You are on page 1of 15

STANDARDS OF PRACTICE

Radioembolization of Hepatic Malignancies:


Background, Quality Improvement
Guidelines, and Future Directions
Siddharth A. Padia, MD, Robert J. Lewandowski, MD, Guy E. Johnson, MD, Daniel Y. Sze, MD,
PhD, Thomas J. Ward, MD, Ron C. Gaba, MD, Mark O. Baerlocher, MD, Vanessa L. Gates, MS,
Ahsun Riaz, MD, Daniel B. Brown, MD, Nasir H. Siddiqi, MD, T. Gregory Walker, MD,
James E. Silberzweig, MD, Jason W. Mitchell, MD, MPH, MBA, Boris Nikolic, MD, MBA, and
Riad Salem, MD, MBA, for the Society of Interventional Radiology Standards of Practice Committee

ABBREVIATIONS

AFP = α-fetoprotein, ECOG = Eastern Cooperative Oncology Group, EASL = European Association for Study of the Liver, FDA =
Food and Drug Administration, FDG = fluorodeoxyglucose, HCC = hepatocellular carcinoma, MAA = macroaggregated albumin,
mRECIST = modified Response Evaluation Criteria In Solid Tumors, QI = quality improvement, RECIST = Response Evaluation
Criteria In Solid Tumors, REILD = radioembolization-induced liver disease, SPECT = single-photon emission computed
tomography, 3D = three-dimensional

PREAMBLE document describes the knowledge base, technical skills, and experience
The mission of the Society of Interventional Radiology (SIR) is to required to perform a specific procedure.
improve patient care through image-guided therapy. The Society was A QI Guideline is produced by the Standards of Practice
founded in 1973, and is recognized today as the primary specialty Committee. The membership of the SIR Standards of Practice
society for physicians who provide minimally invasive image-guided Committee represents experts in a broad spectrum of interventional
therapies. The Standards Division of SIR writes a number of different procedures from the private and academic sectors of medicine.
types of standards documents to reflect the current clinical paradigm Generally, Standards of Practice Committee members dedicate the
and evolution in the treatment of a specific disease state or a procedure. vast majority of their professional time to performing interventional
The Standards Division currently produces four different types of procedures; as such, they represent a valid broad expert constituency of
documents. A Position Statement document reflects the viewpoints of the subject matter under consideration for standards production.
SIR on a specific disease state or procedure. A Reporting Standards
document attempts to codify the key elements required for future
research on a specific procedure or disease process. A Quality DOCUMENT METHODOLOGY
Improvement (QI) Guideline attempts to provide clinical guidelines SIR produces its QI Guidelines documents by using the following
on the application of a specific procedure or treatment of a disease process. Topics of relevance and timeliness are conceptualized by the
process when a significant body of literature is available. A Credentialing Standards of Practice Committee members, Service Lines, SIR

