 
therapy and diagnosis. Aav Prut; I 'eliv Rev 54:631-651 

4.BrittenC, Finn RS, Gomes AS, Armado R.Yonemoto L, Bent- 
ley G, Mass R, Bussutil RW, Slamon DJ (2005) A pilot study 
of IV bevacizumab in hepatocellular carcinoma patients 
:: jj-.H ergi::-:ng chemoembolizarien. 20(15 AS CO Annual Meer- 
ing, Orlando, Florida 

5.Bruix ], Llovet JM, ei al. i l l >/R! Transarterial embolization 
versus symptomatic treatment in paiienrs with advanced 
nepalocelailar carcinoma: results of a randomized, cent rolled 
trial in a single institution. Hepatology 27:1573-1 533 

a. Bruix 1, Sala M, et al. (2304) Chemcemboliznrion for hepa- 
tocellular carcinoma. Gastroenterology 127[Suppl 1 ]:S179- 
S188. 

7.Camma C, Schcpis F, et al. (20D2) Iransarlcrial chemo- 
embohzation ft* jn-i ;c.la"'L iicy.Ua.i llni. • carcinoma: 
meta-analysis ol r.v.>1:<rnijt-i1 i.i-rrc'leJ n. .i. Radiology 
224:47-54 

8.Carr BI (2004) Hepatocellular carcinoma: current manage- 
ment and future ircr.di i is;r.-Liirir:>l.'j;y I2/:S218 

9. Chang JM, Tzeng WS, et al. (1994) lrar,=catheter arte- 
rial embolization with or without cisplatm Ireatmeni ol" 
hepatocellular carcinoma: a randomized controlled study. 
Cancer 74:2449 
10. Cheng HY, Shou Y, et al. (2004) Adjustment of hpiodol dose 
according to rumor olood supply during transcaiheter arte- 
: i.-l chrmoirivifolizai.- c :"..■; large h-p.il- ■Cr.l.i.ai carcinoma try 
multidetector helical i2T. World 1 Gastroenterol 10:2753-2755 
ll.Chiras J, Adem C, et al. (2004) Selective intra-arterial 
chemeembolizarion o: pelvic and spine bone meiasiases. 
European Radiology 14:1774 
12. Chung J W, Park JH.et al. (1996) Hepatic tumors: predispos- 
ing faciors for complicaiions ol" rranscaiherer oily chemo- 
embolization. Radiology 198:33-40 
l3.Clo-.se ME, Perry L. et a I. ( I l / l /4j Hep an." arterial cbemoem- 
boiizalion for metastatic neuroendocrine tumors, inges- 
tion 55[Suppl 3]:92-97 



i. Cons rant in M, rtindueanu G. el al. (2004; Preparation and 
characterisation of po.yi vinyl :". _■,■:■ h ■:■ I .:,'cvcloaexrrin micro- 
spheres as matrix for inclusion and sen a rat ion of drugs. Int 
I Pharm 235:87-96 

a. deBaereT.RocheA.EliasD.Lasser P.Lagrange C.BoussonV 
,; 1 aa ; . ; preoperative portal vein embolization for extension 
of hepa recto my indications. Hepatology 24:1386-1391 

a.Denkbas EB, Seyyal M, et al. (1999) 5-Fluorouracil loaded 
chirosan microspheres f:'i" c h em oem conization. I Microen- 
capsul 16:741-749 

z.Foubister V (2002) Energy blocker to treat liver cancer. 
I 'rug Discovery Today 7:934 

3-Fuiimoto S, Miyazaki M, et al.(1985) Biodegradable mito- 
mycin C microspheres given iiirra-arrerially for inoperan!e 
hepatic cancer. Willi naniciLar reference lo a comparison 
wilh continuous infusion ■:< mitomycin C and ?-fluoroura- 
cil. Cancer 56:2404-2410 

). Geschwind J-F H, Ko YH, et aL (2002) Novel therapy for 
liver cancer: airect imraarteria. imection of a potent inhib- 
itor of ATP production. Cancer Res 62:3909-3913 

3. Geschwind JF, Ramsey DE.et al. (2003) Transcaiheter arterial 
chemoeinbolizaiio-ii o: aver tumors: effects of em> uzai.- ■:". 
prote-ce-. an iitieaablr veh.ime ol" cii em '.■therapy rial suhse- 
quent arterial patency. Cardiovasc Inrervent Radiol 26:111 

I. Geschwind JF, Georgiades CS, et al. (2004) Recently eluci- 
■aalo.l energy catabolism pathways nrovi.re opportunities 
for novel treatments in hepatocellular carcinoma. Expert 
Rev Anticancer Ther 4:449-457 

2. Geschwind JF, Khwaja A, Hong K (2005) New intraarte- 
ria. drug delivery system: Pharmacokinetics ana tumor 
response in an animal model of aver cancer. 200.- ASCO 
Annual Meeting. Orlanae, Florida. 

i.Gobin YP, Vinuela F, et al. (2000) Embolization with radi- 
opaque microbeads of polyacrylonitrile hydrogel: rvalua- 
tion in swine. Radiology 214:113-119 

J.Hanahan D. Folkman I (1996) Patterns and emerging 
mechanisms ol" the angiogenic switch during lamongen- 
esis. Cell 86:353-364 

a. Horst JJ, Ulrich-Martin M, et al. (1996) Sequential transar- 
terial cli em o embolization for unresectable advanced hepa- 
tocellular carcinoma. Cardiovasc Inrervent Radiol 19:388 

S.Kerr DJ, Willmotl N, et al. (1933) Target organ disposition 
ana plasma pharmacokinetics of doxorubicin incorpo- 
rated into albumin microspheres after intrarenal arterial 
administration. Cancer 62:878-883 

7.Kobayashi N, Ishii M, et al. (1999) Co-expression of Be 1-2 
protein and vascular endothelial growth factor in hepato- 
cellular carcinomas t reared by chemoembokzation. Liver 
19:25-31 

3-Konno T, Maeda H, et al. (1983) Effect of arterial admin- 
is ".rat ion of high -mo lee alar- weigh; amicancer a gen I 
SMAN'.rS will: an..: ivmphographic agem on hepatoma: a 
preliminary report. Eur J Cancer Clin Oncol 19:1053-1065 

S.Konya A, Van Pelt CS, et al. (2004) Ethiodized oil-ethanol 
capillary emboiizalion in rabbit kidneys: temporal histo- 
pathologic findings. Radio-log!" 232:147-153 

l.Kovacs AF (200:"l Chemoembolization using Cisplalin 
crystals as neoadiavam treatment of oral cancer. Cancer 
Biother Radiopharm 20:267-279 

I.Lambert B, Praet M, et al. (2005) Radiolabeled lipiodol 
therapy for hepatocellular carcinoma in patients awaiting 
liver transplantation: pathology of Lie explain livers and 
clinical outcome. Cancer Biother Radiopharm 20:209-214 



Research .iiiJ lr 11 T LI : c I'iifi 



n Oncology Embolorherapy 



32. Laurent A, BeaujeuxR.et al. (1996] Trisacryl gelatin micro- 
spheres for iherapeiilic emboiizo noji, 1: development and 
in vitro evaluation. AINU Am I N'euroradiol 17:533-540 

33. Laurent A, Wassef M, et al. (2004] Location of vessel occlu- 
sion of call prs ted iris-acrvl gelatin jis icrospheres for tumor 
and arteriovenous [i:..i 1:'.:t"it..i I . on embolization. ] Vase 
Interv Radiol 15:491-496 

34.Li C, Yang DJ, et al. (1994] Formation and characterization 
■.'if espial .n-]oaded p'":y-:beiizv| l-g.utam.-.le: n 1 ..-.": ■:-'-ph.^r^- 
for chem oembolizat ion. P harm Res 11:1792-1799 

35.LiX, FengGS, et al. (2004) Expression of plasma vascular 
endothelial growth factor m patients wit:; hepatocellular 
carcinoma and effect of iranscatheier arterial chemeem- 
bolizatien therapy on plasma v;i-fcii I:".:' ef.doihel.al growth 
factor level World I Gastroenterol 10:2878-2832 

36.Link DP, Strandberg ]D, et al. (1996) Histopathologic 
appearance of arterial occlusion? w.i;-. hydrogel and poly- 
vinyl alcohol embolic material in domestic swine. 1 Vase 
Interv Radiol 7:897-905 

37.Llovet ]M (2005] Updated treatment approach to hepato- 
cellular carcinoma. 1 Claslreemere-. -Lici? 

38.Llovet JM, Real MI, et al. (2002) Arterial embolisation or 
chemoembolisatien versus symptomatic treatment in 
patients with unresectable hepatocellular carcinoma: a 
randomised controlled trial. Lancet 359:1734-1739 

39. Lo CM.Ngan H, et al. (2002) Randomized controlled trial of 
rransarter.al lipiodo. cheme-embolizaiie-ii tor unresectah.e 
hepatocellular carcinoma. Hepatology 35:1164-1171 

40.Loewe C, Cejna M, et al. (2002) Arterial embolization of 
unresectao-.e hepatocellular carcinoma with use of cyano- 
acrylate and lipiodol. I Vase lnie:v Radiol 13:61-69 

41 . Loewe C, Schindl M, et a I. ■: J333) Permanent transarterial 
embolization o: neuroendocrine metastases of the liver 
using yyrme-acrylate and 1 ip; ocel: assessment of mid- and 
long-term results. Am I Roentgenol 180:1379-1384 

42.MainiCL,ScelsaMG,etal.(1996)Superselectiveintra-arte- 
rial raciic metabolic therapy with 1-131 lipiodol in hepato- 
cellular carcinoma. Clin Nucl Med 21:221 

43. McNair A.V! .:•'-:■: Using cvdrogel pe:;.' me is for drug deliv- 
ery. Med Device Technol 7:16-22 

44.Mondazzi L, Bottelli R, et al. (1994) Transarterial oily che- 
mo em boh z al \.:n tor the treatment of hepatocellular carci- 
noma: a multivariate analysis of prognostic factors. Hepa- 
tology 19:1115-1123 

45.Nakakuma K, Tashiro S, et al. (1983) Studies on antican- 
cer treatment with an oily anticancer drug injected into 
the l.galed feeding hepatic artery for liver cancer. Cancer 
52:2193-2200 

46.Nakao N.Uchida H, et al. (1992) Effectiveness of Lipiodol 
in transcatheter anerial emraohzalien of hepatoceliu.ar 
carcinoma. Cancer Chemother Pharmacol 31[Suppl]: 
S72-76 

47. National Cancer Institute (2002 i C h em o embolization 
ano Revacizumab in treainig patients with liver cancer 
that cannot be removed with surgery. XCI trials daiabase 
(record nrst received November 12,2002) 

48. National Cancer Institute ■: 2033! Bevacizumab in treating 
patients with uiiresectao.e non metastatic .iver cancer. NCI 
trials database i record first received March 6, 2003) 

49.Nijsen IF, Seppenwoolde JH, et al. (2004) Liver tumors: MR 
i mailing ■. ■!" it..: ..■..'i.:v h o I m . u m microspheres - phantom 
and rabbit study. Radiology 231:491-499 

50.Novell JR. Dusheiko G, et al. (1991) Selective regional che- 



moiherapv of un resectable hecatic I urn on is using I. r :■.:■."'■.. 
HPB Surg 4:223-234; discussion 234-236 
Sl.Okamura 1, Kawai S. et a I. ! I 992 ) Prospective and random- 
ized clinical trial for the Ireatmeiit ot hepatocellular carci- 
noma - a comparison of L-TAE with l-'armoi tioicin and L- 
TAH with Adriamvcin (second cooperalive study). Cancer 
Chemother Pharmacol (Historical Archive) 31:320 

52. OsugaK.KhankanAA, etal (2002) Transarterial emboliza- 
tion for large hepatocel hilar carcinoma with use of stipe ra- 
bsorbenl polymer microspheres: imtia. experience. I Vase 
Interv Radiol 13:929-934 

53. Park BH, Lee JH, et al. (2005) Vascular administration of 
adenoviral vecior seul-.ed in absorbable ge.atii; sponge par- 
tic, es . ."' : l') ;■ :■..■.■ uigs tile trans gene expression in itepalo- 
cytes. Cancer Gene Ther 12:116-1121 

54. Peppas NA. Huang V, et al. icTiaT:; Physic o chemical founda- 
tions ano structural design o: hydro-gels in medicine and 
biology. Annu Rev Biomed Eng 2: 9-29 

55. Peter B, Franz P, et al. (1998) Arterial hepatic emboliza- 
tion of unresectable hepatocellular carcinoma using a cya- 
noacrylate/lipicdol mixture. Cardiovasc Intervem Radio! 
21:214 

56.Poon RT, Ng IO, et al. (2001) Serum vascular endothelial 
growth fiic lor predicts venous invasion in hepatocellular 
carcinoma: a prospective study. Ann Surg 233:227-235 

57.Poon RT, Ho JW, et al. (2004) Prognostic significance of 
serum vascular endothelial growth faclor am; encostatin 
in patients with hepatocellular carcinoma. Br J Surg 
91:1354-1360 

58. Ramsey DE, Kernagis LY, et al. (2002 ) Chemoembolization 
o: hepatocellular carcinoma. ■ Vase Interv Radiol i 3:2! 15- 



59. Rand T, Loewe C, et al (2005) Arterial embolization of 

unresectable hepatocellular care. no-ma witi: use of micro- 
spheres, lipiodol. and eyanoacrylate. Cardiovasc Intervem 
Radiol 28:313-318 
60.Raoul JL, Bourguet P, et al. (1988) Hepatic artery injec- 
tion of I -1 31 -labeled lipiodol. Part I. Riodisi rib alien study 
results in patients with hepatocellular carcinoma and liver 



61.Raoul JL, Guyader I 1 , et a I. ! I 997! Prospective randomized 
trial of chemoembolization versus inlra-anerial injection 
of ! 31 l-labeled-iodized oil in the Ireatment of hepatocel- 
lular carcinoma. Hepatology 26:1156-1161 

62.Reid T, Galanis H, ei a I. (2001 ) Imra-arierial administration 
of a replication-selective adenovirus idl].-20! in patients 
with colorectal carcinoma metastatic to the liver: a phase I 
trial. Gene Ther 8:1618-1626 

63. Reid T, Galanis E, et al. (2002] Hepatic arterial infusion of a 
replication-seleclive oncolytic adenovirus idll ?20): phase 
II viral, immune. ogle, and clinical endpomts. Cancer Res 
62:6070-6079 

64. Roche A, Girish B, et al. (2003) Trans-catheter arterial che- 
meemee.ifaiion as firsi-Lne treatment lor itepatic metas- 
tases from endocrine tumors. European Radio.ogy 1 3:1 3d 

65. Siskin GP, Dowling K, et al. (2003) Pathologic evaluation 
of a spherical polyvinyl alcohol embolic agent in a porcine 
renal model. I Vase Interv Radiol 14:89-98 

66. Spenlehauer G, Veillard M, et aL (1986) Formation and 
characterization o- eisplatin loaded poly: d.l-laclide! micro- 
spheres for chemoemraclizalicn. 1 Pharm Sci 75:750-755 

67. Stuart K (2 003 i ChemoemO't-lizanon in the manage mem o: 
liver tumors. Oncologist 8:425-437 



E. Liapi and J.-R H. Geschwind 



68.Tadavarthy SM, Knight L, et al. (1974) Therapeutic trans- 
catheter arterial enibc'kz;:! ion. Radiology 112:13-16 

69.TancrediT, McCuskey PA, et al. (1999) Changes in rat liver 
niiciocirculation after expei .mental hepatic arle::.il embo- 
li nation: comparison ooJifrereni embolic agents. Radiology 
211:177-181 

70.Vallee ]N, Lo D, et al. (2003) In vitro study of the com- 
pal.'n.ily :■!' ins-acryl gelatin microspheies ivhh various 
c-.emnlhi.ra a -tic agents. I Vase Inleiv Radiol 14:t>2 1-628 

71. VhrI I 'J, Wetter A, et al. (2005) Treatment of unresectable 
k*'.i; — i-tastjsis with transpulit'.onaiy chemoeiiioo.izaiioii: 
prehuman . ■ aei aence. Radiology 234:917-922 

72. Winkelbaucr l-W, Niederle B, et al. (1995) Hepatic artery 
e*-r>i: oihernpy of hepatic metastases from carcinoid 
tumors value of using a mixture of cyanoaciylate and 
ethiodized olL AJR Am J Roentgenol 165:323-327 

73. WVnitvhaULT hW, Nierderle B,et al. (1995) Malignant insu- 
l;m;:r.a. per:-., aent hepatic anery embolization of liver 

preliminary results. Cardiovasc Intervent 
8:353 359 



74. YamamotoT, Hayakawa K, et al. (2003} Transcatheter arte- 
ria' embolization asing po.y-L-lactic acid microspheres. 
Radiat Med 21:150-154 

75.Yoo HS, Park CH, et al. (1989) Radioiodinated fatty acid 
esters in the managemenl of hepatocellular carcinoma: 
preliminary findings, dancer C Item ol her Pharmacol 
23[Suppl]: S54-58 

76.Yoo HS, Park CH, et aL (1994) Small hepatocellular car- 
cino:'.;a: high dose inteinal radiation therapy with siioer- 
selective mira-arterial injection of 1-1 31 -labeled Lipiodol. 
Cancer Chemother Pharmacol 33: S128 

77. Yoon CJ.Chung (W.et al.(2004) Transcatheter arterial embo- 
lization with 188Rhenium-HDD -labeled iodized oil in rabbit 
VX2 liver tumor. J Vase Interv Radiol 15:1121-1128 

78. Yu J, Hafeli UO, et al. (2003) 90Y-oxine-ethiodol, a potential 
radiopharmaceutical Ma' The treatment of aver cancer. Appl 
Radiat Jsot 58:567-573 

79. Zielhuis SW, Nijsen JF, et al. (2005) Surface characteristics 
■ji ho.miiim-loaded polv:-. actio acidi microspheres. Hio- 
materials 26:925 



External Carotid 



18 Technical and Anatomical Considerations 
of the External Carotid System 



Paula Klurfan and Seon Kyu Lee 



EGA Anatomy 235 

r: in h' vol; eiirnJ 1'eivlopmeni 2 35 

External Carotid System 237 

Technical Aspects or' Hrad and Neck 

E in 1x0 iz.;t ion 240 

Therapeutic Techniques and Materials 241 

Iviedicaiion 245 

References 245 



The external carotid system (ECS) is a key arterial 
supply for the craniofacial and neck regions. Even 
though the internal carotid artery, the thyrocervi- 
cal, costocervical, and vertebral arteries are also 
supplying these territories, this chapter will focus 
on the anatomical aspects of the ECS and the tech- 
nical implications of the endovascular management 
of these regions. 



18.1 

ECA Anatomy 

The external carotid artery (ECA) is in general the 
smaller branch of the two terminal arteries of the 
common carotid artery. The site of the carotid bifur- 
cation is variable; however, between the C4-C5 and 
the C3-C4 is the most common levels. Despite its 
great anatomical variability, this artery is classically 
described as having eight main branches. The main 



P. Klumjam, MD 

lnterveiiiioiii.il Neuroradiology Conical relkuv. Department 
of Medical Imaging, University o'i Toronto. Toro:i;o Western 
Hospital, University Health Network, Suite 3MC-429, #399 
Bathurst Street, Toronto, Ontario, M5T 2S8, Canada 
S. K. Lee, MD, PhD 

it Professor, Staff Interventional anc. Diagnosis Neil re- 
gion of Neuroradiology, PeparTiiient of Medi- 
cal l:r: aging. University o: Toro:r:o, Toronto Western Hospital, 
University Health Network, Suite 3MC-429, #399 Bathurst 
Street, Toronto, Ontario, M5T 2S8, Canada 



trunk of the ECA de< 
branches to the tongue, deep face, and neck. The 
arterial termination is located at the level of the 
parotid gland, where it is divided into the super- 
ficial temporal artery and the internal maxillary 
artery. Initially the ECA lies anterior and medial 
to the internal carotid artery (ICA), then it courses 
posterolateral^/ as it ascends in the carotid sheath in 
front of the internal jugular vein. During its cervical 
course it is covered by the sternomastoid muscle and 
crossed by the hypoglossal nerve, lying anterolater- 
al^ to the vagus nerve. 



18.2 

Embryological Development 

The wide variety in the anatomical disposition of the 
arterial tree of the head and the neck is explained 
by the embryological development of the vessels in 
these anatomical areas. The specific supply to every 
territory is related to a general hemodynamic bal- 
ance in the whole region. This relationship is estab- 
lished between the territory and several potential 
nutrient vessels. 

During ontogenesis, the arterial supply to every 
territory will be the result of this hemodynamic 
balance, achieved by anastomosis, annexation, and 
involution of blood vessels. 

Initially, at early embryonic stages, the ventral 
and dorsal aortas communicate by a certain number 
of arterial bridges, the aortic arches (1 to 4 in the 
craniocaudal direction). Other embryonic arter- 
ies are the primitive maxillary artery, dorsal oph- 
thalmic artery, ventral ophthalmic artery, anterior 
cerebral artery and the longitudinal neural arteries 
(Fig. 18.1). 

During the subsequent stages, some of these 
arteries undergo modifications through regres- 
sion in the regions of the ventral ophthalmic artery, 
dorsal aorta and the ventral portion of the first two 
aortic arches. 



P. K.urf.tn .tad L=. K.Lee 




Fig. 18.1. EGA embrvontc develi.piuenl at early stage. J, pro- 
atlantai artery; J, hypc-glo^a! artery; .!. third aortic arch; 4, 
second aortic arch; 5, first aortic arch 



The dorsal aorta will give rise to the third aortic 
arch which, subsequently, will reach the remnant of 
the ventral pharyngeal artery, becoming the exter- 
nal carotid artery. As the proximal dorsal aorta 
involutes, the ventral aorta becomes the definitive 
common carotid artery dividing into a primitive 
internal carotid artery and external carotid artery 
(Fig. 18.2). 

The original ventral pharyngeal artery turns 
into the facio-lingual system, while the hyoid artery 
and the stapedial artery will evolve to become the 
internal maxillary artery and the middle meningeal 
artery (Figs. 18.3, 18.4) 

The caroticotympanic artery originates from the 
proximal segment of the hyostapedial trunk, while 
the inferolateral trunk (ILT) is a remnant of the dorsal 



ophthalmic artery. The definitive ophthalmic artery 
originates from the primitive ophthalmic artery. 

Some implications of this vascular development 
rely on the fact that every vascular segment lies 
between the origin of two embryonic vessels, and 
therefore they may be the point of entry of vascular 
rerouting in cases of segmental agenesis of part of 
the ICA proximal to the embryonic vessel: rerout- 
ing via the hyoid artery through the ascending 
pharyngeal artery at a cervical ICA agenesis, otic 
or trigeminal artery in a cervical or petrous agen- 
esis or even primitive maxillary artery originating 
from the carotid siphon on the contralateral side. In 
combined cervical, petrous and cavernous agenesis, 
rerouting through a complex cavernous network 
belonging to the internal maxillary artery has been 
described. 

Anatomic variants result from errors inthe mul- 
tiple steps during the development of the; 
The hemodynamic balance of the intern 
maxillary and external carotid arteries ca 
very different anatomic variants. 

Two types of craniofacial branches aris 
external carotid artery: 

• The arteries that supply the muscular and teg- 
mental structures, arising on the whole from the 
external carotid artery. 

• The arteries that supply the peripheral cranio- 
encephalic system (cranial nerves), which evolve 
from internal carotid to external carotid arteries, 
like the maxillary system that arises originally 
from the stapedial artery. 



al carotid, 

e from the 




Fig. 18.3. Final appearance of the external carotid artery. 1, 
occipital artery; 2, ascending pharvageal artery; 3. inferior 
tympanic: artery; 4, internal carotid artery; 5, lingual artery; 
6, facial artery; 7, posterior auricular artery: S, superficial tem- 
poral artery; 9, petrous branch; 10, middle meningeal artery; 
! !, maxillary arterv: ;_', Iransverse facial artery 



Fig.18.2. EGA embryonic development at a later stage. I, 
occipital artery; 2, a scea da nt phaivagea! arterv: 3. fa cio I in glial 
artery; 4, maxiKoataadihr.lar arterv; 3, supraorbital artery; 6, 
internal carotid artery 



■t" :he !£xrei j : ,i I '.I.inxid Svst 




Fig. 18.4. Angiographic view of the rxiernal carotid artery. I, 
fadolingual trunk; 2, c>ll ipital aneiv; .!, miernul maxillary 
artery; 4, superficial temporal anery; :■, middle meningeal 



involved i 



: of the craniofacial supply is the one 
i the vascularization of the central ner- 

II, and arises on t lie whole from the ICA. 



the lacrimal variant, the orbital supply is limited to 
an anastomosis across the superior orbital fissure, 
while in the meningolacrimal variant; the middle 
meningeal artery is responsible for the supply of a 
portion of the intraorbital territory. 

Other branches of the maxillary artery to the 
orbit include the anterior deep temporal, the infra- 
orbital and the sphenopalatine arteries. These arter- 
ies correspond to remnants of vessels arising form 
the infraorbital artery of the vertebrates, witch gives 
rise to the orbital artery. 

There are multiple anastomoses between the 
intraorbital branches with the external carotid 
system only supplying the periorbital region. This 
includes the internal maxillary, superficial tempo- 
ral arteries and the facial system. 

Supply to the cavernous sinus region may arise 
from the ECA. Arterial branches arise from the 
different segments of the cavernous carotid artery 
and course medially, laterally and in the direction 
of the posterior cranial fossa. These branches will 
anastomose with branches of the external carotid 
artery, which will allow a functional approach to 
this region. The ILT always anastomoses with the 
artery of the foramen rotundum, the middle menin- 
geal artery and the accessory meningeal artery. It is 
also called the inferolateral trunk of the c 



18.3 

External Carotid System 



■ ECA 



It is important to undei 
branches are in a hem 
internal maxillary systen 
of branches, whether theii 
extracranial. 

The ophthalmic artery ; 
supraorbital branch of the stapedial artery win 
later becomes the middle meningeal artery. In addi- 
tion, I he orbital artery will also anastomose with the 
primitive (and later definitive) ophthalmic artery, 
from the supracavernous internal carotid artery. 
Because of the different variants in the involution 
of the proximal segment of the orbital or ophthal- 
mic arteries, the supply to the orbit will finally arise 
exclusively from the ICA, the stapedi 
or both. 

The embryologic hemodynamics i 
to predict the arterial variants that i 



itand tha 
idynamic balance. The 
consists of two groups 
course is intracranial or 

s originally from the 



system (ECA) 



lakes itpossible 
an be observed 
n the orbital region. The role of the middle menin- 
geal artery in supplying the orbit can be seen to vary 
from one individual to another and from one side 
to the other in the same individual (Fig. 18.5a,b). In 



The significance of these various arrangements 
lies in the fact that the anastomoses at the cavernous 
segment of the internal carotid artery constitutes 
the most common pathway of reestablishing blood 
supply in acquired occlusions of the internal carotid 
artery. The acquired constraint re-orients the hemo- 
dynamic balance in order to support and preserve 
the territory distal to the acquired occlusion. 

Embryologically, the dorsal ophthalmic artery, 
the stapedial artery, the trigeminal artery and the 
primitive maxillary artery are involved in this anas- 
tomosis. 

The primitive maxillary artery originates from 
the medial surface of the C5 portion of the carotid 
siphon. It supplies the posterior hypophysis where 
it anastomoses with its counterpart on the other 
side. It may arise from a common trunk with the tri- 
geminal artery and their common remnant is then 
a single artery that arises from C5 and gives off all 



the meningeal, hypophyseal and 
of this region. This variant seems t 
The primitive maxillary artery ah 
meningeal branch for the dorsum s 
artery of the clivus). This 



al branches 
3 be rather rare. 
) gives rise to a 
:llae (the medial 
lastomoses with 



iunterpart on the contralateral side and inferi- 



P. K.LirL-ii .-:'J J. K. L?t 




Fig. 18.5 a-c. Anatomical variation. Orbit suppl; 
ningeal artery (arrow) a. On 
of left EGA angiography, choroid a I blush is clearly shown 
[arrow) b. Intern.il carotid artery angiography demonstrates 
;urysm,but no ophthalmic artery 
lalized from the ICA c 



orly with the clival branch of the hypoglossal branch 
of the ascending pharyngeal artery. The persistence 
or unusual origin of the primitive maxillary artery 
may be demonstrated in certain variants. For exam- 
ple, when there is a cervical internal carotid artery 
agenesis, the internal carotid artery may arise from 
the contralateral internal carotid artery through a 
trans-sellar anastomosis of the persistence primitive 
maxillary artery system. The contralateral intracav- 
ernous origin of the internal carotid artery can be 
developed through the embryological remnants of 
the primitive max il larva ileiy that include posterior- 
inferior hypophyseal artery (anterior to the clivus} 
and medial clival artery (posterior to the clivus). 



Other ICA branches arising from the C5 segments 
are the posteroinferior hypophyseal artery, the lat- 
eral artery of the clivus and the recurrent artery of 
the foramen lacerum. These are constant vessels that 
are anastomosed with the external carotid system 
through the ascending pharyngeal artery and in 
some cases with middle meningeal artery branches. 

The ECA is mainly responsible for supplying the 
supratentorial dura which covers the convexity. The 
anterior ethmoidal artery or the frontal branch of 
the middle meningeal artery supplies anterior aspect 
of dura mater. These arteries are the main suppliers 
for the anterior parietal regional dura of the con- 
vexity, but the blood supply may also come from the 



Technical and A 



is of the External Carotid Svst 



ophthalmic or intraorbital lacrimal artery which 
reaches its territory passing through the superior 
orbital fissure. The parieto-occipital trunk of the 
middle meningeal artery supplies the meninges of 
this region and the petrosquamosal trunk courses 
in the groove of the petrous and squamous portions 
of the temporal bone. These vessels are branches of 
the middle meningeal artery reaching the midline 
where they participate in the supply to the superior 
sagittal sinus and give descending branches to the 
fa lx cerebri. 

Anastomoses to the cortical pial (cerebral) arter- 
ies from these dural arteries are rare and are mainly 
found in occlusive diseases of the ICA. 

The extracranial branches of the maxillary 
artery include arteries that supply the nasal cavity, 
the choana and the nasal part of the pharynx and 
its walls. The blood supply of this region is particu- 
larly rich and presents multiple anastomoses to vital 
structures. 

The accessory meningeal artery is the main 
branch of the extracranial middle meningeal artery 
and anastomoses with the mandibular artery rem- 
nant of the ICA and the superior pharyngeal branch 
of the ascending pharyngeal artery. The accessory 
meningeal artery anastomoses with the Eustachian 
tube meatus arterial branch of the ascending pha- 
ryngeal artery and the ptervgovagntal arteryinfero- 
medially, and with the descending and/or ascending 
palatine arteries interiorly. This variety of territories 
supplied by the accessory meningeal artery makes 
it potentially responsible for the arterial supply for 
numerous lesions arising in this region. 

The pterygovaginal and vidian arteries are also 
supplying these territories with multiple anastomo- 
ses. The pterygovaginal artery anastomoses with the 
accessory meningeal, ascending pharyngeal and the 
mandibular branch of the ICA when it is present. 
The vidian artery is rarely visible during the maxil- 
lary artery angiography, probably due to the bone 
density through which it courses. The vidian artery 
may anastomose with a corresponding branch of the 
petrous segment of the internal carotid artery. 

The sphenopalatine artery, the terminal branch of 
the internal maxillary artery enters the nasal cavity 
where it is divided into a septal, medial branch and 
a lateral branch that supplies the conchae. These two 
arteries have a distinctive appearance on the angio- 
graphic views. These branches usually anastomose 
with the anterior and posterior ethmoidal arteries, 
which arise from the ophthalmic artery system, at 
the anterior and posterior ethmoidal cells, and even- 
tually connects the external and internal carotid 



systems. Other anastomoses are the septal branch 
of the superior labial artery (medially} and later- 
ally with the alar arteries, also branches of the facial 
artery. There is also a septal anastomosis of the ante- 
rior branch of the greater palatine artery with the 
posterior ethmoidal artery and the olfactory artery 
(a branch of the anterior cerebral artery). 

The upper arch of the mouth and the mandibii- 
lozygomatic area branches supply the maxilloman- 
dibular region. This system provides connections 
between superficial (cutaneous) and deep structures 
(bone and mucosa). 

The pharyngo-occipital system consist of an 
occipital artery, witch supplies the cutaneomuscu- 
lar elements and an ascending pharyngeal artery 
that is responsible for the meningeal and neural 
territory. The latter vessel is a metameric artery 
that has numerous anastomoses with the ICA and 
its branches are in hemodynamic balance with the 
suboccipitocervical system. 

The caroticovertebral anastomoses can be found 
as the persistence of the embryonic segmental ves- 
sels in adult. The type II proatlantal artery corre- 
sponds to the second segmental artery. It arises from 
the future external carotid artery and courses poste- 
riorly to the second cervical vertebral canal, where it 
supplies C2. This embryonic artery regresses to give 
the C2 occipito-vertebral anastomosis. However, 
when it persists, it runs into the cervical canal from 
C2 to CI and penetrates the dura at the CI level, sim- 
ilar to the conventional vertebral artery. Additional 
variants can be seen, including the occipital artery 
origin of PICA at C2 that is corresponding to an 
equivalent of the radiculopial artery for the cord. 

Most often, the occipital artery arises from the 
external carotid artery, in some cases as a common 
trunk with the ascend ingpliarv ngea la itery. In other 
cases they may arise from the origin of the internal 
carotid artery. The occipital artery may also arise 
from the vertebral system via the branch of the first 
or the second vertebral body level or, more rarely, 
from the cervical arteries or from the vertebral 
artery at C3 level. 

The ascending pharyngeal artery arises posteri- 
orly from the inferior part of the external carotid 
artery. It may also arise from the occipital artery or 
from the origin of the internal carotid artery. More 
rarely both ascending pharyngeal artery and occipi- 
tal artery may arise from the ascending cervical 
artery. Three pharyngeal branches of the ascending 
pharyngeal aiterv are usually seen including infe- 
rior, middle and superior branches. They supply the 
of the naso- and 



P. K.LirL-ii .-:'J L=. K. Lee 



oropharynx. They anastomose on the midline with 
their counterparts and with the adjacent pharyngeal 
branches on the same side. The superior pharyn- 
geal (or Eustachian) branch reaches the Eustachian 
tube's meatus on its medial side, lateral to the pha- 
ryngeal recess. It anastomoses with the correspond- 
ing branches of the accessory meningeal and ptery- 
govaginal arteries as well as forming a more medial 
and superior anastomosis with the mandibular ves- 
tige of the first aortic arch from the petrous segment 
of the internal carotid artery. 

From the superior pharyngeal branch, the carotid 
branch arises to the carotid canal. This carotid 
branch ascends through the foramen lacerum and 
accompanies the internal carotid artery up to the 
cavernous sinus, where it anastomoses with the 
inferolateral trunk and with the recurrent artery of 
the foramen lacerum, arising from the C5 portion 
of the carotid siphon, which supplies the internal 
carotid artery wall and sympathetic nerve fibers. 

The inferior tympanic branch lias certain distinc- 
tive features. This artery accompanies the tympanic 
branch of the 9th cranial nerve in the inferior part 
of the tympanic cavity, where it usually divides into 
three branches. The ascending branch anastomoses 
with the petrosal branch of the middle meningeal 
artery, accompanying the major deep petrosal nerve. 
An anterior branch joins the caroticotympanic 
artery, following the neural anastomosis between 
the tympanic branch to the ninth cranial nerve 
and the pericarotid nervous plexus. The posterior 
branch that courses towards the facial canal, where 
it anastomoses with the stylomastoid artery. 

The neuromeningeal branch gives rise to hypo- 
glossal and jugular branches. It enters the hypoglos- 
sal canal and supplies the 12th cranial nerve and the 
meanings of the posterior cranial fossa, where it is 
in balance with the other arteries of this region. This 
artery gives off medially a descending branch which 
s with the vertebral artery at the third 
1 space. The jugular branch, which arises 
from the same neuromeningeal trunk as the hypo- 
glossal branch, enters the cranial cavity through the 
jugular foramen, where it supplies the ninth, tenth, 
and eleventh cranial nerves. 

In order to be able to perform endovascvilar 
embolization procedures in the external carotid 
artery it is important to understand the physiologi- 
cal characteristics of the blood flow of this particu- 

The normal hemodynamic characteristics of the 
ECA are the absence of diastolic forward flow. This 
represents an important risk during embolization 



procedure due the possibility of material reflux. 
Thus, it is highly recommended to perform the 
embolization procedure during the systolic phase 
with a small volume on each injection of embolic 
materials. 

Autoregulation mechanisms have been described 
to be more effective in the internal maxillary artery 
and the pharyngo-occipital system, whereas the 
facial artery has not shown to respond as effectively. 
Vasoconstriction as a response for hypertension or 
mechanical trauma is observed in large or medium 
sized arteries whereas small distal arteries seem to 
react with true regulatory mechanisms. 



18.4 

Technical Aspects of Head and Neck 

Embolization 

Preoperative medication is usually not needed, aside 
for anesthesia drugs or those used for other medical 
conditions. A urinary catheter is recommended for 
an accurate measurement of fluids output and will 
prevent any discomfort for the patient in prolonged 
procedures. 

General anesthesia is necessary in most of the 
endovascular procedures for head and neck condi- 
tions, particularly in the pediatric population, in 
ethanol injections {paint til procedures) and patients 
with lesions located near the airway where swelling 
may obstruct the airway. 

Where the lesions are located at or near the a ir.vay, 
and combined procedures are schedule (surgery) a 
prophylactic tracheostomy may be considered. 

Neuroleptic analgesia is a useful tool for diag- 
nostic angiography, when neurological monitoring 
throughout the procedure is necessary and when 
general anesthesia is contraindicated in the older or 
medically compromised patients. Usually a combi- 
nation of an analgesic (opiate derivates) with seda- 
tive drugs (benzodiazepines) is useful to allow the 
patient to tolerate the procedure. 

Conventional Seldinger technique is generally 
used for the majority or the procedures, and a femo- 
ral sheath with hemostatic valve (4 to 9 French) is 
placed into the femoral artery to allow the exchange 
of the catheters, reducing the trauma to the artery. 

Diagnostic procedures are performed with 4 to 5 
French diagnostic catheters: single (Berenstein) or 
double curve (Sidewinder) depending on the tortu- 
osity of the vessels. These catheters are used in com- 
bination with guidewires (0.035") (Fig. 18.6). 



is of the External Carotid Svstem 




Fig. 18.6. Diagnostic citlrieleis vcil; si in:'' lei Rerenstein), double 
curve i Sidewinder: jiii;". guide wire with curved tip 



Therapeutic Techniques and Materials 



A great variety of microcatheters, 
embolic agents and drugs are available for the man- 
agement of a number of indications and purposes. 
The success in the endovascular technique and the 
achievement of the treatment goals not only depend 
on the technical skills of the operator but also in the 
appropriate selection of the endovascular material, 
based on the knowledge and the experience of the 
interventional neuroradiologist. 

Microcatheters are manufactured in different 
sizes: 0.018", 0.014", 0.010" systems allow to navigate 
the arterial branches with different flexibility. These 
microcatheters are used in combination with 0.018" 
to 0.010" outer diameter microwires that gives the 
required support to the system as well as a torque 
control to direct the tip of the catheter. Most of these 
catheters are performed with polyethylene and allow 
shaping the catheter tip. This is usually performed by 
exposing the end of the microcatheter to the steam 
source. Some of these microcatheters are available 
with a pre-shape d tip with different angles. 

Flow guided microcatheters are a second type of 
microcatheters designed to navigate with the blood 
flow with a much smaller outer diameter (0.012" 
- 0.018") achieving a more distal selective catheter- 
ization. The development of these microcatheters 
has permitted interventional neuroradiologist to 
treat a more extensive variety of craniofacial and 
neck lesions that were beyond the reach of the endo- 
vascular techniques with the previous generation of 
microcatheters. 

Balloon catheters are divided into two main 
groups: those used for blood flow control and test 



occlusion, and those used as the embolic agent 
themselves, detachable balloons. 

Single lumen balloon catheters (Hyperform, 
Hyperglide) are available in 4-7 mm in diameter 
and 10 to 30 mm in length. To inflate this balloon, a 
microwire should pass the balloon and stay beyond 
the microcatheter. A 50% iodine contrast material is 
used for the balloon inflation. 

Serbinenko first designed and introduced the 
detachable balloon for the clinical usage in 1974. 
With the evolving designs and materials, they have 
been a good embolic device for single high flow fis- 
tulas of major arteries in the trauma or congenital 
types. These are made of latex or Silicone with an 
Inflation diameter of 4 to 35 mm and are provided 
with a radiopaque marker which permits the bal- 
loon location under fluoroscopy before the inflation 
of the balloon. Detachable balloons can be hand 
assembled and be used with high reliability for clos- 
ing the selected vessel at a precise location. They can 
be removed and changed if the position or size of the 
balloon is incorrect (Fig. 18.7a-e) before its detach- 
ment. Once the desired position is reached, gentle 
and continuous traction is applied on the delivery 
catheter until the detachment occurs. 

The selection of the embolic agents is determined 
by the goal of the procedure, vascular territory and 
type of lesion. Classically they are divided into solid 
or liquid agents. 

Among the solid agents, particles are precut 
agents used for mechanical blockage of a selected 
territory. Several materials and sizes are available 
depending on the result required. 

Absorbable particles consist mainly is Gelfoam 
(gelatine sponge) powder (40-60 um) or particles 
(any size), the use of autologous clot or Avitene has 
also been described. The occlusion achieved with 
this material is not permanent and vascularization 
is reconstituted in 7 to 21 days after the procedure. 
The Gelfoam can be easily cut, and placed in con- 
trast material to be injected through small lumen 
catheters. 

Gelfoam powder can be used for tumor preop- 
erative embolization, in highly vascularized lesion. 
This material is used also tor endovascular manage- 
ment of the epistaxis due to the fact that the embolic 
occlusion occurs at the capillary or precapillary 
level. The Gelfoam is mixed with contrast material 
and is injected under fluoroscopic monitoring with 
a 1-3 cc Luer-lock syringe held in horizontal posi- 

This embolic agent should be used carefully with 
a good positioning of the catheter and continuous 



V. [viiri'.in .nid L=. K.Lee 




Fig. I8.rn -e. Louisas: en: - , a need CT scans a, b in I J-ycLir-old boy with slovdv enlarging rnlsaiile mass legion involving [lie ^:"l 
check region cenionsli-.Uc prom men; vase nl Lit channels lei": pica uric:; l-.ir a ten : iirrpirs). Let': I Liters I cxlc: na! en rod d angiogram 
in early c and Lite arterial phase d demons tilled sing.c hole ti = : u J li li s comniunicadon •:'<iitl?ii , >] between proxan.d interna! 
maxiLai v a • ic: y L ! .::d adiactii; facia, vein. T:aii5aj \t: iai .ateriLi. single b.i !..:■■ .n detLiclinic::: w.is per: oi med at the fistulous 51 :e 
rescuing in iiiinitdi-.iic complete oblilei j\:- 'i: of ilie f:s:uL: and several coils were deposited :n trie proximal ex:ern.;i carotid 
artery for added protection. Po5;-c:i;boliza:io:i lef: lateral coin men carotid angiogram e demonstrated clo5iire oi" die AVF. No:e 
the c.151 of die 5 ingle bnlloon iiinoiisi and ilie discivpancv be: wee:: the grossly enl.-:gec proximal Ir.;nk 01" the external carol id 
artery versus the size of die inlei nal carotid artery 



fluoroscopic monitoring due to multiple anasto- 
mosis between the external carotid system and the 
intracerebral circulation ,i<; well as the cranial nerves 
arterial supply from the ECA branches. 

Gelfoam pellets are mainly used for preoperative 
hemostasis, however, they do not reach the tumoral 
capillary bed, and therefore no necrosis is observed 
in the treated territory. 

When you cannot reach to the target vessel (so 
called superselectivity) but a permanent agent must 
be used, a Gelfoam particle 1x2 or 1x3 mm or even 
larger sized particles can be used to occlude the 
proximal part of a normal vessel. This method will 
protect a normal vessel by preserving its distal part 



and should be able to change the hemodynamics of 
the involved territory so that embolic agents would 
only reach the pathological vessels. Later on, the 
absorption of the Gelfoam particle should restore 
the normal flow into these arteries. In some other 
cases, Gelfoam can be safely used as an occlusive 
agent. The^e include trail malic arterial hemor rhet- 
or traumatic aneurysms. 

Among the nonabsorbable embolic materials, 
Polyvinyl Alcohol particle (PVA) is one of the most 
widely used. This consists of a water-soluble, bio- 
compatible material made by a reaction of polyvi- 
nyl alcohol foam with formaldehyde. When it is 
moisturized, it expands its volume up to 20% more 



Ttvhllk";'.! ond 



.i.m] Coa^deri'.tions of :hr cxijjtuiI ■! ^ : ■: "_ [■,! ?vsi 



Chan its dry volume. PVA suspensions are available 
as dried particles measuring 140-250 to 590-1000 
microns in size. These particles are mixed with con- 
trast material. PVA has a high coefficient of friction 
and its injection may be difficult. For this reason, 
it is recommended to continuously mix the suspen- 
sion in between the material injection and to inter- 
mittent flush the system to prevent PVA deposits on 
the catheter hub. 