From the Division of Interventional Radiology (S.A.P.), Department of Radi- SIR, 3975 Fair Ridge Dr., Suite 400 N., Fairfax, VA 22033; E-mail: spadia@
ology, David Geffen School of Medicine at University of California, Los gmail.com
Angeles, Los Angeles, California; Division of Interventional Radiology (D.Y.S.),
Stanford University School of Medicine, Stanford, California; Section of Inter- S.A.P. is a paid consultant for and serves on the advisory board of BTG
ventional Radiology, Department of Radiology (R.J.L., V.L.G., A.R., R.S.), International (Ottawa, Ontario, Canada) and Guerbet (Roissy, France). R.J.L.
Robert H. Lurie Comprehensive Cancer Center, Northwestern University, serves on the advisory boards for BTG International and Boston Scientific
Chicago, Illinois; Department of Radiology, Division of Interventional Radiology (Marlborough, Massachusetts) and is a paid consultant for Cook (Blooming-
(R.C.G.), University of Illinois Hospital and Health Sciences System, Chicago, ton, Indiana). D.Y.S. receives personal fees from BTG International, Sirtex
Illinois; Section of Interventional Radiology, Department of Radiology (G.E.J.), (North Sydney, Australia), Boston Scientific, EmbolX (Los Altos, California),
University of Washington, Seattle, Washington; Section of Interventional Cook, Covidien (Dublin, Ireland), Amgen (Thousand Oaks, California), and
Radiology, Department of Radiology (T.J.W.), Florida Hospital, Orlando, Codman (Raynham, Massachusetts). M.O.B. is a paid consultant for Boston
Florida; Department of Radiology, Vanderbilt University School of Medicine Scientific. D.B.B. is a paid consultant for Cook, Vascular Solutions
(D.B.B.), Nashville, Tennessee; Division of Interventional Radiology (T.G.W.), (Minneapolis, Minnesota), and Onyx Pharmaceuticals (South San Francisco,
Massachusetts General Hospital, Boston, Massachusetts; Interventional California). R.S. receives grants and personal fees from BTG International and
Radiology, P.C. (J.E.S.), New York, New York; Department of Radiology personal fees from Boston Scientific, Merit Medical (South Jordan, Utah),
(B.N.), Stratton Medical Center, Albany, New York; Department of Diagnostic Terumo (Tokyo, Japan), Siemens (Munich, Germany), and Bayer (Leverkusen,
Radiology and Nuclear Medicine (J.W.M.), Division of Vascular & Interven- Germany). None of the other authors have identified a conflict of interest.
tional Radiology, University of Maryland School of Medicine, Baltimore,
Maryland; Department of Medical Imaging, Royal Victoria Hospital (M.O.B.), & SIR, 2016
Barrie, Ontario Canada; and Department of Radiology, King Faisal Specialist J Vasc Interv Radiol 2017; 28:1–15
Hospital & Research Center (N.H.S.), Riyadh, Saudi Arabia. Received August
16, 2016; final revision received September 18, 2016; accepted September http://dx.doi.org/10.1016/j.jvir.2016.09.024
20, 2016. Address correspondence to S.A.P., c/o Debbie Katsarelis,
2 ’ Quality Improvement Guidelines for Hepatic Radioembolization Padia et al ’ JVIR

members, or the Executive Council. A recognized expert or group of downstaging disease to meet transplantation criteria, controlling
experts is identified to serve as the principal author or writing group for disease while simultaneously inducing hypertrophy of the future liver
the document. Additional authors or societies may be sought to remnant before surgical resection, and treating patients with
increase the scope, depth, and quality of the document depending on macrovascular invasion, and as primary/definitive therapy in patients
the magnitude of the project. with tumors located within a single liver segment (10,12–17).
An in-depth literature search is performed by using electronic Several prospective and retrospective studies have demonstrated
medical literature databases. Then, a critical review of peer-reviewed the safety and efficacy of radioembolization in patients with primary
articles is performed with regard to the study methodology, results, and hepatic malignancies (Table 1) (10,13–25). Studies comparing radio-
conclusions. The qualitative weight of these articles is assembled into embolization versus surgery and percutaneous ablation techniques have
an evidence table, which is used to write the document such that it not been performed because these procedures are typically done in
contains evidence-based data with respect to content, rates, and different patient populations. Several studies comparing chemoembo-
thresholds. Threshold values are determined by calculating the stand- lization versus radioembolization in HCC demonstrated longer times to
ard deviation of the weighted mean success and adverse event reported progression, better quality of life, slightly improved toxicity profiles,
in all relevant trials with a sample size of approximately 50 patients or and fewer days of hospitalization among patients treated with radio-
greater. Calculated threshold values represent two standard deviations embolization compared with patients treated with chemoembolization
above or below the mean for adverse event and success rates, (26–29). However, a significant survival benefit has not been demon-
respectively. strated. Multiple prospective studies are under way comparing radio-
When the evidence of literature is weak, conflicting, or contra- embolization versus systemic chemotherapy in advanced-stage hepatic
dictory, consensus for the parameter is reached by a minimum of 12 malignancy.
Standards of Practice Committee members by using a modified Delphi Cholangiocarcinoma represents the other subset of primary
consensus method (Appendix A). For purposes of these documents, hepatic malignancy. The accepted systemic chemotherapy regimen
consensus is defined as 80% Delphi participant agreement on a value or for unresectable cholangiocarcinoma has a limited survival benefit
parameter. (30). Radioembolization has demonstrated efficacy in the setting of
The draft document is critically reviewed by the writing group intrahepatic cholangiocarcinoma (Table 1), and can therefore play a
and Standards of Practice Committee members by telephone confer- role in improving long-term survival. In survival assessments, several
ence calling or face-to-face meeting. The finalized draft from the factors were found to be good prognostic indicators, specifically patient
Committee is sent to the SIR Operations Committee for approval. The performance status, tumor burden, and peripheral tumor type. Con-
document is then posted on the SIR Web site for SIR membership to versely, factors deemed to be poor prognostic indicators were multi-
provide further input/criticism during a 30-day comment period. These focal tumor, infiltrative tumor, and higher tumor burden (18–20).
comments are discussed by the Standards of Practice Committee, and
appropriate revisions are made to create the finished standards docu-
ment before its publication. SECONDARY (METASTATIC) HEPATIC
MALIGNANCIES
The treatment of cancers metastatic to the liver includes surgical
INTRODUCTION resection and/or systemic chemotherapy. Radioembolization has been
Locoregional therapies have an important role in the management of shown to be safe and effective in patients with liver metastases from
hepatic malignancies. Percutaneous ablation and transarterial therapies primary cancers such as colorectal carcinoma and neuroendocrine
are the two main tools available to interventional radiologists for the tumors.
treatment of liver tumors. Yttrium-90 (90Y) radioembolization is a Radioembolization for the treatment of metastatic cancer to the
transarterial catheter-based technique that is increasingly being used in liver has been evaluated primarily in the setting of colorectal cancer
the management of primary and secondary liver malignancies. metastases, often for chemotherapy-refractory disease (ie, “salvage
This document serves as a guide on hepatic artery radioemboliza- therapy”). Several studies have evaluated the efficacy of radioemboli-
tion for the treatment of primary and secondary liver malignancies. zation for this indication (Table 2) (31–50). In a multicenter prospective
The readers are also referred to the Research Reporting Standards for randomized phase III trial, the addition of radioembolization to
Radioembolization of Hepatic Malignancies (1) with recommendations salvage chemotherapy showed a trend toward prolonged overall
on presenting data regarding radioembolization. survival compared with chemotherapy alone (10.0 vs 7.3 mo; P = .8)
Throughout this document, the procedure under discussion will be (36). Radioembolization as first-line therapy for colorectal metastases
referred to as “radioembolization.” Many other terms have been used has recently been evaluated, with preliminary results showing no
to describe the same procedure in the literature, including selective significant improvement in progression-free survival when radioembo-
internal radiation therapy (or “SIRT”) and transarterial radioemboli- lization is added to systemic chemotherapy compared with systemic
zation (or “TARE”). chemotherapy alone (37). However, an advantage in hepatic
progression-free survival was identified in the radioembolization arm.
Prospective trials assessing the effectiveness of radioembolization in the
PRIMARY HEPATIC MALIGNANCIES second-line setting are currently under way.
There were an estimated 782,500 new primary liver cancer cases and Radioembolization for neuroendocrine tumors metastatic to the
645,500 liver cancer–related deaths globally in 2008 (2). Hepatocellular liver has repeatedly shown excellent tumor response rates with
carcinoma (HCC) constitutes approximately 85%–90% of all primary significant improvement in survival. A retrospective study of 148
hepatic malignancies (2–4). Liver resection, liver transplantation, and patients demonstrated a median survival of 70 months (32). In the
ablation are considered potentially curative therapies in select patients setting of multifocal disease, bland embolization and chemo-
with HCC (5,6). Systemic therapy, including the use of sorafenib, has embolization have also shown efficacy with acceptable safety profiles.
had a limited impact in the management of HCC, and is primarily A systematic review of chemoembolization and radioembolization for
indicated in patients with advanced or metastatic disease (7,8). the treatment of neuroendocrine metastases demonstrated similar
Transarterial-based therapies such as bland embolization, chemoembo- response rates and survival (51). However, the heterogeneity of the
lization, and radioembolization have an established role in the manage- study populations makes this comparison challenging to draw firm
ment of HCC (9,10). Radioembolization has an accepted role in the conclusions. Debate therefore continues as to the optimal transarterial
treatment of HCC in several scenarios, and is endorsed by national treatment regimen for this indication.
guidelines for hepatobiliary malignancies (11). This includes “bridging” Radioembolization has also been studied for several other meta-
patients to liver transplantation by delaying disease progression, static tumors, including breast cancer, uveal melanoma, renal-cell
Volume 28

Number 1

January
Table 1 . Outcomes of Radioembolization for Primary Liver Cancer ([10,13–25)

90
Study, Year Y Device No. of Pts. CTP Score (No. of Pts.) BCLC Stage (No. of Pts.) OR (%) Median (95% CI) TTP* Survival