The PVA has been described to have an embolic 
effect not only by occluding distal small to medium 
size vessels but also by slowing the flow in the 
treated vessel producing stagnation and, therefore 
clot formation. Even though this material is known 
to be non-reabsorbable, recanalization occurs, 
mainly because of clot reabsorption and re-endo- 
thelialization of the PVA deposits on the vessel 
walls. PVA is a useful embolic material for preoper- 
ative devascularization for tumors, some vascular 
malformations such as dural arteriovenous fistulas 
or flow rerouting in preparation for IBCA or etha- 
nol injections. A catheter that was previously used 
for particle embolization should never be used for 
an angiogram of intracerebral arteries (ICA or Ver- 
tebral artery). 

NBCA (N-Butyl-2-Cyanoaciylate) is a vinyl 
monomer of the alkyl-2-cyanoacrikites. This mate- 
rial is the most widely used liquid embolic agent and 
is characterized by being non-reabsorbable, produc- 
ing almost immediate solidification when it reaches 
a polarized fluid such as saline and blood. Polymer- 
ization of the NBCA occurs when it combines with 
an ionic solution such as contrast material, saline, 
blood or endothelium producing an exothermal 
reaction in a few seconds. Polymer retardants are 
generally used to achieve the ideal setting time for a 
particular injection. Iodized Ethyl Esters (Lipiodol) 
are usually a good combination as a polymer retar- 
dant for the NBCA. It has been shown to maintain 
a good dilution retarding polymerization in 4 to 
8 seconds for every 0.2 to 0.5 cc in 1 cc of NBCA. The 
viscosity of this material may be a concern but usu- 
ally it can be injected in the smallest catheters used 
in interventional procedures. 

Due to the radio-opacity of the Iodized Ethyl 
Esters, it can be used with NBCA without the use 
of an opacifying agent unless the NBCA concen- 
tration is over 50% of the mixture. In those cases 
Tantalum powder is recommended using 1 gram 
for every 1 cc of NBCA. Before the injection of the 
NBCA mixture, a 5% dextrose solution should be 
used to rinse the catheter. This will prevent the 
NBCA to solidify before reaching the targeting tis- 



sues. Injection of NBCA must be done under con- 
tinuous fluoroscopic control after the superselec- 
tive placement of the microcatheter. Depending on 
the therapeutic goal, NBCA/retardant mixture will 
be prepared to achieve venous/capillary penetra- 
tion or arterial occlusion (endovascular ligation) 
(Fig. 18.8a-d). When a high flow vessel of fistula 
is to be treated, combined technique may be used 
with using liquid coils. 

Ethyl Alcohol (95% Ethanol) is an effective and 
aggressive liquid embolic neent. Its does not produce 
a mechanical occlusion of the vessels but produces 
an immediate tissue reaction due to the cytotoxic- 
ity on the blood and endothelium. The use of Ethyl 
Alcohol results in a systemic distribution but the use 
of up to 60 cc of alcohol in an adult is below the toxic 
blood concentration. Ethanol is an effective embolic 
material to induce necrosis and endothelial damage 
but may not be indicated for high flow conditions or 
when a mechanical occlusion is expected. The con- 
trol of the Ethanol during its injection is very poor 
especially its distribution in the vascular territory. 
Therefore, its use is generally limited to venous mal- 
formation and some limited territories of the ECA. 
In some cases it can be combined with the use of 
PVA particles or Gelfoam. Ethanol procedures must 
be performed under general anesthesia as it causes 
severe pain during the injections. In addition, spe- 
cial attention should be taken regarding the venous 
outflow of the malformation witch may involve the 
ophthalmic vein, cavernous sinus or the vertebral 
epidural plexus. After the ethanol injection the area 
indurates and swells, starting a few minutes after 
the infusion and lasting 3-7 days. Final result may 
be expected after 1 to 5 weeks. Local skin necrosis 
may be seen after but usually is limited and heals 
spontaneously. 

Other materials have an important role in the 
end ova s cu lar management of the ECA. Detachable or 
pushable coils represent a safe option when arterial 
occlusion is required: arterial ligation, hemorrhagic 
emergencies (carotid blowout), blood flow rerouting 
techniques or combined endovascular embolization 
with other embolic agents. Several types of coils are 
widely available. Detachable bare platinum coils 
(Fig. 18.9) allow very precise coil deployment, and 
can be removed if the size is incorrect or coil place- 
ment. Pushable coils are directly deployed into the 
vessel while fibered coils have filaments that induce 
stagnation and local thrombosis. The 2nd generation 
of coils have been designed with combined material 
that may induce inflammatory reaction in the sur- 
rounding vascular tissue (GDC Matrix® coils) or a 



P. K.LirL-ii .-:-J L=. K.Lee 




Fig. 18.8a-d. Selective occipital :;:'gLOg[aiii in lateriu view a ^emonsmues lugj'.-row scalp AVM which, was also supp.ieJ ov 
bi -.inches 01" ".he ipsilitte; j I ;;nd cont:aljtei-.il stipe; f-.chi! tempc-ial ,11 lenes .-[id com ia lateral occipi:al artery : 110 r slioii-ti). Follow- 
ing tiansa; .^: iai p.ntiiu embi'lizstjon wit:: glue iuid parties 0:' PVA into ti'.ese vessels a yeictit.mtotis approach wjs jerfoi me J 
b,cwith iiijeclioj) of glue (S09d NRCA/ 50% Lipiodol) resulting in complete obliteration of the AVM nidus as shown on the post 
embolization left externa! carotid angiogram d 




Fig. 185. Detachable coils are available in si 
3D coils may be used for vascular occlusio 



Technical and A 



is of the External Carotid Svst 



hydrophilic component that allow the coil to expand 
4 to 20 times its volume to achieve a better vascular 
occlusion (Hydrocoils®). 

The selection of the embolic material will vary 
depending on the vascular territory, the vascular or 
tumoral lesion, the therapeutic goal and the previ- 
ous experience of the interventional neuroradiolo- 
gist. 



18.6 
Medication 

Heparinization is usually recommended when coax- 
ial catheter assembly systems are used to prevent 
fibrin clot formation. Initial prothrombin time, 
partial thromboplastin time and activation coagu- 
lation time is recommended to use as a baseline to 
calculate the dose of the protamine sulfate dosage 
when heparinization is to be reversed. Complete 
heparinization of the patient can be achieved with 
a bolus dose (50 IU/Kg) and a maintenance infusion 
(500UI/Kg in 24 hours) that is initiated once the 
arterial access is obtained. 

Flushing solutions are prepared with heparin 
using 2000 IU/ 1000 ml of 2.5 D/W in children and 
4000 UI/ 1000 ml of 2.5 D/W. 

Corticosteroids are not generally used during the 
endovascular management of the ECS. Its use is lim- 
ited to very prolonged procedures, or when signifi- 
cant inflammation or swelling is expected such as 
maxillofacial alcohol injection. In these cases, dexa- 
methasone 10 mg is given intravenously (bolus) fol- 
lowed by a maintenance dose of 8 mg every 8 hours 
tor the tollowing 3 days. 

Catheter induced vascular spasm can be an unde- 
sirable event during endovascular procedures in the 
ECS and it is usually triggered by mechanical stim- 
ulus. Percutaneous administration of nitroglycerin 



(nitropaste) is a useful treatment without significant 
systemic reaction such as hypot 



References 

Allen WE, Kier EL Rothman SLG (1973) The maxillary 
artery - normal arteriography anatomy. Am ■ jicemgen.ol 
118:517-527 

Berenstein A, Grueo 1 ' l 19S 1 1 Convenient preparation of read y- 
lO-use polyvinyl a loo no. foam suspension :'.:■[■ empo.izauoi":. 
Radiology 145:846-850 

Berenstein A. Kreber C, Edwards ,H, Bank WO. Kricheff II, 
Cromwell L (1980) Complications of therapeutic transar- 
lenal embolization: cooperative sludy. AINR Am 1 Neuro- 
radiol 1:128 

Berenstein A, Kricheff II (1978) Therapeutic vascular occlu- 
sion. J Dermatol Surg Oncol 4:874-880 

Berenstein A, Kricheff II (1981) Neuroradiology interven- 
tional procedures. 'lemni Roentgenol i 6:79-94 

Berenstein A, Lasjaunias P, Kricheff I (1983 [Functional anat- 
omy of the facia; vasculature i:'. putlio.ogicid condilions 
and its therapeutic applications. A]\R 5:149-153 

Connors hWoiak ] 1 1 999) ]ii:et've:i:Lo:iaJ neuroradiology strat- 
egies and practical tech in cues. ;i a tinders, Philadelphia 

Countee TW, Vijayanathan (1979) External carotid artery in 
internal carotid artery occlusion: angiographic, therapeu- 
tic and prognostic considerations. Stroke 10:450-460 

Djindjian R, Meriand II ! I 97SjSuperse!ective arteriography 
of the externa! carotid artery Springer, herkn Heidelberg 
New York 

Kuru Y (1967) Meningeal bi a n.c lies of I lie ophthalmic artery. 
Acta Radiol 6:241-251 

Lasjaunias P (1986) The external carotid artery: functional 
anatomy. In: Taveras }\', Ferruci ]T iec.s : Radiology, chap 
99. Lippincott, Philadelphia 

Lasiaumas P, Berenstein A, Doyen f 1 i 1 "79 i Functional anat- 
omy of the facial artery. Radiology 1 33:631-638 

Lasiaumas P. Doyen I" 1 i 1 97? : The ascenc.mg pharyngeal a.t tery 
and the mood supply of the lower crania! nerves. I Neuro- 
radiol 5:287-301 

Lasjaunias P, Berenstein A, Ter Brugge K(eds) (2001) Surgical 
neuioangicgriipl'.y, clinical vnsctdar anatomy and varia- 
tions, vol 1. Springe:. Heihn l-leiclciberg New York 

OsborneA (ed) il994i I'ntgnosiic nenioriiJiology. Mosby, St 



19 Endovascular Management for Head and Neck 
Tumors 



Paula Klurfan and Seon Kyu Lee 



Nasopharyngeal Tumors 247 

[uvenile Angiofibroma 24S 

Classification 248 

Armiomii." Features 248 

oiiaj ms; rir-.iiu:'e:S 248 

Natural History 249 

Diagnosis 250 

Treatment 250 

Embolization Technique 250 

P:ir;ig;i:;gJomas 251 

Classification 251 

Pathology 251 

Imaging 252 

Treatment 252 

T'lyrolamigeal Tumors 254 

Craniofacial Tumors 254 

Miscellaneous Applications of Embolizatio 

Therapy in the Head and Neck 254 

Preoperative Embolization 254 

f jin M/.nngeraent 255 

Hemorrhagic Emergencies 255 

Cookbook 255 

References 256 



Head and neck tumors consist of primary tumors 
arising from various regional tissues including 
lymph nodes and metastatic lesions. Most of these 
lesions are highly vascularized due to both abun- 
dant vascularities of the head and neck region and 
their histological types. 

Magnetic resonance imaging (MR1) is the most 
useful imaging study for the initial evaluation of 
head and neck tumors. Computed tomography (CT) 
is also helpful in defining the anatomical disposi- 
tion of these lesions. In addition, the CT can pro- 



P. Klumam, MD 

Interventional Net: o:-: ad [.:■;■ >gv 'Iliiiii-.il FcKow, I'vpniimenl of 
Mediciii Imaging, Univer^itv of Toronto, Toronto We^et n Hos- 
pital, 399 Bathurst Street, Toronto, Ontario, Canada, M5T 2S8 
S. K. Lee, MD, PhD 

Assistant Professor a:'.d S i-.i ft' Neiu oj ;i.d :ologist. Department oi 
MedsCLii Imaging, Univei^itv of Toronto, Toronto Ues^et n Hos- 
pital, 399 Bathurst Street, Toronto, Ontario, Canada, M5T 2S8 



vide important complementary information such 
as the presence of calcifications and the extent of 
bony involvement. Diagnostic angiography is hardly 
used as a diagnostic purpose. In fact, its main role 
is a carrier for the endovascular procedures for head 
and neck tumor managements. Endovascular proce- 
dures for head and neck tumors consist of (1) selec- 
tive devascularization procedures of the feeding 
arteries such as transcatheter arterial embolization 
and (2) adjunctive embolotherapy including intra- 
arterial chemoembolization. Functional vascular 
embolization such as intra-arterial chemotherapy 
for malignant head and neck cancers to maximize 
local concentration with minimized toxic effect has 
also been studied, however, further investigations 
are necessary lor its clinical application. 



19.1 

Nasopharyngeal Tumors 

Juvenile angiofibroma (IAF) represents 0.5% of the 
head and neck tumors [1] and 15% of nonepithelial 
tumors of the nasal and paranasal cavities [2]. It is 
the most common benign tumor of the nasopharynx 
and is typically d iagnosed on adolescent males, with 
a peak age of 14-17 years. However, up to 20% of 
these tumors are diagnosed after the age of 20 [3]. 
The most frequent clinical presentations of juvenile 
angiofibroma are nasal obstruction and recurrent 
nose bleeding. These symptoms can be followed 
by sinusitis, otitis, hearing loss, or anosmia. Life- 
threatening massive nose bleeding can occur and 
is often difficult to control with nasal packing. In 
these cases, it requires urgent endovascular treat- 
ment after blood transfusion and establishment ol 
diagnosis. The growth of JAF seems to be influenced 
by hormonal activity, although tumor samples fail 
to show the presence of estrogen, progesterone, or 
androgen receptors. 

Tumors developing in the nasopharyngeal region 
in the adult population are most likely to be malig- 



P. K.LirL-ii .-:-J J. K. L?t 



nant. Infra-arterial embolization may play a signifi- 
cant role when performed prior Co diagnostic biopsy, 
radical excision or palliative treatment. 

The angiographic and therapeutic protocols 
e applicable to other hyper- 
i region. Percutaneous embo- 
i described to be effective in 
:e or recurrent epistaxis from 
is previously irradiated, 
icluding panijifins'lii 



described for JAFi 
vascular tumors of thi 
lization has also 
the treatment of 
nasopharynge; 
Benign 



roblastomas, esthesioneuroblastomas, hemangio- 
pericytomas, and hemangioendotheliomas (most of 
them are considered malignant) may be diagnosed 
in the nasopharyngeal region. These tumors are all 
generally highly vascularized and have a relatively 
benign course. The treatment strategy for these 
lesions is a radical surgery whenever possible and 
therefore they will also be benefited from presurgi- 
cal embolization. 

Extracranial meningiomas are very rare (1%). In 
some cases they may be located in the parapharyn- 
geal space even though most of the tumors within 
this location are benign salivary grand tumors or 
neurogenic tumors like schwannomas. 



19.2 

Juvenile Angiofibroma 

19.2.1 

Classification 

Several grading systems of JAF have been presented 
but theFisch classification [] is the most extensively 
used one. Fisch classifies JAF into four types. Fisch 
type 1 is when the tumor is limited to the nasophar- 
ynx and nasal cavity without bony erosion. Fisch 
type 2 defines a [AF that invades the pterygomaxil- 
lary fossa and the maxillary, ethmoid and parasellar 
region but remains lateral to the cavernous sinus. 
Type 3 is defined as JAF tumors that invade the 
infratemporal fossa, orbit and parasellar region but 
remain lateral to the cavernous sinus. Finally, the 
type 4 tumors are those that sh< 
of the cavernous sinus, the optic chi 
the pituitary toss;). 



19.2.2 

Anatomic Features 



JAF is a highly vascular and locally invasive tumor. 
It is usually originated from the superolateral aspect 
of the choana, near the spheno-palatine foramen, 
but may also arise more medially, near the vomer, 
from the pharyngeal roof where it involves the body 
of the sphenoid bone, or from the adjacent pterygoid 
plates. 



Regardles 
tumor has a significant v 
five activity. It may cause 
even though the JAF does 
sues directly. Attached to 
structures, it extends throi 
into the adjacent oper 



itological origin, this 
ascularity and prolifera- 
significant bone erosion, 
not invade the bone tis- 
the neighboring osseous 
igh the submucosal space 
:es. Macroscopically, JAF 
is reddish-gray or red purple in color and has a firm 
rubbery consistency with a tabulated shape. Multi- 
focal tumors have never been reported. 



19.2.3 

Imaging Features 

Imaging (CT, MRl) studies usually show the pres- 
ence of an expansible lobulated lesion located at the 
nasopharynx. Due to the local extension of the JAF, 
it can show significant bony erosion expanding into 
the surrounding nasopharyngeal cavities, maxillary 
sinus, and sphenoid sinus and infrequently to the 
anterior skull base and the orbit (extra- capsular 
and extraconal). MRI is an excellent complemen- 
tary study for JAF evaluation. Intracranial invasion 
and intradural tumor extension can be evaluated 
and certainly is a decisive factor to determine radi- 
cal treatment. MRI is also useful to differentiate 
tumor extension into the sinuses from sinusitis. 
The contrast enhanced MRI also shows extensive 
enhancement of tumor due to its hypervascularity 
(Fig. 19.1a). Contrast enhancement is essential for 
the JAF CT examination. Reviewing CT scans with 
both soft tissue and bone window setting is nec- 
essary to evaluate the extent of the tumor as well 
as bony erosions. Coronal and axial views provide 
good anatomical information regarding the rela- 
tionships among tumor, nasopharyngeal soft tis- 
sues and osseous structures of skull base. On CT, 
the JAF usually shows displacement and thinning 
of bony structures without definite bony destruc- 
tions considering the size of main mass (Fig. 19.1 b). 
These are useful radiologic findings to differentiate 
the JAF from other malignant tumors in children 



r Management for Head and Neck Tumors 




Fig.19.la-f. TAF:An 18-year-old male presenting wfth epuftaxts. Trie MR] and the CT scan shows orbital 

tempor.U iossM extension of tumor ivi:h t^gnilioan- Pony erosion :.a,b:. Aneiogiam sfiovo; viimof.U ir>l:;sli arising l~r.:- ni Ih.e 
intern.:! maxillary .irtery, ;he facial aioc-rv and the ascrndiisg pharyngeal .irtery li-fj. Preoperative emPo.izaiion with panicles 
wjf perfoimed afler s tie." nvc- cailieteiiz;'.! ion of tiie feeding aneiies. Courtesy of Dr. K. terBrugge 



such as rhabdomyosarcoma that typically destroys 
adjacent bony structures. 

On angiography, the JAF presents with a dense 
tumor blush (Fig. 19.1c}, and this is highly accurate 
to delimit tumor extension. Although it has intense 
hypervascularity, angiosivipluc iiruiings of arterio- 
venous shunting or early venous drainage have not 
been reported. Feeding arteries of JAF are mainly 
external carotid artery (ECA) branches such as 
distal internal maxillary branches, accessory men- 
ingeal artery, superior pharyngeal division of the 
ascending pharyngeal artery and the ascending 
palatine artery. 

Internal carol id artery (ICA) may supply the JAF 
without having intracranial portion of the tumor. 
However, if angiographic tumor blush is located 
above the skull base on AP and/or lateral views and 
has vascular supply from the ICA branches and/or 
from ascending pharyngeal and/or proximal inter- 
nal maxillary arteries, it might represent intra- 
cranial extension of the tumor. However, the sub- 
arachnoid space extension of tumor is exclusively 



supplied by the ICA branches. The evaluation of the 
venous phase of the internal carotid angiography is 
an essential part of diagnostic angiography, since 
it will demonstrate the patency of the cavernous 
sinuses and adjacent venous plexus those will affect 
surgical respectability of the tumor. 



19.2.4 
Natural History 

Osborn and Friedman suggested that repeated 
hemorrhages within the tumor could stimulate the 
formation of granulation tissue and a fibrous reac- 
tion (1). This would explain some cases present- 
ing spontaneous regression and tumors that tend 
to have a slower rate of growth after adolescence 
due to a higher proportion of fibrous tissue and 
less tendency to bleed. As the disease progresses, 
facial deformities, proptosis, blindness and cranial 
nerve palsy may occur. Thus, complete spontaneous 
regression of these tumors should not be expected 



P. K.LirL-ii .-:'J J. K. L?t 



and treatment should not be delayed due to the 
tumor's potential to behave aggressively. 

The recruitment of new vascular supply is proba- 
bly related to the production ofangiogenetic factors 
and it may explain the local i 
types of the JAF. 



19.2.5 
Diagnosis 

The diagnosis of JFA is mainly based on a careful 
clinical history and nasal endoscopic examination 
in addition to the imaging studies (CT and MRI). 
Biopsy to establish liistologic.il diagnosis is not 
indicated, and definitive diagnosis can be estab- 
lished by angiography, which certainly will play 
an important role in the patient management and 
treatment. 



19.2.6 
Treatment 



The objective of the treatment is complete : 
of the tumor. Surgical removal and radiation therapy 
are considered as the best therapeutic options. Con- 
troversy still exists as to how patients with recurrent 
or residual disease should be treated and how large 
tumors of the skull base should be managed. 

Surgical techniques have been mostly successful 
by using advancing endonasal techniques with the 
use of the microscope for limited mid face degloving 
procedures but in a small group of cases: transpala- 
tal, lateral rhinotomy and craniofacial approaches 
can be used. 

Tumoral resection ot I A ]- may be challenging and 
can be limited due to the risk of profuse intraop- 
erative bleeding and the size and extension of the 
tumor. 

To diminish these risk factors and to facilitate the 
surgical resection, preoperative intra-arterial embo- 
lization, radiation therapy and exogenous estrogens 
has been largely used. 

Preoperative exogenous estrogens have shown to 
produce positive effects on the bleeding rate. How- 
ever, the morbidity cause by the administration of 
such drugs to an adolescent male population is very 
high, with consequences on the gonadal develop- 
ment and function, which turns this treatment into 
an undesirable choice. 

Radiation therapy is known to produce partial 
tumoral mass reduction or at least a significant 



arrest of the tumor growth (80%), but functional 
morbidity and the high rate of long term secondary 
neoplasm induction place this treatment option in a 
secondary role. Thus, in general, radiation therapy 
for JNF is restricted to certain cases where intracra- 
nial extension prevents a complete resection of the 
lesion. Its effects are secondary to post radiation 
vasculitis and not the targeting of the cellular com- 
ponent. The use of radiation as a sole treatment has 
shown an overall rate of recurrence of 20%. 

Intra-arterial embolization has resulted in a help- 
ful and reliable technique in preparation for surgery, 
and currently is the preferred combined treatment. 
This procedure, when performed by an experienced 
team carries no significant morbidity or mortality. 
Reported complications or unsatisfactory results are 
likely related to insufficient training or knowledge 
or poor judgment during the procedure itself. 

Based on the classification of Fisch, most sur- 
geons in the international literature tend to operate 
on stages 1 and 2, some on stage 3 and few on stage 
4. The overall mortality of [FA is 3% and the rate of 
recurrence after surgery varies trom 12% to 35% and 
is likely owing to inadequate surgical removal. 

Radiation therapy should be reserved for bilateral 
cavernous sinus involvement and for the situation 
where adequate surgical or therapeutic angiographic 
teams are not available in a given geographic loca- 

The use of surgical tumor resection precede by 
intra-arterial embolization has shown strong evi- 
dence in terms of reducing the number of recur- 
rences and repeated recurrences as a consequence 
of a reduction of the tumoral size and intraopera- 
tive bleeding, and therefore a higher rate of complete 
surgical resection of the JFA. 



19.2.7 

Embolization Technique 

During preoperative angiography, major feeding 
vessels arising from the external carotid system are 
superselectively embolized and subsequent surgery 
can be scheduled within 12 to 48 hours after embo- 
lization. 

The goal of the endovascular procedure is todevas- 
cularize the tumor at the level of capillary bed, not 
just proximal occlusion. Arterial supply of the tumor 
mayvary depending on the site of the tumor and these 
vessels can only be moderately enlarged. The internal 
carotid artery supply to the tumor is usually the most 
important factor to limit the capacity of pre- operative 



Endovascular Man.'.gemear for Head and Neck Tumors 



embolization. Therefore it is recommended to begin 
the study by performing a contralateral injection of 
the ICA with cross compression in the Caldwell view 
followed by an ipsilateral ICA injection. This will 
show the tumor's ethmoidal, sphenoidal and middle 
cranial fossa extension. If the ICA branches can be 
purchased easily and can get a safe position for the 
embolization, there cannot be any restriction for the 
procedure. However, in general, it is technically chal- 
lenging to obtain a safe enough position to embolize 
JNF thru the ICA branches. 

Regarding the ECA branches embolization. Spe- 
cial precaution has to be taken to identify potential 
anastomosis between branches of the internal max- 
illary and ascending pharyngeal arteries and the 
intracranial or intraorbital arteries, and internal 
carotid artery supply to the tumor. 

For the last years, 150-250 urn in size polyvi- 
nyl alcohol (PVA) has been the material of choice 
to reach the tumor capillary bed during the endo- 
vascular procedure. Smaller particles (50 (am) have 
shown to pass through the capillary vessels reaching 
the lungs with no therapeutic impact. Embolization 
can be completed by the injection of Gelfoam pled- 
gets (3 mm in caliber and up to 1 cm in length) to 
achieve a transient devascularization of the region 
(ligation) and in add it ion facilitate the distal throm- 
bosis within the tumor itself. 

Strong evidence indicates that presurgical embo- 
lization facilitates surgical resection of JNF. Fur- 
thermore, either embolization alone or emboliza- 
tion associated with estrogens favors patients that 
present surgically unresectable tumors due to size 

The endovascular management may require the 
usage of other techniques such as injection of fluid 
materials such as NBCA or alcohol with flow con- 
trol thru the internal maxillary artery or ascending 
pharyngeal branches. Sacrifice of the ICA can be 
considered on an individual basis in patients with 
intra caver nous extension. 

In this last case neurologic examination as '.veil as 
a transitory carotid occlusion test should always be 
carried out before the procedure. 

The endovascular treatment results in a sig- 
nificant benefit for the ]AF treatment whether it is 
preoperative or palliative. Usually shrinkage of the 
tumor mass can be observed both radiological and 
clinically within 12 hours after the procedure and 
breathing is usually improved indicating the return 
of nasal patency. 



19.3 
Paragangliomas 



19.3.1 
Classification 



Paragangliomas, glomus tumors, chemodectomas, 
neurocristopathic tumors or nonchromaffin para- 
gangliomas are several names that have been given 
to the neuroendocrine neoplasms arising from the 
neural crest derivates. 

Thesetumorspresentwithawiderangeof locations: 
tympanic, jugular; carotid, vagal, laryngeal, nasopha- 
ryngeal and orbital. Tympanic and Jugular paragan- 
gliomas are classified as temporal paragangliomas. 

Branchial paragangliomas are found in a vari- 
ety of locations in the head or the neck with almost 
one half arising in the temporal bone. These are 
the most common tumors of the middle ear. Its fea- 
tures include multicentricity and frequent associa- 
tion with other neural crest tumors. Other frequent 
locations are: jugular, carotid, vagal, laryngeal, 
nasopharyngeal and orbital, but pure single local- 
ization is uncommon and paragangliomas are usu- 
ally found to extend to multiple regions. Almost all 
of the paragangliomas located in the head and neck 
develop from a pre-existing normal paraganglion. 

The clinical presentation of paragangliomas is 
related to the location of the tumor (mass, bruit, pain 
or cranial nerve palsy] and is usually progressive. 

The malignant potential of paraganglioma is 
reported as between 10% and 18% on the vagal, 
carotid and laryngeal locations while in the tem- 
poral area is about 3%. Spontaneous regression of 
paragangliomas has never been reported. It usually 
has a slow growing rate but in some cases can be 
rapid or associated with additional tumors in other 



19.3.2 
Pathology 

Depending on the location, the paraganglioma may 
be lobulated or oval in shape and histologically it 
consists of a capsulated, highly vascular stroma 
with a paraganglion structure and epithelial cells 
(Fig. 19. 2h). An irregular narrowing of the internal 
carotid artery can Lie seen in some cervical paragan- 
gliomas but is similar to tumoral encasement and is 
probably not specific. 

The ascending pharyngeal artery is a unique 
link be tween parasansliomas in various territories. 



P. K.LirL-ii .-:-J J. K. L?t 



The tympanic, jugular, vagal, carotid and laryngeal 
locations of paragangliomas are supplied by differ- 
ent branches of the ascending pharyngeal artery. 
The internal maxillary artery and the superior and 
inferior laryngeal arteries are also responsible for 
supplying paragangliomas in their respective ter- 



Diagnosis of a carotid cai 
performed by detecting expansive mass and bone 
destruction on the carotid canal on the CT scan 
associated with a narrowing of the intrapetrosal 
carotid artery at the angiography. 



19.3.3 
Imaging 

CT and MRI are important methods for the ini- 
tial evaluation of a paraganglioma. Even though 
the findings are usually not specific for this tumor 
and may mimic other nerve sheath neoplasms, 
these studies are highly effective to demonstrate the 
extension of the tumor into different regions like 
the carotid canal, the inner and middle ear, mas- 
toid process, posterior fossa or pterygoid muscles. 
(Fig. 19.2a,d,e) 

These studies should always be performed with and 
without contrast enhancement and a bone windowed 
as well as a soft tissue windowed evaluation of the 
CT scan of the lesion should be done. Direct coronal 
images are indispensable in the presence of cervical 
or skull base mass lesion and 1 to 3 mm sections are 
required for the petrous temporal bone evaluation. 
Due to the hypervascularity of the paragangliomas, 
intense homogeneous enhancement of these tumors 
occurs following the contrast administration. 

Superselective angiography still represents the 
most reliable study for pre treatment evaluation of 
paragangliomas. If the diagnosis is suspected, the 
angiogram should be performed before any biopsy 
attempt. (Fig. 19.2b,c,f,g) 

The topography of the lesion will always indi- 
cate the arterial supply to the tumor. This could 
consist of a single artery or multiple vascular sup- 
plies. Angiography findings are usually an intense 
tumoral blush, enlargement of the arterial feeders 
and rapid venous filling. Just like in JAF, it is recom- 
mended to perform the endovascular treatment at 
the time of the diagnostic angiogram, whenever it 

The principal arteries that should be studied 
include the ipsilateral vertebral artery, the internal 
carotid artery, the distal external carotid, the pos- 
terior auricular, the occipital arteries and bilateral 
ascending pharyngeal arteries. It is important to 
visualize the venous drainage pattern and recognize 
the presence of a venous thrombosis. 



19.3.4 
Treatment 

Interdisciplinary management has shown to be criti- 
cal for the optimal treatment of paragangliomas. 
Even though complete surgical excision has shown 
to be the treatment of choice, radiotherapy and 
endovascular embolization have become important 
therapeutic options for treating unresectable tumors 
and perform palliative treatment. Preoperative pro- 
cedures such as embolization to reduce the surgical 
procedure timing and the bleeding risk contem- 
plated in surgery have become an important thera- 
peutic tool for these tumors. Endovascular emboli- 
zation, when performed by an experienced operator, 
is a highly efficient and a low-risk method. 

The efficacy of radiotherapy is not clear. Several 
studies have shown a tumor control rate as different 
as 90% and 25%. The mechanism of treatment is due 
to its effect on the vascular component causing vas- 
cular arteritis and fibrosis rather than affecting the 
tumoral cells. Brain tissue necrosis is an undesirable 
side effect related with this method and is usually 
detectable by CT or MRI. 

Embolization of these tumors is usually per- 
formed with 200-350 microns in size polyvinyl 
alcohol (PVA). After particle injection, ligation of 
the arterial supply can be performed with Gelfoam 
strip injection. This will facilitate (he intratumoral 
thrombosis. Liquid embolic agents are usually 
reserved for palliative lesions and only when strict 
flow control can be achieved. 

As in any endovascular embolization of the cra- 
niofacial region, special regards must be taken 
towards the multiple anastomosis channels between 
the ICAand the orbit, middle meningeal or occipital 
arteries. As was previously described, the emboliza- 
tion of the ascending pharyngeal artery with fluid 
agent may induce lower cranial nerve palsy. 



Endovascular Management for Head and Neck Tumors 




Fig. 19.2a-h. Carol id body tumor lii-ti. i^T soan a shows the anatomioal relationship o: the aimoi wiih the oaroitd artery, 
donnas rumor typioally shows enhancement .-tie: oo:i- rast administration. 5eleo:ive ai'.gi' 'giaphv b,e shows aimor blush aris- 
ing l':'.::: siu.il] '-''. A bi anohes. "-!■■ iv.li-. r.:iti!,::r : d -h j Mk! shows liie tui )]■:■: .ooaiioi! in ; car. kinship wj::i Ihe petrous none and 
".he iugiLai bub d,e Seleoiive ai'.gfagiaphv snow? Limo-: plush sapo.ieu by :lic postal ior au: ioular artery i fi a no tlie a so en Ping 
oaa: vngeul ;.: :iriv :g:. baiholoeio e\ami:va:io:'. ih: dciv.onstjaies r.imo; tissue witli the ivpioah oapsulaled si: uor.ire and erpheahV. 
oells sui roui'.dro with a vasoularizcu slroma. Com tesy of Dr. K. terBrugge 



P. K.LirL-ii .-:'J J. K. L?t 



19.4 

Thyrolaryngeal Tumors 

Tumors located in the thyrolaryngeal reg 
usually malignant and highly vascularized. Even 
though cerebral angiography and endovascular 
embolization can offer a significant help for the 
management of these tumors, most of cases are not 
referred for either diagnostic or therapeutic angi- 
ography. 

P ret he rape u tic evaluation of the thyrolaryngeal 
tumors should include conventional radiograms, CT, 
ultrasound and radionuclide scanning. The highly 
vascularized tumors of this region are usually sup- 
plied by the superior and inferior thyroidal arteries 
and are the most likely involved in the endovascular 
treatment. These arteries are usually widely selected 
for endovascular embolization of other head and 
neck tumors due to the collateral supply to the lloor 
of the mouth and the carotid region. 

They even may be the only supply to the floor of 
the mouth when there is a proximal ligation of the 
linguo-facial system. 

Except for large tumors, the non-invasive tech- 
niques have shown little help in anatomical local- 
ization of parathyroid tumors, especially through 
the detection of secreting hormones at the neck 

Endovascular management techniques for these 
tumors include the superselective injection of 2 to 
30 ml of contrast material [] with a higher dose of 
iodine compared to the conventional dose for cere- 
bral angiography. The persistence of the tumoral 
stain indicates the local damage of the tissue, and 
can be noticed for hours or days. After the proce- 
dure, a reduction in the calcium level in the blood 
can be found as a result of the released hormone 
from the cellular granules. Even though a signifi- 
cant result can be obtained from this technique, 
the endovascular procedure should be considered 
incomplete without a surgical excision. Experience 
on particle embolization for parathyroid adenomas 
has shown only a temporary effect and recurrence 
is always noticed. No evidence has been reported on 
the use of selective injection of cytotoxic agents. 

Some of the vascularized tumors that have 
shown to be beneficed by a preoperative emboli- 
zation include: soft tissue and bony hemangiomas 
in adults, capillary hemangiomas in children and 
malignant synoviomas. 

As previously described, some para^anijliom:.^ 
may be presented in the pharyngeal and thyroidal 
regions and may require preoperative endovascu- 



lar embolization for complete excision. Particles of 
Liquid agents may be used as an only treatment for 
palliative management of these lesions. 



19.5 

Craniofacial Tumors 

Endovascular techniques certainly have a role in the 
management of craniofacial tumors. Most of these 
lesions are metastasis or primary malignant tumors. 
The main impact of the procedure is the necrotic 
changes in the tumor territory and this leads to a 
significant size reduction in its mass. Rarely in these 
cases, embolization is used as a single treatment, 
like recurrent or non-surgical lesions, and reaching 
as much of the area of the tumor with minimal side 
effects is the main goal. 

Some techniques can be used to maximize the 
effect of particle embolization, like rearrangement 
of the vascular supply to the lesion. For this tech- 
nique a lateralized tumor may be selectively embo- 
lized to give priority to a single feeder. The feasibil- 
ity of this technique varies depending on the tumor 
extent, accessibility of the vessels and potential vas- 
cular anastomosis. For example, either the ipsilat- 
eral ascending pharyngeal or the internal maxillary 
artery can be embolized with particles. The remain- 
ing feeder is deliberately left open. Repeated angio- 
gram after a few weeks will verify the redirection of 
the tumor supply, witch can now be used for a more 
aggressive treatment: liquid adhesive, powder, cyto- 
toxics, ethanol, loaded microcapsules, etc. Although 
little experience has been reported with these mate- 
rials, significant decrease in the tumor masses has 
been reported. 



Miscellaneous Applications of Embolization 
Therapy in the Head and Neck 

19.6.1 

Preoperative Embolization 

Embolization has been described for the preven- 
tion of hemorrhagic complications horn a planned 
tumoral biopsy. Particle embolization preserves 
the histoarchitecture and inmunohistoanalysis and 
therefore is a safe material for the preservation of 
the tissue for diagnostic purposes. 



r Management for Head and Neck Tumors 



19.6.2 

Pain Management 

Embolization for control of tumor related pain could 
be performed in selected cases where decongestion 
or tumoral mass reduction is thought to decompress 
the surrounding tissues, resulting in a local pain 
relief. 



19.6.3 

Hemorrhagic Emergencies 

Craniofacial and neck malignant tumors usually 
present with a highly aggressive local activity. Bone 
destruction and ulceration are not uncommon find- 
ings especially as a secondary effect of radiotherapy. 
In some cases, invasion of vascular structures may 
lead to a recurrent bleeding or severe hemorrhages. 



In these emergencies initial clinical management 
and manual externalcompression is usually required 
until the patient can be treated (Fig. 19.3a). Diagnos- 
tic angiography is a priority to determine the cause 
for the carotid 'Blow out' syndrome. Aggressive 
tumor invasion or pseudoaneurysm formation of 
the neck or nasopharynx vessels are usual angio- 
graphic findings (Fig. 19.3b-c). These are life-threat- 
ening situations and special consideration must be 
taken for the overall clinical status of the patient 
(shock management, blood replacement, etc.) Mild 
to moderate hemorrhagic event may be temporarily 
treated with particles in the tumor capillary bed. In 
some cases ligation of a terminal branch (nasophar- 
ynx) or even the external or internal carotid artery 
by endovascular means is required (Fig. 19.3). For 
these purposes different kinds of material such as 
detachable or pushable coils, detachable balloons or 
liquid adhesives are highly effective. 




ig.19.3a-c. Nasopharyngeal cancer wiih profuse bleeding ;rea:e; wiili covered s".eni ijonied^ 1 }. A 54-year-old female with 
licv.inced nasopbaivnge-.il ciuicei presented wi:h mtriicuble or:.', .mc n.isal bleeding. Kighl KA ang.-: gram ;i show? pseucoa:'- 
eurysm of disui] oerrosiu segiv.enl i.-i. 1 ivni. Covered sir: 1 .: is introduced and ,idf.:s:ed its posidon across tbe pse.:doane.iivs::i. 
b A ros:-s:e:'.: angiogram, e shows complele ■ irchisjon of pse::doaneurysm 



Cookbook: 


2. Femoral arterial sheath: 5 or 6 Fr, 11 cm. If the 
patient has a very tortuous abdominal aorta, you 




1. Anesthesia: General anesthesia preferred but the 


can use longer arterial sheaths (30 ~ 45 cm). 


tumor embolization can be performed under 




neuroleptic anesthesia if the patient is stable 


3. Diagnostic catheter: 4 or 5 Fr angled Glider 


and cooperative. In case of emergency such as 


(Boston Scientific®) or Berenstein (Cordis®) 


carotid blow-out syndrome or profuse and active 




nose bleeding, you may perform the procedure 


4. Guide wire: 0.035 or 0.038 inch Glider 


under the local anesthesia. 


(Terumo®) 



P. K.uri.;ii .;:'d L=. K.Lee 



5.Guiding catheter: 5 or 6 Fr Envoy (Cordis®) or 
Guider (Boston Scientific®) 90 cm 

6.Microcatheter: 
a.Prowler 14 (0.014 inch) or 18 (0.018 inch) 

microcatheter (Cordis®) for the particle 

embolization, 
b. Elite 1.5 (0.018 inch proximal and 0.011 inch 

distal end) or 1.8 (0.018 inch proximal and 

0.013 inch distal end} (Boston Scientific®) for 

the NBCA embolization. 

7. Microwire: 
a.Tfansend Ex 14 (0.014 inch) (Boston 

Scientific®) for the Prowler microcatheter 
b.Mirage (0.010 inch) (MTI®) for the Elite 

microcatheter 

8. Embolization procedure should always be per- 
formed under simultaneous subtracted fluoro- 
scopic control. 

9. Particle Embolization: 

a.A bottle of 150 ~ 250 urn-sized PVA particles 
mixed with 10~15 cc of contrast media 

b. Inject the particle mixture with lcc Luer-Lok 
syringe. 

c. Intermittent flushing of microcatheter with 
saline using another 1 or 3cc Luer-Lok syringe. 

d.If you have any resistance during the 
embolization or flushing, do not forcefully 
inject the PVA particle mixture or flushing 
saline but remove the microcatheter 
completely and use a new one if needed. 

10.NBCA (N-butyl cya no a cry late) embolization: 
a. Mix the NBCA with Lipiodol according to the 
vascularity and the degree of arteriovenous 
shunting. 
b.For the tumor without significant arterio- 
venous shunting, less than 50% (usually 30%- 
50%) of the NBCA mixture can be used. 
c.For the NBCA embolization of the head 
and neck lesions, you should obtain a 
safe microcatheter position and know 
potential external carotid- internal carotid 
communication pathways to prevent 
disastrous internal carotid territorial 



. Mann WJ, Jecker P, Amedee RG (2004! luvenile Angiofi- 
Ltc'ijs: '.".hanging suigijii! concep: ■over tlie List 20 yens. 
Laryngoscope 114 

. LnsMiinias P. Kej ensiem A. 7er Br.igge K i ! S.irgica! Neu ■ 
lOiingi.igrjphv, Vol. 2, Springer 

. Osborne A (1994! [Hagnc-siic Neuroradiology; Mosby 

. kosenvv.;s:er H ( 1 974! ;"domus iugulaie in mors. Proc Roy 
5oc Med 67:259-1 64 

. Apostol IV, Fiazel! E (196:"; juvenile Niisopi'.aiyngeiil 
Angiofibroma: a clinical study. Cancer 18:869-978 

. Au L: em one TB i I ; !81 .: Hemangioper.cyioma-like rumor 
of the nasal cavity.Arch Otolaryngol 107:172-174 

. Batsakis JG (1979) Tumor of the head and neck, clinical 
.;;id p.'.tho-ogk";'.! c c-j j:ij ;i e r:i tjo n ■= . WiLi.;;V:S & Wilkin's. 2nd 
edn.pp 296-301 

. Batsakis JG, Jacobs JB, Templeton AG (1983] Hemangio- 
periciloma ::if rlie m'.sal c.ivi iy: Electron- on; k Silidy and 
clinical correlation;. I Laryng Otol 97:361-368 

. Berg NO (1 950! Tumors aris.ng from "die tympi'.nic g^aiid 
(gl'i'iv.us iiigiLaiis) .mo (heir different;,] I diagnosis. Act.; 
Pathol Microbiol Scand 27:194-221 

. Casasco A, Herbreteau E, George E, Tran Ba Huy P, Def- 
fresne I '■. Meil.iisd "I : : 934; I 'eviiscii.ai ;?i;iion of cri'.nio- 
f;i.:i;i. in m-.jrs by percutaneous r.imor puncnire. AINK An; 
J Neuroradiol 15:1233-1239 

. Christiansen TA, Duvall AJ, Rosemberg Z, Carley TB 
(1980) Juvenile nasophjiynye.;. Jiigiofibror-.j. Trans Am 
Acad Opdialmol Otolaryngol 78:140-147 

. Cummings BJ (1980) Relative risk factors ir. I he treat mtt;: 
of juvenile nasopharyngeal a iigiollh :v-.;r. 
Surg 3:21-26 

'_"u minings HI, Mend k. i-'it^p.i trick P, Clar i K. HarwouL 
A, Keane T, Beale F, Garrett P, Payne D, R:der W ( I 984] 
PriT.i'.iy r.'.duiion dierjpy for juvenile ;":.iH'['i;i. jngc.u 
.;ngiofi ;"■[::■ ma. Laryngoscope t; 4:l 599-1605 

. Doppman JL (1980) The localization and treatment of 
p:if.Uh.v:i:::d adenomas by Jiigiogr.ipnic :e-.":'.!":iqiirs. Ann 
Radiol 23:253-258 

. Erickson D, Kudva YC, Ebersold M), Thompson GB, Grant 
CS, Van Heerden )A, Young WE (2001] Benign paragan- 
glioma;: '.'Lined present.; ii:: 1 n .;:id l;e.;tmem outcomes in 
236 patients. J Clin Endocrinol Metab 86:5210-5216 

. Ewing JA, Shiyely EH ( 1 981 ] Angiofibroma: a rare case in 
.;;• r.deib" fe:v..;.e. Oioioryngol i-ieod Neck Sing ^• : :::':j- 
603 

. Farrior JB, Hyams V], Benke RH (1980] Carcinoid apu- 
doiVi.i arising in j glomus iug;;!.ire minor: review of endo- 
crine .'.ctivity in glomus iugiil.;:e ;umors. Lmvng. /score 
90:110-118 

. Goncalves CG, Briant TDR (1978) Radiologic findings 
in ii.;sopliary:ige.;l angiofibromas. ■ il.in Assoc kadiol 
29:209-215 

. Harrison K (1 ^74! Glomus uiguli'.re :umors: their clinical 
beh.;viof ;'.nd man.'.gemem. Proc Roy Soc Wed ti7:2i'4- 
267 



. Hertzanu Y, Mendelso 
( 1 ,: S J ! H.;emai":g;.:'pr:.'; 
55:870-873 



3, Kassner G, Hockman M 
a of the larynx. Brit J Radiol 



20 Embolization of Epistaxis 



Georges Rodesch, Ho: 



\ Alvarez and Pierre La 



20.1 Introduction 257 

20.2 Etiologies and Origins of ENT Bleedings 257 

20.3 Clinical Piemen I iu ion and Initio! Management 258 

20.4 Vascular Aniitomv mid Angiographic Protocol 
of the Nasomaxillary Region 259 

20.5 Technique of Embolization 259 

20.6 Idiop nl i'.ii Hyistaxis 262 

20.7 Traumatic Epistaxis 264 

20.8 Tumor-Related Epistaxis 264 

20.9 Epistaxis in Hereditary Hemorrhagic 
Telangiectasia 266 

20.10 Results ■:■:' Embolization Ti'.erapy in Epistaxis 267 

20.11 Conclusions 267 

20.12 "Endovascular management of EPX 
from a Practical Point of View" 268 

20.12.1 What Has to Be Expected from Angiography 
in EPX? 26S 

20.12.2 What Causes of Epistaxis Require Emergent 
Treatment; 268 

20.12.3 When Is Embolization Required in HHT 
Disease? 268 

Cookbook: Embolization of ENT bleeding 269 

20.12.4 Whatls the Place of Surgery in Epistaxis? 259 
References 270 

20.1 
Introduction 

Epistaxis (EPX) is a nosebleed that can result from 
various etiologies. This condition is rather common, 
often benign and self-limit in;; [23], It may however be 
the symptom of a n undc-i'l vi Lis; ].vii hi 'logical condition; 
and it is therefore mandatory to diagnose properly its 
origin in order to propose an adequate treatment [20]. 
Traumatic EPX is another common cause of serious 
epistaxis resulting from maxillofacial trauma, and 
can lead to massive life-threatening intractable hem- 
orrhage from associated vascular tears. 



obi 



EPX has been described as the most frequently 
ymptom leading to emergency i 



ENT clinic [21]. Only 6%-10 % 
of EPX require medical attention [19, 22]. Emboliza- 
tion should only be reserved to patients who have 
been preselected by a proper initial medical and/or 
ENT evaluation and management trial [6] as well as 
a radiological evaluation. 