2017
Hepatocellular carcinoma
Carr, 2004 (13) Glass 65 NR NR NR NR 649 d (Okuda I), 302 d
(Okuda II)
Lewandowski et al, 2009 (14) Glass 43 A (24), B (19), C (0) A (0), B (34), C (9), D (0) 61 33.3 mo (17.8– 33.8 mo) 35.7 mo
Riaz et al, 2011 (15) Glass 84 A (41), B (42), C (1) A (27), B (25), C (31), D (1) 81 13.6 mo (9.3–18.7 mo) 26.9 mo
Salem et al, 2010 (10) Glass 291 A (131), B (152), C (8) A (48), B (83), C (152), D (8) 57 7.9 mo (6–10.3 mo) 17.2 mo (Child–Pugh class A),
7.7 mo (Child–Pugh class B)
Sangro et al, 2011 (23) Resin 325 A (268), B (57), C (0) A (52), B (87), C (183), D (3) NR NR 12.8 mo
Padia et al, 2014 (16) Glass 20 A (11), B (8), C (1) A (2), B (2), C (15), D (1) 95 319 d 90% at 1 y
Mazzaferro et al, 2013 (22) Glass 52 A (43), B (9), C (0) A (0), B (17), C (35), D (0) 40 11 mo 15 mo
Kokabi et al, 2015 (24) Glass 30 A (20), B (10), C (0) A (0), B (0), C (30), D (0) NR 9 mo (6.2–13.1 mo) 13 mo
Saxena et al, 2014 (25) Resin 40 A (30), B (10), C (1), NR 48 NR 27.7 mo
unknown (4)
Gramenzi et al, 2015 (21) Resin 63 A (58), B (5), C (0) A (0), B (26), C (37), D (0) 73 5 mo 13.2 mo
Cholangiocarcinoma
Saxena et al, 2010 (18) Resin 25 NR NR 24 NR 9.3 mo
Hoffman et al, 2012 (19) Resin 33 NR NR 36 9.8 mo 22 mo
Mouli et al, 2013 (20) Glass 46 NR NR 73 NR 14.6 mo

BCLC ¼ Barcelona Clinic Liver Cancer; CI ¼ confidence interval; CTP ¼ Child–Turcotte–Pugh; NR ¼ not reported; OR ¼ objective response; TTP ¼ time to progression.
*TTP is presented as overall time to progression unless otherwise stated.

3
4 ’ Quality Improvement Guidelines for Hepatic Radioembolization Padia et al ’ JVIR

Table 2 . Outcomes of Radioembolization for Metastatic Disease to the Liver (31,33–50)

90
Study, Year Y Regimen Previous Chemotherapy No. of Pts. Objective Response Survival
Colorectal cancer metastases
van Hazel et al, 2016 (37) Resin þ FOLFOX No 267 76.4% 10.7 mo (PFS)
Gray et al, 2001 (31) Resin þ chemotherapy No 35 44% 72% (1 y)
Sharma et al, 2007 (33) Resin þ FOLFOX No 20 90% 9.3 mo (PFS)
Mulcahy et al, 2009 (34) Glass Yes 72 40% 14.5 mo
Van Hazel et al, 2004 (35) Resin þ 5-FU No 11 91% 29.4 mo
Murthy et al, 2005 (39) Resin Yes 12 0% 4.5 mo
Abbott et al, 2015 (40) Glass Yes 68 NR 11.6 mo
Hendlisz et al, 2010 (36) Resin Yes 21 9.5% 10 mo
Kalva et al, 2014 (41) Resin Yes 45 2% 186 d
Saxena et al, 2015 (42) Resin Yes 302 39% 10.5 mo
Kennedy et al, 2015 (43) Resin Yes 606 NR 9.6 mo
Lewandowski et al, 2014 (44) Glass Yes 214 NR 10.6 mo
Neuroendocrine metastases
Rhee et al, 2008 (45) Resin or glass No 42 52% 25 mo
King et al, 2008 (46) Resin þ 5-FU 15% of patients 34 50% 24.2 mo
Kennedy et al, 2008 (32) Resin NR 148 63.2% 70 mo
Saxena et al, 2010 (47) Resin 52% of patients 48 54% 35 mo
Breast cancer metastases
Gordon et al, 2014 (48) Glass Yes 75 35% 6.6 mo
Saxena et al, 2014 (49) Resin Yes 40 31% 13.6 mo
Uveal melanoma
Gonsalves et al, 2011 (38) Resin No 32 6% 10 mo
Klingenstein et al, 2013 (50) Resin 77% of patients 13 62% 7 mo

5-FU ¼ 5-fluorouracil; FOLFOX ¼ leucovorin/5-fluorouracil/oxaliplatin; NR ¼ not reported; PFS ¼ progression-free survival.

carcinoma, and sarcoma (38,48–50,52,53). Current evidence supports Cholangiocarcinoma can present initially as intrahepatic or extrahe-
consideration of the use of radioembolization for these malignancies in patic. Radioembolization has been studied for HCC and intrahepatic
the salvage setting. The incidence of liver-dominant metastases is cholangiocarcinoma.
relatively low with these cancers; therefore, large-scale prospective Secondary liver cancers are those malignancies that originate at an
randomized trials are not feasible. extrahepatic site and then metastasize to the liver. Examples include
metastases from colorectal cancer, neuroendocrine tumors, breast
cancer, lung cancer, uveal melanoma, other gastrointestinal carcino-
DEFINITIONS/TERMINOLOGY mas, renal-cell carcinoma, and sarcoma.
Although practicing physicians should strive to achieve perfect out- Activity is the amount of radiation that is emitted per unit time.
comes (eg, 100% success, 0% adverse events), all physicians will fall The SI unit for activity is the becquerel, which is one nuclear
short of this ideal to a variable extent. Therefore, indicator thresholds disintegration per second.
may be used to assess the efficacy of ongoing QI programs. For the Absorbed dose refers to the amount of energy deposited (in Joules
purposes of these guidelines, a threshold is a specific level of an per kilogram) in matter by radiation, and should not be confused with
indicator that should prompt an internal review. “Procedure thresh- the unit of radioactive activity (becquerels) of the radiation source.
olds” or “overall thresholds” reference a group of indicators for a Exposure to radiation will result in a radiation absorbed dose, which is
procedure, eg, major adverse events. Individual adverse events may dependent on many factors, such as the activity of the source, volume
also be associated with adverse events–specific thresholds. When of exposed tissue, duration of exposure, energy of the emitted
measures such as indications or success rates fall below a (minimum) radiation, and distance from the source. The SI unit for absorbed
threshold, or when adverse event rates exceed a (maximum) threshold, radiation dose is the Gray.
a review should be performed to determine causes and to implement C-arm computed tomography (CT; also commonly referred to as
changes, if necessary. For example, if the incidence of gastrointestinal cone-beam CT) is a three-dimensional (3D) CT-like filtered back-
ulceration is one measure of the quality of radioembolization, values in projection reconstruction after a rotational angiogram is obtained with
excess of the defined threshold (in this case 3%) should trigger a review the use of a flat-panel detector. Potential advantages of c-arm CT
of policies and procedures within the department to determine the include improved tumor detection and extrahepatic perfusion detec-
causes and to implement changes to lower the incidence of the adverse tion, 3D vascular mapping, and real-time 3D guidance during angio-
events. Thresholds may vary from those listed; for example, patient graphic procedures (54).
referral patterns and selection factors may dictate a different threshold Nontarget embolization is defined as the unintended delivery of an
value for a particular indicator at a particular institution. Thus, setting embolic agent to a vascular territory outside the target region. In the
universal thresholds is very difficult, and each department is urged to liver, this includes portions of the liver, possibly free of disease, that
alter the thresholds as needed to higher or lower values to meet its own were not the intended target. Gallbladder, duodenum, stomach,
QI program needs. anterior abdominal wall, and lungs are examples of extrahepatic
Primary liver cancers are those malignancies that originate from nontarget regions. Nontarget embolization can result in radiation-
the hepatic tissue. These include HCC and cholangiocarcinoma. induced damage to these areas and produce symptoms of abdominal
Volume 28 ’ Number 1 ’ January ’ 2017 5