Etiologies and Origins of ENT Bleedings 



The various reported causes of EPX are listed in 
Table 20.1. They can be also be distinguished 
according to the age group in which they develop 
(Table 20.2) 

Broadly speaking, one can categorize EPX into 
two main groups [23]: 

• Instances where an organic lesion is the source of 
the bleed. Different pathological entities can be 
recognized and specific treatments can be offered. 
This requires a multidisciplinaiy approach to 
achieve palliation or cure. Embolization is then 
often only part of the global therapeutic manage- 
ment required. 

• Instances where no underlying wife alar or tumoral 
pathology can be identified [2]. Inconsistent asso- 
ziationwith arterial hypertension, smoking, alco- 
hol consumption, or hypercholesterolemia can be 
found. Most of these patients have been previously 
healthy until presenting with acute EPX requiring 
therapy. In those idiopathic EPX cases, the goal of 
treatment is to control and stop the bleeding. 



Another way to categorize EPX is bas 



G. Rodesch, MD 

Service de Neuroradiologie Diagnostique et Therapeutique, ing site, with two distinct types of EPX: 

Ho pi la J Fixh,40 rue Worth, 92150 Suresnes, France 
H. Alvarez MD; P. Lastalnias, MD PhD 

Service de Neuroi-.idiol ogie Piagncstiq-ie ei 7s'.er;ipeu:iq'.:e, 
Hopital Bicetre, 78 rue du General Leclerc, 94275 Le Kremlin 
Bicetre, France 



lthebleed- 



Antcrior EPX: This is the most frequent type, and 
is usually less severe than the posterior one. It 
often resolves spontaneously. The bleeding site is 



Table20.1.Mo:;i i'reqiii 



Idiopathic 

Heredi:.iiv heiiiorj'h.igic le^ingifi"; 

Arterial hypertension 

TY.uariaasra 

Coagulation disorder 

Benign tumor 

Iatrogenic i*u:'g:c;i!j vascalar tear ■ 

Malignant tumor 

Aneinv;ni 

Arteritis 

Intracranial Va.SClLar niakonn-.i ; iOi' 



Table 20.2. EPX: populations and n 



Pediatric population Trauma 
Tumors 

Maxi Ho -facial surgery 

Coagulation disorders 
k'lderlv ivpiiiiiiio: 1 . Anticoagulant treatment 

Arterial hyper reus ion 
Avei'igr age peculation Idiopathic 

HHT 

P.: in c:s 

Trauma 



located in the anterior septum (on the plexus of 
Kisselbach). If treatment is required, simple pro- 
cedures such as compression, anterior packing, or 
cauterization can usually easily arrest the hemor- 
rhagic episode. 
> Posterior EPX: This type requires more aggressive 
or active treatment. Cauterization, anterior and 
posterior packing, local sclerosis, local injections 
of haemostatic agents, electrocoagulation, surgi- 
cal clipping, and ligation have all been described 
to control the hemorrhage [5, 13, 17, 23, 24]. The 
patients usually poorly tolerate posterior packing. 
They still belong however to the panel of neces- 
sary ENT procedures that has to be attempted 
before performing any embolization. 



20.3 

Clinical Presentation and Initial 

Management 

A recurrent EPX that does not stop spontaneously 
usually leads to consultation with an ENT surgeon 
or at the emergency room staff. Careful endoscopic 
evaluation is then performed in order to delineate 
the site and type of bleeding. A first treatment (cau- 
terization or p;u km;; } is rat tempted. Even such simple 
procedures can control the bleed, even though tem- 



porarily. In severe facial trauma, anterior and pos- 
terior packing might allow stabilization of unsteady 
hemodynamic situations. This allows further clini- 
cal evaluation, correction of the coagulation profile, 
careful history and physical examination, and addi- 
tional radiologic!.]] investigations to be performed 
to better delineate the etiology and precise location 
of the hemorrhage [23]. EPX can have other origins 
than the nasal tossa: lesions in the frontal, sphenoi- 
dal (Fig. 20.51, or maxillary sinuses can present with 
EPX; middle ear hemorrhage can also be external- 
ized via the Eustachian tube. Initial proper clini- 
cal examination is thus mandatory combined with 
evaluation of the angiographic findings and careful 
analysis of the potential collateral circulation. Care- 
ful preliminary evaluation will be more likely to lead 
to a satisfactory diagnosis and treatment. 

Packing stops the EPX but is often poorly tolerated 
by the patient and, if prolonged, can lead to serious 
complicated such as aspiration, sinus infection, and 
necrosis by intranasal balloon pressure [23]. It has to 
be therefore limited in duration. Packing is however 
often a necessary step in the therapeutic manage- 
ment of nosebleed and further therapeutic proce- 
dures should be performed only if these initial ENT 
treatments fail, and EPX recurs. Failure rates up to 
25%-50% and complications rates of 20%-60% have 
been described [17, 23, 24]. In these situations, one 
considers the EPX to be intractable and mandates 
other managements. External carotid artery ligation 
has been proposed in the past but should currently 
be avoided as the rich max illo- tana I collateral supply 
will rapidly reconstitute distally the sacrificed arte- 
rial trunk, leading then to clinical recurrences [1, 2, 
10]. Moreover, further endovascular treatment will 
then be more complex or impossible because of indi- 
rect arterial supplies. Internal maxillary clipping or 
ligation have also been reported [5,23], but with a 
complication rate ol tip to 47% and clinical failures 
of 15 % [13,17,23]. Rebleed can easily be explained by 
the developments of anatomical collateral pathways. 
Anterior and posterior ethmoidal arteries (aris- 
ing from the ophthalmic artery), internal carotid 
artery branches (inferolateral trunk, vidian artery} 
and nasopharyngeal vessels (ascending pharyngeal 
artery, descending palatine artery, ascending pala- 
tine artery, accessory meningeal artery) will partici- 
pate to restore the flow distal to the ligated internal 
maxillary artery. However, the mere fact that proxi- 
mal disconnection alone might be successful sug- 
gests that the decrease of pressure and indirect flow 
to the nasal mucosa might play a role in the physi- 
ologic hemostasis that will occur [23]. 



i-.niLx'kziition of Epistaxis 



Endovascular therapies have gained much popu- 
larity nowadays. Embolization of EPXhas to be per- 
formed according to strict angiographic protocols 
taking in consideration the clinical situation and the 
cause of the bleed [9]. This will be further illustrated 



Vascular Anatomy and Angiographic 
Protocol of the Nasomaxillary Region 

The cavum and the nasal fossa, both located on 
the midline, need bilateral explorations even if 
the symptom or the disease seems to be unilateral 
(Fig. 20.1). Exploration of adjacent territories will 
help in delineating with accuracy the vascular limits 
of a given lesion. A unilateral vascular approach 
should therefore be avoided [1, 9, 10]. 

The distal internal maxillary artery is the main 
arterial trunk supplying lesions in the nasomaxil- 
lary area (9, 10, 23]. Vascular supply is via the sphe- 
nopalatine artery of the internal maxillary artery, 
the alar and septal bran-: lies of the facial artery. 

The sphenopalatine artery originates from the 
pterygopalatine segment of the internal maxillary 
artery. It exits the pterygopalatine fossa through 
he sphenopalatine foramen and penetrates in the 
nasal fossa behind and above the middle concha. 
It divides then in two trunks: a posterior lateral 
nasal artery (or concha! artery) supplying the tur- 
binates and parts of the maxillary, ethmoidal and 
sphenoidal sinuses, and a posterior medial nasal 
artery (or septal a itei v) supplying the nasal septum. 
Anterior and posterior ethmoidal arteries arising 
from the ophthalmic artery beyond its second intra- 
orbital portion also supply this midline structure. 
Ethmoidal arteries supply the superior parts of the 
septum at the level of anterior and posterior eth- 
moidal cells; they will build a rich anastomotic net- 
work with septal arteries. 

In its distal portion, the septal artery of the sphe- 
nopalatine artery abandons the nasopalatine artery 
that anastomoses with the terminal branches ot the 
greater palatine artery through the distal hard palate. 
The arterial supply to the inferior and anterior por- 
tion of the nasal cavity is thus accomplished. 

The contribution of the distal facial artery to the 
v;i<,.t]l;in;:,ri]onoltbe nn^iiiia^ilkuy region depends 
from alar arteries, and from anterior septal arteries 
(branches of the superior labial arteries). This ante- 
rior and inferior portion of the septum (the plexus of 



Kisselbach) is thus an arterial junction between the 
facial, nasopalatine and descending palatine arter- 
ies [23]. 

Accessory supply to the nasal fossa and to the 
cavum has been described in vascular lesions or 
after proximal ligation via the accessory meningeal 
arteries and ascending phan ngeal arteries [3, 23]. 

The nasal cavity is therefore a difficult area to 
control by endovascular procedures because of the 
complex arterial supply belonging to two systems: 
the internal carotid artery via the ethmoidal arter- 
ies, and the external carotid system mainly via 
the sphenopalatine artery. Transarterial approach 
to this region will mainly use the external carotid 
artery vascular channels. 



20.5 

Technique of Embolization 

Diagnostic and therapeutic angiography is per- 
formed in the angiography suite, the nasal pack- 
ing being kept in place and always after thorough 
clinical examination of the pat lent by an ENT doctor 
[23]. The whole procedure is performed under gen- 
eral anesthesia (GA) for the patient's comfort. GA 
also allows suppression of breathing during angio- 
graphic runs, and eliminates agitation and uncon- 
trolled motion. In addition, the patient airway and 
ventilation are secured despite the nasal packing. 
Indirectly, these measures lower the risk of vaso- 
spasm due to intra-arterial catheter manipulation. 
Conscious sedation and neuroleptic analgesia are 
less frequently used because they lack the above 
advantages of GA; it will be reserved mostly to 
patients with contraindications to GA. 

An introducer sheath is placed into the femoral 
artery; its size will depend on the pathology sus- 
pected: in case of rupture of the internal carotid 
artery in adults, a large 6 or 7 F introducer sheath 
should be initially placed to allow the use of large 
size guiding catheters for the manipulation of 
detachable balloons. In children, the smallest pos- 
sible introducer size is initially inserted. In our 
experience, all major nasomaxillary arteries and 
main branches, which invariably represent the 
pathology responsible for the EPX, can be catheter- 
ized with a 4F or 5F catheter [23] (Fig. 20.1). Direct 
puncture of the arterial branches or of the internal 
or external carotid artery should not be considered 
[1]. The embolization (usually with particles) is per- 
formed through the same catheter, except if specific 



G. Rodesch et al. 





Fig.20.la-j. Twenty-live-year-old ivoiVj.ui with von Willebrand disease 
suffering from recurrent EPX mainly originating from the left nasal 
fossa despite medicjl treatment to correct her .loagulopiithy, and nasal 
packing. Embolization was indicated because of failure of all other treat- 
ments. The angiographic protocol for EPX is followed. The procedure 
begins with injection of the right internal carotid artery a that shows a 
faint blush in the .interior etljiiioiii.il region I ,; no ■•■■ j . The right internal 
maxillary artery is catheienzed and contrast iniection in lateral b and 
AP projections c only shews, a normal fain; mucosal blush on the turbi- 
nates i asterisk) and nasal septum i sniitll -.incws). The internal maxillary 
artery is emboli zed will: micro particles until distal disconnection of the 
arterial territory is obtained d. A large snip of Gelfoam is then injected 
to enhance the regional devasculanzation i noi shown I. The right facial 
artery is then opacified, e: AP view! showing its contribution to the 
vascularization of the alar (iin-Lnr) and anterior septa! regions [double 



Embolization of Epistaxis 




arrow). The right facial artery is embolized widi the same material fol- 
lowing the same ruies. The nasomaxillary region on the left side is then 
studied, with injection of the internal carotid artery showing faint septjl 
hyperemia supplied ] cy die anterior ethmoid.;, arteries [f (lateral view) 
and g (AP view): arrow]. The left intern. 1 . 1 maxillary artery supplied the 
left nasal fossa [U (lateral view) an.: i !AP view): ijsh'nsk], and is then 
embolized in the same manner. The left facul artery ij, lateral view) is 
not embolized because or its hypoplastic territory th;:t does not partici- 
pate to the vascularization of the alar zone. Note that embolization was 
carried out despite the fact that no clear-ctii abnormality was detected 
in the vascular tree or at the level of the nasal fossa. The ENT surgeon 
was told that he might have to clip the ethmoidal arteries in case of 
e of EPX because of the prominent r.spect or" these vessels. The 
1. oack Lng was removed c4 his .ate:; the patienl did not present with 
it EPX anymore 




G. Rodesch et al. 




Fig.20.2a,b. Young woman with a uimor o: the cheekbone. Angic-gi aphy was pel formed in orc!er to disclose- any hypervascu- 
lariiv oefore surge! v. The >sio:'. was iivosluLli/ but opacif.ca;io:i of the internal maxillary ,n tei v in the Af a and la lei jI views 
l> showed iniensie nasal rossa mucosal o!ush (<iit':>;.iki corresponding :t: ,1 no: ::.:.. appearance 1:1 o woman in the premenstrual 



embolic materials are necessary (balloons or coils} 
or if superselective catheterization is required in 
case of anatomic variations that have to be respected 
(e.g. meningo- ophthalmic artery). Other investiga- 
tors [23] recommend routine superselective embo- 
lization after catheterization of the sphenopalatine 
artery; however, no differences in the immediate 
outcomes and clinical follow-ups have been noted 
between these techniques. Undoubtedly, the super- 
selective approach is associated with higher cost, 
and more time and labor, which is why we have 
continued to favor the simpler regionally selective 
approach since 1979. 



20.6 

Idiopathic Epistaxis 

The initial study in idiopathic EPX should involve 
the internal carotid artery ipsilateral to the bleed, 
using both lateral and AP views in order to detect 
any lesion of the petrous or cavernous segment of 
this vessel. This also allows the evaluation of nasal 
fossa vascularity that originates from the ethmoidal 
arteries. Angiography ol internal maxillary arterv is 
then performed in lateral views to depict any culprit 
anastomoses with the internal carotid system, as the 
external carotid origin of the ophthalmic artery. These 



anatomical variations donot contraindicateendovas- 
cular therapybutwillrequire superselective catheter- 
izations in order to avoid any erratic emboli in the 
internal carotid territory. Embolization is performed 
with small size particles (250-350 micron) that will be 
injected within the flow at each systolic pulsation until 
stagnation of contrast is detected in the vessel. Large 
strips of Gelfoam are secondarily injected in the inter- 
nal maxillary artery in order to increase the distal 
devascularization. Embolization should always begin 
first with the most distal territory [2]. Second, angi- 
ography of the ipsilateral facial artery is performed. If 
contribution to the nasomaxillary territory is noted, 
the facial artery is then embolized with large strips of 
Gelfoam in order to reduce the flow in its distal region 
and participate to the hemostasis. 

The same studies are repeated in the contralat- 
eral vessels, and embolization performed accord- 
ing to the same rules. As a rule, proximal ligation 
or occlusions with coils must be avoided, as they 
will not properly control the bleed and will favor the 
development of collaterals. 

In our experience, the nasal packs are usually 
kept in place for 24 hours and then withdrawn by 
the ENT surgeon. Some teams advocate removal of 
the packing at the end of the procedure in the angio- 
graphic suite [23]. The patient will be seen for follow 
up in consultation both by the referring physician 
and by the interventional neuroradiologist. 



Embolization of Epistaxis 




Fig.20.3a-f. Young male with .in history of progressive nasal obstruction 
it nosebleed. CT a and MR b conn mi a nasopharyngeal angiofi- 
broma. Presurgical embolization is perfoimed: ang.oei aphy of the internal 
carotid artery (c: lateral view) shows faint participation to the ethmoidal 
vase L La i ity via the pos:erio: ethmoid a I arteries (si'.wr iiiracsj. The 
nal maxillary arteiy (d. lateral vievvi and ascenon'.g pharyngeal artery (e, 
is) injections confirm the typical hyperintei'.se capilhn v blush c:' 

.il angiohbioiv.a (.inc. ir.O. Embolization was car: iec 
o particles and stiips or Gelfoam. Como!e".io:' ai'.giogiaphv of .he 
n carotid artery (f. lateral view: confirms the devascularizi 
n all its compartmeiivs 



Persistence of prominent ethmok 
seen on the post embolization nnsiogiv 
diet further bleeding, requiring surgi 
of these arteries, as this type of recurre 



il arteries usually shows a normal blush of the nasal 

nmaypre- Embolization is performed according to the : 

il clipping rules than described above combined with co 

n not tion of the underlvmg 1 0,11:11 lop;] thy will help 



be managed properly by additroti.il endovascular trol the bleeding [11]. Partial In r at ten! ion has to be 

therapy. paid to the orbital anastomosis, usually patent in 

EPX is a frequent complication of coagulopathy children, in order to avoiding erratic embolus that 

mainly in the pediatric population. Angiography could give rise to visual complications. 




Fig.20.4a,b. Patient with HHT disease suffering from recur- 
rent HVX. Nasal to:;* J telant;ie."1..isias ;i:e v.isoiil.nized by both 
septal and turbinate brnnohes or the internal ma.xilkiry artery 
(lateral view a, snaill ,-m'i>irj) and by ethmoidal branches of 
the onhl;',;iJiik" ar:e:y i later a I view b, small . 



20.7 

Traumatic Epistaxis 

Traumatic EPX requires initial CT imaging to rule 
out any skull base or maxillo-facial fracture that 
could be linked to a vascular tear responsible for 
the bleed (Fig 20.6 and 20.7). Damage to the internal 
carotid artery could be diagnosed if the fracture 
involves the carotid canal, or ii a subarachnoid hem- 
orrhage is associated to the osseous trauma, where 
rupture of the suprac aver nous portion of the inter- 
nal carotid artery should be suspected. Traumatic 
EPX is a life-threatening emergency, and can be 
associated with severe hemodynamic instability. If 
a history of serious vascular trauma is evoked in 
the setting of severe EPX, superselective diagnostic 
angiography should be avoided in order to prevent 
rupture of a potentially associated arterial false 
aneurysm, which is effectively the unclotted portion 
of the hematoma associated with the vascular injury. 
This represents a weak point that could rupture if 
subjected to sudden increased pri 
forceful injection. Instead, we recommend 
situations that a nonselective runs 
carotid artery territory be obtained in order to 
depict angiographic signs of a vascular tear on the 
internal or external carotid artery, including pres- 
ence of a pseudo-aneurysm, localized vasospasm of 
the affected artery, or truncation or even no filling of 
the traumatized artery. It is important to know that 
the exact localization of the rupture is best visual- 
ized via the collateral circulation: an angiographic 
protocol studying internal carotid, vertebral and 
external carotid artery branches helps to built up 



the regional vascular cartography and points thus 
to the arterial leakage. 

If the external carotid artery territory is affected, 
sacrifice of the traumatized arterial segment is best 
performed with glue in our experience. N-butyl 2- 
cyanoacrylate (NBCA) is gently deposited proxi- 
mally to the arterial tear or at the level of the stump 
of the traumatized artery after superselective micro- 
catheterization. Traumatic rupture of the internal 
carotid artery is usually treated by sacrifice with 
balloons and coils [7]. The recent availability of cov- 
ered stents offers another endovascular alternative, 
especially when occlusion of the internal carotid 
artery is poorly tolerated [4]. 



20.8 

Tumor-Related Epistaxis 



types have been associated with 
EPX, both benign (juvenile angiofibromas (Fig. 20.3), 
angiomatous polyps, aipiilriry hemangiomas etc.) 
and malignant (primary head and neck carcino- 
mas, epitheliomas, metastatic lesions etc.). Tumor- 
related bleeding tends to be recurrent and moderate 
in severity. It may occur at night leading to anemia [2], 
or be can associated with breathing difficulties due 
to nasal obstruction. Life-threatening hemorrhage 
is rare in these conditions but has been reported [2]. 
In the diagnosis of recurrent tumor-related EPX, the 
patient usually first undergoes an MRI in order to 
delineate the tumor location and extension. Angiog- 
raphy and embolization are performed usually as a 



Embolization of Epistaxis 



pre-surgical procedure, or to control hemorrhage in 
the rare occasion? ol lite-threatening EPX associated 
with head and neck tumors. The goal of the endo- 
vascular procedure is to devascularize the tumoral 
capillary bed. All the vessels supplying the lesion 
are studied according to an angiographic protocol 
[9]. Angiographic patterns of primary and collateral 
vascular supply are predictable in most cases, and 
the location of the arterial feeders depends on the site 
of origin of the lesion [2]. The moderately enlarged 
vessels supplying the tumoral hypervascularization 
should be emlx'lized with particles and Gelfoam: we 
currently use small size particles (150-350 microns) 
to embolize the tumor bed, as smaller particles may 
pass through the tumoral capillaries and reach the 



lungs [18]. The embolization is concluded by injec- 
tion of large strips of Gelfoam in order to produce 
a transient deva.si.Tili.in/.aiion of the region ,md 
enhance distal thrombosis in the tumor. In case of 
intracranial extension of the tumor, glue can be used 
in order to selectively occlude any tumoral feeders 
originating from the internal carotid artery. Balloon 
occlusion of the internal carotid artery (to be con- 
sidered only after proper evaluation of the collateral 
circulation on arterial and venous phases} can be 
utilized if radical surgical exclusion is planned in 
large otherwise unresectable lesions [2, 23]. 

In our practice, we perform catheterization and 
embolization using preset procedural sequences 
that are applied in every external carotid artery 




Eia. 20. >a t i. A !■ i .=■:.! <\.\ •■: jr.: -= . ni.: -: i L . < iir.ii;.,: by .-■::& .oi.." ,; ;h-j .i py ivh.o i.leve.opr.:. -lid. In; rl-'X. i. ":' a .;r.d \1 K b -honed 
e marge mem of i he ieii cavernous sinus with ;'. slructure bulging hiio the spher.oio si mis i ■>: iiiioir: jii.i m irregular aspect of 
ihe inlenj.il cr.rotio .irtery :b; iiriviri!. An infectious false aneiirvsivi was suspecied and ti'.e patieni underwent eiv.ergem .'nci- 
ogri'.pi'.y. A left common carotid artery injection it, lateral view: confirmed the lesion i.e. -.uroif) on the c.vernous portion of 
ihe .mei'iiiil carotid r.riery, which wr.s also stenosed. Sacrifice of ti'.e internal carotid artery Iwitj trapping of die oathologica. 
zone) was performed with occlusion oalloons lij.'foii'Si after a satisfactory toierance test d 




Fig. 20.6. Intractable HI-'X following ir.axi.ki facial and skull 
base trauma. A common carotid ortery injection i lateral view! 
shows r u pin re of the intern;'. I carotici a:: cry [large arrow) and 
a spastic posttraumatic aspect or" the di;tal external carotid 
artery Uinall arrows) 



endovascuhir embolization procedure [2]. If several 
branches must be embolized, one should always 
tackle the most distill target first to avoid later loss of 
access due to vasospasm. The next artery (arteries) 
to be catheterized should include possible sources 
of collateral circulations to the region embolized in 
order to provide an alternate route to reach the ter- 
ritory in case the embolization was too proximal. 
Superselective catheterization of distal pathological 
arteries is in our daily practice only performed if 



Epistaxis in Hereditary Hemorrhagic 
Telangiectasia 

Hereditary hemorrhagic telangiectasia (HHT) or 
Rendu-Osler-Weber disease (ROW) is a disorder that 
presents with recurrent episodes of EPX. These epi- 
sodes are difficult to manage because of the recur- 
ring nature of telangiectasias (Fig. 20.4). 

HHT is ,i dominant autosomal disease with vari- 
able expressivity Ihat evolve 1 , usually in four stages: 
latency, hemorrhages, telangiectasias, and anemia. 
There are two genotypes recognized to date, HHT1 



and HHT2, with a third one suspected (HHT3). 
Endoglin is the gene that maps to chromosome 9q33, 
which is mutated in HHT1, and which is thought to 
be responsible for the telangiectasia, pulmonary 
arterio-venous fistulas (AVPs) and cerebral AV 
shunts [12]. Lack of endoglin, a binding protein for 
transforming growth factor [3 protein, compromises 
normal vascular remodeling in endothelial cells. 
EPX is the commonest presentation of HHT both 
in adults and children; however it may be not given 
enough importance in H HT families and it needs to 
be inquired about specifically. Cutaneous lesions are 
accountable for 66.7% of the adults and 27.3% of the 
pediatric population in our series, which is keeping 
with its acquired nature and presentation in the 2nd 
and 3rd decade [14]. Usually by the 4th decade the 
telangiectasias will be visible [8]. The localization 
in the mucous membrane is usually the one giving 
rise to hemorrhages. The hemorrhagic manifesta- 
tions in HHT have the following d istributions: EPX 
85%, oral 20%, digestive 20%, genitourinary 10%, 
lungs 17% [11]. Epistaxis is the major cause of death 
in 4%-27% according to the series. The diagnosis 
of the disease is mainly clinical, two of four criteria 
being required: spontaneous recurrent EPX, typi- 
cal mucocutaneous telangiectasias, positive family 
history and typical visceral AVMs (cerebral, pulmo- 
nary or gastro-intestinal [14]. Telangiectasias have 
to be searched for carefully as they may be located 
under the nails or on the tip of fingers or toes [11]. 
The family history may be poor as the penetrance 
of the disease is variable, some families being not 
affected by the disease but only transmitting it. 
Appreciation of the evolution of the disease is dif- 
ficult but of importance for the selection of the best 
treatment. Complete cure is not possible nowadays 
for this systemic disease; the only therapeutic objec- 
tive is the stabilization of the bleedings. 

We generally distinguish three stages in HHT, 
which primarily reflect the disease impact on the 
patient's daily life [11]. Stage 1: episodic EPX that 
resolve spontaneously. Stage 2: periodic EPX some- 
times following mechanical trauma, requiring no 
more than one hospitalization and transfusion per 
year, and allowing normal professional activity, 
Stage 3: frequent spontaneous EPX requiring mul- 
tiple hospitalization and transfusions per year with 
resulting incapacitation or inability to lead a normal 
lite. The therapeutic approach will depend on the 
stage of presentation.. Stage 1 does not require any 
treatment. Stage 2 should be treated whenever blood 
transfusions are necessary. Stage 3 represents an 
indication for therapy and usually requires a combi- 



i-.mbokzation of Epistaxis 



nation of both conventional EN'T procedures (local 
treatments and scleroses, clipping of ethmoidal 
arteries, etc..) and embolization that is currently 
generally considered the better therapeutic alter- 
native, albeit still palliative in nature, unless it has 
been proven that collateral supply (mainly via eth- 
moidal arteries) has developed. 

Non symptomatic telangiectasias should not be 
treatedln symptomatic HHT-reiated telangiectasias, 
embolization with particles gives a satisfactory imme- 
diate result with in) mediate hemostasis, last my tn:!in 
3 weeks to 2 years in our experience. This reflects the 
high angiogenetic activity of the disease and the con- 
tinued development of new telangiectasias. Failure 
to stabilize stage 2 patients, or to transform stage 3 
patients to stage 2, will require the addition of more 
aggressive treatments [1 1]. Estrogen therapy (at a dose 
of 1-2 mg/day) has proven to be effective in the treat- 
ment of EPX in this disease because of the specific 
nature of nasal mucosa. Careful clinical and biologi- 
cal follow up has to be performed in order to prevent 
the side effects or complications of the treatment such 
as gynecomastia in men, cancers of the endometrium 
in women, phlebothrombosis, alterations of choles- 
terol levels, etc... 

Treatment of associated lesions in the brain, cord 
or lungs has to be debated taking in consideration 




Fig. 20.7. Massive H!-X wit:', hear 'dymuuic iiiS'.iihiatv following 
n'.iixillofacial tra ■.:::': a. A large false aneurysm due to a vascular 
laceration of the aista! :nlv:n.il maxilla j y artery (iiirari.'Aj is 
detected. After microca:liele: ization of me pa:bologk\\l :uterhl 
segment, it is embolized wi;h pio\i:::al glue deposition, with 
rapid hemodynamic stabilization and control of the EPX 



the natural history of the disease linked to HHT. 
For example, the risk for hemorrhage seems to be 
less for unruptured brain AVMs linked to HHT 
(0.7%/year) compared to non-syndromic brain 
AVMs (2%-4%/year}[12]. Endovascular treatment 
of HHT-associated spinal cord a rterio -venous 
fistulae (AVF) in the pediatric population has 
to be offered, because of potential for neurologic 
sequelae that can be linked to these lesions [15, 16]. 
Pulmonary AVF also require treatment because of 
the hemodynamic symptoms they create, and the 
potential for devastating neurological deficit asso- 
ciated with thromboembolic diseases (strokes and 
abscesses). 



20.10 

Results of Embolization Therapy 

in Epistaxis 

If properly performed, embolization allows imme- 
diate hemostasis (about 97% of all patients in 
our series) [1]. Recurrences occurred more com- 
monly in HHT patients, requiring either addi- 
tional endovascular treatments, or surgical clip- 
ping of the ethmoidal arteries. Satisfactory stable 
long-term results with no EPX are achieved in 
86.3% of patients [1]. "Therapeutic Failures" with 
recurrence of EPX is seen in 13.7% of embolized 
cases, 75% of them being HHT patients, which 
only reflects the aggressive angiogenetic potential 
in HHT. 

Procedure-related morbidity is rare, as are neu- 
rological complications. Proper anatomic evalua- 
tion can minimize the risks of stroke [23]. Careful 
fluoroscopic monitoring, low pressure injection 
of the embolic material and consideration of the 
potentially dangerous anastomoses and vascular 
supply to cranial nerves are mandatory to obtain 
safe and effective results. In our series, complica- 
tions occurred during the phase of diagnostic angi- 
ography in older patients (0.4% and 1% permanent 
and transient deficits, respectively) but were never 
related to erratic emboli due to poor technical con- 
trol during delivery of the embolic material, or to 
lack of recognition of dangerous arterial anasto- 
moses. Necrosis of the nasal septum occurred in 
0.4% of cases. Non-neurological complications 
include pain, trismus or facial edema that can be 
sometimes described after these interventions but 
have always regressed rapidly with analgesics and/ 
or corticosteroids. 



20.11 

Conclusions 

Management of patients with EPX should involve 
a multidisciplinarv approach. An escalating cas- 
cade of treatment modalities offers these patients 
a tailored approach yielding a high success rate 
with minimal recurrences and low complication 
rates [23]. Proximal vascular occlusion has to be 
avoided, as are proximal surgical clippings. They 
may reduce perfusion pressure to the nasal mucosa 
but will not avoid reconstitution of the flow by the 
rich collateralcircula t ion. Distalbilateralemboliza- 
tion to achieve blockage at the arteriolar-capillary 
bed level are essential for successful endovascular 
managements of EPX. The effectiveness and safety 
of the embolization procedure in trained hands has 
currently rendered it the preferential modality of 
treatment in intractable EPX. In idiopathic EPX, 
devascularization of the posterior septal area is 
emphasized, as this is the usual source of bleeding 
hardly reachable for examination and cauteriza- 
tion [23]. 



necessary to assess them properly in order to 
manage appropriately these patients, by either 
endovascular or surgical methods. 

• Less frequently, mucosal hyperemia can occur in 
women during the premenstrual period (Fig. 20.2). 
EPX in those cases is usually modest and does not 
require therapy most of the time. Intracranial 
vascular malformations draining towards the 
cavernous sinus and secondarily through orbital 
veins may give rise to EPX: treatment of these 
malformations relieves the bleedings thanks to 
the secondary venous decongestion. 

• In traumatic EPX the imaging signs are usually 
more helpful and may help localize the bleed- 
ing site. One has to look for a pseudoaneurysm, 
,i] renal spasm or a missing artery. Hndovasailar 
treatment should be targeted in priority towards 
the bleeding zone. 



20.12.2 

What Causes of Epistaxis Require Emergent 

Treatment? 



20.12 

"Endovascular management of EPX from a 

Practical Point of View" 

20.12.1 

What Has to Be Expected from Angiography in 

EPX? 

• Angiography will rarely show active bleeding or 
its cause. This should not precludes embolization 
where clinical indications exist. 

• A unilateral bleed, or a unilateral angiographic 
localization of bleed should not preclude bilateral 
embolization. 

• The clinical, biological and imaging data orient 
the diagnosis and will help to choose the appro- 
priate material to use. 

• In idiopathic EPX secondary to hypertension or 
coagulation disorders, angiography is most often 
negative. The aim of an angiography is then to 
study the vascular 1 a natomy ot the skull base and the 
inter- territorial and inter- regional anastomoses. 

• In the case of HHT disease, telangiectasias are 
detected at the level of the nasal mucosa. Bilat- 
eral embolization will also need to be performed 
in these conditions. Ethmoidal arteries are often 
well developed in these patients and it is always 



• The clinical suspicion of a ruptured internal 
carotid artery (by trauma, surgical damage or sec- 
ondary to a cavernous aneurysm) is an absolute 
emergency, as is also the rupture of the external 
carotid artery, even if the patient is hemodynami- 
cally stable. 

• In older patients suffering ol hypertension or 
coagulation disorders (due to hepatic or hemato- 
logical disorders or to anticoagulation therapy), 
nasal packing and correction of the causative dis- 
ease are usually sufficient to achieve hemostasis. 
Failure of initial treatment requires complemen- 
tary endovascular therapy, usually performed in 
a less urgent context. 

■ Presentation ol idiopathic HPX will often allow a 
deferred embolization procedure. 



20.12.3 

When Is Embolization Required in HHT Disease? 

Repeated embolization is usually necessary. In our 
experience clinical remissions can last from few 
weeks to years, and recurrences will usually require 
new treatment. We perform embolization during 
the acute phases of hemorrhage, when the bleeds 
are too frequent and interfere with the quality of 
daily professional or personal life, or when blood 
transfusions are necessary [1, 11]. The long-term 



Cookbook: 




Embolization of ENT bleeding 


Common EPX 


(Idiopathic, coagulopathy, tumors, HHT..) 




SFTerumo (children) 




6F Terumo (adults) 


Catheter: 


4F Vertebral (Terumo) 




In the case of failure (tortuous vessels e.g.): 4F Simons type II (Terumo) 


Guide: 


Glide wire 35 (Terumo) 


Embolic material: 


Contour particles 250-350 microns (Boston Scientific-Target) 




Gelfoam (cut in strips) 


If sup erselec five cc 


Ihere.rization is needed: 




(.".uidiiig call'.eiei : Hr.voy (Cordis) 5 or 6F 




Guider (Boston Scientific-Target) 5 or 6F 




Microcaiheter: Excelsior 18 or Excel 14 




Microguide: Terumo 12 (45° or 90° angulation) 




Mirage 008 (MTI) 




Embolic m uteri ni: Contou: ;; a nicies l Bos ".en Scientific -Target) 




150-250 urn 




250-350 urn 


Rupture of ICA 




Introducer: 


5F Terumo (children) 




6F Terumo (adults) 


Guiding catheter: 


Envoy (Cook) 5 or 6F 




Guider (Boston Scientific-Target) 5 or 6F 


Guide: 


Glide wire 35 (Terumo) 


Micro catheter: 


Minitorquer CIFN 130 (Mynvasis) 


Embolic material: 


Detachable Gold Vulve Bulbous 16 (Mynvasis) 


If to be used in children, accciding tc age, weight, and size: 




HvesiiiKiliy 4B inlioducei and Ar guiding catheter 




If not: 5F introduce! and ?r guiding catheter 




Preload system wiili jiiir.iioique: and dciachable balloons 


Traumatic false ar 


eurysm on external carotid artery 


Introducer: 


5F Terumo (children) 




SFTerumo (adults) 


Guiding catheter: 


Envoy (Cook) 5 or 6F 




Guider (Boston Scientific-Target) 5 or 6F 


If needed in child ft'ii, aceidmg re age. ■■•■ eight, and size: 




Possibly 4F introducer and Ar guiding cuke It; (4r Vertebral, Terumo) 


Guide: 


Glide wire 35 (Terumo) 


Micro catheter: 


Magic 1.8 or 1.5 (Bait) 




Excelsior 18 or Excel 14 (Boston Scientific -Target) 


Micro guide: 


Terumo 12 (45° or 90° angulation) 




Mirage 008 (MTI) 


Embolic material: 


NBCA (Braun Aesculap, Germany) 



s of embolization is unpredictable. Emboli- 
zation will be repeated as often as necessary and 
possible. Clipping of the ethmoidal arteries and 
estrogen therapy will be proposed when the ves- 
sels responsible for the bleeds are inaccessible to 
endovascular therapy. 



20.12.4 

What Is the Place of Surgery in Epis taxis? 



> Clipping of the ethmoii 
tive and relatively easy procedure to perform. It 
should however be offered only after emboliza- 



tion, or when the technical or anatomic condi- 
tions are inadequate for success in endovascular 
therapy, such as when the size of the ophthalmic 
artery does not allow safe distal catheteriza- 

> Ligature of the internal maxillary artery should 
be avoided. The unilateral arid proximal aspect of 
the procedure will allow the development of ipsi- 
and contra -lateral anastomoses that will distally 
reconstruct the vessel, and will continue to favor 
rebleeding but without allowing subsequent selec- 
tive auheteriz.itiom. 



References 

1. Alvarez H, Theobald ML, Rodesch G, Attal P, Magufis G, 

Robin S,L;;sj.;uni;;s P i I W$\ Tranemenl en a. ova sen la ire ces 

episuixis. I Nearoradiol, 25: 15-18 
2.Berenstein A, Lasjaunias P, Terbrugge K (2004) Nasopha- 

ryngeal Illinois. 1:1: Surgical neuroraiigiograpliy, vol 2.1. 

Springer, Berlin Heidelberg New York, pp 201-226 
3.Breda SC, Choi IS, Persky NS, Weiss M (1989) Embolization 

:n die Ireuinient of epis:axis after failure oi" internal ira'.xil- 

..ii y .nery kg:-. I .on. Laryngoscopy ;,il : 809-81 5 
4.Celil G, Engin D, Orhan G, Barbaras C, Hakan K, Adil E 

!2004! intnictuble epistaxis rel.'.led to cavernous carotid 

artery pseudoaneurvsm: treatment of a case with cevereo 

stent. Auris Nastis Larynx 31: 275-278 
5.Chandler JR. Serrins A] (1965) Transantral ligation of the 

intern;;! ni.ixi!la:y artery for epislaxis. Larvngosccpe 7.-: 

1151-1160 
6. Evans AS, Young I '.Ac! a:'.; son R 12004! Is die nasal ; a mo on 

a suitable treatment for epistaxis in accident and emer- 
gency; A comparison of outcomes for ENT and A&E 

packed p.'.tients. 1 L.;ryngo! C;ol 118:12-4 
7.Hadfield P], Gane SB, Leighton SE (2002) Epistaxis due to 

traumatic internal enrol id .irtery aneurysm. Int ; Pediatr 

Otorhinolaryngol 66: 193-196 
8-Guttniacher A, Marchuk D, White RI [r (1995) Hereditary 

haemorrhagic telengiectasja. N Engl | Med 333:913-924 
a . Lasiamiias P, Berensiein A :. 1 '''87 1 Angiographic protocol of 

die nasomaxillary region. In: Surgica. neuroai'.g.ograpk.v. 

v::il I . springer. Reran He.aelbrrg New York, pp 371 -3S2 

10. Lasjaunias P, Berensteiri A, Terbrugge K (1987) Skull base 
ami maxillo-facial region. !n: Surgical neuroai'.g.ograpkv. 
vol 1, Clinical v.iscidar anatomy and variaiiens. springer. 
Berlin Heidelberg New York, pp 261-385 

11. Lasjaunias P, Berenstein A (1987) Craniofacial hemangio- 



mas, v;;scuiar nialform.;tions and angiomatosis; specific 
a so eels. In: Surgical iieuroangiogiapliy. vo! 2, !- ndovascu.ar 
treatmen: of craniof.'.cia! lesions. Springer, Berlin He. del- 
berg New York, pp 341-397 

2.Mahadevan I, Ozanne A, Yoshida Y, Weon YC, Alvarez H, 
Rodesch '.j, Lasiaunias P. (2004: Herediiary nemo it;: agio 
lelangirct.isia cerebrospina! loan iza lion in adults and chil- 
■j iirii. Kevirwof 3* c.ises. intervenuonal Neuroradiology i .':; 
27-35 

i. Met son R, Lane K i 1 ' :, 88j ! me in;; I maxil.arv ligation for epi- 
staxis: an analysis of ia'l'.;re c ! nrvi-R'-scr-pe 50- 760 764 

l.Plauchu H.DE Chadarevan |l', Kideau A.Koben )M (1989) 

Afe :e.air.: ."I. II. oa. ' ' '! :-:-lt: r r h I j;.. 1 

lelengieclasia in an epoetin.' ..ii;..i-..lv "t.r. 'id p::pu"n 
tion.Am] Med Genet32.29l 297 

3. Rodesch G, Hurth M, Aivaro^ II. \adie V, lasjaunias I' 
i2T:r:2; Classification ol .pir.al t:ir;'. arlLnirivroux %hunls; 
proposal for a reap p rai;„.. 1 r.^ b..a:e esper.en.L «i± '..<• 
consecutive oaiienrs treated oerween i ,: 81 and : 399. Neu- 
rosurgery 51: 374-3S0 

j. Rodesch G, Hurth M.Alvarez H, Ducot B, Tadie M, Lasjau- 
nias P i20r:4; Angioarcliiieclure of spin;;! cord arteriove- 
nous shiinis a i present;;! ion. Clinic. ;1 correlations in adults 
and children. Acta Neurochir (Wien), 16; 146: 217-227 

7.SchaitkiriB, Strauss M, HouckJK, Hershey PA (1987) Epi- 
siaxis: meilical surgical dier;;pv-a comparison o: efficacy 
co nip licit io ns j\-\:. s^onomw. ■: o n s id era i ions. Laryngoscope 
97: 1392-1397 

H.Schroth G, Haldemann AK. Wr.riani L. Remonda L, Raveli 
I ! 199!!! Preoo native embolization, oi" paragangliomas and 
..lngiot.'oromas. Measurements o: intruiumoial arteriove- 
nous shunts. Arch Otolaryngol Head Neck Surg 122:1320- 
1325 

). Small M, Murray J, Maran A (1982) A study of patients 
will; rpi- : ;axis requi; .ng ad miss. on to- hospnal. Health Bull 
(Edinb) 40:24-29 

].Sp;;racino LL (2000) Epistaxis management: what's new 
una what's noieworiliv. Lippi neons !-'!im Care j- rit-ot 4: 498- 
507 

I.Timsit CA, Bouchene K, Olfalpour B, Hermann P, Tran 
Ba Huy P (2C01 : S.pidemiology and clinical findings in 
20563 patients attending Lariboisiere Hospital ENT Adult 
Emergence Clinic. Ann Otolaryngol Cliir Cervioofac. 1 1 S: 
215-224 

2. Vaamonde L;;go P. Martin Martin C, Lecituga Garcia MR, 
Mi:;gue7. I. Laoellu Caballere T 12334: Epioemiological 
notes on nasal bleeding. An uionnolaringol Ibero Am, 
31:123-132 

i. Valavanis A. Sen on A ( I 003.: Embolization of epistaxis. in: 
Valavanis A ieo: Intervention;;! neuron;.:. ologv. Springer. 
Berlin Heidelberg New York, pp 55-62 

4. Wane L, Vogr. I"'M :. '. '"SI i Posierior epistaxis: comparison 
t. Otolaryngol Head Neck Surg 89: 1001-1006 



21 Diagnosis and Endovascular Surgical Management 
of Carotid Blowout Syndrome 



., Walter S. Lesi 



, and Shik-Wei Hsu 



Background 271 
Definition 273 
Patho etiology 274 

Diagnostic Evaluation 277 
Endovascular Techniques 277 
"Deconslr active" Techniques 27t> 
"Reconstructive" Techniques 279 
Outcomes 285 
Future Directions 289 
References 290 



21.1 

Background 

The apoplectic consequences of a ruj 
artery have been well recognized 
dating back to antiquity. In both the distant and 
near past, this catastrophe was exclusively the result 
of a penetrating injury derived from an act of war- 
fare or accident. Surgical intervention for treatment 
of carotid rupture is also historically relatively old, 
predating "modern" neurosurgical practice for more 
than 100 years [1], John Abernathy, a former pupil 



I.C.Ch. 



i. Y I ■■ 



Director of Interveiilioii-.il NirUi-.iuidio.ogy. t'rofessor of Radi- 
ology and Neurosurgery, University of lovva Hospitals ana 
Clinics, University of Iowa Carver College of Medicine, 200 
Hawkins Dr, 3893 JPP, Iowa City, IA 52242, USA 
W.S. Lesley, MD 

Chief, Section of Surgical Neuroradiology, Assistant Profes- 
sor of Radiology, The Texas A&M University Health Science 

M. Hayakawa, MD 

Visiting Assistant Professc-i of Radiology. University of Iowa 
Hospitals and Clinics, University oi Iowa Carver College of 
Medicine, 200 Hawkins Dr, 3893 JPP, Iowa City, IA 52242, 
USA 

S.W.Hsu.MD 

Visiting Scholar. University of Iowa Hospitals and Clinics, Uni- 
versity of Iowa Carver College of Yedicme, jOO Hawkins Dr, 
3893 JPP, Iowa City, IA 52242, USA 



t Professor, Department of Diagnostic Radiology, 
Chang C-ting Manorial Ho-soita!, Kaonsiiing, Taiwan 



■ of the famed surgeon [ohn Hunter, 
probably can be credited with the first well described 
published account of successful surgical treatment 
of a ruptured carotid artery in the late eighteenth 
century [2]. The case involved a patient who suffered 
a laceration of the internal carotid artery after being 
gored in the neck by a bull's horn. Dr. Abernathy suc- 
cessfully arrested the hemorrhage by simple ligation 
of the artery, which fortunately was well tolerated 
by the patient. 

Although penetrating mechanisms of injury to 
the carotid arterial tree still occur today, in more 
recent times, physicians have been increasingly 
confronted with carotid rupture or so-called "blow- 
out" as an iatrogenic complication of surgical man- 
agement of cervical neoplasms [3, 4]. The first well 
described cases of carotid rupture occurring in 
association with head and neck surgery date back to 
isolated reports in 1962 by Borsdany [5], followed 
by the first large case series published by Ketcham 
and Hoye in 1965 [6]. The term "carotid blowout" 
also was first coined during this time period by 
Rutledge and Cagle [7]. 

The reasons for developing carotid blowout in 
patients with head and neck cancer are potentially 
multifold, being closely linked to the advent of 
aggressive operative resections, flap mobilizations, 
and adjuvant therapies for both primary and recur- 
rent neoplasms. Occasionally, these interventions 
may produce either direct or indirect trauma to the 
carotid arterial tree, resulting in a cascade of events 
that produce progressive structural fatigue and 
eventual rupture of the affected artery. Owing to 
the usual loss of anatomic fascial planes and barri- 
ers from surgery, catastrophic hemorrhage through 
various external and oropharyngeal pathways ulti- 
mately ensues [4-6, 8-12]. 

Despite anecdotal reports of technical and clini- 
cal success ol simple operative ligation for the treat- 
ment of carotid rupture, most open surgical series 
reported over the last few decades have shown 
exceedingly high rates of mortality and morbidity 
associated with this complication. Emergent opera- 



J. C.Chaloupkai 



tive ligation or attempts at primary repair/recon- 
struction of the common or internal carotid artery 
had been traditionally the only therapeutic maneu- 
ver available lor 1 treating carotid blowout [3, 5, 7, 13- 
20]. These approaches, however, generally resulted 
inunacceptably high rates of major stroke and mor- 
tality. A review of cumulative published outcomes 
whereby carotid blowout was managed by surgical 
ligation or repair/reconstruction revealed an aver- 
age mortality ot approximately -40°o, and an average 
major neurologic morbidity of approximately 60% 
[3,4,6,11,12]. 

Several general limitations of operative manage- 
ment of patients with carotid blowout may explain 
this high morbidity and mortality. Patients suffer- 
ing a carotid blowout often are hemodynamically 
unstable, particularly in situations of uncontrol- 
lable or protracted hemorrhage. In such scenarios 
severe reductions in mean arterial blood pressure 
and cardiac output may be encountered, which in 
turn can lead to regional and global cerebral isch- 
emia in susceptible patients (e.g. dysfunctional 
autoregulation, lack of anatomic and/or physi- 
ologic collaterals}. Adverse reductions in cerebral 
perfusion pressure can be further exacerbated by 
induction of general anesthesia, particularly in 
patients with depleted intravascular volumes who 
not infrequently will develop profound transient 
hypotension in response to anesthetic. Extreme 
blood loss from a carotid rupture not infrequently 
produces a depletion coagulopathy that leads to 
further uncontrolled bleeding (particularly in a 
fresh operative wound). Extensive, often multiple, 
previous surgeries in combination with either 
external beam or intraoperative brachytherapy 
radiation can make operative dissection extremely 
challenging [4, 12, 15). 

Another important limitation of past operative 
series managing carotid blowout syndrome (CBS) 
is that they were invariably performed without the 
benefit of diagnostic angiography. Consequently, 
the type and precise location of the hemorrhage 
was often unknown, being mostly inferred from the 
physical examination. Based upon our past cumula- 
tive experience, a wide spectrum of anatomic loca- 
tions for pseudoaneurysm formation involving the 
carotid circulation likely would have been encoun- 
tered in these series [4, 21]. This likely would have 
resulted in frequently inadequate therapy of the 
actual pathoetiologic mechanisms of hemorrhage 
by empiric common carotid ligation. 

An additional compounding proble 
with conventional proximal carotid li 



sk of thromboembolic complications 
that may occur from the phenomenon of propagat- 
ing thrombus within long arterial segments. Dandy 
was one of the first to recognize that thromboem- 
bolic stroke may occur after carotid ligation from a 
growing tail of thrombus that often develops within 
the large intravascular "dead space" distal to the 
site of occlusion [22]. Historically, extensive clini- 
cal experience with both open surgical and endo- 
vascular parent artery occlusion techniques (mostly 
for treatment of giant intracranial aneurysms) has 
shown that "proximal" occlusions of the carotid 
artery within the neck are associated with a rela- 
tively high risk of thromboembolic stroke [23-27]. 
The risk of stroke from such a proximal occlusion of 
the carotid artery also is likely to be increased with- 
out the use of systemic anticoagulation for some 
period of time immediately after carotid occlusion 
[27, 28]. In the past it was likely that systemic antico- 
agulation was not used in the previously published 
surgical series for postoperative management of 
therapeutic carotid occlusion. 

Fortunately, an excellent alternative to open sur- 
gical management of CBS has become available, 
consisting at first of various endovascular tech- 
niques for occluding major brachiocephalic arteries 
[25, 27, 29-36]. Furthermore, within the last several 
years, endovascular revascularization techniques 
utilizing stent technology also have become more 
readily available, providing additional options for 
the successful treatment of CBS without sacrifice of 
the carotid circulation [37-43]. 

There are several distinct advantages of endovas- 
cular versus open surgical management of carotid 
blowout. Although patients managed by endovascu- 
lar approach also may be hemodynamically unsta- 
ble, there is usually no need to place the patient 
under general anesthesia with the additional inher- 
ent risks of diminished cerebral perfusion pres- 
sure. Because only a relatively small arteriotomy 
is used for many types of endovascular operations, 
there are generally less difficulties with iatrogenic 
hemostasis that frequently complicates a large sur- 
gical wound. Since the therapeutic interventions are 
directed through endovascular navigation, dissec- 
tion through scarred and deformed tissue planes 
altered by prior radical neck dissection and irradia- 
tion is completely avoided. 

As shown by earlier publications by our group [3, 
4], precise localization of the pathoetiology of hem- 
orrhage can be readily identified in most patients 
through meticulous diagnostic angiography per- 
formed just prior to endovascular surgery. This 



Di„gnc 



if Siirgkol M ;'.::.! gem ent o: Cj;'o:id Blowout Synorc 



permits specific targeting of the most likely site of 
potential or actual bleeding, as well as better selec- 
tion of the most appropriate endovascular device 
and/or technique. Finally, the added risk of post- 
operative thromboembolic stroke after carotid sac- 
rifice can be substantially reduced by use of both 
intra- operative and postoperative systemic antico- 
agulation, which can be done more safely inpatients 
undergoing endovascular operations. 

Interestingly, the application of these endovas- 
cular techniques speciticully lor the management of 
carotid rupture is a relatively recent development. 
The first reports of successful treatment of CBS 
using endovascular therapeutic techniques date 
back to the mid-1980s [29, 33, 34]. However, a rigor- 
ous evaluation of the specific indications and tech- 
nical approaches to this problem did not occur until 
1995, when our group at Yale published a prelimi- 
nary clinical series on the first 15 patients managed 
by an interdisciplinary protocol centered on endo- 
vascular surgery [3]. At that time we also recognized 
that the various scenarios of carotid rupture actu- 
ally manifest in a few discrete and well recognizable 
patterns of clinical presentations, which we coined 
the "carotid blowout syndrome" [3, 4]. This permit- 
ted the creation of a simple classification scheme 
that provided a means of rapidly communicating 
the nature, severity, and urgency of a given clinical 
presentation, which greatly facilitated triage and 
therapeutic planning [4]. Our subsequent work and 
that of others have shown that CBS can be effectively 
managed by a well-coordinated, multidisciplinary 
protocol centered on deconstructive endovascular 
techniques [3, 4, 21, 29, 35, 36]. 

Although our results with deconstructive endo- 
vascular management of CBS have been overall 
significantly better than previous open surgical 
series, there is considerable room for improvement, 
owing to a combination of acute complications, 
technical limitations, and recurrent disease. Prob- 
lems such as delayed collateral failure resulting in 
stroke, acute intolerance to balloon occlusion of the 
internal carotid artery, and recurrent hemorrhages, 
remain as significant challenges to still overcome 
when utilizing so called deconstructive techniques. 
Consequently, we have begun to aggressively pursue 
an alternate management strategy using so-called 
endovascular reconstructive techniques centered 
on stenting with or without adjunctive measures. 
Our recently published case series experience with 
this approach has been indeed very favorable, and 
has now has led us to consider fully adopting this 
change in treatment paradigm [43]. 



This chapter will review some of these issues 
and present both diagnostic and therapeutic strat- 
egies for managing carotid blowout using modern 
endovascular approaches as the centerpiece of a well 
coordinated multidisciplinary paradigm. 



21.2 

Definition 

Based upon a combination of our previous review 
of the literature and cumulative clinical experience, 
our group proposed that the varying presentations 
of actual or potential hemorrhage arising from the 
carotid circulation can be conveniently classified 
within a broad clinical diagnostic scheme termed 
the "carotid blowout syndrome" [3, 4]. CBS can be 
operationally defined as either an episode of acute 
hemorrhage (usually trans-oral or trans-cervical}, 
or exposure of a portion of the carotid arterial 
system (e.g. wound dehiscence, devitalized mus- 
flap}, that occurs in a patient who 
;ly had undergone a surgical resection of a 
neoplasm. Although this classifica- 
tion scheme derives mostly from the commonly 
encountered clinical scenarios of carotid hemor- 
rhage in head and neck cancer patients, it is equally 
valid for accidental, intentional, or other iatrogenic 
penetrating injuries to the cervical carotid circula- 
tion [4]. 

The clinical spectrum of CBS can be sub-classi- 
fied into three distinct groups of affected patients. 
This nosologic scheme originally was created out 
of a need for enhanced clarity and uniformity in 
describing certain clinical scenarios that would 
frequently arise, which in turn could permit more 
efficient communication and triage between the 
clinical services responsible for the care of these 
patients. Based upon extensive review of the litera- 
ture, these group definitions are equivalent to many 
previously described variations of carotid blowout 
[3,12,18,29]. 

The first distinctive type of presentation of CBS 
is in patients who develop wound dehiscence from 
prior radical neck dissection or flap mobilization, 
resulting in a visibly exposed carotid artery. In such 
a setting it is well recognized that the exposed artery 
will inevitably rupture if it is not promptly covered 
with well-vascularized, viable tissue (e.g. free ped- 
icle flap, rotated musculocutaneous I lap, etc.). For 
such a scenario, although rupture of the carotid 
artery has not occurred at this point in time, there 



}. C. Chaloupka 



is a considerable threat of eventual rupture, leading 
to the designation: threatened carotid blowout (or 
Group 1 CBS). 

A second commonly encountered group of CBS 
patients are those who present with a short lived 
acute hemorrhage that resolves either spontaneously 
or with simple surgical packing. The hemorrhage is 
typically either transoral or transcervical through 
a surgical wound or fistula. These events may be 
episodic, and are considered sentinel hemorrhages 
occurring from a ruptured vessel with a pseudoa- 
neurysm that intermittently leaks. Because there 
is no real wall with supporting structural elements 
around the pseudoaneurysm, unconfined rupture 
is almost always certain to eventually occur. Such a 
scenario maybe described as an impending carotid 
blowout (or Group 2 CBS). 

The final group of patients with CBS is those 
who present with an apoplectic, profuse hemor- 
rhage that is not self-limiting and is not well con- 
trolled with surgical packing. This presentation 
often, but not always is preceded by the above 
mentioned sentinel hemorrhage of impending 
carotid blowout. In this scenario, there has been 
complete rupture of the affected artery and lack 
of confinement by the organized hematoma sur- 
rounding the associated pseudoaneurysm. These 
patients rapidly deteriorate from exsanguination, 
unless intensive resuscitation measures and ther- 
apeutic occlusion of the ruptured artery is imple- 
mented immediately. It is this most catastrophic 
scenario that is best designated as acute carotid 
blowout (or Group 3 CBS). 



21.3 

Path o etiology 

There are a variety of causes of carotid rupture, 
which can be broadly grouped into the following 
three categories: traumatic, non-traumatic (related 
to "spontaneous" carotid dissection), and those 
related to head and neck surgical management 
of cervical cancers. Traumatic carotid rupture or 
blowout can occur from various penetrating insults 
(e.g. ballistic missiles, knives, impalements, etc.) or 
blunt trauma (e.g. dissecting aneurysm). Iatrogenic 
traumatic injury to the carotid not associated with 
head and neck cancer surgery is also an occasional 
cause of delayed rupture, typically occurring in the 
setting of carotid endarterectomy (CEA). In fact the 
original term "carotid blowout" was probably first 



applied in the well-known complication of delayed 
rupture of a venous graft used for patch angioplasty 
after CEA [7]. 

It has also become increasingly recognized that 
non-traumali.: or "spontaneous'' carotid dissection 
can occasionally result in significant pseudoaneu- 
rysm formation and growth, which arguably may 
pose a subsequent risk of a catastrophic, life-threat- 
ening hemorrhage. In the past conventional teach- 
ing has contended that in the absence of disruption 
of the normal fascial planes within the neck (as in 
the case of penetrating trauma or surgical inter- 
vention), the expanding hematoma that consti- 
tutes a pseudoaneurysm should stay well contained 
within the carotid sheath, and therefore pose an 
unlikely hazard of exsanguination. However, our 
group and others have noted rare anecdotal cases 
of "breakthrough" hemorrhages occurring in non- 
traumatic carotid dissection that resulted in Group 
3 CBS. This has in part lead to more of an impetus 
toward active intervention when dissecting pseu- 
doaneurysms are now diagnosed, although admit- 
tedly there is little known about the true natural 
history and risk of these lesions to provide strong 
evidence-based support of such a management 
strategy. 

Although recently our group at UIHC has seen 
an unexplained rise in traumatic and spontaneous 
etiologies of CBS, our overall cumulative experi- 
ence has revealed that most cases are an iatrogenic 
complication of aggressive multimodality treatment 
of malignant cervical neoplasms. It has been previ- 
ously hypothesized that there are numerous putative 
mechanisms of injury to the carotid artery that ulti- 
mately lead to CBS in head and neck cancer patients 
[3, 4-6, 8-12, 27]. Both the cellular and extracellu- 
lar matrix components of the carotid arterial wall 
may be directly damaged from a wide spectrum of 
insults commonly encountered with contemporary 
head and neck surgical management of cervical car- 
cinomas. These include iatrogenic surgical manipu- 
lation during functional neck dissections or radical 
tumor resections, radiation induced necrosis (from 
either an external beam source or intra-operative 
brachytherapy), exposure desiccation after break- 
down of a reconstructive flap, secondary wound 
infections producing fascitis, direct tumor invasion, 
and enzymatic degradation horn oiopharyngocuta- 
neoin iistulae. 



if Siirgkol M ;'.::.! gem ent o: O.;odii Blowout Synoro 



21.4 

Diagnostic Evaluation 

Many patients with CBS present in an unstable con- 
dition, which obviously requires emergent triage 
and immediate basic resuscitative measures. How- 
ever, at some point early in the evaluation of any CBS 
patient who is to be considered for possible interven- 
tion, a focused, yet detailed history with particular 
attention to the specific types and anatomic sites of 
previous surgical intervention, is critical for consid- 
ering not only where the likely source(s) of bleed- 
ing may be located, but also what treatment options 
may be most realistically considered. For example, 
patients with previous bilateral functional neck dis- 
sections and flap reconstructions are more likely 
to harbor multi-focal (and unfortunately bilateral} 
disease, making deconstructive intervention less 
favorable, if not impossible. This is also the case in 
patients with a previous history of CBS that required 
deconstructive intervention, who return with so- 
called recurrent carotid blowout syndrome (rCBS). 
Again, consideration of realistic treatment options 
will largely depend on whether an internal carotid 
artery already has been therapeutically occluded, 
and where the suspected source of recurrent bleed- 
ing is originating. 

A quick and directed otolaryngologic examina- 
tion is also an essential preliminary step for evaluat- 
ing any patient with CBS. Of particularly paramount 
importance is the ability to localize and/or lateralize 
the site of bleeding, which will enable a more focused 
search for the precise pathoanatomic substrate when 
the patient is subsequently studied by catheter angi- 
ography. Furthermore, prospective identification of 
the most likely potential pathoetiology of the CBS, 
such as a flap breakdown with carotid exposure, 
presence of a pharyngo- cutaneous fistula and/or 
infection, or recurrent tumor can facilitate thera- 
peutic decision making when treatment options are 
considered. 

Often in the setting of Group 2 and 3 CBS, the use 
of cross sections I imaging studies are of limited util- 
ity, owing to the emergent nature of these presenta- 
tions and need for prompt and definitive interven- 
tion. However, in patients who are clinically stable or 
who present with threatened CBS, CT or MR imag- 
ing may serve the dual purpose of not only helping 
to elucidate the likely source of active or potential 
bleeding, but also assessing open surgical options 
for the overall management of the patient's disease. 
In our experience, close scrutiny of either contrast 
enhanced CT or MRI scans of the neck in the set- 



ting of CBS not infrequently shows evidence of one 
or more of the following features: pseudoaneurysm 
formation, pockets of extravasation, fistulae, tumor 
encasement of a large carotid vessel, or prominent 
tumor enhancement (secondary to neoangiogenic 
hypervascularity). These cross-sectional imaging 
studies may be supplemented with three-dimen- 
sional reconstruction CT angiography or MR angi- 
ography to better define the relevant cervical and 
cranial vasculature. These non-invasive imaging 
modalities have improved substantially over the last 
few years, and are increasing supplanting catheter 
angiography for initial diagnostic work up of a vari- 
ety of vascular diseases. However, in our experience 
we have found that both CTA and MRA have lim- 
ited capability in detecting the likely pathoetiologic 
sources of bleeding in the setting of CBS, particu- 
larly lesions involving the external carotid system. 
Therefore, at our institution we do not routinely 
utilize either of these modalities in the preoperative 
evaluation of CBS. 

As in the case lit obtaining a good directed clini- 
cal history and physical examination, recognition 
of the critical imaging findings relevant to opti- 
mal multidisciplinary management can greatly 
facilitate the localization ot pathology to target for 
intervention. With regards to open surgical plan- 
ning, demonstration of the presence and extent 
of recurrent neoplasia by cross-sectional imaging 
will to a large degree determine whether this is not 
only a therapeutic option, but also what endovas- 
cular approaches may be preferable to manage the 
CBS. For example, a patient who has been previ- 
ously "disease-free" or "disease-stable", but devel- 
ops a flap breakdown that results in threatened 
carotid blowout, may also harbor an unrecognized 
primary or nodal recurrence that is amenable to 
salvage resection. In such cases (particularly if the 
mass is attached to or encasing the carotid artery) 
it may be preferable to perform therapeutic occlu- 
sion of the artery (providing that the patient tol- 
erates balloon test occlusion) above and below the 
expected margins of resection in order to simplify 
such an operation. 

Ultimately, the gold standard for diagnos- 
tic evaluation of a patient with CBS is a meticu- 
lously performed catheter angiography study of 
the head and neck, using modern high resolution 
(1024x1024 pixel matrix) and preferably biplane 
digital subtraction imaging (DSA) of the cervi- 
cal and intracranial carotid circulation. This 
requires selective catheterization and injection of 
each common carotid, external carotid, and not 



infrequently internal carotid artery performed in 
at least three different projections. Frequently the 
pathology is obvious [4, 27], such as large pseu- 
doaneurysm formation, active extravasation, or 
fistula formation arising from the ICA, ECA or 
CCA (Figs. 21.1-21.3). In some cases the source of 
bleeding is not from a ruptured vessel within the 
carotid tree, but rather from a recurrent tu 
arising from primary or nodal metastatic s 
The mechanism of such bleeding may be ei 
secondary to invasion and/or erosion tl 
oro-pharyngeal or cutaneous anatom: 
or from stimulation of tumor neoangiogenesis 
resulting in hypervascularity [27]. 

It must be emphasized, however, that obvious 
pathology is not always apparent, requiring one to 
maintain a very high index of suspicion when scru- 
tinizing the angiographic images. In such cases we 
have frequently observed very subtle, and/or non- 
specific alterations in endoluminal contour on close 
inspection of selective (and occasionally superse- 
lective) injection DSA runs that have ultimately 
turned out to be the source of bleeding in patients 
with Groups 2 and 3 CBS (Fig. 21.4a). The areas that 
require the closest scrutiny are around the common 
carotid bifurcation, including the external carotid 
trunk and its lower branches (e.g. superior thyroid, 
facial and lingual branches), and the internal carotid 
bulb. 

If bleeding is encountered within the lower 
neck (particular when involving a tracheostomy 
site), bilateral selective arteriography of the sub- 
clavian, costocervical, and thyrocervical arter- 
ies also must be performed. Angiographic study 
of these vessels usually can be closely correlated 
with the actual site of bleeding (which should be 
marked by a radio-opaque object). In our experi- 
ence, the tracheostomy site or underlying distal 
margins of a surgical neopharynx are the most 
commonly affected structures, which typically 
bleed from tumor recurrences, erosions, and radi- 
ation-induced necrosis. 

The anatomic substrate of potential collat- 
eral pathways (particularly involving the circle 
of Willis) must also be assessed, which requires 
angiographic study of the intracranial circula- 
tion from selective carotid and vertebral injec- 
tions. Prospective identification of major breaks 
within the circle of Willis that result in partial 
or complete isolation of the cerebral circulation 
[e.g. ipsilateral absent Al segment and posterior 
communicating artery (PCoA)], is often enough 
to dissuade one from performing a balloon test 




Fig.21.1. Angiogram shows a Ia:ge I'seudo.ineurysm of the 
common carotid o j lei y iltrgt' ,n rot; }. Narrowing of the inter- 
i'.ii] c.U'xid ane:y \:ii:..iil ,ifty-.;s) is a!so piesen;, mos: likely 
related to postoperative cicatrization, radiation -induced 
changes, or tumor ei 




Fig. 21. 2. Left common carotid ane:v iO_"A :■ angiogram shows 
a small amount of extravasation from the CCA into the oro- 
pharynx (ijjtch-) from a ris-nla That developed after rupture 
of a pseud oa neurysm 



occlusion in consideration for a deconstructive 
intervention. Other anatomic variants, such as a 
persistent trigeminal artery are also likely to dis- 
courage consideration of a therapeutic occlusion 
of the ipsilateral carotid artery. 



Diagnosis and Endorascu.ar Surgical Maaagemenl of O.rotid Blowout Syndror 




Fig. 21..*. a Ria. - .: C'.:v.;>: n i - ,i ;■'.:■; :■,] .jrlei'y : - l : a l ■■ ■ a : n n l j.'.ows :i iai gc c^rad. -.i^e.i i yj;ii ..■[' ;l;- :a.." -Cc-:vical i ighi nii-ri :".al caood 
;:: :ery (ICAi (/dijy .'n/j:/ ,ii/ii;.!. Small concurrent p s en d o p. lieu rysjns of the proximal right ICA tirui'ivti iinon-i) and riglil 
externa I carotid artery iKCA! in/H-'" itt'irorj are alio pre ; enl. Ii jiigat .:o ::".::". on caivi.d anerv laieciion after :■ alio on occlusion 
of ::g::: o-.'A sj-.ow lire preiidoaneurysm o: the ::g:\: !-_'." A :.iii ran :■ ■= r i . I ■;..[■■. c Angiogram ■ :■■ l -1 : :i j :~. c ... af>r composite pertv.ao.ea.t 
balloon occlusion show- embolization ::-i the crucaid systeiv.. A oalloon had oeen oosil loned across i tie origin o: the riglil E'.IA 



(<" 



*) 



21.5 

Endovascular Techniques 

Over the last decade, dramatic technical and techno- 
logical progress in endovascular surgery has greatly 
expanded its role in the management of many types 
of intracranial and extracranial cerebrovascular 
diseases. These advances have been particularly 
significant in two categories: (i) new micro-cath- 
eterization technologies and (ii) new endovascular 
stent systems. Both have yielded devices that are 
safer, easier, and ultimately more effective to use. 
For now nearly two decades it has been increas- 
ingly recognized that the acquired arterial injuries 
of CBS are often readily amenable to endovascular 
intervention. However, the specific indications and 
appropriate selection of techniques and technology 
had only recently become better defined in the past 
decade [3, 4, 21]. This definition is already undergo- 
ing substantial revision and evolution in response to 
inherent limitations and complications of various 
endovascular approaches [21, 43] and our expanded 
technical and technological capability. 

Endovascular management of CBS can now be 
broadly defined into the two following categories: 
"deconstructive" and "reconstructive" approaches. 
The former consists of various techniques and tech- 
nology used to produce therapeutic occlusion or 



"sacrifice" of an affected artery. In contrast, the 
latter category consists of various techniques and 
technology for eliminating bleeding while preserv- 
ing the patency ot an affected carotid artery. 



21.5.1 

"Deconstructive" Techniques 

Therapeutic occlusion of major brachiocephalic 
arteries is one of the oldest neuroendovascular thera- 
peutic techniques, originally having bet 
for the treatment of inoperable aneur 
cranial base using simple open ligation or g 
clamping (e.g. Seleverstone clamp) of the ca 
arteries. Both Serbinenko [30, 31] and Debru: 
44] are credited with pioneering the endovas 
equivalent of such an approach through thei 
of detachable balloons for the treatment of a 
types of giant aneurysms (e.g. cavernous segment 
in the mid and late 1970s). Today, besides detach- 
able balloons, a wide variety of embolic devices 
and endovascular techniques may be employed for 
deconstructive therapy of CBS. The selection of a 
particular device or technique for therapeutic arte- 
rial occlusion may be based upon a variety of fac- 
tors, including: the specific pathoetiologic lesion 
identified on angiography, prevailing cerebrovas- 



> of the 

rotid 
*[32, 



J. C. Chaloupka 



cular hemodynamics, perceivt 
nal carotid artery occlusion 
test occlusion (BTO), access ti 
for rapid and definitive occlu: 



d tolerance to inter- 
based upon balloon 
the lesion, urgency 
lability 






and/or comfort level with certain techniques/tech- 

Detachable latex (Goldvalve, Nycomed) or sili- 
ichable (DSB, Boston Scientific-Target) bal- 






e of thee 
g rapid o 



loons remain as one of the easiest and most reliable 
in of a large vessel, 
1 carotid arteries, 
and therefore in my view continue to be essential 
embolic devices tor such applications. These devices 
can be readily mounted on various conventional 
microcatheters (e.g. Tracker 18 HiFlo Unibody, 
Boston Scientific-Target, or Prowler 14 (Cordis Neu- 
rovascular, Miami Lakes, FL), and once navigated 
into the targeted vasculature, rapidly inflated and 
deployed by simple traction detachment (Figs. 21.3, 
21.4) As a general rule it is most desirable to place 
the first balloon into a fairly distal purchase within 
the ICA (typically the petrous segment), owing to 
the risk of thromboembolism that may occur from 
the creation of a long propagating thrombus in case 
of more proximal placement of the balloon within 
the cervical carotid [4]. A second important rule 
for therapeutic carotid occlusion is to always use a 
second so-called "safety" balloon (Figs. 21.3c, 21.4b) 
to ensure permanent occlusion, since detachable 
balloons on occasion are well know to prematurely 
deflate. Optimal placement of the second balloon 
will largely depend upon the location of the breach 
within the carotid artery, since trapping of the 



defect is the effective means of stopping/preventing 
further hemorrhage [4]. In some cases, therapeutic 
occlusion of the entire carotid system (ICA, CCA, 
and ECA origin) is necessary or desirable, such as in 
cases of multifocal pseudoaneurysms and fistulaeor 
in patients with Group 1 CBS who are candidates for 
salvage surgical resection of local recurrent disease. 
In such case performance of a so-called composite 
carotid occlusion is indicated [4, 21, 27], 

Unfortunately, (he use and availability of detach- 
able balloons has been steadily on the decline over 
the last few years, which just recently were mark- 
edly amplified by the abrupt discontinuation of the 
only FDA approved detachable balloon in the United 
States (DSB; Target-BSC). Currently, it remains 
unclear if this device will be reintroduced, which 
has created a new impetus in utilizing alternative 
techniques and technology. 

Many centers have been using various types of 
embolic coils (e.g. non-retrievable fibered plati- 
num, detachable bare platinum) as an alternative or 
adjunct to balloon occlusion of the carotid arteries. 
Our group was first to propose using detachable coils 
in combination with detachable balloons in cases 
where composite carotid occlusion was needed [4]. 
Although these conventional embolic coils can pro- 
duce therapeutic occlusion of large caliber vessels, 
such as the common and internal carotid arteries, 
there are several intrinsic disadvantages in using 
them as the sole embolic agent. These disadvantages 
include: (i) lack of precision in deposit ion within the 
distal targeted vessel segment; (ii) frequent instabil- 
ity of the first "framing coils" in large caliber arter- 




a Right common carotid artery angiogram shows a subtle pseudoai 
tow), b Lateral radiograph siiows L'.ree .:. era ."liable balloons used i: 
distal balloon cove::; [he of. lice o: the oseiuloaneurysm (arrow) 



? ! .: its m of the cavernous inter mil carotid artery 
permanent balloon occasion of the right ICA. 



Di.gnc 



ir Surgical Management o: O.;odd Blowout Syndrome 



ies that are typi.allv carrying high volumetric blood 
flow; (iii) need to use a relatively large number of 
coils to achieve occlusion; (iv) increased procedure 
time and expense owing to the typically long times 
required to achieve arterial occlusion; and (v) pos- 
sible increased risk of artery to artery thromboem- 
bolism as progressive thrombosis within the coils 
occurs when there is still antegrade flow within the 

A new detachable platinum coil coated with a 
hydrogel polymer recently has been introduced 
for intracranial aneurysm therapy (HydroCoil, 
Microvention), which our group and others have 
found to be also useful for carotid occlusion, at least 
in potentially decreasing She number of coils needed 
to occlude the targeted artery. Unfortunately theses 
coils still suffer from many of the same above-men- 
tioned disadvantages. Although no definite proto- 
types have been revealed, a few device companies 
have indicated some interest in creating more novel 
large vessel occlusion devices that may incorporate 
certain already existing technologies, such as down- 
stream embolic filtering devices, stent grafts, and 
hydrogel polymers. 

In cases requiring smaller vessel occlusion, such 
as pseudoaneurysms or tears to the ECA and bra- 
chiocervical branches, both nonretrievable plati- 
num microcoils (e.g. Tornado, Cook, Indianapolis, 
IN; Vortex, Boston Scientific-Target, Freemont, 
CA), and various retrievable/detachable platinum 
microcoils (e.g. GDC, Boston Scientific-Target) can 
be used. Owing to the enhanced control and safety of 
the latter type systems, our group has long switched 
over to using them exclusively. Occasionally, our 
group has utilized careful injections of small ali- 
quots of cyajionavhle-iod-inized oil embolic mix- 
tures (Trufill-nBCA, Cordis Neurovascular) to very 
effectively obliterate small distal pseudoaneurysms 
of the ECA circulation [45] (Fig. 21.5). 

Occasionally some cases of CBS are ultimately 
found to be the result of hemorrhage from a hyper- 
vascular neoplasm (typically in our experience 
from transformed squamous cell carcinomas that 
have exuberant induction of angiogenesis after mul- 
timodality treatment). Endovascular therapeutic 
devascularization can be achieved with superselec- 
tive transarterial embolization using either con- 
ventional embolic agents such as polyvinyl alcohol 
(PVA) particles (Contour, ITC/Target Therapeutics/ 
Boston Scientific, Freemont, CA} or more recently 
with the slow polymerizing cyanoacrylate 2-octyl 
cyanoacrylate (e.g. Dermabond) [46]. Additionally, 
in cases where superselective catheterization of sup- 



plying arteries is not feasible, we have successfully 
utilized direct puncture tumor embolization with 
either absolute alcohol or cyanoacrylate embolic 
mixtures [47]. 



21.5.2 

"Reconstructive" Techniques 

In lieu of the inherent risks of unilateral therapeutic 
ICA occlusion, the unfeasibility of bilateral ther- 
apeutic ICA occlusion in most patients who have 
already undergone a previous carotid occlusion, and 
the high rate of recurrent disease resulting in future 
episodes of CBS, there has been a growing impetus 
to alternatively consider endovascular repair of the 
damaged arterial segment, in an effort to preserve 
flow within the vessel. Therefore an alternative 
strategy tor (he management of CBS is to reconstruct 
the damaged artery through the various mechani- 
cal and biological effects of endovascular stents [38, 
40-43, 48]. 

With the increasing use of stent-assisted angio- 
plasty for the treatment of extracranial carotid 
occlusive disease, it is becoming more widely 
acknowledged that such endovascular reconstruc- 
tive techniques can be performed with a relatively 
low rate of peri-operative cerebral ischemic compli- 
cations [42, 49-52]. These ischemic complications, 
on average appear to be lower than those for thera- 
peutic occlusion of the carotid artery using either 
open or endovascular techniques. 

There are clearly a few important theoretical 
advantages afforded by stent reconstruction of the 
carotid artery that may ultimately enhance the over- 
all efficacy of endovascular management of CBS. 
First and foremost, such techniques are aimed at 
preservation of the affected carotid artery, thereby 
having the combined effect of broadening applica- 
tion of endovascular intervention (for example in 
patients who outright fail carotid BTO or possess 
an isolated circulation from an incomplete circle 
of Willis), and reducing delayed ischemic compli- 
cations (which can be both thrombo-embolic and 
hemodynamic) Second, the scaffolding and posi- 
tive remodeling force induced by placement of one 
or more self expanding stents not only maintains 
endoluminal patency, but likely reinforces the 
mechanical integrity of the arterial wall, which in 
the case of CBS has been damaged by the previously 
described processes developing as a consequence of 
therapy. This ultimately should result in an artery 
that is more resistant to future spontaneous rup- 




Fig. 21.5a-f. Iatrogenic pseuooanenrysm .:■[' an exle:"ji..i I carol id artery branch af:er '."'-guided biopsy, a Axial noiier.fianced 
'".".' image snows 1 lie tip of a .ii 2 -gauge :Ili ib.i needJe within li'.e aiiierior aspect of a sort- i it sue abnormality icniivi/ iinvirj that 
Wiii concerning for recurrent sqa anions ceil carcinoma. Histologic analysis showeri only iiiflamma:o:y changes, b.c Two weeks 
Liter li'.e patient presents ■.villi artenalizeri epistaxis. l> 'selec:ire righ: external carotid arteriogram, lateral proieciion, shows 
mi Id iiTegu.ar.ty •:■: the distal internal maxiilary artery and focai dil.italion of die distill buccal branch idrii'n ). c Peiayed image 
from same injection shows ri|j.;g . .;' a 5-::.::'. pser.doaneurysm : inToir:. d-f iindovasculii: surgical management. .1 vhiperseleci.ve 
angiogram, lateral oro:ertion afier catheter. zation :■( I he coninion I run I, o: ti'.e descending pa.aiine and bucca. aiinies, slic'ws 
filling of the p sen doa neurys in : /,?iyr am' no and adiacenl ex:ravasation i.stiiiill iirrano. e '. -e laved image iiins-ihiracted ) from 
same iniection slic'ws persistent tilling of li'.e psetidoaneurysm. I Selective r.giii external carotid arte no gram immediaiely afler 
embolization shows complete occlusion of The buccal nranch pseudoaneurysin 



iiisii.il Wi'.ii.i^cmenl '.:■:' C.::'o:ii1 ivowoiii Jyiiifo 



Cure. Another putative benefit of stent-assisted 
reconstruction of an affected carotid artery is the 
favorable alteration in flow mechanics that lead to 
a reduction in disturbed blood flow and eddy for- 
mation that has been linked Co cerebral artery-to- 
artery thromboembolism [42,52]. Such favorable 
alterations in hemodynamics may translate into a 
significant risk reduction of thromboembolic isch- 
emic complications. 

Consequently, over the last 6 years our group has 
progressively moved from a primarily "deconstruc- 
Cive" to a primarily "reconstrucCive" paradigm for 
the endovascular management of CBS. Thisapproach 
first originated in Che mid 1990s at Yale, when we 
occasionally encounCered paCients who were noC 
candidates for therapeutic carotid occlusion owing 
Co ouCright failure of a balloon test occlusion or 
angiographic demonstration ot an isolated anCerior 
hemispheric circulaCion (i.e. hypoplasia/aplasia of 
Che ipsilaCeral Al segment and posterior communi- 
cating arCery). During this time our experience with 
carotid scenting was starting Co evolve, providing an 
opportunity Co try this alternative technique in a few 
selected patients [39, 43]. Over time we broadened 
inclusion criteria to all patients presenting with CBS 
who were at high risk of a stroke from therapeu- 
tic carotid occlusion. This included patients with 
advance age (>65 years), significant cardiovascu- 
lar and/or pulmonary co-morbidities, significant 
relative reduction of CBF (> 20%) based on cere- 
bral SPECT imaging during BTO, and contralateral 
carotid occlusive disease. Within the last 3 years at 
UIHC, we have now shifted to attempting a recon- 
structive procedure whenever feasible if the affected 
arterial segment involves the common or internal 
carotid artery, with the expectation that such a shift 
in therapeutic strategy could serve the primary 
purpose of reducing overall risk of peri-operative 
stroke, while at the very least keeping the rates of 
immediate and delayed hemorrhagic arrest at the 
same level of our previous experience with decon- 
sCrucCiveCechniques [43], Arterial injuries involving 
Che exCernal carotid tree or rarely branches arising 
from the subclavian arteries continue to managed 
by deconstructive techniques as was originally con- 
ceived in our early experience. 

We have successfully utilized a few different tech- 
niques of stent-reconstruction for the management 
of CBS, which can be broadly classified as follows. 
First, in a similar fashion to techniques developed 
for the treatment of wide-neck aneurysms of the 
intracranial circulation [37,53-56], endovascu- 
lar stenting across a pseudoaneurysm can be per- 



formed for neck-bridging scaffolding to eventually 
support endosaccular embolization [38,39,43,48] 
(Figs. 21.6, 21.7). The rationale tor such an approach 
originally derived from animal studies of experi- 
mentally constructed side-wall aneurysms, which 
showed that stent placement alone could result in 
sufficient flow diversion and altered recirculation 
that ultimately produces thrombosis and neointi- 
malization of the aneurysm sac and ostium, respec- 
tively [53-55,57]. However, the time required for 
aneurysm obliteration can be substantial in such 
circumstances, which prompted the idea of trying 
to conjointly use thrombogenic coils placed within 
the aneurysm after deployment of the stent, in order 
to promote rapid thrombotic occlusion [37,55], 
This strategy has indeed been used successfully in 
an increasing number of clinical cases [39,43,48], 
although its application in the treatment of large 
pseudoaneurysms is questionable [39]. We have 
also experimented with use of alternative embolic 
agents, such as liquid adhesives [39], which can be 
delivered either by endovascular catheterization or 
direct puncture (Fig. 21. 7e) of the pseudoaneurysm. 
Although we have had success with such techniques 
in a small number of cases, widespread adaptation 
has been limited owing to concerns of proper con- 
trol and stability of the liquid embolic agent. 

The second technical strategy that we have suc- 
cessfully employed for management of CBS is the 
use of overlapping, self- expanding stents deployed 
across the neck of a pseudoaneurysm or along a 
weakened segment of the affected artery [43,58], 
The major advantage of such a strategy is that the 
effective porosity of Che stent cell configuration is 
substantially decreased, resulting in a combination 
of additional flow diversion away from the aneu- 
rysm sac, and markedly reduced bulk flow out of the 
aneurysm sac. Such alterations in fluid mechanics 
may diminish growth and re-rupture of the pseu- 
doaneurysm, as well as promote its thrombotic 
occlusion (both spontaneously and iatrogenically 
with the assistance of embolic agents). Further- 
more, the added relative coverage and scaffolding 
afforded by placement of a second stent allows for 
better bridging of a pseudoaneurysm neck, which 
in turn facilitates more secure placement of embolic 
agents (particularly coils). In fact this approach has 
overall been so successful that it has now become 
essentially a standard strategy in all our cases when 
utilizing uncovered stents for endovascular recon- 
struction (Fig. 21.6). 

For reconstruction of the common carotid or 
proximal cervical internal carotid artery, we have 




Fig. 21.6a-f. Steni-assisleo 
projection) shows two larg 
the internal carotid artery 
navigation confirms propei 
conlroi angiogram after pla 
and moderately reduced til 
carotid injection 46 h after 
gram shows persistence of 
with a microcatheter, and 
pseudoaneurysm. No 
syndrome in whom a large 



coiling of pseudoaneurysm. a Pre-rreatmeiii angiogram, right common carotid injection (lateral 
e pseudoaneurysms of the right cent r.; on carotid anery :n a patient with Group 2 CBS. Note that 
had been previously occluded from an earlier episode -.A 'ZhS. b Control angiogram during stent 

positioning of the stent over the diseased segment or trie right common carotid artery, c Delayed 
cement of a single Smart Stent. Note i It e markedly oi;v. mi shed f.llmg of the ventral pseudoaneurysm 
ling of die dorsal aneurysm. Active bleed .ns; stopped sti'.'itly after ceployment. e-f Right common 
first intervention, d The patient developed a seconc episode or '.I.KS 1 days later, in which the angio- 
the dorsal aneurysm after sient placement, e The pseudoaneurvsm is. entered through a stent cell 

tr anting detachable coil is p.ace.s. f The 'ins. con I :o. anjtioftram s stows cotr.p.ete ooltteral ion o: the 
bleeding was subsequemly encotintered. A !-!-vear-olc woman with recurrent carotid blowout 
pseudoaneurysm developed 



Diagnosis and Endov 



if Siirgicjl M ;'.:!.! gem ent o: 0.;e.dd Blowout Syndic 



favored using Nitinol self- expanding stents (e.g. 
Precise, Cordis tndovascular or Acculink, Guidant), 
owing to a combination of factors that include: ease 
of access with a long (90-cm) 6-F sheath, high pre- 
cision and accuracy of delivery, good conformity 
within tapering segments, and relatively high posi- 
tive remodeling force after deployment. Our pre- 
ferred techniques for carotid stenting are similar to 
those used for extracranial atherosclerotic revascu- 
larization [42]. For initial access into the carotid cir- 
culation, there are two basic techniques commonly 
employed. In both cases, a large inner diameter 
guiding catheter or sheath must be positioned into 
the common carotid (or occasionally the innomi- 
nate artery). It has been our experience that place- 
ment of a long arterial sheath from a percutaneous 
common femoral artery puncture is preferable from 
multiple perspectives, including enhanced stability, 
ease of coaxial delivery of PTA and stent catheters, 
and minimization of the size of arteriotomy. This 
technique is performed as follows. A standard 6-F 
arterial sheath is inserted into the common femo- 
ral artery after single wall percutaneous puncture 
and guide wire insertion. A 5-F diagnostic cath- 
eter is carefully positioned within the appropriate 
great vessel (usually the external carotid artery 
origin during internal carotid artery PTA/Stenting), 
for subsequent placement of an exchange length 
(260 cm) 0.035" or 0.038" (e.g. Amplatz regular or 
extra-stiff) wire. The diagnostic catheter and sheath 
are carefully removed, while maintaining the distal 
purchase of the guide wire. For extracranial stent 
cases where a 0.018" SmartStent (O.D. = 2.3 mm) or 
Wallstent (O.D. = 2.4 mm) will be deployed, a 7-F, 
90 cm Shuttle Sheath (I.D. = 0.10", O.D. = 0.131") 
is carefully advanced over the stiff exchange wire, 
until a stable position within the parent lesion vessel 
(distal common carotid artery) is obtained. We 
prefer the final sheath position to be at least 1-2 cm 
below the lowest planned stent placement to mini- 
mize potential stent deployment difficulty. We also 
find that placing a wide arcing curve on the Shuttle 
inner dilator and distal sheath, as well as removal of 
the intervening rotating hemostatic valve facilitates 
navigation of tortuous aortic arches and brachioce- 
phalic vessels. Care must be taken to hold both com- 
ponents (dilator and sheath) together during initial 
advancement. Subsequently, the outer sheath is 
independently advanced into position over the dila- 
tor and guide wire within the proximal brachioce- 
phalic vessels. This latter maneuver is critical, since 
the dilator is poorly seen on fluoroscopy (without a 
marker band), and therefore may be inadvertently 



advanced too far distally into the stenotic lesion if 
both components are advanced in tandem. An alter- 
native approach that may be employed requires a 
coaxial technique utilizing a diagnostic catheter (5- 
F, 120-cm) and guide wire (0.035" or 0.038") within 
the 90 cm Shuttle sheath, and subsequent staged 
advancement of each into the appropriate position. 
The diagnostic catheter and its associated curves 
may assist in distal navigation, while also minimiz- 
ing wire/catheter/sheath transitions. Following sta- 
bilization, the sheath is then connected to a large 
bore rotating hemostatic valve, and continuous pres- 
surized heparinized saline infusion. In some cases, 
smaller delivery devices may be utilized, such as 
coronary PTCA and stent microcatheters, in which 
case a 6- or 7-F thin walled guiding catheter with 
larger internal diameters (e.g. Envoy, Guider, Plat- 
form) may be placed primarily or over an exchange 
guide wire. Appropriate testing of device/catheter 
compatibility is recommended before attempting 
therapy. 