pain, bleeding, or dyspnea with the possibility of temporary or for Organ Sharing tumor/node/metastasis, and Hong Kong staging
permanent disability. It should be recognized that, depending on the systems. Although one study suggested that the Cancer of the Liver
angiographic anatomy, gallbladder, anterior abdominal wall, and lung Italian Program system outperformed other staging systems in HCC,
deposition within limits may be clinically acceptable. However, non- no single staging system can be recommended in view of the limited
target deposition involving the duodenum and stomach are never data comparing the various staging systems (57–59).
clinically acceptable. Given the numerous types of liver metastases that may be treated
Postembolization syndrome is the occurrence of abdominal pain, with radioembolization, it is impractical to mandate a specific staging
low-grade fever, nausea, vomiting, loss of appetite, and/or malaise in system based on all tumor types (eg, Dukes/tumor/node/metastasis
the first few days after radioembolization. Although this is an expected staging for colorectal cancer). However, baseline data regarding
part of recovery, its frequency and severity may be lower than seen with presence of extrahepatic metastases should be obtained.
chemoembolization. This process should not be considered an adverse
event of radioembolization unless unplanned medical therapy or
hospitalization is required. Indications and Contraindications
Technical success is the administration of 90Y microspheres into
If feasible, therapy selection is recommended by consensus in a
the target artery supplying the intended region of treatment. On multidisciplinary team, for example, including hepatologists (in the
occasion, a patient may not be a candidate for 90Y infusion based on case of primary liver cancer), medical oncologists, radiation oncolo-
nonsuitable vasculature identified on the mapping angiogram or
gists, surgeons, and interventional radiologists.
because of a high lung shunt fraction. This should not be considered For HCC, size, number, and distribution of tumors play pivotal
a technical failure because the treatment plan changed before the
roles in patient selection. Other locoregional options should also be
infusion procedure.
considered, including transarterial chemoembolization and ablation.
Radioembolization-induced liver disease (REILD; often described There is considerable overlap in the indications and patient selection of
as “radiation-induced liver disease” because of subtle differences
each treatment modality. The choice of therapy must take multiple
caused by external-beam irradiation) consists of a constellation of factors into consideration, such as best potential tumor response,
symptoms related to radiation-induced liver dysfunction in the absence
lowest degree of toxicity, patient preference, and availability of local
of objective tumor progression. These include jaundice and/or ascites,
expertise. Degree of cirrhosis (measured by Child–Turcotte–Pugh class)
usually with associated increases in serum alkaline phosphatase and must be considered, as well as trends in patients’ liver function over
bilirubin levels (unlike radiation-induced liver disease from conven-
time. The albumin-bilirubin grade is being investigated as an adjunct to
tional external-beam radiation, which is often associated with normal the Child–Pugh score (60). In general, radioembolization may be safely
bilirubin levels). REILD usually occurs 1–2 months after radioembo- offered to patients with borderline liver function if performed in a more
lization, but its onset is highly variable, making the diagnosis often
selective (ie, segmental) fashion, even though this depends on stability
challenging (55). of the patient’s liver function over time, goals of treatment (eg,
palliation vs downstaging to transplantation), and the expertise and
experience of the performing interventional radiologist.
PATIENT SELECTION For intrahepatic cholangiocarcinoma, patients should have sur-
A comprehensive understanding of the patient’s oncologic stage, gically unresectable disease at the time of treatment. Presence of
performance status, laboratory test results, treatment history, and extrahepatic metastases should not be considered an absolute contra-
potential future treatment options is critical in patient selection. indication, as several studies have shown promising overall survival in
Interventional radiologists should be aware of (i) clinical parameters this scenario (18,20). Rather, efforts at combining radioembolization
such as the Eastern Cooperative Oncology Group (ECOG) perform- with systemic chemotherapy should be considered in this situation.
ance status; (ii) laboratory assessment of liver function, coagulation Caution should be taken in patients with biliary obstruction or in
status, renal function, and tumor markers; and (iii) radiologic staging patients with a history of biliary manipulation, even though this does
of disease, including distribution, number, and size of tumors and the not represent an absolute contraindication and is associated with a
presence of vascular invasion or extrahepatic metastases. The Coui- lower risk for hepatic abscess formation than seen with chemoembo-
naud liver segment classification system is preferred for describing the lization (61). There is currently no role for radioembolization in the
anatomic location of hepatic tumors. setting of hilar/extrahepatic cholangiocarcinoma.
A thorough clinical history, including previous therapies (surgical/ In the setting of hepatic metastases, indications vary widely and
medical/interventional), should be obtained. The previous administra- are often institution-dependent. For colorectal cancer metastases, the
tion of certain systemic chemotherapy agents (especially patients metastases should be considered unresectable and not amenable to
receiving biologic agents such as bevacizumab) is relevant because of ablation. However, the timing of radioembolization with regard to a
the association with hepatic vasculature adverse events, such as vaso- patient’s chemotherapy regimen remains controversial. Most centers do
spasm, dissection, or inability to deliver the microspheres. Other not perform radioembolization as first-line therapy, but rather in a
systemic agents may cause subclinical hepatic toxicity and compromise salvage setting. It is unclear when the maximum benefit from radio-
liver reserve. Finally, the interventional radiologist should decide embolization may be derived, such as after failure of first-line or second
whether to proceed with radioembolization in the context of other line chemotherapy. Although survival times tend to be longer in
available therapies (eg, surgical resection, ablation, external-beam patients receiving radioembolization earlier in the disease course, this
radiation, systemic chemotherapy, chemoembolization). In patients may reflect selection and lead-time bias. In certain situations when a
who are already receiving systemic chemotherapy, the interventional patient has exhibited toxicity from systemic chemotherapy, radio-
radiologist should also assess whether radioembolization should be embolization can allow for a chemotherapy holiday while controlling
performed concurrently or during a chemotherapy “holiday” (56). tumor growth.
Patients with metastatic disease to the liver should have accept-
able performance status, ie, ECOG performance status 0 or 1. Poor
Disease Staging performance status should be addressed before treatment, as this may
Oncologic staging should be performed by using accepted staging reflect a patient whose condition is rapidly declining. In these cases,
systems for the particular malignancy. In the case of HCC, in which radioembolization would be of no benefit, and could even be poten-
there is often coexisting liver cirrhosis, assessment of liver function with tially harmful by accelerating the decline in performance status.
the Child–Turcotte–Pugh score should be calculated. Some commonly Hepatic function should be within normal limits, and any liver function
used staging systems for HCC include the Barcelona Clinic Liver abnormality before treatment must be addressed and managed. Abnormal
Cancer, Cancer of the Liver Italian Program, Okuda, United Network liver function is often a sign of toxicity from systemic chemotherapy or
6 ’ Quality Improvement Guidelines for Hepatic Radioembolization Padia et al ’ JVIR