Generally, self-expanding stents (Smart Stent, 
Wallstent, Precise stent) are preferred within the 
extracranial vessels, especially at the level of the 
common carotid bifurcation. The Wallstent has 
a greater strut density, but has a tendency to fore- 
shorten significantly during deployment, which 
makes accurate sizing and placement more dif- 
ficult. Although more porous, the Smart Stent 
deploys with relative precision and minimal fore- 
shortening. The Precise and Acculink stents have 
a much lower profile than the Smartstent, allow- 
ing facile, less traumatic passage through tortu- 
ous anatomy or stenotic segments. Of the two, the 
Acculink has somewhat less radial force (positive 
remodeling force), which may be an advantage for 
treating pseudoaneurysms(i.e. less risk of iatrogenic 
rupture). Appropriate measurements of the parent 
vessel diameter and length of desired coverage are 
required. For the management of CBS, we prefer to 
use a stent of 1- to 2-mm greater diameter than the 
parent vessel (e.g. a 6-mm ICA would receive a 7- to 
8-mm diameter stent). Slight oversizing of the stent 
ensures adequate radial tension and approximation 
ot the device to the eiuioluminal surface, preventing 
migration and early thrombosis. This oversizing also 
may promote a delayed mechanical and biological 
remodeling of the diseased segment through grad- 
ual continued dilatation, and stimulation of various 
growth factors and extracellular matrix proteinases 
(occurring over days to weeks). Excessive oversiz- 
ing of the self-expanding stents should be avoided, 
since this may result in vessel wall necrosis, and pro- 




Fig. 21.7. a Oblique view or a : ighr common cai'oiiti drier v injection shows ].■ :ge pseud oancuysm isriuigiit ii-noui. Note evi- 
dence of prior surgery, including ligation or rhe ex rem.;! carotid artery (cm ivi/ ,l^^. |, ■■. i. b Angiogram afrer deploy mem of an 
8x_0-mm Wallsrent across ihe rem in lire carotid arrery shows good disra! runor'r our persistent rilling or" rhe pseud oan.eurysm. 
i' Repeat angiogram arlei oeplovnient of a second. overlappi::gi:>!*i'0- mm : IVallstenr shows Aw: tilling of the pseud oan.eurysm. 
d I 'elayed ::::age snows siag::aiion ;:if coiirrast material wit in:; the pseud oaneurysm (iji/pi; }. e rluoroscopic spc-r III in shows .: 
balloon occlusion carherer within rhe slenrs. wit!; the balloon pai lially mil a red {stt\i:glit iiiio-. i. This j] Jo wee. ioiik ret n. 'grade 
llc'W .;if opacilied blood ro rill ihe p send oaneurysm, which was percriraneously punctured with a h-Ci-gauge needle ici/iiyJ 
iPi'1'pi! }. f Control angiogram after I wo dnvct-puncture acrylic emoolirraiion^ shoots complete obi i term ion ;;if the pseud oar.cr;- 
rvsn: with patency of lire carotid at tery. Subtraction artifact is seen where NBC A was injected ar Ihe site of i upture (di 101; ). 
End ova scalar treatm^nr of a head and neck cancer- rehired CF6 oy use or' the self- expanding, covered VV'allgraft stenr 



Di.gnc 



if Snrgicjl M ;'.::.! gem ent o: O.;odii Blowout Synorc 



tracted carotid body stimulation of varying severity 
and clinical sequelae. We also prefer to use a stent 
5-10 mm longer than the lesion, to ensure adequate 
coverage of both the distal and proximal stenotic 
segments. Often it is necessary to cross the exter- 
nal carotid artery with a stent, owing to extension 
of disease into the common carotid bifurcation that 
needs revascularization. Our group and others have 
observed that this can be done with impunity, since 
the ECA in most cases remains patent on follow-up 
examinations. In the rare cases when the ECA does 
occlude (usually from severe coexistent atheroscle- 
rotic disease), patients always remain asymptomatic 
secondary to the presence of abundant collaterals. 
If a second stent is required, a self-expanding stent 
of equal or greater diameter is overlapped with the 
primary stent and appropriately deployed. If a self- 
expanding stent of a smaller diameter is positioned 
within a larger self- expanding stent, the risk of 
delayed dilatation and subsequent migration of the 
inner stent arises. On occasion, sell-expanding cor- 
onary stents (e.g. Radius, Magic Wallstent, Boston 
Scientific), balloon expandable coronary stents (e.g. 
S7-Medronic AVE, Vision-Guidant), or the recently 
FDA approved self-expanding neurovascular stent 
(Neuroform, Boston Scientific) may be required for 
use in smaller arteries (e.g. distal cervical or skull 
base portions of the internal carotid) or in chal- 
le using anatomy (e.g. excessive tortuosity or loops). 
Great care in proper sizing is requiring preventing 
excessive trauma (e.g. oversizing balloon expanding 
stents) or stent migration (undersizingboth balloon 
expandable and self-expanding stents). On post- 
deployment angiography, the stent margins should 
approximate the luminal diameter and geometry of 
the parent vessels, proximal and distal to the dis- 
eased segment. If poor approximation of the stent 
margins is observed, post stent dilatation or "flar- 
ing" using PTA balloons may be required, although 
in the setting of CBS may entail additional risk of 
carotid rupture. An alternative strategy is to use tele- 
scoping "bridging" stents (typically self-expanding) 
to remodel variations in contour and diameter of an 
arterial segment [58]. 

Finally, our group has slowly begun to utilize 
covered stents or "stent grafts", as a third alterna- 
tive technical strategy for endovascular reconstruc- 
tion of CBS (Fig. 21.8). These devices have slowly 
evolved over the last several years in which both bal- 
loon expandable and more recently, self- expanding 
designs have been devised using either native venous 
grafts or synthetic materials (e.g. PTFE) sewn into 
the inner portion of the metallic stent frame. Such 



devices have the major advantage of being more 
likely to produce immediate exclusion of an aneu- 
rysm from the parent arterial blood flow, which 
effectively results in an endoluminal bypass of the 
aneurysm. As with bare stent reconstruction for the 
management of CBS, proper sizing and precision 
of deployment are critical for technical and clini- 
cal success. However, unfortunately there remain 
significant major disadvantages of covered stents at 
this time. In general both balloon expandable and 
self-expanding designs are inherently larger in pro- 
file for a given targeted vessel diameter, therefore 
requiring larger access sheaths and delivery systems. 
These larger profile systems can be very difficult, if 
not impossible to safely navigate into the cervical 
carotid system. The balloon expandable stent grafts 
also suffer from problems related to the need for an 
open surgical harvesting of a vein graft, increased 
danger of pseudoaneurysm/parent artery rupture 
from high pressure inflation, poorer flexibility for 
navigation into carotid arteries, and less availabil- 
ity. Finally, all current stent-graft designs suffer 
from inherent increased thrombogenicity, resulting 
in increased risk of immediate or delayed in-stent 
thrombosis or thromboembolic cerebral ischemia. 
Unfortunately, although aggressive ant i- thrombotic 
adjunctive therapy using systemic anticoagulation 
and anti-platelet drugs diminishes this risk, there 
is a commensurate increased risk of perioperative 
hemorrhage (often catastrophic) from the CBS. 

Despite these limitations our group [43] and 
others [59-63] have successfully treated a limited 
number of pseudo-aneurysms of the common and 
internal carotid arteries. It is anticipated that future 
advances in material sciences and engineering will 
result in lower profile and more flexible self-expand- 
ing stent-graft systems that will pose comparable 
technical demands to bare stents. Partially porous 
synthetic graft materials also hold the promise of less 
acute and subacute thrombogenicity, since they will 
be composed or more biologically inert compounds 
and may more readily permit neointimalizalion. 



21.6 

Outcomes 

Since 1993, the senior author has managed over 150 
CBS events by various endovascular approaches, 
likely representing the largest single operator expe- 
rience in the world. With this greater than a decade 



of ci 






:ch ha< 




Fig. 21.8a-f. a Initial angiogram jwe-.ils a slump : iinou) ,ii the proximal ex;ernai i! rot id ai tery. b Microc.itheler i mention of 
the 5: limp during e nd ova sci: .a r exploration confirms a pse. doaneiiiysni as die source of hemo'ii huge. <: An s?. ?M-mm Wallgraft 
stent is positioned ■.villi in the common carotid artery and ICA pi notion, oridgnig _ .i 1 ■=- exit; nal carodd artery origin, d After 
deploying die sieni. angiography -hows exclusion 01 die pseucoan.t-ii ysm and normal cahtv: of die parent, stenled ail^iy. 
e 1 ngna; subtraction aiigiogiam mask :s shown fo: detail, f ? ho login pli oc die YVallgraf; stent is shown for detail 



recognizing the relative strengths and weaknesses of 
various endovascular strategies. 

Deconstructive techniques founded in sound 
basic endovascular principles rapidly proved to be 
quite effective in achieving short-term hemorrhagic 
arrest in patients with Group 2 and Group 3 CBS, 
which approached a rate of over 95% for the first two 



case series reported by our group at Yale [3, 4]. In 
particular, composite occlusion of the internal and 
common carotid segments affected by pseudoaneu- 
rysm formation using multiple detachable balloons 
and embolic coils immediately proved to be a supe- 
rior approach to historical cumulative experience 
with open surgical repair of the carotid artery. Peri- 



Di„gnc 



iiisii.il Milii.igcmeiil o:' C.::'o:ii1 ivowoiii L : yn.:;c 



operative major morbidity (neurologic and hemor- 
rhagic) and mortality was surprisingly low, initially 
in the range of 10% and 2%, respectively. With 
increasing experience, the types of deconstructive 
techniques applied to the management of CBS nar- 
rowed Co mostly combined detachable balloon and 
coil embolization of the larger arterial segments (i.e. 
composite parent artery occlusion of the common, 
internal and external carotid arteries), liquid or coil 
embolization of pseudoaneurysms of the external 
carotid artery branches, and tumor embolization 
using either particulates or liquid adhesives. 

Ou [ group's first large cumulative case series [21] 
evaluated 46 consecutive patients with a clinical 
diagnosis of CBS between 1993 and 1997, who were 
referred for a total of 62 events for evaluation and 
intervention. Of these patients 43 had undergone 
extensive primary and/or salvage radical resec- 
tions and had received additional adjuvant therapy/ 
intervention (e.g. external beam and brachytherapy 
irradiation, and chemotherapy) that is believed to 
be associated with increased risk of CBS. Again, 
overall, the short-term safety and efficacy of endo- 
vascular management of CBS remained excellent, 
consisting of 100% initial hemorrhagic arrest, 7% 
minor neurologic morbidity (0% major neurologic 
morbidity) and 2% mortality. However, we discov- 
ered a disturbing new problem of additional events 
of CBS occurring in previously treated patients that 
we coined "recurrent carotid blowout syndrome" 
(rCBS). Our group was indeed the first to describe 
rCBS, having recognized this previously unde- 
scribed complication while reviewing our cumula- 
tive first 4 years of experience with endovascular 
management of CBS [21]. We specifically defined 
rCBS as either a repeat episode of self-limited or 
uncontrollable bleeding (i.e. Groups 2 and 3) occur- 
ring within 12 h of completed therapy for a previous 
episode of CBS, or a newly exposed portion of the 
carotid system (Group 1} occurring any time after 
completed therapy for a previous episode of CBS. 
In this series we found an astonishing 26% rate of 
rCBS in treated patients, with an average number of 
episodes of almost three per affected patient. Also 
interestingly, there was considerable variability 
between episodes of rCBS , ranging from 1 day to as 
long as 6 years. 

Upon reviewing patterns of recurrence in CBS 
patients, we observed two readily discernible cat- 
egories that could be linked to either proven or pre- 
sumptive pathoetiologic mechanisms of recurrent 
hemorrhage. The first category could be broadly 
defined on the basis of what we hypothesized to 



be so-called progressive disease (PD). All patients 
within this group developed new events of either 
potential or actual hemorrhagic recurrence, attrib- 
utable to one or more putative etiologies: (1) surgi- 
cal wound dehiscence, (2) free pedicle or mobilized 
musculocutaneous flap necrosis, (3) iatrogenic 
mechanical vascular injury, (4} radiation induced 
arteriopathy, (5) tumor invasion of a major arterial 
segment, and (6) recurrent tumor growth and inva- 
sion into adjacent mucosal surfaces. 

In terms of prognosis and management strategy, 
rCBS attributable to PD could be further classi- 
fied into bleeding arising from vasculature that is 
either ipsilateral or contralateral to the pathoetio- 
logic lesion causing the index event (i.e. original CBS 
episode). This was the most commonly encountered 
type of rCBS, occurring in approximately 65% of 
cases. In the PD group 54% had recurrent ipsilateral 
disease and the remaining 46% had recurrent con- 
tralateral disease. 

The second category of rCBS was best defined 
simply as events attributable to treatment failures 
(TF) (i.e. rCBS attributable to same affected arte- 
rial segment or territory that had been previously 
treated}. This included recurrent bleeding from a 
previously treated arterial pseudoaneurysm, arte- 
rial fistula, or tumor neovasculature. Interestingly, 
the time course between recurrent events attrib- 
utable to TF tended to be relatively short, varying 
between 1 and 10 days. In our cumulative series we 
found that approximately 32% rCBS could be attrib- 
utable to treatment failu res. 

Despite our short-term ability to arrest or prevent 
hemorrhagic complications in patients presenting 
with CBS, the various [>atlioloetiolo;;k" substrates 
that originally may have been responsible for the 
index event often persist chronically in this patient 
population. This persistence of pathoetiologic fac- 
tors, which frequently occur bilaterally, would 
explain the frequent occurrence of rCBS attributable 
to both ipsilateral and contralateral PD. It is inter- 
esting to note that within the PD group of patients, 
nearly half had an episode of rCBS attributable to 
disease occurring contralateral to the index event. 
This surprisingly high rate of contralateral events 
has the potential of creating serious longitudinal 
complications, particularly in situations where a 
new pseudoaneurysm develops on the common 
and/or internal carotid artery contralateral to a 
previously occluded internal carotid artery. In such 
cases, therapeutic parent artery occlusion of the 
remaining carotid artery carries a very high risk of 
catastrophic cerebral ischemia. 



J. C Ghalonpka 



Although less common than PD, TFs also consti- 
tute a major cause of rCBS. This group of patients 
develops recurrent events that are directly attribut- 
able to repetitive bleeding occurring in the same pre- 
viously treated arterial segment or territory. Unlike 
the PD group, the time intervals between events 
for rCBS attributable to TFs are usually fairly short 
(typically in the range of 1-10 days). Several specific 
limitations of therapy may explain the associated 
high incidence oh CBS attributable to TF. Occasional 
technical failures (e.g. inability to cross the ostium 
of apseudoaneurysm for endovascular trapping) or 
device failures (e.g. premature balloon deflation) 
may occur. More commonly, endovascular TF maybe 
the result of recurrent tumor hemorrhages that were 
previously treated by partial transarterial emboli- 
zation with particulates. This technical approach to 
tumor devascularization has a higher probability of 
short-term failure due to the frequent inability to 
achieve ideal superselective microcatheterization of 
all arterial pedicles supplying the tumor. This type 
of limitation may be overcome with the application 
of direct puncture embolization techniques using 
either cyanoacrylate or ethanol [27, 47]. 

The apparent high incidence of recurrent CBS 
observed in our patient population raised impor- 
tant issues regarding assumptions of durability and 
optimization of endovascular therapeutic manage- 
ment, and served as the fundamental impetus to try 
alternative endovascular management paradigms. 
Unfortunately, we are more frequently encounter- 
ing situations in which patients may not be candi- 
dates for conventional therapeutic ICA occlusion, 
owing to one or more of the following factors: (1) 
clinical or CBF imaging failure of BTO, (2) sponta- 
neous occlusion of the contralateral ICA/CCA sec- 
ondary to concurrent occlusive arteriopathy, and 
(3) prior contralateral therapeutic ICA occlusion 
in rCBS. The option of extracranial to intracranial 
bypass in such individuals is usually not favorable 
due to the combination of technical limitations in 
performing this surgery inpatients with prior radi- 
cal neck surgery and irradiation, and the frequent 
urgency of presentation [21, 43]. Our group first 
described one such patient [39] in whom we initially 
attempted to treat a large pseudoaneurysm by endo- 
luminal exclusion using overlapping self-expand- 
ing stents. Although this was initially successful in 
arresting the index event of acute CBS (Group 3), 
the patient rebled 24 h later. This prompted us to 
attempt an unconventional heroic intervention (i.e. 
direct puncture acrylic embolization), which for- 
tunately was successful. However, such a technical 



approach has potential limitations and risks that 
make it unsuitable for a routine primary therapeu- 
tic modality [43]. 

Our center's preliminary experience with selec- 
tive use of stent-assisted carotid reconstruction 
consisted of 16 CBS events in 12 patients who ful- 
filled one or more of the above defined case selec- 
tion criteria. These included seven patients with an 
incomplete circle of Willis on catheter angiography, 
two patients with a contralateral carotid occlusion, 
and three patients who failed BTO. Ten patients had 
underlying head and neck cancer (83%), whereas 
two suffered post-traumatic CBS (17%). Acute hem- 
orrhagic arrest was initially achieved in all Group- 
2 and -3 patients; however, there were four patients 
(33%) who each had one episode of rCBS that neces- 
sitated additional endovascular intervention. Fortu- 
nately in all cases of rCBS, durable hemostasis was 
ultimately achieved after re-treatment. A total of 26 
stents were used, in which 22 (85%) of the stents were 
uncovered, and the remaining four stents were cov- 
ered. There was a 96% technical success rate in plac- 
ing the stents; the sole technical failure occurred 
when an autologous vein- cove red Palmaz stent 
failed to deploy in the carotid artery. In six of the 16 
procedures (37.5%), pseudoaneurysm embolization 
was also performed using adjuvant acrylic glue or 
platinum coils. 

One patient developed post- procedural tran- 
sient ischemia (TIA), but no permanent neuro- 
logical complications occurred in this series. No 
deaths were attributable to ERCA although one 
death occurred within the 30-day perioperative 
period. Specifically, one patient with advanced 
HNC (Group III) died from complications of dis- 
ease progression (pulmonary embolus) on the 
fourth post- operative day. These complication 
rates were comparable or lower than those previ- 
ously reported for endovascular deconstructive 
techniques and stent-assisted carotid angioplasty 
of atherosclerotic disease [4, 21, 51]. 

Unfortunately the rate of rCBS in this series 
(33%) was similar to that of our cumulative experi- 
ence using deconstructive techniques, highlighting 
the continued major shortcoming of endovascu- 
lar management of CBS in completely preventing 
recurrent hemorrhages. In all cases, rCBS occurred 
in the same carotid vasculature that was originally 
targeted for reconstruction, thus representing ini- 
tial treatment failures as previously defined. Of 
interest however, is that these events of rCBS only 
occurred when either uncovered stenting or adju- 
vant embolization was performed alone. When 



Diagnosis and Endov 



ir Surgical Management o: '.].'.;■ odd Blowout Syndrome 



these techniques were combined together, efficacy- 
appeared to be relatively better in preventing future 
hemorrhages. 

Based upon our experience in the current series, as 
well as extensive cumulative experience with endovas- 
cular revascularization of carotid occlusive disease in 
general [42], we strongly believe that a self-expanding 
stent is highly preferable to a balloon-mounted stent 
for the treatment of CBS, owing to a variety of techni- 
cal and biomechanical factors. Self- expanding stents 
(e.g., Waligraft, Wallstent, Precise, SMART) easily 
accommodate varying diameters of the carotid tree, 
especially at the transition from CCA to ICA, and are 
more forgiving when determining the proper diam- 
eter size needed. These stents tend to have decreased 
porosity compared to balloon mounted stents, which 
improves vessel support and promotes pseudoaneu- 
rysm thrombosis. They also have superior flexibility 
in conforming to tortuous segments of the carotid 
anatomy, permitting more facile and less traumatic 
deployment. Furthermore, the selection of a self- 
expanding stent is preferable to a balloon-mounted 
stent owing to the potential for arterial injury result- 
ingfrom the requisite high balloon inflation pressures 
needed for deployment. This is especially relevant in 
the setting of CBS, since the target arterial segment by 
definition has been weakened from a variety of pro- 
cesses that predispose to CBS. These concerns have 
indeed been confirmed recently by Kwok et al. [64], 
who presented an example of HNC-related CBS in 
which a covered stent (fostent) offered no protection 
to high-pressure balloon-stent expansion; extravasa- 
tion at both ends of the stent with persistent transoral 
hemorrhage required endovascular occlusion of the 
common carotid artery [64]. 

We recently have experienced excellent results with 
the currently available VValkratt in, rapid deployment 
ofthispolytetrafluoroet hylene -coveredstentproduced 
immed iate exclusion of the targeted pseudoaneurysm, 
while simultaneously providing reconstructive pres- 
ervation of the carotid artery (Fig. 21.8). However, it 
must be emphasized that the safe and effective use of 
the Wallgraft stent for CBS requires a variety of favor- 
able mwvomii: niiii palhophviioloiik lacior^ that were 
present in both of our patients. These factors include 
common femoral and iliac arterial anatomy that 
will permit placement of a large caliber (9- or 10-F) 
vascular sheath, simple curvature of the aortic arch, 
relatively straight carotid and brachiocephalic arter- 
ies, and the lack of coexistent atherosclerotic or post- 
radiation stenoses. Therefore, more widespread use of 
self- expanding, covered stents for the management of 
CBS will not be possible until significant technologi- 



cal improvements are realized, such as the creation of 
lower profile delivery systems, increased overall flex- 
ibility, and enhanced deliverability. 



21.7 

Future Directions 

Endovascular management of CBS has evolved 
considerably over the last 15 years, paralleling the 
evolution of the specialty as a result of various 
technical, technological, and intellectual inno- 
vations. Over this period of time we have seen a 
paradigm shift emphasis from deconstructive to 
reconstructive techniques. It must be stressed, 
however, that the full armamentarium of endovas- 
cular techniques should alivays be considered when 
confronted by each case of CBS, since often the 
"less elegant" deconstructive techniques may still 
be the best option for definitive management. This 
is especially true when dealing with pseudo-aneu- 
rysms orfistulae of the external carotid system and 
tumor hemorrhages, which really remain prefer- 
ably managed by conventional or unconventional 
embolization techniques. Unfortunately, extensive, 
multi-focal disease of the common and internal 
carotid arteries also remains difficult to recon- 
struct, and as such may be only amenable to com- 
posite parent artery occlusion procedures, using 
current techniques and technology. 

However, clearly our group and others have shown 
the feasibility in managing more cases of CBS with 
reconstructive endovascular surgery than what was 
originally predicted. As stent-assisted angioplasty 
has become increasingly utilized for a variety of 
diseases affecting the extracranial carotid system, 
major advances in technology and technique have 
permitted more facile, safe, and effective placement 
of stents into the carotid arteries. Consequently, 
techniques described previously such as stent- 
assisted coiling and overlapping stent remodeling 
are now being routinely used at our center, with 
at least equivalent (and possibly superior) clinical 
efficacy compared to conventional deconstructive 
techniques. We continue to modify and refine these 
approaches as increased experience and improved 
technology is gained. 

As noted earlier, stent-grafts will likely become 
more "user-friendly" for management of CBS, and 
maybe the better technological solution to the prob- 
lems and limitations that remain with current endo- 
vascular reconstructive techniques. Inparticular the 



promise of lower profile, more flexible and trackable 
devices will enable broader application of this tech- 
nology. We can also expect further enhancements 
in the mechanical and biologic properties of future 
graft materials, which will likely translate into better 
short- and long-term outcomes. 

Finally, it is predicted that modification/manipu- 
lation of biological response to implanted stents will 
also improve technical and clinical efficacy olencio- 
vascular reconstruction of CBS. As drug eluting 
technology has already made an enormous impact 
on diminishing neointimnl hvperplnslic restenosis 
instented coronary arteries, we are likely to see sim- 
ilar technology using synthetic pharmaceuticals, 
growth factors, cytokines or other biologic agents 
that will be applied to bare or covered stents. These 
bioactive agents will be utilized to manipulate spe- 
cific molecular and/or cellular processes that could 
promote certain desirable responses, such as rapid 
healing, neointimalization, and enhanced struc- 
tural support of the affected artery. 

With the advent of these improvements, it is 
almost certain that constructive endovascular 
approaches, without adjuvant embolization, will 
become the treatment of choice for repairing both 
the common and internal carotid arteries affected 
by CBS. We predict that the use of such devices will 
not only substantially diminish the immediate peri- 
operative complications, but also decrease the rate 
of rCBS attributable to treatment failure that has 
a problem with uncovered stents. 



References 

l.Hamby WB (1952) Intracranial aneurysms. Charles C. 
Thomas, Springfield, II, p 45 

l.A'c crn.it liy [ ( i 8) :■■) Surgical observaiioiis. Longmans, 
London, p 193 

3.Citardi MJ, Chaloupka JC, Son YH, Sasaki CT (1995] Man- 
agemeiu •:■< c;i:"o: id artery rupture by monitored endovas- 
cular therapeutic occlusion ! 1388-1994). Laryngoscope 
105:1086-1092 

4. Chaloupka JC, Putman CM, Citardi MJ, Ross DA, Sasaki CT 
! ] aa ;i) Endovascular dierapy o'i carond b-owcut syndrome 
.ii hrad .11.:. neci. surgical pa; i en Is: cvi-lvaig di.:gnosl.c ond 
niaiiageniem considerations. Am ' Xeuroradiol l7:S4.i-8?2 

S.BorsanyiSJ (1962) Rupture of the carotids following radi- 
cal neck surgery in irradiated patients. Evol Psychiatr 
(Paris) 41:531-533 

6.Ketcham AS, Hoye RC (1965) Spontaneous carotid artery 
:". it mo nil age ,if;er head and neck surgery. Am J Surg 
110:649-655 

7.Ru tledge RH.Cagle JE (1966) Vein wrap for carotid blowout 
in endarterectonnzed carotid artery. Arch Surg 92:94-95 



S.Marchetta FC, Sake K, Maxwell W i)9o7j Complications 
after radica' head and neck surgery performed through 
previously radiated tissues. Am I Surg 114:835-838 

9.SwainRE,Bi!lerHF,OguraJH (1974) An experimental anal- 
vsis of causative ractors and pioreclive methods in carol id 
artery rupture. Arch Otolaryngol 99:235-241 

l.Huvos AG, Learning RH, Moore OS (1973) Clinicopatho- 
logic study of -he resected carotid artery: analysis of o4 
cases. Am J Surg 126:570-574 

I.Ariyan S, Marfaggi RA, Harden G, et al (1980) An experi- 
ment! mode, to dei ermine the effects of adjuvant I he ra.'y 
■.in the incidence or' postoperative wound in fee: ion. I. Eva hi - 
..uiiig l-'ivoperalive Kaoialion TLeraoy. Pkisi Reconsir Sing 
65:328-337 

2. Sanders EM, Davis KR, Whelan CS, Deckers KR (1986) 
Tinea teo.ee carotid tupture: a comulicalion oi radical neck 
surgery.J Surg One 33:190-193 

i. Shumrick I 'A ! 1971; Carotid artery lupiure. Laryngoscope 
83:1051-1061 

i. Heller KS, Strong EW (1979) Carotid artery hemorrhage 
after radica. head and neck surgery. Am I Surg L ; ?:::-37- 
610 

j.Razack MS, SakoK (1982) Carotid artery hemorrhage and 
ligation in head ami nee-, cancer. I Surg u n co.ogy 1 9:1 S9- 
192 

o.Coleman Ij (198?) Treatment of the iiipiured or exposed 
carotid artery: a latiomil approach. SoutO: Med I 7S\li:.l- 
267 

7. Porto DP, Adams GL, Foster C (1986) Emergency manage- 



8.Maran AGD.Amin M, Wilson JA (1989) Radical neck dis- 
section: a 19 year experience. | Laryngol Otol 103:763- 
764 

9.Koch WM (1993) Complications of surgery of the neck. 
In: Eisele I" 1 ied! Complications in head and :teck ■airgny. 
Mosby, Philadelphia, PA, pp 393-413 

0. Ditmars ML, Klein SR, Bongard FS (1997) Diagnosis and 
management of zone III carotid injuries, itmiry 23:515- 
520 

1. Chaloupka JO. Kolh TC, I'jtman CM, Mitra S, Koss DA, 
Sasaki CT (1999) Kf;urronl carutid Mtiwout syndrome 
diagnostic a pcnl.. .'C| t ni;i.s in ;■. newly -vc 
■ognizeo subi'/.cr ;>r pancr-.N A.I Mi A— I Ntiir:r..di:v 
20:1069-1077 

2. Dandy WE I ■ l-.i. r,.~.i„ n-t.-jrys::i :i the jriierna" 

caroi id aiter.. ..uicd u; ... 1 t...i.u..;. Aim ouig i .... .u.-,-u.^ 

o. Handy WE :. : t; 42 ) Res u. is fo. lowing .ig.cioii of the internal 
carotid artery. Arch Surg 45:521-533 

4. Chaloupka JC, Awad IA (1995) Strategies and armamen- 
tarium -:A~ treatment option:;. In: Awad I A. Harrow PL iedsj 
Giant intracranial aneurysms. AAN'S Publications, Park 
Ridge, IL.pp 91-116 

5.FoxAJ,V"ifiuela F, Pelz DM, et aL (1987) Use of detachable 
balloon for proximal artery oc-.mis.oii in die treatment o\ 
unclippable cerebral aneurysms. ■ Neurosurg 66:40-46 

i:. Berenstein A, Kunsouor: I, Kupersniith M, Flamni E and 
Ciaeb D ! : 984iTraiisvascular i tea I men I of g. a in aneurysms 
oi die caver nous carotid and vertebra! arteries. Functional 
investigation and emoonzatioit. Surg Neurol 21:3-12 

7. Chaloupka JC, Putman CM (1995) Endovascular therapy 
o\ surgical diseases oi the crania! base. Clin Puisiic Surg 
22:417-450 



Diagnosis and Endov 



ir Surgical Management of Carotid Blowout Syndrome 



28. Dare AO, Chaloupka JC, Putman CM, Fayad PB, Awad IA 
(1998) Failure of the hypotensive provocative test 

during :empo]";iry balloon lest occlusion of the internal 
carotid artery to predict derived hemodynamic ischemia 
a fief I herapeatic carotid occlusion. Surg Neurol 50:1 47- 

29. Zimmerman MC, Mickel RA, Kessler DJ, et al. (1987) 

Treatment of impending carotid rupture with detachable 
balloon embolization. Arch Otolaryngol Head Neck Surg 
113:1169-1175 
lO.Serbineitko FA ;".974i Balloon catheterization and occlu- 
sion of m:\io: vessels. " Neurosurg 4i :125-i 45 

31 . Serbinenko FA, Faltov JM, Spallone A, et al. (1990) Manage- 
menl of giani intracranial ICA aneurvsms with combined 
extracranial intracranial anastomosis and en.dovascular 
occlusion. J Neurosurg 73:57-63 

32. Debrun G, Lacour P, Vifiuela F, et al. (1981 ) Treatment of 54 
traumatic carotid-cavernous fistulas. 1 Neurosurg 55:::-7B- 
692 

33.0sguthorpe JD, Hungerford GD (1984) Transarterial 
carotid occlusion: case report and review oi the literature. 
Arch Otoloaryngol Head Neck Surg 110:694-696 

34. Khoo CTK, Molyneux AJ, Rayment R, Saad MN (1986) The 
control of carotid arterial haemorrhage in head and neck 
sargery ay ."'.il.oon c; I. trier tamitonaae and detachable oal- 
loon embolisation. Br J Plast Surg 39:72-75 

35.Morrissey DD, Andersen PE, Nesbit GM, Barnwell SL, 
Everts EC. Cohen II (1997) Endovascular management of 
hemorrhage in patients with head and neck cancer. Arch 
Otolaryngol Head Neck Surg 123:15-19 

36.Macdonald S, Gan I, McKay A], Edwards RD (2000) Endo- 
vascula: treatment o: acute carotid blow-out syndrome. 1 
Vase Interv Radiol 11:1184-1188 

37.Szikora I, Guterman LR, Wells KM, Hopkins LN (1994) 
Combined use of sients and coils to treat experimental 
wide-neck carotid aneurysms: a re li m in a :y results. A1XR 
15:1091-1102 

3S.Matsuura JH, Rosenthal D, Jerius H.Clark MD, Owens DS 
; 1 997! Traumatic carotid artery dissection .uvi pseudoan- 
eurysm treated with encovascular coils and stent, i Endo- 
vasc Surg 4:339-343 

39. Roth TC, Chaloupka JC, Putman CM, Weaver EM, Tarro J, 
Wecht DA, Sasaki CT (1998) Percutaneous direct puncture 
acrylic embolization of a pseudoaneurysni after failed 
carotid stenting for the trealnsent of acute carotid blowout. 
Am I Neuroradiol 19:912-916 

40. Simionato F, Righi C, Melissano G, Rolli A, Chiesa R, Scotti 
G (2000) Stent-graft treatment of a common carotid artery 
pseudoaneurysni. i End ova sc Ther 7:136-140 

41 . Coldwell DM, Novak Z, Ryu RK, et al. (2000) Treatment of 
posttraumatic internal carotid arterial nseudoaneurysms 
with endovascular stents. ■ Trauma 48:470-472 

42. Chaloupka JC.Weigele JB, Mangla S, Lesley WS (2001) Cere- 
brovascular angioplasty and stenting lor the prevention of 
stroke. Curr Neurol Neurosci Rep 1:39-53 

43. Lesley WS, Chaloupka JC, Weigele JB, Mangla S, Dogar MA 
i^DDa; Preliminary experience with encovascular recon- 
struction for the management ::<;' carotid blowout syn- 
drome. AJNR Am J Neuroradiol 24:975-981 

44. Debrun G, Fox A, Drake C (1981) Giant unclippable aneu- 
rysm';: treatment w.ta detachable bad.:-' ns. A1XR Am 1 
Neuroradiol2:167-173 

45. Walker AT, Chaloupka JC, Putman CM, Abrahams JJ, Ross 



DA (1996) Sentinel trans oral hemorrhage from a pseudoa- 
neurysm of an .menial maxillary artery oranch: a compli- 
cation of CT guided biopsy of the masticator space. A|NR 
Am J Neuroradiol 1 7:377-381 

i. Weigele JB, Chaloupka JC, Lesley WS, Dogar MA, Bempo- 
rad IA i J 00 J ) C'Ctyl-2-cyanoacrylate: .^n effective agenl for 
head ana neck tumor embolization [abstract]. 401 It Annual 
Mrel ing of the American Society oi Neuroradiology, May 
Vancouver, BC, Canada 

7.Chaloupka JC, Mangla S, Huddle DC, Mitra S, Ross DA, 
Sasaki CT (1999) Evolving experience with direct punc- 
ture therapeutic embolization for adjunctive and palliative 
maitayemeni of head and neck Irypervascular neoplasms. 
Laryngoscope 109:1864-1872 

3. Horowitz MB, Miller G, Meyer Y, Carstens G, Purdy PD 
( [99;ii Use of intravascular s ten Is n: the treatment of inter- 
nal carotid ana extracranial vertebral artery pseadoaneu- 
rysms.AJNRAm J Neuroradiol 16:693-696 

J.Diethrich EB, Ndiaye M, Reid DB (1996) Stenting in the 
carotid artery: initial experience in I I 1 " 1 patients. | Endo- 
vasc Surg 3:42-62 

l.Yadav JS, Roubin GS, Iyer S, Vitek J, King P, Jordan WD, 
Fisher WS (1996) Elective stenting of the extracranial 
carotid arteries. Circulation 5:1-6 

I.Wholey MH, Wholey M, Mathias K, et al. (2000) Global 
experience in cervical carotid arterv sienl placement. Cath- 
eter Cardiovasc Interv 50:160-167 

2.Phatouros CC, Higashida RT, Malek AM, et al. (2000) 
Carotid artery stent placement for atherosclerotic dis- 
ease - rationale, technique, and current stauis. Radiology 
217:26-41 

S.Wakhloo AK, Schellhammer F, de Vries J, Haberstroh 
], Schumacher \i (1994) Self-expanding and balloon- 
exnand able sients in tite treatment ot carotid aneurysms: 
ait experimental study in a canine model. AJNR Am | 
Neuroradiol 15:493-502 

4. C.eiemia G, Hakim M. Brennecl.e i. :. '. 994! Embolization of 
experimentally created aneurvsms with intravascular stent 
devices. AJNR Am J Neuroradiol 15:1223-1231 

j. Turiman F, Massoud TF, Ji C, Guglielmi G, Vinuela F, Robert 
] [1994! Combined stent imalantation and endosaccular 
coil placement for treatment of experimental wide-necl. 
aneurysms: a feasibility study in swine. AINR Am J Neuro- 
radiol 15:1087-1090 

i. Higashida RT, Smith W, Gress D, Urwin R, Dowd CF, 
Balousek PA, Halbach W (1997) Intravascular stent and 
endovascular coil placement for a ruptured fusiform aneu- 
rysm of the basilar artery. Case report ano review of the 
literature. I Neurosurg 87:944-949 

7.Graves VB, Strother CM, Partington CR, Rappe A (1992) 
Flow dynamics of lateral carotid arterv aneurysms, and 
iheir effects on coils and balloons: .^n experimental study 
in dogs. AJNR Am J Neuroradiol 13:189-196 

3. Lesley WS, Weigele |B, Chaloupka JC (2004) Outcomes for 
overlapping stents, in the extracranial carotid artery.Cath- 
eter Cardiovas Interv 62:375-379 

). Parodi JC, Ferreira M, Estol CJ (1996) Treatment of carotid 
arterv disease with an endoiuminal stent-venous graft. I 
Neurovasc Dis 1:27-31 

]. Ruebben A, Merlo M, Verri A, et al. (1997) Exclusion of an 
internal carotid aneurysm by a covered sient. 1 Cardiovasc 
Surg 38:301 -303 

I.Marotta TR, Buller C, Taylor D, Morris C, Zwimpfer T 



(1998) Autologous vein-covered stent repair of a cervical 63. Alexander MI. Smith TP, Tucci PL (2002) Treatment of a i 



1 carotid artery i^eiidoiiiiciit'ysm: technical c 
report. Nemo surgery 42:408-412 
62. Scavee V, De Wispelaere JF, Mormont E, Courier B, Trigaux 



ia:rogemc pes rou 5 oa:orid artery pseudo.meurysm with ,t 
iymbiol covered stent: technical c.se report. Neiio'-iireeiv 
50:658-662 



|P, Schoevaerdts)'.: (2001) Pseudoaneurysm of the internal 64.Kwok PC, Cheung lY.T.ing KVV, VVcng VVK (2001) Endov 
I'ith a covered stent. Cardiova* 



Intervent Radiol 24:253-235 



Gene Therapy and Pediatrics 



22 Embolotherapy Applications in Gene Therapy 



CONTENTS 

22.1 Introduction 295 
22.1.1 Background 295 

22.2 Embolization and Gene Therapy 29fi 

22.2.1 Embolization by [nn;s:on o:' Genelii.illy 
Altered Cells 296 

22.2.2 Embolization as a Prelude to Gene Therapy 298 

22.2.3 Embolization as an Adjunct to Gene Therapy 300 

22.3 Future Directions for Embolization 
and Gene Therapy 300 

22.3.1 Cell Transplantation 300 

22.3.2 Embolization to Promote Gene Therapy 301 

22.3.3 Gene Therapy to Promote Embolization 301 
References 301 



22.1 

Introduction 

Gene therapy is an emerging technology that will 
likely yield a wide variety of treatments. Initially 
gene therapy focused on rare diseases such as inborn 
errors of metabolism, but it is increasingly being 
applied to common conditions such as cancer and 
peripheral vascular disease [4, 12, 13, 22]. Human 
gene therapy trials in the United States can be 
reviewed online at www.gemcris.od.nih.gov, and 
many of these trials include interventional proce- 
dures. As summarized in Table 22.1, interventional 
procedures include direct arterial injection, direct 
injection into tumors, stent placement, and infusion 
through a central venous catheter. While Interven- 
tional Radiology will certainly play a role in this 
field, this chapter will concentrate on the interplay 
between embolization and gene therapy. 



tfj.MD.PhD 

it Professor of Radiology jn: Surgery, Mallinckrodt 
Institute of Radioj.'gv mvj W;ish ingtoij University School 
of Medicine, 510 S. Kingshighway Blvd. St. Louis, MO 63110, 
USA 



22.1.1 
Background 

By its nature, gene therapy is an indirect treatment 
method. In most cases, the gene (DNA} must be 
transcribed into RNA, the RNA translated into pro- 
tein, and it is the protein product that achieves the 
desired clinical effect. The transformations from 
DNA to RNA and then RNA to protein require access 
to the internal machinery of the cell. Delivering the 
gene to the cell's interior has been a significant bar- 
rier to successful gene therapy. A series of different 
strategies have been used; these are summarized in 
Table 22.2. 

The clinical cond it iond relates what met hod might 
be used to deliver the gene to the cell's interior. For 
example, correcting an inborn error of metabolism 
by gene replacement would ideally entail a single 
treatment that confers lifelong production of a pro- 
tein. This requirement excludes the brute force and 
hybrid strategies, because these only result in short- 
term expression of the therapeutic protein. This 
requirement also excludes most viral vectors and 
leaves only retroviral and adeno-associated viral 
vectors. On the other hand, short-term expression 
(days to weeks) may he sulticient lor other therapies 
such as combating atherosclerosis by angiogenesis 
or killing tumor cells. Since a wide variety of vectors 
confer short-term expression, choosing a vector then 
requires balancing factors such as level of expression 
with untoward attributes such as immunogenicity. 



22.2 

Embolization and Gene Therapy 

Three different gene therapy st rategies have employed 
embolization (Table 22.3). In the first, cells contain- 
ing the desired gene are infused and this method can 
lead to embolization since the altered cells are large 
enough to occlude vessels. In the second, emboliza- 
tion is used to facilitate gene therapy by preparing 



techniques used for gene therapy 



Selected references 



lurea injection inio lesion Inject ,ide:io , v r i:".i I vector con Liming gene wlncl; selectively .-Uls N:-:mvnaitis et al. [14] 

Direct injection into tissue Inject adenoviral vector wit!: gene :'oi vasculai growth factor Rajagopalan [16, 17] 

Selective intra-arteiul injection Iniecl adenoviral vedor containing gene which Selectively kills Sze [21] 



Attach adenoviral vi 



Table 22.2. Gene delivery vehicles 
Strategy Premise 



Brute When cells .ire bathed in Luge None - inject DNA 

force amounts of DNA, some DNA directly into tissue 

crosses cell's membrane 
Elegant Genetically engineered viruse: 

provide a highly evolved 

method of inserting genetic 

material (DNA or RNA) into 



Construct agents which mimic Canonic liposome: 
the biologic methods used by peptide fusion dor 



Easy to produce the 
gene in large quantnie- 
low immunogenicity 

hi':'. ceil I; 

long term expression is 

possible with selected 



Very inefficient means of trait 
ferring gene into cells; gene is 
expressed for dav-weeks 
Large-scale production of 
the viral vectors can be dif- 
ficult; viruses can induce or b 
destroyed by immt 
immune response i 
systemic illness 



Low immunogenicity, 
s potential Cell specific 
de.iverv 



Adapted from Giat 



3 [6] 



the tissue to receive the gene therapy vector or by 
helping target the gene therapy vector to the cells of 
interest. Examples are portal vein embolization as 
a method of stimulating hepatocyte replication in 
the nonembolized segments of the liver and using 
Ethiodol embolization to help target a viral gene 
therapy vector to hepatocellular carcinomas. In the 
third case, gene therapy is used as an adjunct to 
embolization. An example is tethering adenoviral 
particles to embolization coils. 



22.2.1 

Embolization by Infusion of Genetically Altered 

Cells 

The first human gene therapy experiments used 
this approach [18]. Cells were collected, genetically 
altered and then infused back into the patient. The 
advantage of this approach is that the difficult task 
of inserting the gene into cells was performed in 
the laboratory where conditions could be optimized. 
This approach also minimized the possible risks 
since the patient was not directly exposed to the 
gene therapy vector. In these experiments, circulat- 
ing lymphocytes were collected, modified and then 



infused. Subsequent studies have used a wide variety 
of cell types. 

An early therapeutic trial that involved emboli- 
zation was a complex protocol that is summarized 
in Fig. 22.1. In this protocol, patients with familial 
hypercholesteremia caused by mutations in the low 
density lipoprotein receptor (LDL receptor) were 
treated with the hope of normalizing cholesterol 
metabolism [8, 9]. For treatment to be successful, 
long term expression of LDL receptors was desired 
and this requirement prompted the investigators 
to use a retroviral vector because these vectors had 
the advantage of inserting the LDL-R gene into the 
genome of the harvested hepatocytes (Fig. 22.2). 
Other vectors offered only a short-term solution 
because genes that are not incorporated into a 
cell's chromosomes are lost over a period of days to 
weeks. 

In this experiment, gene expression was assessed 
by several methods. Liver biopsies found hepato- 
cytes which expressed the LDL receptor. In addi- 
tion, small changes in cholesterol metabolism were 
found. However, the clinical effect was negligible 
and the investigators concluded that the ex vivo 
method was limited by the "low efficiency of genetic 
reconstitution". 



'-. iiil>. "■ I .:■ liier.i pv Applicai 



Table 22.3. Interplay heiween embolization and g^jje theiajiy 



Miaiegy 



i-X/LLLple 



infusion of gene:icaliv altered :::;:::::::;: nan j e 1 1 > 
Portal vein eml> 'lizalion siimulales hepaiocy te lepiicadoi; 
Elhiodol embolization helps taigei viral vector to tumor 
Gene therapy as adjunct to embolization :imho!ize vsing bifu::c:io::a! agent - ;';iii embolization ono pa:: gene liter 



Part and parcel 

'-. n'.b-.'kz:-, t :.:■[! fac.lnates gene :he:apv 




Fig.22.1. Protocol schematic t'.:ir ex vivo hepatic gene therapy. 
The lateral segment of ihe patient's live: was removed by a 
subcostal incision. ,n\d a Hickman catheter was placed by 
sacrificing the inferior mese[i;eric win. The liver specimen 
: ■■ !:•!) gmj was :hen per fusee. wi:h co II agenase to release hepa- 
tocytes ( -3 billion) which were then divided into 800 culture 
dishes. Two days later, a letrovh us containing the sequence for 
the LDL receptor was added lc Ihe culture media. The hepa- 
tocytes were harvested !_!-] 8 bis I a lei and divided into three 
aliquots.Each aliquot w : as infused slowly (J cc/min) back into 
the patient's portal circulation using the Hickman catheter. 
Adapted from Grossman [8] 









j 




to 


■-I 


— 


© 


© 



Fig. 22.2. Schematic for retroviral geiie iheiapv. Retroviral par- 
ticles containing RN'A thai encodes the gene of interest are 
prepared and added to ihe recipieiv. cells. The retroviral vector 
is internalized and the:: releases iis ge::e:ic material into the 
cytoplasm. Reverse :ranscnptase c.ses the viral template to 
produce a DNA coj.iv of ihe desired gene and :bis gene can then 
he integrated into the recipient cell's gen-' ir.e. The integrated 
gene then serves as a template fo: lianscription into mRNA 
and translation into protein. 



Numerous factors limited the efficiency of 
genetic reconstitution. First was the number of 
cells that were recovered from the liver specimen. A 
250-gm liver specimen was resected, and from this 
only 3 billion hepatocytes were isolated for culture. 
A typical liver contains approximately 100 mil- 
lion hepatocytes per milliliter and thus the 250-gm 
Id have provided approximately 25 bil- 
lion hepatocytes. Second, only 1 billion hepatocytes 
were viable, and only 20% of them expressed the 
LDL-R after transfection with the retrovirus. Even if 
these 400 million hepatocytes expressed the normal 
number of LDL receptors, they would have only 
the number of receptors typically tottnd in 4 ml of 
liver. Since the average liver volume is slightly more 
1600 ml [24], even if the methods were improved 
100-fold, the result would be a LDL receptor levels 
that would still only be 25% of normal. Cholesterol 
metabolism is clearly sensitive to the number of LDL 
receptors since patients with one half the number of 
LDL receptors (heterozygotes for familial hypercho- 
lesterolemia) have markedly elevated levels of LDL 
cholesterol and develop premature atherosclerosis 

pi. 

Even if more hepatocytes could be isolated by 
either resecting more liver or improving the yield 
per gm of resected liver, infusing more of the geneti- 
cally altered hepatocytes into the portal vein carries 
real risks. Hepatocyte infusion causes occlusions at 
the microvascular level and transient increases in 
portal venous pressure were observed during the 

Ex vivo gene therapy followed by infusion of the 
altered cells clearly illustrates the promise and prob- 
lems of gene t herapy for inborn errors of metabolism. 
While it is difficult to conceive how the transplanta- 
tion method might provide more than a few percent 
of normal levels for any protein, there are diseases 
where restoring low levels of the missing protein 
significantly ameliorates the clinical condition. One 
such example is hemophilia A where achieving 1% 
of normal levels for Factor VIII substantially lowers 
the risk of severe hemorrhage [3]. 



Even with integration of the target gene into a 
cell's genome, the durability of the treatment will be 
limited by the lifespan of the altered cell unless the 
cell divides and passes the desired gene to its prog- 
eny. Hence there is interest in genetic manipulation 
of stem cells. If the genetically the altered stem cell 
has a survival advantage over its endogenous coun- 
terparts, this could lead to increasing levels of gene 



22.2.2 

Embolization as a Prelude to Gene Therapy 

The limitations of cell transplantation for hepatic 
gene therapy prompted Kathy Ponder, Marshall 
Hicks, and this author to study how embolization 
might be used as a preparatory step tor gene therapy. 
This project stemmed from the fact that retroviral 
vectors could integrate genes into the hepatocyte 
genome only if the hepatocytes were dividing. In 
adult animals, far fewer than 1% of hepatocytes 
were replicating under normal conditions. However 
based on data from partial hepatic resections and 
hepatic embolization, we postulated that occluding 
the portal vein branches supplying two thirds of the 
liver might be a safe and effective means of stimulat- 
ing hepatocyte replication in the remaining third of 
the liver (Fig. 22.3). 

A series of animal experiments confirmed that 
embolizing two thirds of the portal branches stimu- 
lated hepatocytes in the spared portion of the liver 
to divide [5]. These experiments also determined 
the timing and timing and extent of hepatocyte rep- 
lication. The embolized liver segments atrophied 
and the atrophy was not accompanied by evidence 
of hepatocyte lysis. Instead, it was found that hepa- 
tocytes underwent apoptosis. Finally, these experi- 
ments also demonstrated the feasibility of maintain- 
ing ,i catheter in the portal vein tor several davs. 



Attempts to build upon this groundwork were 
less encouraging. Large doses of the retroviral 
vector were prepared and infused into the liver after 
embolization using the portal venous catheter. Sub- 
sequent blood samples revealed only low levels of 
the gene product. Later experiments in a dog model 
encountered additional technical issues. Specifically, 
it was more difficult to obtain and maintain portal 
vein access. Finally a severe immune response was 
encountered during retrovirus infusion. These and 
other issues prompted Kathy Ponder to stud)' the 
feasibility of neonatal hepatic gene therapy. Those 
experiments found that a sizable fraction of hepa- 
tocytes are dividing in neonates without any added 
stimulus. Subsequent work by her group has found 
that intravenous infusions led to successful gene 
therapy in neonates and the immune response seen 
in adult animals was not reproduced [15, 20, 23]. 

We next hypothesized that portal vein infusion 
in neonates might have additional advantages over 
intravenous infusion. For these experiments, we 
attempted to use the umbilical vein for access and 
then select the portal vein. Infusing the vector into 
the portal vein might prove more efficient than 
intravenous infusion. The approach is summarized 
in Fig. 22.4. The small size of the animals and sharp 
angles between the umbilical vein and main portal 
vein forced us to abandon this model. However, we 
still expect that there may be a role for this method 
of portal vein in ins ion that might be best studied in 
future human trials. 

Since embolization alters tissue blood flow, 
embolization could potentially be used to facilitate 
gene delivery to cells of interest. Embolization could 
improve the target to background ratio by altering 
blood flow (Table 22.4). Embolization could also 
increase the likelihood that the vector will interact 
with the individual cells in the target tissue. This 
dwell time argument is often cited to explain the 
efficacy of chemoembolization. 



bb bb BB BSi 



BBBB ®& — BB 



Fig. 22.3. Portal vein embolization stim- 
ulate? hepntocyte replication. Hepato- 
cytes and their porta! vein branches are 
shown. If the portal vein branch supply- 
ing a portion of the liver is occlude,!, the 
hepatocytes supplied by that branch 
undergo apoptosis. The embolized seg- 
ment of the liver atrophies while hepa- 
tocytes in nonembolized liver replicate. 



'-. iiil>. "■ I .:■ liiei.i pv Apr- 1 ion 




i 200-gm pup was delivered 
i section, and the umbilical 
vein was cannulated with a 20-g Angio- 
cath. Diluted contrast was injected and 
images recorded in either anteropos- 
terior (a) or lateral fb) planes. Vessels 
are labeled as follows: umbilical vein 
(UV), main portal vein fPV), left portal 
vein (LPV), ductus venosus (DV), right 
hepatic vein (RHV). a A digital sub- 
traction image; b is nonsubtracted. 
Attempts to select the portal vein from 
the umbilical vein access were hindered 
by the angles between the left portal 
vein and umbilical vein as well as the 
angle between the left portal vein and 



Table 11. -I. S l : . i : e t: i e s :oi improving ui^i io background :. 



Possible ckmca! example 



Flow to 



Flow to Target/ 

Nontarget background 



Selectively embolize v< 
nontarget tissue 



Embolize vessels supplying 
lesion and surrounding tisst: 



Selectively embolize gastroduodeni'E 

nor n; a I tissue, the;; infuse vector via 

artery catheter 

EmboJize i'.epalic artery orancE using H tar:- ..(.:■. 



protect No change Decreased Increased 



"Increasing delivery io the target tissue by embolizatio 
cause an increase in blooc f.ow to the target tissue. 



nsidered because it is unlikely that emboliza 



iiBA et al. [19] tested whether embolization 
i improve gene delivery Co hepatocellular car- 
i. They induced spontaneous hepatocellular 
a rats using chemical carcinogens and 
performed hepatic artery embolization using Lipi- 
odol. It is noteworthy that most animal studies have 
used models where tumors are implanted in the liver 
and it is a considerable leap of faith to believe that 
the blood flow to a tumor 5-21 days after implanta- 
tion will accurately simulate the blood flow of pri- 
mary or metastatic tumors in humans. Shiba et al. 
then tested gene transfer using an adenovirus car- 
rying a marker gene. The adenoviral vector could be 
mixed with Lipiodol without destroying the vector 
because adenovirus lacks a lipid membrane. This 
mixture was injected into the hepatic artery and the 
efficiency of gene transfer was assessed by staining 
for the marker protein and calculating the percent- 
age of stained tissue in normal liver, small tumors 
and large tumors. In control animals, the adenoviral 
vector without Lipiodol was injected into the hepatic 



A substantial increase in tumor to background 
ratio was found when the adenoviral vector was 
coinjected with Lipiodol (Table 22.5). This reflected 
an increase in the percentage of tumor cells which 
expressed the gene therapy marker as well a 
decrease in the percentage of normal hepatocytes 
that expressed the marker. The latter is an expected 
consequence of embolization. The increase in tumor 
cell staining suggests that the dwell time effect more 
than overcame the decrease in blood flow caused by 
embolization. 

The finding that tumor cell staining exceeded 
staining in normal liver by approximately sixfold is 
remarkable because the available data indicates that 
tumors are almost uniformly underperfused [2]. 
While hepatomas appear hypervascular on angiog- 
raphy, at the microscopic level, the tumor vascula- 
ture is disorganized. Furthermore, even if a tumor 
possessed a well organized microvascular network, 
tumor interstitial pressure would reduce perfusion. 
The mechanism by which Lipiodol improved the 
target to background ratio is uncertain. Prior work 



Table 22 5. Marker gene express 


on following gene 


ransfer 






IllieCiion 


Tumors <5 




Tumors >5 mm 


No:' if, a I liver 


Vector after iodized oil (n=9) 
Vector alone (n=15) 


37.4 +/- 6.0 
14.0 +/- 3.6 




35.6 +/- 9.3 
7.0 +/- 1.9 


5.7 +/- 1.3 
14.4+/- 2.3 



The adenoviral vector ti'-r.r.'-Ljiir.t; 7 lie gene :or |'i-ga.aclosidasr was injected .mo The hepatic arlerv o: tumor oea ring ao.imals ei trier 
by itself or im mediately following injection o: iociizeo oil. Animals were sacrificed 1 days later and liver sections were .'re oared. 
Expression of :iie maraer gerie was dele: milled by staining tor |i-gr lac iosioase. Tire .irea of staining i:i small In mors (-011:11: ), .a rye 
tumors i '."■5:1:111) ana normal liver was determined. rLe ,;l .: Irs are exaressir.i as mean pnaemages -■■/- s::i:idara error of the me.in. Nine 
animals received the veer:)!" arler iodized 01 a I f animals received rl'.e verier alone. Adapted from Shtba et al. [15] 




Fig.22.5. Attaching adenoviral particles 
:o embolization coils. Platinum and bio- 
degradable coils were coated with colla- 
gen to provide a surface for subsequent 
attachment of an anti-adenovirus anti- 
body. The derivatized coils were then 
incubated with the adenoviral vector 
which contained tire gene for green fluo- 
rescent protein 



has shown that injection of Lipiodol into hepatic cyfr 

arterial branches results in Lipiodol deposition in to t 

both normal liver and hepatic tumors. The initial gene transfer to smooth 

deposition in tumors is felt to reflect the large extra- o 
cellular spaces within tumors and the different pres- 
sures within the arterial and portal systems [10]. 



were embedded in the thrombus adjacent 

. Interestingly, there was no evidence of 

icle cells or endothelial 



22.2.3 

Embolization as an Adjunct to Gene Therapy 



22.3 

Future Directions for Embolization and 

Gene Therapy 



Endovascular treatment of aneurysms has focused 
on the mechanical aspects of the procedures but the 
prevalence of endoleaks and incomplete obliteration 
of intracranial aneurysms indicates there is ample 
room for improvement. An integrated approach 
where the mechanical device also serves as a scaf- 
fold for drug delivery has been proposed. In this 
scheme, the coils deployed within the aneurysm 
might also be coated with a drug which promotes 
intimal hyperplasia and the intimal hyperplasia 
helps seal the aneurysm neck. 

Abrahams et al. [1] recently reported linking an 
adenoviral vector to embolization coils. A diagram 
of the attachment strategy is shown as Figure 22.5. 
These initial experiments tested the feasibility of 
this approach both in vitro and in an animal model 
of intracranial aneurysms. A marker gene was used 
and expression of the marker protein was found in 
cells which were in contact with the coil. The animal 
experiments detected the marker protein in leuko- 



22.3.1 

Cell Transplantation 

Research in cell based gene therapy represents the 
leading edge of how gene therapy and embolization 
can overlap. The field remains in its infancy and 
the research thus far has focused on the feasibility 
of transferring genes into cells and gene expression 
after infusion of the altered cells. The nuances of 
embolization are largely ignored because most infu- 
sions are by an intravenous route and also because 
it is desirable to keep these early experiments as 
simple as possible. As the technology matures, 
there will be increasing interest in embolization 
techniques. Catheter directed infusion into specific 
tissue beds could be used to improve the therapeutic 
window by both by concentrating the cells in the 
target tissue and sparing nontarget organs. Most of 
these targeted embolizations would be from arte- 
rial access but it is also likely that infusion into 



I- iiil>. "■ I .:■ rhera pv Applicai 



n Gene Therapy 



the portal venous system and selected branches of 
the pulmonary vasculature will be advantageous in 
certain circumstances. 

Delivering these cells to the site of interest is only 
the first step. The duration ot therapy will be depen- 
dent upon the survival of cells following injection. 
That survival will depend upon adequate delivery 
of nutrients and removal of waste products. This 
might seem trivial since the cells will initially reside 
in the intravascular space but this location does not 
guarantee perfusion especially since the cells them- 
selves will likely occlude the vessel in which they 
reside. This mechanical occlusion would likely lead 
to thrombosis, especially if the transplanted cells 
lack the cell surface moieties found on normal endo- 
thelial cells. A transplanted cell within a cocoon of 
thrombus is unlikely to survive. Ifthis is true, anti- 
coagulation either by inhibiting the coagulation 
cascade or platelet deposition might improve sur- 
vival of the transplanted cells. 

22.3.2 

Embolization to Promote Gene Therapy 

The need to induce hepatocyte replication by portal 
vein embolization is fading into the past. Neona- 
tal infusions, new vectors and other strategies cir- 
cumvent the need for embolization. Further work 
is needed to determine if hepatic embolization with 
oil based agents can indeed improve target to back- 
ground ratios for gene therapy vectors. Ifthis is pos- 
sible, it will be critical to establish the mechanisms 
responsible so that the technique might be applied 
to other organs. 



22.3.3 

Gene Therapy to Promote Embolization 

Gene therapy could be used to improve embolization 
outcomes. Interventional Radiology has tradition- 
ally concentrated on the mechanical aspects of our 
procedures but it is clear that biological coatings 
that alter tissue response will become increasingly 
important. The same strategy that has been used 
to link adenoviral particles to embolization coils 
could easily be adapted to link gene therapy vectors 
to embolization particles. This technology clearly 
illustrates that the key to a successful marriage of 
embolization and gene therapy will be understand- 
ing both embolization and gene therapy at the cel- 
lular and molecular levels. 



1. Abrahams JM, Song C, DeFelice S ef a!. (2002) Endo- 
vasc.ular mic recoil gene delivery using immobilized 
anti-adenovnus antibody for vector tethering, stroke 
33:1376-1382 

2.Carmeliet P, Jain RK (2000) Angiogenesis in cancer and 
other diseases. Nature 407:249-257 

S.Chuah MK, Collen D, VandenDriessche T (2004) Clinical 
gene transfer studies for hemophilia A. Strain Threimb 
Hemost 30:249-256 

1. Co 11 in son 1 ■■■, Donnelly H (2004) Therapeutic aitgiogeiies.s 
i;i peripheral arterial disease: ojii biotechnology produce 
;"ii"; effective co llai era I circiilauoii? Eur 1 Vase End ova sc Surg 
28:9-23 

S.Duncan IR, Hicks ME, Cai SR et al. (1999) Embolization 
'.:' poita! vein branches mcltices hepatocvi^ replication in 
swine: a potential step in hepatic gene therapv. Kadiof'gv 
210:467-477 

i.Giannoukakis N,T:u ccc M : 2003) Cu: rent si. it us and pros- 
pec is rot geiiT and cell therapeutics for type : diacetes nir'- 
litus. Rev Endocr Metab Disord 4:369-380 

7. Goldstein IL, Hobbs HH, Brown MS (1995) Familial hyper- 
cholesierolemi.;. In: Scriver Cji.. Fieatidet AL. Sly VVS et al. 
(eds) The metabolic -.inc. molecular bases of inherited dis- 
eases. McGraw-Hill, New York, pp 1981-2030 

3. Grossman M, Raper SE, Kozarsky K et al. (1994) Success- 
ful ex vivo gene therapy directed to livt-r in a patient with 
familial hypercholesterolemia. Nat Genet 6:335-341 

S.Grossman M, Rader DJ, Muller DW et al. (1995) A pilot 
study of ex vivo gene therapy for homozygous familial 
liypcicholcStefO'Licniia. Nat Med 1 :1 148-1154 

i.Kan Z. Wallace S i:ji L J4) Sinusoidal embolization: impact 
of iodized oil osi hepatic microcirculation. I Vase Inlerv 
Radiol 5:881-886 

t.Klugherz BD, Song C, DeFelice S et al. (2002) Gene deliv- 
eiv io pig coronary arteries from stents carrying antibody- 
tethered adenovirus. l-linr: Gene Ther 13:443-454 

2. Manninen l-ll. Makmen K (2002 i Gene therapv lechniqttes 
for peripheral arterial disease. Card i ova sc Int^rveni Radiol 
25:98-108 



l. Mulligan RC (1 993). The basic 

ence 260:926-932 
4. Nemunaitis J, Ganly I, Khuri F t 

tion and oncolysis in p53 mut 

an ElB-55kD gene-deleted at 

advanced head and neck cancel 

Res 60:6359-6366 
3. Ponder KP, Melniczek [R, Xu L 

neonatal hepatic gene therapy 

VII dc 



ace of gene therapy ?ci- 

(2000) Selective replica- 

umorswith ONYX -01 5, 

patients with 

a phase II trial. Cancer 

t al (2002) Therapeutic 
. mucopolvsaccharidosis 
i.Proc Natl Acad Sli USA 99:13102 i 3 07 

ii S, Mohler ER 3rd, l.edcrman :*| <r, al. (2003a) 

Regional aiig...'C.eiir- : .s wit ial growth 

factor in peripheral arterial ilsusi .i pl'.«c :l randoni- 
izr.:.. double-blind, control. t\l .1 . v • • ...U- viral deliv- 
ery of vascular endothelial it> v.:"-. :. . i..| . in patients 
with disabling intermittent dtiu-icjiiin Circulation 
108:1933-1938 

(.Raiagopalan S, Mohler E 3rd, l.ederman H\ et al. (2003b) 
Regional Align: genesis with V.iv n ..: I : .1 ;.-. : ial Growth 
Factor (VEGF) in peripheric ■.-.. lenal dadoae. design of the 
RAVE trial. Am Heart J 145:1114-1118 

i. Rosenberg SA, Aebersold P, Cornetla K el al. (1990) Gene 



transfer into humans-iT.munolherripy or patients with 
advanced melanoma, using I u trior-; null rating lympho- 
cytes modified by retroviral gene transduction. N Engl I 
Med 323:570-573 

19. Shiba H, Okamoto T, Futagawa Y et al. (2001 ) Efficient and 
cancer-selective gene transfer to hepatocellular carcinoma 
in a rat using adenovirus vector with iodized oil esters. 
Cancer Gene Ther 8:713-718 

20. Sleeper MM, Fornasari B, Ellinwood NM et al. (2004) Gene 
therapy amelioraies cardiova scu.ar disease in dogs with 
mucopolys.icc.haridosis VII. Circulation 110:815-820 

21. See DY, Freeman SM, Slonim SM et al. (2003) Dr. Gary J. 



Becher Young invesugaior Award: intraanerial ader 

for metasratic gastrointestinal cancer: activity, radiographic 

response, and survival. I Vase Interv Radiol 14:279-290 

22.Wadhwa PD, Zielske SP, Roth JC et al. (2002) Cancer gene 
therapy: scientific basis. Arum Rev Med 53:437-452 

23. Xu L.Gao C, Sands MS et al. (2003) Neonatal or hepatocyte 
growth :'::.:!■ u polem-atec .nl ah iterie therapy with a rerro- 
viral vector results :n therapeutic levels or canine factor IX 
for hemophilia B. Blood 101:3924-3932 

24. Yu HC, You H, Lee H et al. (2004) Estimation of standard 
l.ve: vol u n'.e for aver transplantation in lire Korean popula- 
tion. LtverTranspl 10:779-783 



23 Embolotherapy in Pediatrics 



nd Laurent Gar 



Introduction 303 
Embolotherapy 303 

Pre-procedure 303 

Indications 303 

Consent 304 

Prior Laboratory Tests 304 

Preparation 304 

Anesthesia 304 

Setting - Equipment 304 

Procedure 305 

Fluids 303 

Contrast Medium 305 

Arterial Access 305 

Arterial Spasm During Catheti 

Arterial Spasm Following Withdrawal 

of the Introducer 305 

Embolizing Agents .''05 

Post-procedure 306 

Main indications in Pediatrics 306 

Trauma 306 

Pelvic fracture 306 

Vascular anomalies 306 

Hemangiomas Rem;: lory 

to Medical Treatment 308 

Kaposiform Hemangioendothelioma 308 

Liver Hemangiomas 308 

Arteriovenous Malformations 311 

Renal Embolization 311 

Epistaxis 31] 

Hemoptysis 315 

Gastrointestinal Bleeding 316 

Embolization or Sclerotherapy of Varicoceles 

Hvpei splenism 317 

Thrombolysis 317 

Cfiemoembolization 318 

Conclusion 319 

References 319 



J.Dvbois.MD 

Professor of Radiology, Pediatric and Interventional Radi- 
ologist, Peportiv.ent of Mec.ic.il imaging, Hopital Ste-lustine, 
3175 Cote Ste-Catherine Road, Montreal, Quebec H3T 1C5, 
Canada 
L. Garel, MD 

Professor of Radiology, Pediatric and Interventional Radi- 
ologist, Pepartiv.ent of Mec.ical imaging, Hopital Ste-lustine, 
3175 Cote Ste-Catherine Road, Montreal, Quebec H3T 1C5, 
Canada 



23.1 
Introduction 



Pediatric interventions differ from adult i 
tions in several ways: both the setting and the equip- 
ment must be adapted to infants and children. The 
disease processes and the indications for treatment 
are clearly distinct in this age group. 

Interventional procedures have been slower to gain 
acceptance inpediatrics because physicians were more 
conservative, training centers were few in number, 
and the equipment not designed for small patients. 

Xoivadays, pediatric embolotherapy has become 
feasible, thanks to the availability of microcathe- 
ters. Such procedures must be performed in tertiary 
pediatric centers, because newborns, infants, and 
children require special attention in the choice of 
general anesthesia versus sedation, control of tem- 
perature, fluids, radiation hazards, and dedicated 
equipment. These procedures rely upon a team of 
trained nurses, radiology technicians, interven- 
tional radiologists, and anesthesiologists. 

The aim of this chapter is to outline our approach 
regarding the environment (setting, sedation, 
equipment), and to share our experience in pediatric 
endovascular procedures. 



23.2 
Embolotherapy 



23.2.1 
Pre-procedure 



23.2.1.1 
Indications 



It is important to ensure the relevance of the proce- 
dure. The advantages of embolization by arterial route 
over a surgical procedure must be well established. The 
potential hazards must be discussed with the attend- 
ing staff. A clinician and surgeon must be available as 
back-up to face any potential complication. 



J. Dubois and L.Garel 



23.2.1.2 
Consent 



23.2.1.5 
Anesthesia 



Informed consent is obtained from the parent or 
guardian. In Quebec (Canada), a child over age 14 
has the legal right to sign for him/herself. The pro- 
cedure and ill I potential complications must be fully 
explained and discussed. 



23.2.1.3 

Prior Laboratory Tests 



Blood tests, including a standard c 
up, are mandatory [1]. 



The majority of simple interventions - biopsy, 
venous access, and drainage - are performed under 

With the exception of embolotherapy for varico- 
celes, the majority of embolotherapies by arterial 
route are performed under general anesthesia for 
the following reasons: (a) to prevent the child from 
moving during a long procedure, (b) to focus fully on 
the technique while the patient is monitored by the 
anesthesiologist. Besides, embolization with alcohol 
carries definite risks, which are well described in the 
literature, and for which general anesthesia is man- 
datory. 



23.2.1.4 
Preparation 

Patients must be fasting for the appropriate dura- 
tion as determined by age and health condition. 
No prophylactic antibiotic is administered before 
embolotherapy except in children with congenital 
heart defects for bacterial endocarditis prophylaxis 
[2]. Sedation at the bedside is prescribed by the anes- 
thesiologist for anxious children and adolescents. 



23.2.1.6 

Setting - Equipment 

The angiography room must be warm, especially 
for newborns and infants. Pediatric anesthesiolo- 
gists must have the necessary means to maintain the 
child at a constant body temperature: covers, hat, 
Bear Hugger, heat lamp, and warming blanket. The 
solutions as well as the contrast medium are heated. 



Table 23.1. Materials for pediatric angiography and embolotherapy 

Introducer sheath: 4Fr (for paiienis weighing less than S kg) 
[nlrodt-cer sheath: .~Fr i for paiienis weighing over 8 l-:g) 

Catheters - 5Fr and 4Fr i selected .;cco:diiig to I he introducer sheath!: 

Celiac trunk catheterization: Hook (Cook! i RIM - second choice) 

Superior mesenteric artery: Hook (Cook] (RIM - second choice) 

Inferior mesenteric artery: RIM (Cook] 

Bronchial artery: Hook (Cook] 

Inferior and superi'.'r limbs: Hook (second choice: Tracker, Boston Scientific; 

Common carotid: Harwood Nash (Cook) 

Microcatheters ! used with the following none t.tpered catheters) 
Fast Tracker 18 (Boston Scientific]: Coaxial 5Fr 

Renegade 18 (Boston Scientific): Coaxial 5Fr 

Excel 14 (Boston Scientific): Coaxial 4Fr 

Fast Tracker 10 (Boston Scientific]: Coaxial 4Fr 

Terumo guidcwire (the csost often used in otir institution): 

Available in different angle shapes :4? :: and W) selected according to arteries angulation. 

Transend 14 (Boston Scientific): 

The tip of this guidewire has the dislinctive feat tire of being mal. cable and therefore it can be 

preshaped before calheterization. For this reason, this guideivire is often used to perform dif- 

ficull catheterization. 

Mizzen Soft (0.012) (Boston Scientific): 

The tip of this guide is smaller than Transend and is sometime useful for distal 



'-. nil> "■ I .:■ iiie:.i py .n t-'ed i:i 



Pulsed fluoroscopy, low mA, filters, 
and coning are mandatory in pediatr: 



23.2.2 
Procedure 



ideocapture, always used: 4-F for under 10 kg and 5-F above. 

: practice for Occasionally, when there are specific technical 
demands, a 5-F introducer maybe used for children 
under 10 kg. Heparin therapy of 50-100 units/kg 
is recommended for all infants weighing less than 
10 kg, with the exception of those with bleeding 
problems [4]. 



Although partially modified for children (mainly 
for shortening their length) the catheters used are 
basically the same as those used for adults. The cath- 
eterization technique and the embolizing agents are 
similar. However, certain details specific to children 
must be discussed: fluids, contrast medium, and 
equ ipment. 



23.2.2.4 

Arterial Spasm During Catheterization 

The medications used are intraarterial papaverine 
(1 mg/kg) or nitroglycerin (2-3 |J.g/kg - which may 
be repeated 3 times, maximum 20 Lig/kg), and xylo- 



23.2.2.1 
Fluids 

The fluids administered must be closely monitored 
in order to avoid rapid pulmonary overload, partic- 
ularly in an infant weighing under 10 kg. It is impor- 
tant to inform the anesthesiologist of the quantity 
and nature of the fluids that are injected during the 
procedure. Specific attention must be given to the 
infusion via the introducer, which must be regulated 
by a counter to 20 cc/hr. The quantity of rinsing fluid 
must be minimised. 



23.2.2.2 
Contrast Mediu 



n of 5 cc/kg must be the rule. It is better 
to perform an embolotherapy in several steps than 
to exceed the maximum dose of contrast. Aspirating 
surplus contrast medium remaining in the catheter 
helps to reduce the volume administered. Hydration 
should be maintained. Reported rate of minor reac- 
tions is 0.9% for non-ionic contrast media [3]. 



23.2.2.3 
Arterial Access 

Arterial spasm is frequent in children, especially 
when having difficulty with the arterial puncture. 
In children, it is preferable to puncture an artery 
with a Cathlon rather than a metal needle to lessen 
the risk of arterial spasm. A 20-G Cathlon is used 
for children weighing less than 10 kg, and an 18-G 
Cathlon for children over 10 kg. An introducer is 



23.2.2.5 

Arterial Spasm Following Withdrawal 

of the Introducer 

We recommend keeping the limb warm. A nitro 
ointment can be applied to the puncture site. Occa- 
sionally, a nerve block may be done. The patient 
must be on heparin and, it necessary, an intravenous 
infusion of rtPA can be used under the supervision 
of a hematologist. 

Thrombolysis by arterial route is seldom rec- 
ommended, particularly for newborns and babies 
weighing less than 5 kg, given the risk of damaging 
other vessels. Infants have a well-developed collat- 
eral network which, in most cases, enables r 
larization of the affected limb. 



23.2.2.6 
Embolizing Agents 

The embolizing agents are the same as those used 
for adults. The gelatin sponge particles (Surgifoam, 
Ethicon, Johnson & Johnson Co., Somerville, New 
Jersey) are used for a temporary occlusion, and 
the polyvinyl alcohol particles (Contour, Boston 
Scientific Corp., Fremont, CA) for a permanent 
occlusion. Tissue adhesive: N-butyl-2-cyanoacry- 
late (Indermil, Tyco, Norwalk, USA) or enbucrilate 
(Histoacryl, Braun, Aesculap) opacified with oily 
contrast media and alcohol can also used in pedi- 

Specific precautions are needed for alcohol use. 
A maximum dose of 1 ml/kg (or 60 ml) per 
should never be exceeded [5]. 



J. Dubois and L.Garel 



23.2.3 
Post-procedure 

As soon as the interventional procedure is over, the 
pediatric patient is taken to the recovery room and 
then transferred to his/her room. He/she must be 
monitored and the catheter entry site checked every 
15 minutes for the first 2 hours, and then every 30 
minutes for an addition;! I 2 hours. Bed rest for eight 
hours is recommended with special attention given 
to the limb involved. 



23.3 

Main Indications in Pediatrics 



For pseudoaneurysm accessible by percutaneous 
approach, the procedure can be performed under 
Doppler guidance (Fig. 23.3a,b). The adequate 
needle placement is confirmed by contrast injection. 
Thrombostat (Thrombin, Parke-Davis, Scarbor- 
ough, On., Canada), 1000 units/cc, is the most com- 
monly used agent. We start with an initial bolus of 
200 units. If the flow in the pseudoaneurysm is still 
present on Doppler ultrasound, we repeat the injec- 
tion up to a maximal dose of 1000 units. Since there 
is a possible risk of contamination with Thrombo- 
stat, we elected to use human thrombin 500 which 
is included in a kit available at the blood bank of 
our institution (Tissel Kit VH, Baxter, USA). Tissue 
adhesive, Gelfoam, or coils can also be used percu- 
taneouslv. 



23.3.1 
Trauma 

Organ injuries occur following a blunt or a pen- 
etrating trauma, including biopsy. Hematuria, 
hemobilia, or intraabdominal bleeding are the rel- 
evant clinical symptoms indicating traumatic inju- 
ries. Delayed or recurrent hemorrhage is the most 
common complication of trauma occurring in 3-8% 
of hepatic injuries, 1.5% of liver-spleen injuries, 6% 
of liver-spleen and pancreas injuries, and 31% of 
isolated pancreas injuries [6]. Pseudoaneurysm with 
expanding hematoma and subsequent rupture is the 
most serious evolutive complication. CT-scan is able 
to identify arterial injuries or fistulae. Angiogra- 
phy is indicated in unstable patients with dropping 



n patients it 



ular lesion 



i[7]. 



: the fum 



hemoglobin oi 
is questioned 

The embolization should be 
possible to the injury site to ( 
tional parenchyma. The emboli 
formed by an endovascular route with a coaxial 
microcatheter system (Tracker 18, Target Thera- 
peutics, Fremont, Ca) to reach the vascular lesion 
or by a percutaneous approach under Doppler 
ultrasonography guidance. There are several 
options regarding the embolic agents: polyvinyl 
alcohol particles, isobutyl-2-cyanoacrylate, alco- 
hol, and microcoils (Figs. 23.1a-d, 23.2a,b). The 
first choice for the occlusion of pseudoaneurysms 
is microcoils deposition on both sides of the pseu- 
doaneurysm neck [8] followed by in-situ deposi- 
tion of tissue adhesive, alcohol, or particles. We 
do not use Gelfoam because it is a temporary 
occluding agent and it carries the risk of future 



23.3.2 

Pelvic fracture 

Although rare in pediatric patients, severe hem- 
orrhage is a significant complication of pelvic 
fractures and pelvic crush injuries, and a lead- 
ing cause of early mortality. Angiography and 
embolization of bleeding vessels have been rec- 
ommended for the management of pelvic bleeding 
in patients in whom hypotension is unresponsive 
to resuscitation and/or surgical exploration [9]. 
The pelvic vessels are accessed by femoral or axil- 
lary approach using Seldinger technique. Once the 
extravasation is identified and selectively cannu- 
lated, the embolization is performed with Gel- 



23.3.3 

Vascular anomalies 

Mulliken and Glowacki [10] proposed a classifica- 
tion that was accepted by the Workshop on Vascular 
Anomalies in Rome infune 1996. The vascular anom- 
alies are divided into vascular tumors (hemangioma, 
hemangioendothelioma, and other vascular tumors) 
and vascular malformations. Hemangiomas are the 
most frequent tumors in infancy. They are character- 
ized by initial rapid growth of endothelial cells and 
subsequent slow involution. Vascular malformations 
are made of malformed or dysplastic vessels. They 
never regress. These vascular malformations are sub- 
categorized based on the type of channel abnormal- 
ity (arterial, capillary, venous or lymphatic) and flow 
rate (hi«h- or Ion- 1 low malformations). 



E iiil>. "■ I .:■ rlie:a pv in Pedia 



n 


. 



Fia.2.1.! n-d. Liver trauma in a 15-year-old boy. a CT-sc.an revealed the u;ese;ice of an arleria. pseudoa:ieii:ysni. h Selec;ive 
cadieierizjuon of I lie hepaUc a;:ety confirmee [lie pseudoaneiirysiii. c With a coaxial -vs;e::\ (Trac^e:: iic-nc-gade i we overpass 
die pse'jcloaneurysiii and occlude the exii hiancb with a coil. The: 1 ., we occlude ihe pseiidoanemvsiii wit/, y.ue fed owed by ihe 
occlusion of die afferenl aioery by a coii. d Conl;el a;ig.og:am revealed a compleie occlusion -.A' ti'.e pseudoaneurysm 




Fig.23.2a,b. A 10-year-old girl 
with severe hematuria post renal 
transplant biopsy, a Angiogram 



shoi 



the 



e of a 



doaneurysm. b With a coaxial 
system, a selective catheteriza- 
tion of the pseu doaneurysm 
was performed. The occlusion 
was performed with a coil 



J. Dubois audi. Garel 




Fig.23.3a,b. A 10-year-old boy with a p 

sonography shows the presence of a ps< 
performed with a 22-G Cathlon. 300 im 



:udOBneurysm of the femoral artery related 
idoaneurysm. b Under u Urn sonography, dire 
5 of Thrombostat permit to occlude the pseu 



catheterization, a Color ulti 
e of the pseud onne'jrysm h 



The indications of embolotherapy in vascular 
anomalies are: (1} hemangiomas refractory to medi- 
cal treatment, (2) hemangioendotheliomas with 
Kasabach-Merritt phenomenon, (3) liver heman- 
gioma with cardiac failure, and (4) arteriovenous 
malformations. 



23.3.3.1 
Hemangio 



s Refractory to Medical Treatment 



Ten to 20% of hemangiomas need to be treated 
[11]. Medical treatment is the first choice using ste- 
roids, interferon, or vincristine. Embolotherapy is 
only indicated in cases of ineffective medical treat- 
ment. Embolization is mostly performed in cases of 
hepatic hemangioma with cardiac failure, heman- 
gioendothelioma complicated L>v Kasabach-Merritt 
phenomenon, and uncontrolled proliferative hem- 
angioma wilii functional ilisorcler (e.g. tongue with 
feeding problem). 

The embolization provides the control of the 
growth of hemangioma in its proliferative phase. 



23.3.3.2 

Kaposiform Hemangioendothelioma 

Mueller [12] and Enjolras [13] reported that 
Kasabach-Merritt phenomenon (KMP) is caused 
by kaposiform hemangioendothelioma (KHE) or 
tufted angioma. The KMP consists of thrombocy- 



topenia, microangiopathic hemolytic 
localized consumption coagulopathy in association 
with rapid evolutive hemangioendothelioma. This 
syndrome requires an aggressive treatment, and 
carries a mortality rate of 20 to 30%. 

Aspirin, dipyridamole, antifibrinolytic agents, 
aminocaproic acid, corticosteroid, interferon, 
embolization, cyclophosphamide, pentoxifylline, 
radiotherapy, and antiplatelet aggregating agents 
have been tried with variable success [12-14]. Hepa- 
rin has been shown to boost the growth of KHE [12] 
and worsens the clinical situation [15, 16]. 

Embolization aims at reducing the high flow. 
Embolization is performed by arterial approach 
using polyvinyl alcohol particles or alcohol 
(Fig. 23.4a-d). 



23.3.3.3 

Liver Hemangiomas 

The differential diagnosis of hemangioma of the 
liver include hepatic angiosarcoma, hepatic epithe- 
lioid hemangioendothelioma, or metastatic disease 
like neuroblastoma. No treatment is required incase 
of asymptomatic hepatic hemangiomas. The main 
indications for treatment of hepatic hemangiomas 
are congestive heart failure, patients who requires 
mechanical ventilatory support, feeding problem, 
or Kasabach-Merritt phenomenon. Steroid is the 
initial drug. Interferon or vinblastine is reserved 
for refractory cases. We believe that embolization 



'-. ml>. "■ I .:■ thera pv .n t-'ed i:i 






Fia.2.i.-lii-t). A : 3 -year-old boy with rerraclory scroral mid perineal heniangi:: iriii.1o;he.ioma wit;: recc.rreni r-crota I bleed n'.y. 
a Selective cath^eri/aiion 01" die perinea I branch from I he inlci na! pudenda! artery through an anastomosis tVoin the common 
re mora i arrei y was per to i into. The opaciticadon. c!emonsira:es an impo; '::.:•:. stagiiado:' of contrast m Ihe ".umor. b The embo- 
lization was pei formed with Gelfoan'.. c Calfielci nation of [he i'.yy, ;-ga stric a; terv wit:: selective caiheici iz.Uion o:' die perineal 
branch from [he interna] pudenda: artery shows a tumor blush, d (Control) angiogram after devasculariziUion of the tumor 
with particles 



in combination with medical treatment is the best 
alternative in symptomatic liver hemangiomas. 
Pre-embolization mapping is mandatory, assessing 
the potential involvement of intercostal or phrenic 
arteries in addition to the hepatic artery and the 
portal system [17-19]. 

Five patterns of angiographic ti tidings had been 
described by Kassarjian et al. [20]. Embolic mate- 
riel 1 is selected depending on Hie vascular type. Tile 
first type, the most classical appearance, is early 
filling of abnormal vascular channels, stagnation of 
contrast material, and no evidence of a direct shunt- 
ing (Fig. 23.5a-h). Type 2 shows high-flow nodules 



without direct shunts. Large particles can be used in 
type 1 and 2. Type 3 is made of arteriovenous shunts, 
type 4 of portovenous shunt, and type 5 of both arte- 
riovenous and portovenous shunts. 

The embolization is performed by arterial 
approach for types 1,2, 3, and 5, and by transhepatic 
transvenous approach for portovenous shunts in 
type 4. Platinum fiber microcoils are generally safe 
in types 3, 4 and 5, and permit the occlusion of the 
shunts. Glue (n-butyl-2-cyanoncryhte) is the most 
effective material in patients with direct arteriove- 
nous and arterioportal shunting arising from mul- 
tiple sources [21]. Medical antiangiogenesis drugs 



J. Dubois and L.Garel 




s. 



Fig.2.i."ii -h. A _-:"cntii-oid girl 0:1 steroio neatnim; i v l t :: progressive .'we: hemangioma .-[id reTOn'.g L'roHcr.:, a MK imagiiig 
shows a I urge live: iv.Liss iii the right lobe of the liver, b-d The a 01 log ram demons; rates j vascular turn 01 supplied oy "die rig Is I 
branch ot' the hep.itic artery with stasmition 0: die conlras; and normal di fining vvm. i!Uis;rnlive of type 1 live: iiemu 11 gi oiy.ii. 
c Selective ■."iitiietei izatic:' of the light hep-aide a: :eiv was pel formed. f,g With ,1 coaxiid sysiem, m .duple feeders were catheter- 
ize-ci to! I owed by a:' embolizadcn widi pji licles. h i-d-st embolization opacification of the right heoadc ai iery shows a significant 
devuscdanzadon of die tumor 



should be maintained after embolization 
nearly complete regression of the lesions. 

Vascular malformations of the liver are 
except for the venous malformatic 
hemangioma in adults. Most arteriovenous malfor- 
mations of the liver are seen in hereditary hemor- 
rhagic telangiectasia (Rendu-Weber-Osler) with 
hepatic ischemia, congestive heart failure, and 



1 portal hypertension. Because of the risk of increas- 
ing the hepatic failure, embolization is not recom- 

:, mended in diffuse lesions. Liver transplantation is 

i indicated in such instances. 

Arterioportal fistula in cases of hereditary hem- 
orrhagic telangiectasia, Ehlers-Danlos syndrome, 
or patients with biliary atresia and cirrhosis can be 
treated by embolization. 



'-. nib, "■ I .:■ iiie:.i py .n t-'ed i:i 



Pure venous malformations are rare and asymp- 
tomatic in children. Most of them are seen specifi- 
cally in patients with blue rubber bleb nevus syn- 
drome which is a familial condition with multiple 
venous malformations of the skin, musculoskeletal 









23.3.3.4 

Arteriovenous Malformations 

Arteriovenous malformations (AV.VI) are .in impor- 
tant challenge for interventional angiographers. 
These his;li I low vascular malformations are abnor- 
mal communications between arteries and veins. 
We do not understand exactly why some AVMs 
respond well to embolization while other AVMs 
progress ineluctably despite embolotherapy. That 
is the reason why we favor a conservative manage- 
ment for quiescent, stable, and non-bleeding AVM 
with an annual MRI and cardiac ultrasound. If the 
AVM progresses, bleeds, or disfigures, angiography 
is essential to provide the road-map necessary for 
embolization. The angiographic characteristics of 
AVMs are dilatation and lengthening of afferent 
arteries, with early opacification of enlarged effer- 
ent veins [22]. To destroy the AVM and reduce the 
risk of recurrence, a superselective catheterization 
with microcatheters is necessary combined with 
percutaneous puncture when feasible [23]. The best 
agent to destroy the nidus is dehydrated alcohol. 
The amount of ethanol needed and the pressure 
of injection are evaluated with contrast media test 
injections. The maximum dose is 1 ml/kg. Over 
1 ml/kg, the elevated serum ethanol levels put the 
patients at risk for respiratory depression, cardiac 
arrhythmias, seizures, rhabdomyo lysis, and hypo- 
glycemia [24]. The ethanol penetrates to the cap- 
illary level and totally devitalizes normal tissues. 
Balloon occlusion, tourniquets, blood pressure 
cuffs inflated above systolic pressure, or a combi- 
nation of these can be useful if vascular occlusion 
is necessary to induce stasis. Temporary compres- 
sion of the venous drainage during the injection 
slows the blood flow, but one should relieve the 
occlusion slowly to avoid a significant modification 
of the pressure within the AVM. Coagulation dis- 
turbances have been reported in response to dehy- 
drated alcohol thai could increase the risk of bleed- 
ing, thrombosis, or hematoma. In these patients, in 
which the embolization is followed by surgery, the 
use of glue or coils as a substitute for dehydrated 
alcohol is recommended [25]; further studies are 



needed to evaluate the specific changes that occur 
with the dehydrated alcohol. N-bu ty 1-2- cy a noa cry- 
late or coils are used for large AVM or to avoid 
neuropathy when the AVM is close to nerves. 

Many complications are reported particularly 
with alcohol embolization, such as pulmonary 
embolus, cardiovascular collapse, neuropathy, skin 
blisters, radiculopathy, finger numbness, and focal 
skin necrosis [23, 24]. Arterial line monitoring and 
Swan-Ganz catheters are recommended for large 
AVM embolization [24] (Figs. 23.6a-d, 23.7a-d). 

AVM can be associated with hereditary hemor- 
rhagic telangiectasia (HHT), also known as Osler- 
Weber-Rendu syndrome. It is an autosomal domi- 
nant inherited disease of the vascular connective 
tissue characterized by epistaxis, telangiectasia, 
and visceral arteriovenous malformation. The 
organs mostly affected are the lungs (Fig. 23.8a-c), 
liver, brain, and the gastrointestinal tract. HHT is 
difficult to treat and requires a multidisciplinary 
approach tor its management. 



23.3.4 

Renal Embolization 



Renal scarring secondary to vesicoureteral reflux 
may be the cause of renovascular hypertension. 
Renal ablation is an alternative to nephrectomy to 
remove to involved kidney. The selective emboliza- 
tion should be performed with alcohol to prevent 
collateral revascularization. The efficacy is debated 
considering that embolization may delay the defini- 
tive treatment [26]. Gelfoam and coils are less valu- 
able than alcohol because of collateral revascular- 
ization (Fig. 23.9a,b). 

Selective renal embolization can be useful and 
effective in cases of refractory urinoma following 
partial nephrectomy or blunt trauma. The goal of 
the embolization is to ablate the fragment of renal 
parenchyma that is producing and leaking urine 
[27]. The embolization is done through selective 
catheterization of the vessel adjacent to the leakage. 
We recommend the use of particles (polyvinyl alco- 
hol) as embolic agent. 



23.3.5 
Epistaxis 

The differential diagnoses of epistaxis in childre 
includes trauma, foreign-body impaction, bleedir 
diathesis, vascular disorder, vascular anomalii 



J. Dubois and L.Garel 




Fig.23.6a-d. A 1 7-year-old boy wsth p. rcerjiiiver.o'.LS mo I for ma; ion i AV'V ) o:' die ;hird Jig. I. si ^eleouve o.uneiei izo:ion of die br.i- 
ohial a;:ery was peiformeo and showed jii AVM or' :lie ihii\i digit. b,o Perouran.e :■ n s oppiviooo w.is perfo: med. "Juder fhiorosoo.'y, 
wilh prior mopping ■.:■:" il'.e AVM from I lie ;i:oeria; side, we pu no; lire oneolly the AVM with o l.'-G hi'.lerrlv needle n two siles. 
The joqu I si :i o:'. CiOWS I .: t- :p.:0.roo:io:\ :T L'.e AVM. We m r.Med _ oo ■ ■:' aloohol. d : ■. ■ n L : ■. I .ngi ogre, m oe or lei ;.-l approoo.'. i.".'-ws 
;i signir.eant d ev .■ so : ..lonza:i on of [he AVM but o severe orler.ol spas:v.. We mon.iloreo, the ooloration 0110 skin ;empe:oiure. No 
arleiial isohen'/o wjs seen: beea : .,se of :lie sigmfieaiiL venous eon.gestion. o noil reseouon was pert"' Milled wrho.ii any icqueior 



(reported but rare in Sturge-Weber syndrome), and 
nii.i.il hemangioma. The most common indication of 
embolotherapy in children presenting with epistaxis 
is nasopharyngeal angiofibroma. A benign tumor, 
most often seen in the adolescent boy, juvenile 
angiofibromas originate from the sphenopalatine 
foramen and extend into the paranasal sinuses, the 
infratemporal fossa, the middle fossa, and the orbit. 
The optimal treatment consists of preoperative 
embolization followed by radical surg 



CT-scan and MR are essential both for the diagnosis 
and the assessment of the tumoral extent. 

The arteries supplying juvenile angiofibromas 
arise from branches of the external carotid artery 
(ECA). The tumor blush is intense and persistent. 
There is no arteriovenous shunting within the 
lesion. The contralateral ECA should be explored in 
all cases that reach the midline. The distal internal 
maxillary artery is the first vessel to investigate. 
Large tumors are also supplied by other branches 



!-_ mbo I o therapy in Pedia 




Hg.2.i.7a-ii. A:i i ! -year- old boy widi an arteriovenous malformation i AVM) o: ".he lei": ^h or. Id er. ;i ielec live ca:hcteri/.tlioii oi' 
".he posterior circumflex artery from the humeral artery was performed with a Trucker }■?. Angiogram rieiru 'nslrir.ed mukiple 
dilated arteries with early venous drainage. l> A Tracker 1 3 was introduced through the Tracker 33 for select ire catheterization 
of the arterial feeders. Alcohol was used for embolization. The control angiogram revea.ee. a complete devascdarizauon of the 
AVM. c S weeks later a residual AVM was see;: at the control ar;g.ogram. d We used the same coax. a I -vsirm and alcohol tor the 
oevasculanzation -:A the residual AVM w.ta a good result 





b 


i 



Hg.2. l .Ba c. A S-vear-cl.i girl with jiendr.-Wener-Osler syndrome. ,i At pulmortarv angiogram, -mall aneurysms din-flirs) are 
seen in the left cr.iig. Multiple areas of telangiectasia w.li'. diffuse arteriovenous shuming are seen predomirtanllv in both lower 
lobes liinoir/icii:/!. l> On selective injection of the right renal artery, several small aneurysms din-ciu'/icir/i associated with 
:elai;g. ectasias [ay >■:■■;. i\ are seen, c After selective imection o: the superior mesenteric artery, a small aneurysm uiiraH and 
diffuse area of telangiectasia are demonsl rated in the proximal jejunum 



J. Dubois and L.Garel 




Fig. 2 3.9a, b. Akohol e:-LL"iojzo;io:i :■( o nop hio kidney :oi rct":oo:o:y hvpenei'.s lor., a Se.eoiiv- ooihetensni of .e:'l je:".;il 0Ke:y. 
h Reno] .uteiy opLiiilkjlion of:ei okohol embolization shows o complete devosaLiinzation of [lie kidney. NoiT.iuizoiio: 1 . o:" 
blood pressure was noted 



such as the accessory meningeal, the ascending revascularization) is usually performed with par- 

phai vngeal, and the ascending palatine arteries. In tides or tissue adhesive (Fig. 23. 10a, b). When the 

case of intracranial tumor extension, the hemody- tumor has invaded the cavernous sinus, the pitu- 

namks (anastomoses) between the internal max- itary fossa, the suprasellar area, or the intracranial 

illarv system and the ipsilateral internal carotid intradural area, permanent preoperative balloon 

artery must be analyzed carefully. Distal emboli- occlusion of the internal carotid artery could be 

zation (proximal occlusion would result in tumor helpful [28, 29]. 





flg.l ■-lOo.ti. I ::■■■.-.' i.- pli.i! ■:.)!-. . .::;g;oii : o . I v r .n ...-,|,- ; .y. ;-!-■ pei ot.ve =■ j j"i L" ■ :■ . i ^ o : l ■; ■ n wi:l'. portholes of trie oi Le- 

na I bronohrs oi f I he inlemol maxillary artery, a s elective ex lei no I corolid angiogrom shows o vascular tumor le-ci by multiple 
branches :■( ijie in;ernul moxillory orlerv. I> A fie: mukiple s^c live caihelc: izalion \vi:h .: cooxia! system i Tracker o- ond Trackei 
18), embolization wos penormed kilIi poi ucies. Pos'.embokzoiioii -.i n g i i:-gi :.:•:. shows sigmficoiit de vascularization 



'-. nil>. "■ I .:■ theta pv in t-'ed i:i 



23.3.5 
Hemoptysis 

Most patients who may benefit from bronchial artery 
embolization are teenagers with cystic fibrosis (CF) 
and major hemoptysis. The hazards of general anes- 
thesia with positive pressure ventilation inCF patients 
have been addressed recently in the literature. A 
fatal pulmonary hemorrhage during induction was 
reported by Mcdougal and Sherrington [30]. The 
technique is well established: femoral artery access, 
5-F catheter, precise vascular mapping, coaxial tech- 
nique with Tracker 18 catheters, secure catheter 
placement prior to performing embolization, and use 
of polyvinyl alcohol particles (size: 355-500 microns 
or 500-710 microns if microfistulas are observed) 



(Fig. 23.11a-d). Coil embolization or surgical ligation 
of bronchial arteries should be avoided because they 
hinder subsequent catheterization of the proximally 
occluded vessel. Careful attention should be paid 
for the identification of spinal arteries arising from 
the bronchial or intercostal arteries. The localizing 
value of emergency bronchoscopy or multidetector 
CT scanning prior to embolization remains to be 
evaluated. 

Bronchial artery embolization in CF patients is 
very effective immediately and on short-term basis. 
Many patients will require repeated embolizations 
during the follow-up [31], 1 Vspite the reported severe 
complications of bronchial artery embolization, the 
procedure has proved to be safe if performed by 
experienced angiographers. 




Fig. 2.V I ] a-il. Hemolysis in .'. i 7-year-old boy wiih -."y~:ii" flnrosis. a Selective catheterization, o: tite light crouch i a I ;iririy with 
a 5-F hook catheter shows dilated anci tortuous arteries in the superior .'.ad middle pulnioaaiv lobes, b With a coaxial system 
:. kvnegiv.e 1 Pi, an embolizai.oi": was perfoimed with particles 1 Contour 500-733 microns). Postembol.z.tiion angiogram shows 
olevasculaf.zi'.tion ct trie bleed, ng site, c Seleot ive c.'.theteriziition. o'i Ihe broncho-intercostal Ir.inL \iulliple ciliated and tortu- 
ous arter.es are seen, d selective catheterization wiih tiie same coaxial system. Embolization was peiformeo with pi'.rucles. 
i-rsiemholiziU.on angiogram shows complete- devaso.ilarization 



J. Dubois and L.Garel 



23.3.6 

Gastrointestinal Bleeding 

In children, localized gastrointestinal bleeding is 
usually secondary to duodenal ulcer and less fre- 
quently to gastric ulcer, Meckel's diverticulum, and 
vascular malformations. Diffuse bleeding can occur 
in vasculitis and coagulopathy. 

Gastrointestinal vascular anomalies are often 
associated with known syndromes such as Klippel- 
Trenaunay, Rendu- Osier-Weber, blue rubber bleb 
nevus, and Proteus syndromes. 

In our experience, venous malformations are the 
more common vascular anomaly encountered in 
cases of bleeding (Fig. 23.12a,b). 

In children with gastrointestinal arteriovenous 
malformations, the angiographic examination con- 
firms the diagnosis and the extent of the AVMs. 
Embolization is usually not recommended in small 
bowel and colonic lesions because of the risk of 
necrosis [32, 33]. Embolotherapy is sometimes con- 
sidered preoperatively to lower the risk of operative 
bleeding. 



23.3.7 

Embolization or Sclerotherapy of Varicoceles 



Varicoceles are present in 15%-20% of preadoles- 
cents and adolescents. The treatment of varicoceles 



in this age group is controversial. The treatment 
can be surgical, endoscopic or radiological (sclero- 
therapy or embolization of the internal spermatic 
vein). Most of the varicoceles are seen on the left 
side. Bahren et al. [34] described five types of left 
varicoceles according to the anatomy of the internal 
spermatic vein (ISV). 

Irrespective of the type of varicocele, the sclero- 
therapy procedure is the same. Our technical pro- 
tocol for percutaneous endovascular occlusion 
of the ISV is as follows: IV sedation by Ketamine/ 
Midazolam, femoral vein approach, 7-F Cobra 
catheter with coaxial 3-F or Tracker 18 for distal 
sclerotherapy by sodium tetradecyl sulfate (STS) 
followed by more proximal coil occlusion, bed-rest 
for 4 hours, and discharge 6 hours post-procedure 
(Fig. 23.13a,b). Results are assessed by the referring 
surgeon 2 months later. 

In our series, we have found a high incidence (-[■[%) 
of anatomical variants in the pediatric population. 
Technical difficulties of retrograde sclerotherapy 
were seen in type IVb. collaterals from segmental 
renal veins to the internal spermatic vein with a com- 
pete nt ostial valve (12% of our cases, failure rate 50%) 
and in type V, double renal veins (14% of our cases, 
failure rate 33%). Our overall results (failure rate 10%) 
are comparable to the recently reported pediatric 
series [35, 36], in the radiological and surgical litera- 
ture. The issue of radiation related to interventional 
procedure has been addressed in the literature. If the 




if the light colli: .iiiriy shows dyspljs 




Fig.23.13a,b A 12-year-old boy with left varicocele, a Selecrive catheterization of the left spermatic v 
coaxial 3-F catheters. I> iniection of 4 cc of sodium r e r j" .1 J e ■." y J sulfate- with proximal coil occlusion 



t-f cobra and a 



usual principles of radioprotection a 
gonad-dose (0.01 mSv) is negligible. 



23.3.8 
Hypersplenism 

The causes of Hypersplenism in children are cirrhosis 
secondary to cystic fibrosis or biliary atresia, portal 
vein thrombosis, thalassemia, and idiopathic throm- 
bocytopenic purpura. Hypersplenism is treated by 
surgical resection with subsequent increased risk of 
infection in the pediatric age group. 

Partial splenic embolization is an alternative to 
splenectomy. Preprocedural and postprocedural 
antibiotic prophylaxis is recommended. Under gen- 
eral anesthesia, the embolization is performed with 
a femoral 5-F catheter. The catheter is advanced 
through the femoral artery to the mid splenic artery. 
Subsequent catheterization into the intrasplenic 
arterial branches is performed either with the 5-F 
catheter or if necessary, coaxially with a microcath- 
eter. The embolization is done with an iniection 
of polyvinyl alcohol particles and antibiotic solu- 
tion containing 0.2 mg of ampicillin (Fig. 23.14a,b). 
Splenic embolization is monitored angiographically. 
The procedure is completed after approximately 
70% of the splenic parenchyma isdevascularized [6]. 
Aggressive pain control is needed for 7-10 days post- 
procedure. In children, Harned [37] demonstrated 



that 30%-40% embolization of splenic blood flow 
is enough to improve the platelet count and white 
blood cells with a shorter hospitalization, faster 
recovery, and fewer complications. Complications 
of splenic embolization include fever, leucocytosis, 
pain, pleural effusion, splenic abscess, and peri- 
tonitis. The long-term results after partial splenic 
embolization have not been well established [38]. 
Up to now, partial splenic embolization as treat- 
ment of Hypersplenism in children has not gained 
wide acceptance in western countries. 



23.3.9 

Thrombolysis 



Experience with thrombolysis in children i 
ited but the need for this procedure I 
because of the need to treat complications of cardiac 
catheterization and systemic arterial intervention. 
Agents used include urokinase and rtPA. Effective 
dose schedules for children have been extrapolated 
from adult studies. Coagulation and fibrinolysis are 
probably different in pediat 1 ics, particularly in neo- 
nates. Plasminogen levels are known to be low in 
neonates, and it has been proposed that plasmino- 
gen or fresh plasma be given to enhance fibrino- 
lytic therapy. Most centers favor rTPA, and this may 
be locally delivered via a selective catheter. Local 
low-dose therapy is unlikely to produce systemic 



J. Dubois and L.Garel 




Fig.23.14a,b.A /-yeor-old hoy with spheno.cytosk on..: ■".yi'eisn.eaism. a Selei":iv=? L.iihcici'iz.ii.oij •:■< ti'.e -^leen shows die j 
div ;y, i ;gr. '.;•.; of the sn.een. b Pcs:.fmbol.i.ai::oi .1 ngii:-gr;i:r. will', i.'.e lesiduol n or en c by ma 



changes in coagulation, whereas systemic therapy 
will and therefore has greater risk and more contra- 
indications. In all systemic therapies, the fibrinogen 
level, thrombin time, prothrombin time, and acti- 
vated partial thromboplastin time are monitored at 
regular intervals, and the children are observed in 
an intensive care unit or neonatal nursery. Systemic 
therapy with heparin is recommended for indwell- 
ing catheters, but its role in the neonate is uncertain. 
The trend in thrombolytic therapy is toward higher- 
dose, shorter-duration treatment given locally, 
which often produces rapid clearing of thrombus. 
Failures may be related to delays in implementing 
therapy, going to maturation of the thrombus. 

The most common lesion treated in the authors' 
center is femoral artery thrombosis complicating 
catheterization, especially for balloon angioplasty 
of the aortic arch or aortic valve. These procedures 
require the insertion of a large balloon, which is 
currently mounted on large shafts. Initially a local 
low-dose approach from the opposite groin was pre- 
ferred, but now systemic therapy is frequently used 
if there are no contraindications. Local low-dose 
thrombolysis is used for thrombosis of Blalock- 
Taussig shunts, dialysis fistula, pulmonary artery 
thrombosis, iliofemoral thrombophlebitis, aortic 
thrombosis in neonates, and brachial artery occlu- 
sion after supracondylar fracture. 

Bleeding is the most serious complication of 
thrombolytic therapy. Cerebral hemorrhage is a 
concern in the neonate, and in this age group the 
authors elect local low-dose treatment. Bleeding 
from recent surgical sites is also a well-known com- 
plication of thrombolysis. 



23.3.10 
Chemoembolization 

We have no personal experience of chemoembolization 
and the pediatric literature on the topic is scarce. 

According to the International Society of Pediat- 
ric Oncology and the Pediatric Oncology Group, the 
standard therapeutic protocol for childhood primary 
malignant tumors of the liver is based on the associa- 
tion of systemic chemotherapy and surgery [39-44]. 
Such an approach has improved signilicantly the out- 
come of hepatoblastomas, especially when the tumor 
is unresectable at presentation. Some case reports 
and a few series have outlined the interest of preoper- 
ative hepatic artery int 11 sion ot eisplat in and/or doxo- 
rubicin and of TACE (transcatheter arterial chemo- 
embolization) in advanced hepatoblastomas [45-51]. 
According to these reports, the results were more than 
encouraging, with a much lower toxicity than con- 
ventional systemic chemotherapy. To the best of our 
knowledge, however, no study has demonstrated the 
superiority of preoperative TACE over preoperative 
systemic chemotherapy in increasing the resectabil- 
ity of initially inoperable hepatoblastomas. Besides 
hepatoblastomas, TACE has been reported also in 
pediatric cases ot hepatocarcmoma [?2] and even 
hepatic metastases [53]. According to the proponents 
of the technique, TACE appears feasible in children 
and can be performed in cases of unresectable tumor 
confined to the liver, when the portal vein is patent, 
and in the absence of biliary obstruction. 

Inadvertent pulmonary lipiodol embolism during 
TACE has been reported in adults [54] and children 
[55]. 



E iiil>. "■ I .:■ rhera pv in [-' e J i ;i 



Transarterial catheter chemotherapy and/or 
rinb''! i.-'.ii lion in the management ot advanced 
hepatic malignancies is still a work in progress and 
has not been endorsed by the international pediat- 
ric oncologic societies. On the other hand, the future 
development of gene therapy in children delivered 
through a vascular route can already be antici- 



23.4 
Conclusion 

Embolotherapy of small vessels in small patients 
needs well- trained pediatric radiologists and a ded- 
icated environment. Networking between the few 
centers performing such procedures in children is 
paramount for continuously optimizing both their 
indications and their techniques, and for assessing 
their effectiveness. 



l.MacPherson DS ( 1 993 ) Preoperative laboratory testing: 
should any tests be "routine" before s : ..j gery? Med Clin 
North Am 77:289-308 

2.Saker MC, Ueiima T (2002] Management of the pediatric 
patient for intervention;;! radiologic procedures, Semin 
Inter vent Radiol 19:3-12 

3. American College .:■[ Radio. ogy Commit:ee on Drugs and 
Conlras: Media i ] 99S) V a mi a I on con: rast media, 4:h edn. 
Reston,VA 

4.Freed MD, Keane JF, Rosenthal A (1974) The use of hepa- 
tization to prevent a::e:ia! thrombosis after percu:a ne- 
ons cardiac cathe:e: ization in in fa ins. Ci:cula;ion 50:565- 
569 

5. Burrows PE, Mason KP (2004) Percutaneous treatment of 
low flow vascular malformations. ! Vase [mervenl ji.ad.ol 
15:431-445 

6.SpigosDG,TanW5, MozesMF, etal. (1980] Splenic embo- 
lization. Cardiovasc Intervent Radiol 3:282-287 

7. Vane DVV (2002) Imaging of the imured child: important 
questions answerer! quickly and correctly. Surg Clin N An; 
82:315-323 

S.Coffer:e PP, La:erre PF [1001] Trauma- ic injuries: imaging 
,m.:: intervention in post-traumatic so nip licit ions (delayed 
intervention) Eur Radiol 12:994-1021 

9. Cook RE, Keating JF, Gillespie I (2002) The role of angi- 
ography :n the management of haemorrhage from maior 
fractures of the pelvis. I Bone Joint Surg 84-8:178-182 
10. Mulliken JB, Glowacki I (1982) Hemangiomas and vascu- 
lar malformations in infanis and children: a slass.fication 
based on endothelia! characleiisiics. Plast Keconsir Surg 
69:412-422 
1 1 . Enjolras O, Riche CM, Merland JJ, et al. (1990) Management 



o: alarming heman.g.omas ;n infancy: a review of !-.< cases. 
Pediatrics 85:491-498 

2. Mueller BU, Mulliken JB (1999) The infant with a vascular 
tumor. Semin Perinatalo 23:332-340 

3. Enjolras O, Wassef M, Mazoyer E, et al. (1997) Infants with 
Kasabach-Mern:; syndrome do not have "true" heman.g.o- 
mas.JPediatr 130:631-640 

4. Hu B, Lachman R, Phillips J, et al. (1998) Kasabach-Merritt 
syndrome-associated kaposiform hemangioendothelioma 
successfully ireaied ivi-h cvclophosphamide. vincristine, 
and actinomycin D. | ! J edia:r Hema:ol Oncol 20:567-569 

5.Folkman J, Klagsbrun M, Sasse J, et al. (1988) A hepa- 
rm-binding angiogenic pro: tin - basic fioroblasi gro\v:li 
factor - is stored within basement membrane. Am ) Pathol 
130:393^00 

fi.Folkman J, Mulliken JB, Ezekowitz RAB (1997) Antiangio- 
genic :herapv of hemangioma: wi:h in;er:eron-alpha. in: 
Stuart -Harris R, ! J enny HP teds; Clinical applications of rhe 
interferons. Chapman Hal! Medical. London, pp 255-265 

7.Burrows PE (1991) Variations in the vascular supply to 
infantile hepatic hemangioendotheliomas. Had io logy 
181:631-632 

8. Fellows KE, Hoffer FA, Markowitz RI, O'Neil JA Jr (1991) 
Multiple collaterals to hepatic infanti.e hemangioeiidothe- 
liomas and arteriovenous malformations: effect of embo- 
lization. Radiology 181:813-818 

9.McHugh K, Burrows PE (1992] Infantile hepatic heman- 
gioendotheliomas: significance of ponal venous and sys- 
lem.ic collateral ar:e:ial supply. I Vase interven; Radio! 
3:337-344 

0. Kassar.ian A, Dubois J, 
classification of hepat. 
ogy 222:693-698 

1. Burrows PE, Dubois J, Kassariian A (2001 ) Pedial 
vase a la r anomalies. Pediatr Radio. 3: :=32-'-4s 

2. Burrows PE, Mulliken JB, Fellows KE, el al. (1983) Child- 
hood hemangiomas and vascular malformations: angio- 
graphic differentiation. A I k Am ! Koemgenol [41 :4Sa-4?B 

3.¥akes WF, Luelhke )M, Parker SH, et al. (1990) Ethanol 
embolization o: vascular malforma:ions. Had.ogiaphies 
10:787-796 

4. Mason KP, Michna E, Zurakowski D, et al. (2000] Serum 
etrianol levels in. children ju: aduks after eihanol emboli- 
zation oi s otero; hei apy for vascular anomalies. Hadiologv 
217:127-132 

5. Mason KP, Neufeld EJ, Karian VE, el al. (2001 ) Coagulation 
aonormaliiies in pediatric and adul: patients after sclero- 
therapy or embolization of vascular anomalies. AJR Am J 
Roentgen 177:1359-1363 

6. Ognianovic MV, Richardson D, de la Hung M, et al. (2002) 
Selective renal embolization for renovascular hyperten- 
sion? Arch Dis Child 86:127-129 

7. Horikami K.Matsuoka Y, Nagaoki K, et al. (1997) Treatment 
of post-traumatic urinoma by means -:A selective arteria! 
embolization. J Vase Interv Radiol 8:221-224 

S.Lasiaumas P tlPPOi Nasophamigeal angiofibromas: haz- 
ards of embolization. Radiology 136:119-123 

9. Valavanis A ! 1993! Embolization of intracranial and skill! 
base tumors. In: Va.avams A iedi Interventional neurora- 
diology Springer- Vertag. Berlin: New York, pp 77-83 

O.MeDougall RJ, Sherrington CA (1999) Fatal pulmonary 
haemorrhage daring anaesthesia for bronchial anery embo- 
lization in cystic fioros.s. k'aediatr Anaesth 9:345-348 



I, Burrows PE (2302; Angiographic 
e hemangiomas in infants. Radii;!- 






I. Dubois and L. Gar 



31.Barben I, Robertson D, Olinsky A, et al (2002) Bronchial 
artery embctizaiion tor hemoptvs.s in young patients wit.: 
cystic fibrosis. Radiology 224:124-130 

32. Fremond B, Yazbeck S, Dubois J, et al. (1997) Intestinal vas- 
cular anomalies in children. I Pediatr Surg 32:873-877 

33.Fishman S J, Burrows PE, Leichtner AM, et al. (199B) Gas- 
troinlesLinal manifestations of vascular ,i noma lies in 
childhood: v.ined etiologies recti. re muliiple iherapeui.c 
modalities. J Pediatr Surg 33:1163-1167 

34.Ba'hren W, Lenz M, Porst H, et al. (19B3) Side effects, com- 
plications and contraindications for percutaneous sclero- 
therapy of the internal spermatic vein at the I rea intent of 
idiopathic varicocele. ROFO 128:172-179 

35. Lopez C, Serres-Cousine O.Averous M (1998) Varicocele in 
adolescents. Treatment by s c le tori: era pv and percutaneous 
embol.zution: reflect ion- on the method. A propos of 23 
leases. Prog Urol 8:382-387 

36,Ficarra V, Porcaro AB, Righetti R, et al. (2002) Antegrade 
scrota, scleroiherapv m the treaimenl o: varicocele: a pro- 
spective study. BJU Int 89:264-268 

37.Harned RK 2nd, Thompson HR, Kumpe DA, et al. (1998) 
i-'artial splenic embolization in five children with hyper- 
splenism: rffirCis >.■:' reduced-volume em conization on effi- 
cacy and morbidity. Radiology 209:803—806 

38.KimuraF, ItohH.Ambiru S.et al. (2002) Long-term results 
of LiiLti.il and repeated parl.al srlenic embolization for 1 he 
treatment of chronic idiopathic thrombocytopenic pur- 
pura. AJR Am I Roentgenol 179:1323-1326 

?'\ ;." a reel let A, H.anchai J !-!,' Ir. air. p ague l.rt a.. Surgical resec- 
tion anu chemoiherapy improve sarvivai rate for patients 
with hepatoblastoma. J Pediatr Surg 36:755-759 

40. Czauderna P, Macldnlay G, Perilongo G, et al. (2002) Hepa- 
toce:lu!ar carcinoma in children: results of the first pro- 
spective study of the international Society of Pediatric 
Oncology group. J Clin Oncol 20:2798-2804 

41.Pritdiard I, Brown J, Shafford E, et al. (2000) Cisplatin, 
doxorubicin, and delayed surgery for chilcihood hepaio- 
biastoma: a successtnl approach - results of the first pro- 
spective study of [he international Society of Pediatric 
Oncology.J ClinOncol 18:3819-3828 

42. Sasaki F, Matsunaga T, Iwafuchi M, et al. (2002) Outcome 
or" hepatoblastoma neaieo with the [PIT-! (Japanese Sltidv 
Group for Peril at: ic Liver Tumor) Protocoi-1 : A report from 
the lapanese Study Group for Peril abac Liver Tumor. | Pedi- 
atr Surg 37:851-856 

43.Schnater JM, Aronson DC, Plaschkes I, et al. (2002) Surgi- 
cal view -:.n the treatment ■.:'!" pa I. ems wilh fierr,t:tblast:tma: 



results from the first prospective trial of the In lei national 
Society of Pediatric Oncology Liver Tumor Study Group. 
Cancer 94:1111-1120 

44. Suita S, Tajiri T, Takamatsu H, et al. (2004) Improved sur- 
vival outcome for hepatoblastoma baser, on an optimal 
chemotheraaeiitic regimen - a ten or t front the suidv group 
for pediatric solid ntaiignant tumors in the Kyushu area, i 
Pediatr Surg 39:195-208 

45.Arcement CM, Towbin RB, Meza MP, et al. (2000) Intra- 
hepatic chemoemoo.izuiioi: in unresectable reu.airic .iver 
malignancies. Pediatr Radiol 30:779-785 

46. Gerber DA, Arcement C, Carr B, et al. (2000) Use of intra- 
hepatic ch en: o therapy to ]:c.)i advanced pediatric hepatic 
malignancies. | Ped ia lr ijastroenterol Ntitr 30:137-144 

47.Han YM, Park HH, Lee JM, et al. (1999) Effectiveness of 
preoperative t ra ns a rteri a i chemoembolization in pre- 
sumeo inoperable hepaioo.astoma. i Vase Interv Radiol 
10:1275-1280 

48. Malogolowkin MH, Stanley P, Steele DA, et al. (2000) Feasi- 
bility M':t.i toxicity of chemoemboiizatior. for children with 
liver tumors. J Clin Oncol 18:1279-1284 

49.0htsuka Y, Matsunaga T, Yoshida H, et al. (2004) Optimal 
slrategy of preoperative transcatheter arterial chemoem- 
bolization for hepatoblastoma. Surg Today 34:127-133 

50.Oue T, Fukuzawa M, Kusafuka T, et al. (1998) Transcath- 
eter a rteri a: ch err. o embolization in the treatment ot hepa- 
toblastoma. J Pediatr Surg 33:1771-1775 

Sl.Tashiian DB, Moriarty KP, Courtney RA, et al. (2002) Pre- 
operative ch err: o embolization tor unresectadie hepatooias- 
toma. Pediatr Surg Int 18:187-189 

52.Uemura S, Todani T, Watanabe Y, et al. (1993) Successful 
left hepatectomy for hepatocellular carcinoma in a child 
after Iransca .' ' oem. conization: report of 

a survival. Eur I'inIui: S.rjj ):54-56 

53.Mutabagani KM, KloplVnslem K[, Hogan MJ, et al. (1999) 
Metastatic p .: ii'.-*:iv.:i[i-.a .-.::.■ paraneoplaslic-induoed 
anemia in an ndnieMte-l ireatir.ent with hepatic arterial 
ciiemoembo../:rr.n ' IVdi.it- Hematol ijncol 21:544- 

54.Tajima T, Honda II, Kunnwa I', et al. (2002) Pulmonary 
complication- il'lt" ncpaiic a-tiry chemoembolization or 
infusion via iI:l inferior prire-i. artery tor primary liver 
cancer. J Vase Inlerv Kadi.ii ' W3-900 

55. Yamaura K. i-iigash. M. Akiyoshi K, et a.. (2000) Pulmonary 
lip.odo. embol.sm riar.ng transcatheter arteria. chemoem- 
bolization for hepatoblastoma unaer genera, anaesthesia. 
Eur J Anaesthesiol 1 7:704-708 



Subject Index 



-aorta 103 

itbsolute 

-alcohol 11,192 
-ethanol 169,204,205 
acalculous cholecystitis 185 
accessory 144 

- meningeal artery 239, 259 

- renal artery 113 

- rid'.l hepatic artery 151 
active bleeding 103 
acute pancreatitis IH5 
adenoviral vector 251 
aetmtisclerol 2H 

airv. :v 

compromise 35 

ma::ajjerr.cnl 
equipment iH 

|v-;-*-,i.:-.i:pi r/'irapy for intubjt 
alar artery 259 
ale: ■:-..:. .;.-.. HI 
allopurinol IBfl 
;;!p!i:i-fe:. /protein i 2 9 
i'.iiiiramy 52 
ancillary vessel 152 

- arterial wall 100 

- configuration 101 
-fusiform 100 

- mycotic 100 

- pseudoaneurvsin 100 
-rupture 109,114 
-saccular 100 

- true aneurysm 100 

I'.IlgioiTl.itOllS 

- polyps 264 

- "vascularization 192 
angiomyolipoma 101,113,202,210 



- spasm 30.i 



angn 



225 



angioneogenesis 225 
anosmia 247 

- cerebral artery 235 
-EPX 257 

- septal artery 259 
amiangiogenesis agent 222 
anticoagulation 123 
apoptosis 164 



.j failure i 



- fistula (AVF) 3,11,12,64,6 

- ma [formation 3,311,313 

- clinical stages 8 

- - iTongestivi 
--CT 9 

- MR imaging 9 

- percutaneous manag 

- - soft tissue mann S 

- shunting 134 

a tli ero sclera sis 101 
autopsy 113 
autosomal dominant dis- 
AVE 285 
AVF, Si 



avitene 242 
AVM 9,243,311 



bacteria! endocarditis. 
balloon 273 

- balloon- mounter siei 

- catheter 241 
-occlusion 273 

- tamponade 87 
bare metal stent 104 

bevacizumab 222 
biliary 
-atresia 317 

- indocyanine green 

biochemical pump 224 
biopsy 90 
birthmark 3 
bleeding diathesis 311 
blue rubber tieb nervus 
bone 

- healing 196 



- embolization 191 

- - estimated blood loss 193 

- - obliteration of tumor blush 193, 195 

- - post-emboliz.ttion syndrome 196 
bowel 

- ischemia 112 
-preparation 135 



breast 149 




-perfusion 82 


3-bromopyruvate (3-BrPa] 


222 


colorectal carcinoma 149 


bronchial artery ernbolizaii 


on 315 


common iliac 71 


Budd-Chiari disease 164 




complication 212 

congestive heart failure 202 
connective tissue disorder 111 


C 




contour SE 205 


c-.iucirr ai'.giogenesis 132, 1 


44 


com ra s 1- in c! : .; ceo nephropathy 210 


capecitabine 150 




corpuscular element 21 1 


capillary 




corticosteroid 5,245 


- hemangioma 264 




- therapy 196 


-malformation 16 




covered stent 285 


carcinoid 131,139,169 




cranial nerve 236,267 


- liver metastases 182 




craniofacial tumor 254 


-syndrome 178 




critical FRL 171 


-tumor 177 




cryotherapy 158,201 


- - metastatic 133 




CV'l. see lower-extremity chronic ver 


care! j ;;c arrhythmia 1 1> 




cyano; cry late 167, 168, 180 


caroticotympanic artery 236 


cystic 


caroticovertebral anastomose 239 


-fibrosis 315,317 


carotid 




- media! degeneration 1 11 


-blowout 244,271 




cytokine 164,170 


-dissection 274 




cytolysis 170 


- occlusion 272 




cy to reductive procedure 178 


-rupture 271 






- tumor 253 






causative agent 51 




D 


cauterization 258 




debulking ::ip":-rectomy 201 


CCC, see c hula *:niin:dl Jar 


carcinoma 


iliTiyilrstt-d .ilc.ih.ol 311 


celiacaxis 133 




d:':.l,' il 1. 


cell cycle enzyme 222 




balloon 114,241,277 


cell transplant n:n ,10 n 




r>ari: plaii::_m coil 244 


chemical i ir^iru^t:-: 293 




-latex 273 


chemodeclnma 251 




-platinum coil 279 


chemotaclic factor ! 32 




diabetes mellitus 172 


chemothi-apy : .1" 




diaphragmatic artery 132 


- mixture 




direct thrombin injection 105 


- - admission orders 136 




cisseminated intravascular coaguiaii 


--cisplatin 134 




cislal internal I'.iaxklaiy ,u lei v _?■ 


— complications 136 




oiverdculitis III 


- - doxorubicin 134 




dominant autosomal disease 266 


— hydrochloride 134 




dorsal 


— lipiodol 134 




-aorta 236 


— materials 135 




- '.';~ii:hal:n;c artery 235 


— mitomycin C 134 




doxorubicin 180,222 


cholaiigioceliular carcinoid 


a (CCC] 129 


- doxorubicin-eluting bead 228 


chronic idiopathic thrombi. 


■cvv-penic purpura 215 


drug-carrier 224 


cicatrization phase 125 




drug-eluting 226 


circle of Willis 276 




drug-loaded embolic agent 228 


circulatory shock 38 




ouodenal ulceration 100 


cirrhosis 21 1 




duplex ultrasound 70 


cisplatin 222 




DVT 124 


- cisplatin-londe-d polyid.l-iacddei microsphere 228 




classification of Pisch 250 






clinical toxicity 157 




E 


clival artery 238 




ecchymosis 123 


CNS depression 16 




Ehlers-Danlos syndrome 101,310 


coagulopathy 263 




electrolytic detachment 114 






hmboGold microsphere 205 
embolic agent 192 


-deposition 106 




-embolization 107 




embolization 


collateral 




- equipment list 84 


- pathway 82 




- of the superior gluteal artery 66 



embosphere 93,130,134,192 

- microsphere 226 
enbucrilate 305 

encapsulated microorganism 211 
endoglin 266 

end organ supply 1 14 
endothelium 204 
endovenous thermal ablation 

- clinical success 123 
■n rates 124 



- e\'Lnephrii 



123 
iS thermal injury 122 
-laser 121 
-RP ablation 121 

venospasm 122 
HNTbleeding 257,269 
en terobihary anastomosis 185 
or'eri chromaffin cell 177 

■ y ] 77 
o;.vdu:ai m. sthesia 213 
eprubicin 222 
episodic KPX 266 
ep:staxis 257,311 
epitbelioid angiomyolipoma 202 
:..-.' < 264 

e:-anl tnn :lization 193 
erythrocyte 204 

poh/amine 164 
esthesioneuroblastoma 248 
estimated blood loss 192,209 
e:han:il 2H.30,203 

ablation 113 
eti'.anolainme oleate 2S 
ethibloc 29-31 
ethiodol 223,225 
ethyl alcohol 243 
eustachian tube 258 
extern.;! carotid artery 23?, 236,264 
extracranial meningioma 248 
exti ahepalic 



72 



familial hypercholesteremia 296 
fatal toxicity 157 
IS FDG-PET 159 
femoral artery 73 

- - percutaneous ernbolizaiio 

- - postcatheterizalion 69 

- surgery 70 

- techniques 71 

- - thrombin injection 71 

- transcatheter 71 

- - ultrasound-guided compr 

- pse-;doaneurysm 84 
fibromuscular dysplasia 101 
fibrosis 170 

i-isch Classification 1-1% 
flap breakdown 275 
floxuridine(FUDR) 149 



fluoro-fade 191 

fluorouracil (5-FU) 158 

flush syndrome 169 

foam 30,31 

fracture of the pubic ramus 63 

frequent spontaneous EPX 266 

fresh frozen plasma I 91 

FRL, see future remnant liver 

5-FU, see fluorouracil 

FUDR.seefloxuridine 

future remnant liver (FRL) 163 



■"""TV -galactosyl serum albumin 172 

gangrenous cholecystitis 185 

gastrin 181 

gastrinoma 177 

gastroduodenal 

-artery 103,151,185 

-necrosis 134 

gastroepiploic 212 

gastrointestinal bleeding S3 

gasiroieiiinostomy S3 

-tube 

— bleeding 83 

— complication 83 
gastrostomy tube 
-bleeding 83 

- complication 83 

GCS, see graduated compression stocking 

GDC matrix" coil 244 

gelatin sponge 192 

-powder 180,223 

gelfoam 64,86,91,130,205,224,262,265,311 

- embolization 49 



-powder 64,242 
gene(DNA) 295 

- therapy vector 296 

- transfer 299 

general anesthesia _40 

genotype 266 

Giacomini vein ablation 122 

giant hemangioma 6 

gigabec-querel 1 42 

Gilbert's syndrome 151 

glomeruli 204 

glomovenous malformation 22 

glucagon 164,181 

glue 309 

graduated compression stocking (GCS) 123 

grays 142 

great saphenous vein (GSV) 119 

groin bleeding 69 

growth factor 170 

- inhibitor 150 

GSV, see great saphenous vein 
Guglielmi detachable coil 93 



H 

HCC, see hepatocellular a 
hemangioendothelioma 248, 306 
hemangioma 306, 30S 

- arterial embolization 5 

- complete resolution 4 

- complications 4 
-CT 5 

-MR 5 

- proliferating hemangioma ."■ 

- strawberry lesion 4 

- superficial 4 

- surgical removal 5 
hemangiopericytoma 248 
hematologic purpura 211 
hematoma 111 
hematuria 202 
hemorrhagic shock 35 

- severity class 39 
hepatectomy 170 
hepatic 103 

- - iatrogenic embolization Si 

- artery aneuryms 

- common hepatic artery 105 
--multiple 105 

- solitary 105 

- artery embolization 48, 50, 52 

- biological tolerance 169 

- encephalopathy 1 80 

- epithelioid hemangioendothelioma 308 

- hemangioendothelioma 7 

--CT 49 

- - hepatic injury severity scale 49 

- pseudoaneurysm 71 

- tumorigenesis 22 1 

hepatico-enteric arterial communication 15 
hepatitis 129 

-B 129 

hepatoblastoma 31 8 

hepatocellular carcinoma (HCC) 129, 141 

hepatocyie 

- genome 298 

- growth factor (HGF) 164,223 
hepatojejunostomy 131 

hep a to trophic 
-content 164 
-factor 163 
hereciian" 

- spherocytosis 211 
-syndrome 201 

hereditary hemorrhagic telangiectasia 8, 11 
hexokinase 11 

- expression 222 

- specific inhibitor 111 

HGK ,; ;r hepaiocyte growth facior 
5-fflAA 225 
histoacryl 29,167,305 
holmium 166(-166-Ho] 227 
hormonal syndrome 177 
human yene therapy 29? 
hyaluronate 172 



hydrocoil 244 
hypercalcemia 202 
Hypersplenism 211 
hypertension 190,210 
hypertrophy 163 
hyper vascular tumor 190 
hyper vascularity 149 
hypocalcaemia 190 
hypophyseal artery 238 
hypotension 202 
hypo-vascular tumor 190 
hypoxia 223 



iatrogenic 34 

- injury 79,101 

- arterial puncture 91 

- arterial rupture 91 

- carotid arteries 91 

- femoral artery 69 

- gastrointestinal bleeding 83 
--hepatic 88-90 

- - hepatic veins 86 

- - intrahepatic pseudoaneurysm SO 
--renal 85,90,93 

-- renal PTA 91 

- renal transplant 90 

- subclavian 89 

- - subclavian arteries 91 

- pseudoaneurysm 100 
idiopathic 

- epistaxis 262 

- thrombocytopenic purpura 317 
iliolumbar artery 63 

!MA, see inferior mesenteric arterv 
immunologic response 239 
immunotherapy 201 
incidental aneurysm 103 
mcependent indicator 224 
indium 111 pentetreotide 179 
indocyanine gteen retention rale 172 
indolent cyst 147 
infarction of the bowel 111 

- capsular artery 203 
-gluteal 63 

- artery 191 

- mesenteric artery (IMA) 103 
-polar artery 212 

- tympanic branch 240 
inflow artery 10 

inhibitor of growth -factor-signaling 222 

in-iine stabilization 37 

insulin 164 

intercellular disjunction 164 

intercostal artery 132 

interferon 1/^.189,308 

interlcukin 2 189 

internal 

- carotid artery 235,238,249,273 

- iliac anery 62, <i4, 65 



Subject Index 



- maxillary artery 235 

- maxillary system 237 

- pudendal 63 
intestinal angina 110 
intraarterial brachy therapy 216 
intrahepatic rupture 106 

:nt i a ope: alive blooc loss 1 89, 2 1 ' 11 ' 1 
:■ 'CizeJ ethyl ester 242 

- i r inotec an -elu ting bead 228 

irreversible failure ] 55 
ischemia 111 
ischemic neuropathy 



167 



.v.i. on 205 



J 



n-line stabilization 
juvenile 
-angiofibroma 247,264,312 

- nasopharyngeal angiofibrom. 



kaposiform hemangioendothelioma 5, 6 
Xasabach-Merritt phen 
Ki-67 labeling index 
kidney 

- fracture 53 

- renal embolization 

- renal injury 

- embolization 51 
Klippel-Trenaunay s; 



landingzone 108 
laser 29 
lateral 

- circumflex femoial ai 

- sacral 63 

LDL receptor 296 
'.t'A gastroepiploic arrei 
leucovorin 158 
leukemia 211 
lidocaine 181 
ligation 62 



lipiodol 130,180,222,299 
- ]3l lipiodol 224 
liver 106,149 

- abscess 185 

- embolization 50 

- trophicity 164 

longitudinal neural artery 235 
lower- extremity chronic venous i 
-GSV 119 
- sphenous space 120 



-SSV 120 

- superficial wnous insufficiency 
LSF, see lung shunt fraction 
lung 149 
-shunt fraction (LSF) 152 

lymph j iic ma lie-: mat ion 12, 14, 15 
lymphoma 211 



M 

"TmMAA 143 
-lung shunting 151 
^Tc-MAA 150 
macro-aggregated albumin 143 
macrophage cell 170 
Maffucci's syndrome 22 
magic wallstent 285 
malignant hypertension 113 
Mai Ian'; syndrome 101 
mechanical skeletal failure 190 

- circumflex femoral artery 191 

- emergency 35 
metaiodobenzylguanidine (MIBG 

microcirculation 150 
microcoil 49 
microph.ebeciomy 1 21 
middle capsular artery 203 
mitomycin 222 

mucocutaneous familial venous n 
multiple renal artery 203 
myec-iic aneurysm 105 



N 

nanotechnology 228 

nasc'palatine artery 259 
nasopharyngeal angiofibroma 312 

NBCA, see n-butyl cyanoacrylate 

n-butyl cyanoacrylate (NBCA) 87, 93, 242, 251 

- N-butyl-2- cyanoacrylate 225,242, 305, 309 
neomtimal hyperplastic restenosis 290 
nephron-sparing procedure 113,201 

-block 305 

- injury 66 

neural compression riemaloma I [4 
neuroblastoma 248 
neurocristopathic tumor 251 
neuroendocrine tumor 177 

- complication 184 

- e\ciusion criteria 1 SO 

- hepatic aioery emooiotherapy 1 79 

- inclusion criteria 180 
-pitfalls 185 
-results 184 
-survival 184 
-tools 181 

- treatment algorithm 179 
neuroform 285 



neuroleptic analgesia 2-10 

neuropathy 16,70 

neuropeptide 178 

neutropenia 185 

nidus 16 

nitroglycerin 245,305 

nitropaste 245 

nonchromaffin paiaganglio 

nontarget 

- embolization 114,144,11: 

-radiation 155 



obstructive jaundice 166 
obturator 63 
occipital artery 239,243 
occlusion balloon 205 
-catheter 204 
ontogenesis 235 

ophthalmic artery 237,259 

-ablation 201 

- injury 306 

oropharyngeal obstruction 37 
Osier- Weber- Rendu syndrome a 
outflowvein 10 



packing 255 

palliative cnrnsiization 209 
piir.i-cniii ivc* if. I tumor 177 
pancreatitis 100, 111,114 
papaverine 101i 

..'jjlJiifclii'md 248,251 
paraneoplastic •.■yr-iptom 202 
■;ar<ithyri"d adenoma 254 

paresthesia 124 

partial 

- hepatic resection 163 
-splenectomy 211 

- splenic embolization 212 
patient work up 101 
patient-controlled -anesthesia fPCA) 16 
PGA, see patient-controlled -anesthesia 
PGoA, see posterior communicating artery 
pediatric intervention 303 

- arteriogram 63 
-fracture 63,65,306 

- - algorithm tor t jj e management 62 

- classification 61 

- - external fixation 62 

- - Kane's classification 61 

- laparotomy 62 

- - mechanism of injury 59 

- - radiographs 61 

- sources of bleeding 60 

- trauma 

- hemorrhage 59 

- - mortality 59 



- -shock 60 

— surgery 60 

— therapy 63 

-biliary drainage 80,81 

- venography 26 

.•erfoiaimg artery 203 
perfusionMRI 137 
ye miliary fibrosis 170 
periodic EPX 266 

peri procedural antibiotic 



103 



peroneal 71 

pharyngo-cutaneous fistula 275 

piperacillin 185 

platinum fiber microcoil 309 

pledget 223 

polidocanol 28 

polyfbenzy! 1-glutamate) (PBLG) microsphere 22 

polyacrylonitrile (PAN) hydrogel 227 

polycythemia 202 

polyethylene 241 

poly-L- lactic acid (PLLA) microsphere 227 

polymerization 1 67 

polyvinyl alcohol (PVA] 49,86,223,225,226,251 

- hydrogel drug-elutingbead 228 
-particle 192,214,242,305 

- hydrogel 227 

- particle 6 

-hypertension 211 

- pressure 167 

- vein embolization I 63 

- embolic agenls i *<9 

- - growth rate of liver 1 70 

- - long-term survival 173 

- surgical transileocolic approach 168 

- vein patency 106 

-vein thrombosis (PVT) 132 

per re -anastomosis 1 69 

post catheterization j3 

postembolization syndrome 103, 145, 196,210 

- communicating a: ;ery (PCoA) 276 
-EPX 258 

- lateral nasal 259 

- medial nasal artery 259 
postoperative 

- bleeding 88 

- liver failure 172 
posttraumatic AVF 12 
precise stent 283 
primitive maxillary artery 237 
profunda femoris artery 191 
picgnoslic factor 177 
prominent ethmoidal artery 263 
prophylactic 

- embolization 



202 

Vioie'.is syndrome 3b.' 
protracted hemorrhage 272 
pseudoaneurysm 69,70,80 
-of the common carotid artery 276 



"ir: ygcvaginal artery 239 


- - organ injury score 54 




pulling sensation 125 


- vascular injury 52 




pulmonary 


renal 




- AVM year 8, 1 1 


Rendu-Osler- Weber syndrome 266,310,313,31 




- Upiodol embolism 31 8 


Renegade 145 




- lyiriph-.iiigio.eioiTivomaiosis 202 


ien:n-.i:'giotensin 190 




PVA. see polyvinyl alcohol 


replaced 




PVT, see portal vein thrombosis 


- leii hepatic artery ; 44 




pyrexia Is] 


- right hepatic artery 144, 185 
resin-based microsphere 141 
respiratory obstruction grade 36 




R 


resuscitation 39,40 




radiation 


reticuloendothelial hyper plasm 21 1 




- dosimetry 145 


retroperitoneal bleeding 53, 69 




-hepatitis 159 


RF ablation 124 




- pneumonitis 142, 150, 155 


lii.ibdomvos.iicoma 249 




- therapy 250 


right 




:adical nephrectomy 201 


- gastric artery 144 




:ailioen:oojzauon ! 39 


- hepatic artery 144 




- calculation of lung dose 155 


RNA 295 




-complications 146,159 


road mapping 191 




- discharge instructions 146 


rtPA 317 




- duodenal ulcers 157 






- follow up 147 


S 




- gastric ulcers 157 


saccular 




-HCC 141 


- microaneurysm 




- insoluble sl.-ss microspheres 153 


- - amphetamine abuse 101 




- liver metastases 149 


- - polyarteritis nodosa 101 




- materials 146 


- pseudo.tneurysm 100 




resin based microspheres 154 


sacral fracture 63 




results 1 [16 


sandwich technique 167 




raci:if:t\|ui.ncy ablation 140, 170,201 


saphenous 






-nerve 119 




radiotherapy 252 


-vein reflux 119 




radius 285 


sarcoma 139 




rCBS, sec ■>.', urrent carotid blowout syntonic 


sciatic nerve 193 




RCCmetastase 190 


sclerosing agenl 28 




recanalization 106 


sclerotherapy 27,30,316 




- ofpseudoaneurysm 106 


seizure 114 




recombinant 


selective internal radiation (SIR) 141 




- adenoviral vector 223 


-sphere 141,149,154 




- P-galactosidase 223 


self- expanding stent 281,283 




recurrent carotid blowout syndrome (rCBS) 275 


sepsis AS 




refractory hypertension 314 


serotonin 178 




renal 


- serotonin- sec re ting tumor 1 77 




- abscess 210 


sev.ia! dysfunction 66 




- artery 103 


shock 35,60 




- artery aneurysm 


silicone detachable 278 




- - iingiomyolipoma 112 


single photon emission ccnip.i'.ed :onioguphy 


150 


— balloon occlusion catheter 113 


SIR, see selective internal radiation 




— classification 112 


skeletonization 172 




— iatrogenic 112 


skin 






-burn 124 




— natural history 1 12 


- ulceration 16 




— neurofibromatosis 112 


SMA 103 




- - posl-embolization syndrome 1 1 3 


small saphenous vein 119 




— surgical repair 113 


smart stent 283 




— traumatic 112 


sodium tetradecyl sulfate 316 




- arte j y embolization 51, 52, 207, 216 


solid hepatic tumor 129 




- cell carcinoma 201 ,207 


solitary lesion 190 




--bone metastases 189 


somatostatin 




-failure 113 


- analogue 178 




- trauma 


-receptor 178 





sotradecol 28 



learlery 239,259 
sphere 226 
spherical PVA 227 
sphincter of Oddi 131 
sphincterotomy 131, 185 
spidervein 121 
splanchnic, aneurysm 109 
spleen 46,49 
splenic 103 

- angiogram 46 
-artery 212,214 

- artery aneurysm 

-- calcified masses 107 

- -rupture 107 

- .'.rtery embolization 43,45,47,48,21 
-infarction 48,109,185 

--scale 45 

- trauma 

- -AV fistula 47,48 
GT grades 47 

- -CT-based classification 44 

- pseudoaneurysm 45,47 

- splenic injury scale 45 

- pathologic fracture 197 

- retroperitoneal hem orr luge 202 
stem cell 298 

- angioplasty 279 

- fracture 71 

-graft 71,88,89,104,113,114,285 

strawberry hemangioma 4 

streptozocin 179 

St'ji'ge- Weber syndrome 312 

subclavian artery injury 89 

sunburst 203 

superficial temporal artery 235, 243 

superior 

- capsular artery 203 

- gluteal 63 

- gluteal artery 191 

- mesenteric artery aneurysm 1 0° 
-polar artery 212 

surgical metastaseclomy 178 
sf.i viva! benefit 139 
synthetic materia! 25.- 
systemic steroid tr 



TAC, see transarlei lal chemotherapy 

TACE,see transarteriul chemo embolization 

TAE, see transarterial embolization 

tamponade 114 

tazobaclam 185 

tehiiigieciasia 121,267 

TGF-a, see transforming growth factor-alpha 

thalassemia 317 

-major 211 

TheraSphere 141,149,154 



topography 252 
torpedo 49 
total splenectomy 



- -dual phase 130 

- - follow up 137 

- - procedure 132 

- -results 138 

- chemotherapy (TAC) 139 

- embolization (TAE) 139 

transcatheter aneria! chemoemLiojzaiion 221 

transforming growth factor-alpha (TGF-a) i 64 

transient ischemia 2PB 

transitory carotid occlusion 251 

transplant kidney 90 

trauma 35,101,306 

-blunt 48 

- iatrogenic 48 

- kidneys 52 
-liver 48 

- pelvic 59 

- solid organ 43 
-splenic 43 

- visceral 43 
pistaxis 264 



- of saccular aneurysm 106 
-ofSVI 120 

Trendelenburg position 123 
tris-aciyi gelatin microsphere 
truncalvein 120 
tuberotis sclerosis 202 
tufted angioma 308 
tumescent anesthesia 122 

- antigen 209 



- growth 165 

- lysis syndrome 135 

- marker 142 

- tumor-relaled epistaxis 264 

- vessel 203 
-viability 137 
-volume 165 

U 

uncovered stent 87 
urate-induced renal failure 180 



V'AA, ice vi~jcr.iL arieri: 
v;'.lri.i';if insufficiency 
varicocele 316 



121 



:ular 



- anomaly 21 

- endothelial growth factor ( VEGF] 222, 223 

- injury <>9 

- malformation 3, 83, 306 

- supply to the kidney 113 
-tumor 30fi 

VBCtH.scc sasculai endothelial growth factor 
venous malformation 3, 12,23 

- arteriography 2-1 

- classification 21 

- coagulopahies 22 

- computed i:im: g-.iphy 24 
-diffuse 22 

- direct percutaneous venography 25 

- endovascular approach 30 
-focal 22 

- genetics 21 

- magne:ic resonance .mag.ng 24 

- management 26 
-MRI 25,27 

- percutaneous approach 30 

- peripheral venography l:< 

- predominantly venous malformation 2! 

- sclerotherapy 26 

- ultrasound 23 



vei'.n;":. 
-aorta 236 

- ophthalmic artery 235 

- pharv.ige.U artery 236 
vidian artery 239 
viral hepatitis 172 
visceral arteni'.l a usury flu i 

- incidence 99 



vitamin K 191 
v.in llippel : mj,!ii syndror 
v:in Kcck!mt>"~itt:M':'.'s disea: 
von Willehrand disease 26i 
walljiraf: 2S^,2K9 
wallstenl 2S3 
Wallmar: l<i:ip 104 
W:: pp tprnt'Lurt: 88 
vvhoiied appearance 203 



yttrium-90 (-90-Y] ] 
- glass microsphere 1 
-microsphere 149 



List of Contributors 
Volume 2 



KamhanAhhar.MD 

Section of Interventional R;t dun logy 

Division of Diagnostic Imaging 

The Univerfity or" Tex a s 

MD Anderson Cancer Center 

1515 Hokombe Boulevard, Unit 325 

Houston, TX 77030-4009 

USA 



Hortensia Alvarez, MD 

Service de Neuroradiologie 

et Therapeutique 

Hopital Bicetre 

78 rue due General Leclerc 

94275 Le Kremlin Bicltre 

France 



John C. Chaloupka, MD, FAHA, FACA 
Director of Interventional Neuroradiology 
Professor of Radiology and Neurosurgery 
University of Iowa Hospitals and Clinics 
University of Iowa Carver College of Medicii 
200 Hawkins Dr, 3893 JPP 
Iowa City, IA 52242 
USA 



Michael D. Darcy.MD 

Professor of Radiologv .-no Surgery 
Division or Ui.ignosiic Radiology 
Chief, Intervention ill Radiology Section 
Washington University School of Medicin 
Miillinckrodt Institute of R.idiologv 
:" 10 South Kingshighway, 6th Floor 
St.Louis,MO 63110 
USA 



Josee Dubois, MD 

Professor of Radiologv 
Pediatric and Interventional ] 
Department of Medical Imag 
Hopital Ste-Justine 
3175 C6te Sle-Catherine Roat 
Montreal, Quebec H3T 1C5 



James R. Duncan, MD, PhD 

Assist.;]")! Professor or Radio-ley and Surgery 
Mallinclcrodt Institute or' Radiology and 
VV.isoington University School of Medicuice 
510 S. Kingshighway Blvd 
St.Louis,MO 63110 
USA 

Dominique Elias.MD 

Head of Pige stive Surgery Section 
Institut Gustave Rousssv 
39, Rue Camille Desmoulins 
94800 VillejuifCedex 
France 

Laurent Gakh. M ', '■ 

Professor or' Radiology 

Pediatric and Interventional Radiologist 

f'eur-.rtmenr of Medical k'rusiiiH 

Hopital Ste-Justine 

3175 Cflte Ste-Catherine Road 

Montreal, Quebec H3T 1C5 

Canada 

Christos S. Georgiades, MD. PhD 

Assistant Professor or' Radiology and Surge: 1 ;" 
Johns Hopkins Medical Institutions 
Blalock 545, 600 North Wolfe Street, 
Baltimore, MD 21287 
USA 

Jean-Francois H. Geschwind, MD 

Associate Professor of Radiologv. surgery and ( 
Division of Vascular and 
The Russell H. Morgan Dep; 



and Radiological Sci 

Johns Hookins \' ed io 

Blalock 545, 600 North Wolfe Street 

Baltimore, MD 21287 

USA 

Craig B. Glaiberman, MD 

instructor, Radiology 

Division of Interventional Radiology 

Washington University School of Medicine 

Malhnckrodt Institute or' Radiology 

510 South Km.gshighvvav. :-th Floor 

St.Louis,MO 63110 

USA 



of Contributors 



,N,MD 
Professor of Radielogv 
Director, Vascular and Interventional Radio 
University of Iowa 
Department of Radiology 
200 Hawkins Drive, 3957 JPP 
Iowa City, IA 52242 
USA 

Minako Hayakawa, MI 1 

Visiting Assistant Professor of Radiology 
University of Iowa Hospitals and Clinics 
University o:' low.; Carver College of Med:; 
200 Hawkins Dr, 3893 JPP 
Iowa City, IA 52242 
USA 



.i Hf 



,MD 



Department of Radiology 
University Hospitals Gasthuisberg 
Herestraat 49 
3000 Leuven 
He.g.um 

Shih-Wei Hsu, MD 

Visiting Scholar, University o'i low.! Hospita 

and Clinics 

University of low.; Carver College of Medic 

200 Hawkins Dr, 3893 JPP 

Iowa City, IA 52242 

USA 

Assistant Professor, 1 iepartiuent .:■:' Piagnost 

Radiology 

Chang Gung Memorial Hospital 

K;l' ■l"i- : i.;:ic 

Taiwan 



Matthew S. Johnson, MD 

Associate Professor .:■:' Radiology 

I :■ ject-.j- 1 . Section of Interventional Raciioiogv 

Indiana University Hospital, UH0279 

Department of Radiology 

550 Umversiiy Boulevard 

Indianapolis, IN 46202-5253 

USA 

John R. Kachura, MD, FRCPC 

Division of Vascular and Interventional Radio 

I 'epartiiien: oi Medical Imaging 

Toronto General Hospital 

200 Elizabeth Street, Eaton South l-454d 

Toronto, ON M5G 2C4 

C a :i a d a 

NeilM.Khilsani.MD 
Cornell Vascular 

Weill Me.aical Co Lege of Corned Universitv 
416 East 55th Street 
New York, NY 10022 

USA 



Paula Klurfan.MD 

interventional Neuroradiely Clinical Fellow 
1 Vpar;:i-:ii ::' Medical I maging 
University of Toronto 
Toronto Western Hospital 
399 Bat hurst Street 
Toronto, Ontario M5T 2S8 



Pierre Lasjaunias.MD, PhD 

Service de Neuroradiology 1 'nagn.ostique et Therapet 

Hopital Bicetre 

78 rue due Genera! Leclerc 

94275 Le Kremlin Bicetre 

SeonKyu Lee, MD, PhD 

Assistanl Professor and Staff Neuroradiologist 
1 Vparnieni :■:' Medical I maging 
University of Toronto 
Toronto Western Hospital 
#399 Bathurst Street 
Toronto, Ontario M5T 2S8 



Walter S. Lesley, MD 

Chief, Section of Surgical Neuroradiology 

Assistani Profess;:'!" .:■:' Radiology 

The Texas A&M University Health Science Center 

USA 



Eleni Liapi, MD 

The Russell H. Morgan 
and Radiological Scene 
lohns Hopkins Medical 
Baltimore, MD 
USA 



David C. Madoff, MD 

Section of Interventional Radiology 

Division of Diagnostic Imaging 

The University of Texas 

MD Anderson Cancer Center 

1515 Holcombe Boulevard, Unit 325 

Houston, TX 77030-4009 

USA 

Geert Maleux, MD 

Department of Radiology 
University Hospitals Gasthtusberg 



Francis Marshalleck, MD 

Assist.i ni Profess;:'!" of Radiology 

Indiana University School of Median 

Indiana University Hospital 

Room 0279, 550 North University Bot 

Indianapolis, IN 26202 

USA 



parrment of liadiolgv 



List of Contribute 



Robert J. Min.MD 

Cornell Vascular 

Weil] Medical College of Cornell University 

416 East 55th Street 

New York, NY 10022 

USA 

Anne C. Roberts, HD 

University of California. 5 an 1 ! iego Medical Center 
Division of Vascular and Interventional Radiology 
200 West Arbor Drive 
San Diego, CA 92103-8756 
USA 

AlainJ.Roche.MD 

Head of Interventional Radiology Section 

Professor, Institut Gustave Rousssy 
39, Rue Camille Desmoulins 
94800 Villejuif Cede* 

Georges Rodesch, MD 

Service de Neuroradioloie I 'iagiios:L]ue et Ta.eraoeuiique 

Hopit.il Foch 

40 rue Worth 

B.P. 36 

92150 Suresnes 

France 

Riad Salem, MD, MBA 

Assistant Profess;;!" .:■!' Radiology and Oncology 

Northwestern Memor.r.l Hospital 

Department of Radiology 

676 North St. Claire, Suite 800 

Chicago, IL 60611 

USA 

Melhem J. Sharafuddin, MD 

University of Iowa Hospitals and Clinics 

Department of Radiology 

200 Hawkins Dr, 3957 JPP 

Iowa City, IA 52242 

USA 

Gary Siskin, MD 

Albnnv Medical College 
Vascular Radio iogv. Alia 
47 New Scotland Avenue 
Albany, NY 12208-3479 
USA 



gSun.MD 
Associate Professor of Radiology 
Department of Ri-diology 
University of Iowa, College of Medicine 
200 Hawkins Dr., 3955 JPP 
Iowa City, IA 52242 
USA 

Kong Teng Tan, MB, BCh, FRCS, FRCR 
Division of Vascular and Interventional R 

r'ear.rtmen: ot Medioal i:':aaiiia 

Toronto General Hospital 

585 University Avenue, NCSB 1C-563 

Toronto, ON M5G 2N2 

Canada 

Maria Thijs,MD 
Department of Ri-diology 
Univers.itv Hospital:". Gasthaisberg 
Herestraat 49 

3000 Leuven 



17 Bramble Lane 
West Grove, PA 19390 

USA 

Rajiv Verma, MD 

Section of Interventional Radiology 

Division of Diagnostic Imaging 

The University of Texr.s 

MD Anderson Cancer Center 

15 15 Holcombe Boulevard, Unit 325 

Houston, TX 77030-4009 

USA 

Lucy A. Wibbenmeyer, MD 

University of Iowa Hospitals and Clinics 

Department of Ri-diology 

200 Hawkins Dr. 

Iowa City, IA 52242 

USA 

Jeffrey J. Wong, MB ChB, BMedSc 

University of California, ion Pi ego Medical Center 

Division of Vascular and Interventional Radiology 

200 West Arbor Drive 

San Diego, CA 92103-8756 

USA 



Contents - Volume 1 



General Principles 



Em bolo therapy: Basic Principles and Applic 
Melhem J. Sharafuddin.Shiliang Sun, and Jaf/ 

Embolization Tools 

Jafar Golzarian, Gary P. Siskin, Melhem ]. Sha 

Hidefumi Mimura, and Douglas M. Cols 



Controlled Delivery of Pushable Fibered Coils for Large Vessel Embolotherapy 
Robert I.White Jr. and Jeffrey S. Pollak 35 



Work-up and Follow-up after Embolization 
Jim A. Reekers 



5 Upper GI Bleeding 

Luc Defreyne 49 

6 Embolization for Lower GI Bleeding 

Michael Darcy 73 

7 Haemobilia and Bleeding Complications in Pancreatitis 

Tony A.Nicholson 87 

8 Balloon-occluded Retrograde Transvenous Obliteration ol Gastric Varices 
in Portal Hypertension 

Koji Takahashi and Shiliang Sun 99 

Gynecology and Obstetrics 105 

9 Interventional Management of Postpartum Hemorrhage 

Hicham T.Abada, Jafar Golzarian, and Shilliang Sun 107 

10 Fibroids 

Gary P. Siskin, Jeffrey J.Wong, Anne C. Roberts, Jean Pierre Pelage, 

Arnaud Fauconnier, Pascal Lacombe, Alexandre Laurent, and 

Jafar Golzarian 119 

10.1 Uterine Fibroid Embolization: Practice Development 

Gary P. Siskin 119 

10.2 Pre-op Work-Up and Post-op Care of Uterine Fibroid Embolization 

Jeffrey J.Wong and Anne C.Roberts 125 

10.3 Fibroid Embolization: Anatomv and Technical Considerations 

Anne C.Roberts 141 



336 Contents -Volume 1 

10.4 Results and Complications 

Jean Pierre Pelage,Arnaud Fauconnier, and Pascal Lacombe 157 

10.5 How to Minimize Failure after UFE 

Jafar Golzarian and Jean Pierre Pelage 177 

10.6 Perspectives 

Alexandre Laurent, Jean Pierre Pelage and Jafar Golzarian 187 

1 1 Pelvic Congestion Syndrome 

Lindsay Machan 199 



Genitourinary 213 

12 Varicocele Embolization 

David Hunter and Galia T.Rosen 215 



13 Embolization Therapy for High-Flow Priapis. 
Jim A. Reekers 



Aortic-Iliac 233 

14 Endoleak: Definition, Diagnosis, and Management 

David Valenti and Jafar Golzarian 235 

15 Internal Iliac Artery Embolization in the Stent-Graft Treatment of 
AortoLliac Aneurysms 

Mahmood K.Razavi 253 



Respiratory Syster 



16 Bronchial Artery Embolizatio 
Jos C. van den Berg 



17 Pulmonary Arteriovenous Malformations 

Jean Pierre Pelage, Pascal Lacombe, Robert I. White, Jr., 

and JeffrayS.Pollak 279 



Subject Index 297 

List of Contributors 303 

Contents and List of Contributors of Volume 2 305 



List of Contributors -Volume 1 



Hicham T.Abada.MD 

! Vp.i rl::~.c-jj! a: Imaging and Interve:i1ic-jial Rac. 
Centre Hospitalier Rene Dubos 
6, Avenue de L'lle-de-France 
95303 Cergy Pontoise Cedex 

France 

Douglas M. Coldwell, MD 

Professor of Rao:.\.:gv 

University of Texas Southwestern Medical Ce 

5323 Harry Hines Blvd. 

Dallas, TX 75390-8834 

USA 

Michael D.Darcy.MD 

Professor of Radiologv and Surgery 
\iallinckrodt Institute of Radiologv 
Washington University School of Medicine 
5 10 South Kingsbighway Boulevard 
SaintLouis,MO 63110-1076 
USA 



LucDefreyne.MD 
Department of Vascular and 
Interventional Radiology 
Ghent University Hospital 
De Pintelaan 185 
9000 Ghent 
oe^giu:;". 

Arnaud Fauconnier, MD, PhD 

Department of Obstetrics and Gynecologv, 
Centre Hospitalier de Poissy, 
10, rue du Champ Gaillard, 
78300 Poissy Cedex, 

Iafar Golzarian,MD 

Professor of Radiologv 

Director, Vascular and Interventional Radiol 

University of Iowa 

Department of Radiology 

200 Hawkins Drive, 3957 JPP 

Iowa City, IA 52242 

USA 

David W. Hunter, MD 

Department of Radiology 

"2-447 Fairvjew-Lniversity Medical Center 

University of Minnesota 

500 Harvard Street S.E. 

Minneapolis, MN 55455 

USA 



Pascal Lacombe, MD 

Department of Radiologv H ■ : y i t n I Amhroise : 
9, Avenue Charles De Gaulle 
92104 Boulogne Cedex 
France 

Alexandre Laurent, MD, PhD 

Associate Professor 

Center for Research in 

Interventional Imaging (Cr2i APHP-INRA) 

Jouy en Josas, 78352 

France 

Lindsay Machan, MD 

Department of Radiology 

University of British Columbia Hospital 

2211 Wesbrook. Mall 

Vancouver, BC V6T 2B5 



HlDEFUMl Ml.MURA, MD 

Associate Professor of Radiologv 

University of Iowa Hospitals and Clinics 

Department of Radiology 

200 Hawkins Dr, 3957 JPP 

Iowa City, IA 52242 

USA 

Tony A. Nicholson, BScM, Sc, MB, ChB, FRCR 
Consultant Vascular Radiologist & Senior Lecture 

Leeds Teaching Hospitals NHS Trust 
Great George Street 
Leeds, LS13EX 

UK 

[ean-Pierre Pelage, MD, PhD 

Department of Radiology 

HopitalAmbroisePare 

9, Avenue Charles De Gaulle 

92104 Boulogne Cedex 

France 

TeffreyS.Pollak.MD 

Yale University School of Medicine 

Department Diagnostic Radiology 

PO Box 20842 

New Haven, CT 06504-8042 

USA 



Mai 



d K. Ra: 



ri, MD 



Center tor Research and Clinical 
St. Joseph Vascular Institute 



LJ := ; of Contributors -Volume 1 



Jim A. Reekehs, MD, PhD 
Department of Radiology, G 1-207 

Academic Medical Center 
University or' Amsterdam 
Meibergd reef i_; 
AZ 1105 Amsterdam 
The Netherlands 

Anne C. Roberts, MD 

University of California, San Diego Medic; 
Division of Vascular and 
Intervention.;! Radiology 
200 West Arbor Drive 
San Diego, CA 92103-8756 



GaliaT. Rosen.MD 

Department of Radiology 

J2-447 Fair view-University Medical Center 

University of Minnesota 

500 Harvard Street S.E. 

Minneapolis, MN 55455 
USA 



Melhem J. Sharaflddin, MD 
Departments of Radiology and Surgery, 3JPP 
University of iowa Hospitals and Clinics 
200 Hawkins Drive 
Iowa City.IA 52242-1077 
USA 



Gary P. Siskin, MD 

Associate Professor of Radiology a: 

Obstetrics fx Gynecology 

Albany Medical College 

47 New Si". :~..,\ nci Avenue, MC- 1 i. ; 

Albany, NY 12208-3479 

USA 



Shiliang Sun.MD 

Associate Processor of Radi.ikgv 
University of Iowa Hospitals and Clii 
200 Hawkins Dr, 3955 JPP 
Iowa City, IA 52242 
USA 

KojiTakahashi.MD 

Department of Radiology 
Asahikawa Medical College 
2-1-1-1 Midorigaoka 
Asahikawa, 078-8510 



los C. VAN DEN Bebg, MD, PhD 

Head or' Service t-A inter vein ion.il Rao iology 
Ospedale Regionale di Lugano, sede Civico 
Via Tesserete 46 
6900 Lugano 
Switzerland 

David A. Valenti, MD 

Royal Victor;.; Hospital 

McGill University Health Centre 

McGill University 

687 Pine Avenue West, Suite A451 

Montreal, Quebec H3A 1AI 



Robert I. White, Jr., MD 

Yale University School of Medicine 

Department of Diagnostic Radiology 

333 Cedar Street, Room 5039 LMP 

New Haven, CT 06520 

USA 



Jeffrey J. Wong, MB ChB, BMedSc 

Senior House Officer 

Royal National Orthopaedic Hospital 



MEDICAL RADIOLOGY Diagnostic Imaging and Radiation Oncology 



Titles in the series ,i/rv<i,fi published 



Diagnostic Imaging 



Radiology of the Upper Urinary Tract 

Edited by E.K.Lang 

The Thymus -Diagnostic Imaging, 

functions, and Pathologic Anatomy 

Edited by E. Walter, E. Willich, 

andW.R.Webb 

Interventional Neuroradiology 

Edited by A. Va lava n is 

Radiology of the Pancreas 

EditedbyA.L.Baert, 

, _ o-ediied by G. Pelorme 

Radiology of the Lower Urinary Tract 

Edited by E.K. Lang 

Magnetic Resonance Angiography 

Edited by I, P.Arlart, G. M. Bongartz, 

andG.Marchal 

Contrast -Enhanced MRI of the Breast 

S. Heywang-Kobrunner and R. Beck 

SpiralCToftheChest 

Edited by M. Remy-Jardin and J. Remy 

Radiological Diagnosis of Breast Diseases 

Edited by M. Friedrich and E. A. Sickles 

Radiology of theTrauma 

Edited by M. Heller and A.Fink 

Biliary Tract Radiology 

Edited by P.Rossi, 

co-edited by M. Brezi 

Radiological Imaging of Sports Injuries 

Edited by C.Masciocchi 

Modern Imaging of the Alimentary Tube 

Edited bv A. R. Margulis 

Diagnosis and Therapy of Spinal Tumors 

Edited bvP.R.Algra, ]. Vaik, 

andJ.J.Heimans 

Interventional Magnetic 

Resonance Imaging 

Edited by J.F.Debatin and G.Adam 

Abdominal and Pelvic MRI 

Edited by A. Heuck and M. Reiser 

Orthopedic Imaging 

Techniques and Applications 

Edited by A.M. Davies 

and H. Pettersson 



Radiology of the Female Pelvic Organs 

Edited by E.K.Lang 
Magnetic Resonance of the Heart 
and Great Vessels 

Clii)k\il Applications 

Edited by J. Bogaert, A.J. Duerinckx, 

andF.E.Rademakers 

Modern Head and Neck Imaging 

Edited by S.K.Mukherii 

andJ.A.Castelijns 

Radiological Imaging 

of Endocrine Diseases 

Edited by J.N. Bruneton 

in collaboration with B. Padovani 

andM.-Y.Mourou 

Trends in Contrast Media 

Edited bv H. S. T horns e n. 

R.N.Muller.andR.F.Mattrey 

Functional MR] 

Edited by C. T. W. Moon en 

andP.A.Bandettini 

Radiology of the Pancreas 

2nd Revised Edition 

Edited by A. L. Baert. Co-edited by 

'.j. ', ! elorme ,u:d L.Van Hoe 

Emergency Pediatric Radiology 

Edited by H.Carty 

Spiral CT of the Abdomen 

Edit;," by r. Terrier, X. Crossholz. 

and C.D.Becker 

Liver Malignancies 
Diagnostic and 
Interventional Radiology 

Edited by C. Rai'talozzi 

Medical Imaging of the Spleen 

Edited by A.M. De Schepper 

and F.Vanhoenacker 

Radiology of Peripheral Vascular Diseases 

Edited by E.Zeitler 

Diagnostic Nuclear Medicine 

Edited oy C. ^ohiepers 



Portal Hypertension 

Diagnostic Imaging-Guided Therapy 

Edited bv P. Rossi 

Co-edited by P. Ricci and L. Broglia 



Recent Advances in 
Diagnostic Neuroradiology 
Edited by Ph. Demaerel 

Virtual Endoscopy 

and Related 3D Techniques 

Ed.:ed bv ?. Rog.^Kj, I. Terwisschj 
Van Scheltinga, and B.Hamm 

Multislice CT 

Edited by M. F. Reiser, M. Takahashi, 
M. Modic, and R. Bruening 

Pediatric Uroradiology 
Edited by R. Fotter 

Transfontanellar Doppler Imaging 
In Neonates 

A. Couture and C.Veyrac 
Radiology of AIDS 

A Prarlical Approach 

Edited by J.W.AJ. Readers 

and P.C.Goodman 

CT of the Peritoneum 

Arm.iiido Rossi and Giorgio Rossi 

Magnetic Resonance Angiography 

2nd Revised Edition 

Edited by I. P.Arlart, 

G.M. Bongratz, and G. Marchal 

Pediatric Chest Imaging 

Edited by Javier Lucaya 

and Janet L. Strife 

Applications of Sonography 

In Head and Neck Pathology 

Edited by J. N. Bruneton 

in collaboration with C. Raffaelh 

andO.Dassonville 

Imaging of the Larynx 
Edited by R.Hermans 

3D Image Processing 

Techniques and Clinical Applications 

Edited by D. Caramella 

and C. Bartolozzi 

Imaging of Orbital and 

Visual Pathway Pathology 

Edited by W. S. Miiller-Forell 

Pediatric ENT Radiology 

Edited by S.J.King 
and A. E.Boothroyd 

Radiological Imaging of the Small Intestine 
Edited by N. C. Gourtsoyiannis 



MEDICAL RADIOLOGY Diagnostic Imaging and Radiation Oncology 

Titles in the series already published 



Imaging of the Knee 
Techniques and Applications 

Edited by A.M. Davies 

andVN.Cassar-Pullicino 

Perinatal Imaging 

From Ultrasound (a MR Imaging 

Edited by Fred E.Avni 

Radiological Imaging of the Neonatal Chest 

Edited by V.Donoghue 

Diagnosticand Interventional 

Radiology in Liver Transplantation 

Edited by E. Biicheler, V. Nicolas, 

C.E.Broelsch.X-Rogiers, 

and G. Krupski 

Radiology of Osteoporosis 

Edited by S. Grampp 

Imaging Pelvic Floor Disorders 

Edited by C.I. Bartram 

and J. 0. L.DeLancey 

Associate Editors: S. Halligan, 

F.M. Kelvin, and J.Stoker 

Imaging of thePancreas 
Cystic and Rare Tumors 

Edited by C.Procacci 
and A.J. Megibow 

High Resolution Sonography 
of the Peripheral Nervous System 

Edited by S. Peer and G. Bodner 

Imaging of the Foot and Ankle 
Techniques and Applications 

Edited by A.M. Davies, 
R.W.Wh'itehouse, 
and J. P. K. Jenkins 

Radiology Imaging of the Ureter 

Edited by F. Joffre, Ph. Otal, 
and M. Soulie 

Imaging of the Shoulder 
Techniques and Applications 

Edited by A. M. Davies and J. Hodler 

Radiology of the Petrous Bone 

Edited by M. Lemmerting 

andS.S.Kollias 

Interventional Radiology in Cancer 

Edited by A.Adam, R. F. Dondelinger, 

andP.R.Mueller 

Duplex and Color Doppier Imaging 

ofthe Venous System 

Edited by G.H.Mostbeck 



Mu It i detector-Row CT of the Thorax 

EditedbyU.J.Schoepf 

Functional Imaging ofthe Chest 

Edited by H.-U.Kauczor 

Radiology of the Pharynx 

and the Esophagus 

Edited by O.Ekberg 

Radiological Imaging 

tn Hematological Malignancies 

Edited by A.Guermazi 

Imaging and Intervention in 

AbdomlnalTrauma 

Edited by R. F. Dondelinger 

MultisliceCT 

2nd Revised Edition 

Edited by M. F. Reiser, M. Takahashi, 

M.Modic, and C.R.Becker 

Intracranial Vascular Malformations 

and Aneurysms 

From Diagnostic Work-Up 

to Endovascular Therapy 

Edited by M.Forsting 

Radiology and Imaging ofthe Colon 

Edited by A.H. Chapman 



Dynamic Contrast -Enhanced Magnetic 
Resonance Imaging in Oncology 

Edited by A. Jackson, D. L. Buckley, 

and G.J. M.Parker 

Imaging in Treatment Planning 

forSinonasalDiseases 

Edited by R. Maroldi and P. Nicolai 

ClinicalCardlacMRI 

With Interactive CD-ROM 

Edited by J.Bogaert, 

S. Pym;"! j' ho wjki, ai:d A. M. Taylor 

Focal Liver Lesions 

Detection, Characterization, 

Ablation 

Edited by R. Lencioni, D. Cioni, 

andC.Bartolozzi 

Multidetector-RowCT Angiography 

Edited by C.Catalano 

andR.Passariello 

Paediatric Musculoskeletal Diseases 

With an Emphasis on Ultrasound 

Edited by D.Wilson 



Contrast Media in Ultrasonography 
Basic Principles and Clinical Applications 

Edited by Emilio Quaia 

MR Imaging in White Matter Diseases of the 

Brain and Spinal Cord 

Edited by M. Filippi, N. De Stefano, 

V. Dousset, and J. C. McGowan 

Diagnostic Nuclear Medicine 

2nd Revised Edition 

EJite." bv C. Sohieper:; 

Imaging ofthe Kidney Cancer 

Edited by A.Guermazi 

Magnetic Resonance Imaging in 

Ischemic Stroke 

Edited by R. von Kummer and T. Back 

Imaging of the Hip S Bony Pelvis 

Techniques and Applications 

Edited by A. M. Davies, K. J. Johnson, 

andR.W.Whitehouse 

Imaging of Occupational and 

Environmental Disorders ofthe Chest 

Edited by P.A.Gevenois and 

P.DeVuyst 

Contrast Media 

Safety Issues and ESL'R Guidelines 

Edited by H. S. Thorn sen 

Virtual Colonoscopy 

A l'i .uTiiiil Guide 

Edited by P. Lefere and 5. Gryspeerdi 

Vascular Embolotherapy 

A Comprehensive AppiuaJi 

Volume 1 

Edited bv i. Golzarian. Go -edited bv 

S.SunandMJ.Sharafuddin 

Vascular Embolotherapy 

A Cuinpn'liensive Approach 

Volume 2 

Edited bv i. Golzarian. Go -edited bv 

S.SunandMJ.Sharafuddin 

Head and Neck Cancer Imaging 

Edited by R. Hermans 

Vascular Interventional Radiology 

Current Evidence in End ova Millar 

Surgery 

Edited by H. G. Cowling 



4Q Springer 



MEDICAL RADIOLOGY Diagnostic Imaging and Radiation Oncology 

Titles in the series ahead) published 



Radiation Oncology 



Lung Cancer 

Edited by C.W.S. 

Innovations in Radiation Oncology 

Edited by H.R. Withers 
and L. J. Peters 

Radiation Therapy 

of Head and Meet Cancer 

Edited by G.E.Laramore 

Gastrointestinal Cancer- 

Radlatlon Therapy 

Edited by R.R. Dobelbower, Jr. 

Radiation Exposure 
and Occupational Risks 

Edited by E. Scherer, C. Streffer, 
and K.-R. Trott 

Radiation Therapy of Benign Diseases 

A Clinical Guide 

S.E. Order and S.S.Donaldson 

Interventional Radiation 

Therapy Techniques- Bra chytherapy 

Edited by R. Sauer 

Radlopathology of Organs andTissues 

Edited by E. Scherer, C. Streffer, 

and K.-R. Trott 

Concomitant Continuous Infusion 

Chemotherapy and Radiation 

Edited by M. Rotman 

and C. J. Rosenthal 

Intraoperative Radiotherapy - 

Clinical Experiences and Results 

Edited by R A. Calvo, M. Santos, 

and L.W.Brady 

Radiotherapy of Intraocular 

and Orbital Tumors 

Edited by W.E.Alberti and 

R.H.Sagermar, 

Interstitial and Intracavitary 

Thermoradiotherapy 

Edited by M. H. Seegenschmiedt 

and R. Sauer 

Hon-Disseminated Breast Cancer 

Controversial Issues in Management 

Edited byG. H.Fletcher and 

S.H, Levitt 



Current Topics in 

Clinical Radloblology of Tumors 

Edited by H.-P.Beck-Bornholdt 

Practical Approaches to 
Cancer Invasion and Metastases 
A Compendium of Radiation 
Oncologists' Responses to 40 Histories 

Edited by A. R.Kagan with the 

Assistance of R. J. Steckel 

Radiation Therapy in Pediatric Oncology 

EditedbyJ.R.Cassady 



Carcinoma of the Kidney andTestis, 
and Rare Urologic Malignancies 
Innovations in Management 
Edited by Z. Petrovich, L. Baert, 
and L.W.Brady 



the 



Late Sequelae In Oncology 

Edited by J.Dunst and R. Sauer 

Mediastinal Tumors. Update 1995 

Edited by D.E.Wood 
and G.R. Thomas, Jr. 

Thermoradiotherapy 

andThermochemotherapy 

Volume 1: 

Biology, Physiology, and Physics 

Volume 2: 

Clinical Applications 

HJili." :'v M.H. ?e egensihi'iieJi. 

P. Fessenden, and C.C.Vernon 

Carcinoma of the Prostate 
Innovations in Management 

Edited by Z. Petrovich, L. Baert, 
and L.W.Brady 

Radiation Oncology 
of Gynecological Cancers 

Edited by H.W.Vahrson 

Carcinoma of the Bladder 
Innovations in Management 



Blood Perfusion and 
Mlcroenvlronment of Human Tumors 
Implications for 
CI in ical Ra d i o o n cology 

Edited by M.Molls and P.Vaupel 

Radiation Therapy of Benign Diseases 
A Clinical Guide 

2nd Revised Edition 

S.E. Order and S. S. Donaldson 



Progress and Perspectives 
Treatment of Lung Cancer 
Edited by P. Van Houtte, 
J. Klastersky, and P. Rocmans 

Combined Modality Therapy of 
Central Nervous System Tumors 
Edited by Z. Petrovich, L. W. Brady, 
\!. L. Apuzzo, and M. Bamberg 

Age-Related Macular Degeneration 
Current Treatment Concepts 
Edited by W. A. Alberti, G. Richard, 
andR.H.Sagerman 

Radiotherapy of Intraocular 
andOrbitalTumors 
2nd Revised Edition 
Edited by R.H.Sagerman, 
andW.E.Alberti 

Modification of Radiation Response 
Cytokines, Growth Factors, 
and Other Biolgical Targets 
Edited by C.Nieder, L. Milas, 
andK.K.Ang 

Radiation Oncology for Cure and Palliation 

R.G.Parker,N.A.Janjan, 

andM.T.Selch 

Clinical Target Volumes In Conformal and 
Intensity Modulated Radiation Therapy 
A Clinical Guide to Cancer Treatment 
Edited by V. Gregoire, P. Scalliet, 
and K. K. Ang 

Advances in Radiation Oncology 
In Lung Cancer 

Edited by Branislav Jeremic 

Hew Technologies In Radiation Oncology 
Edited by W. Schlegel, T. Bortfeld, 
and A.-L.Grosu 



4Q Springer