rapid tumor infiltration. In these situations, radioembolization would be away from routine prophylactic embolization of the gastroduodenal
contraindicated. This is in contrast to patients with HCC, who usually artery and right gastric artery (73). Many cases of collateral vessel
have underlying cirrhosis, typically resulting in some baseline decline in formation have been observed following gastroduodenal and right
performance status and/or hepatic function abnormality. Furthermore, gastric artery embolization, and these collateral vessels are exceedingly
trends in hepatic function should be reviewed if available, as patients with difficult to embolize (74,75). The degree of anticipated arterial stasis
declining liver function may still have laboratory values that are during radioembolization is an important consideration when deciding
considered within normal limits. Isolated elevation of alkaline phosphatase if prophylactic embolization is necessary.
is typically not of concern and is more a marker of metastatic foci in the Several techniques (in addition to coil embolization) have been
liver rather than hepatic dysfunction. used to decrease the incidence of extrahepatic microsphere deposition.
The absolute contraindications to radioembolization for any A thorough evaluation of the technetium-99m (99mTc) macroaggre-
malignancy include severely compromised hepatic function, poor gated albumin (MAA) images should be performed to assess for
performance status (ECOG 4 2), active hepatic infection, significant extrahepatic uptake. Free technetium should be excluded as a cause
extrahepatic metastases, and pregnancy. After mapping angiography, of uptake in the stomach. In certain cases, a second mapping angio-
the inability to administer 90Y microspheres without nontarget embo- gram may be necessary if scintigraphic findings are concerning. The
lization to the bowel is also considered an absolute contraindication. A concept of vascular redistribution (ie, occluding intrahepatic vessels
high lung shunt fraction (LSF) that would lead to a greater than 30-Gy before radioembolization to induce redistribution of flow) has been
dose to the lungs with a single treatment is an absolute contraindication developed to simplify treatment and potentially avoid gastric vessels
for radioembolization. (76,77). For example, in the case of a replaced left hepatic artery
Relative contraindications to radioembolization include uncorrect- originating from a left gastric artery, the gastrohepatic trunk is
able coagulopathy, severe radiographic contrast medium allergy, and embolized to redistribute flow to the left from the right hepatic artery.
renal impairment, all of which can often be medically addressed. Increased This is commonly done for radioembolization with higher particle
bilirubin level and significant ascites are relative contraindications; loads. Finally, antireflux catheters (eg, Surefire, Surefire, Westminster,
individualized patient decision-making is recommended in these settings. Colorado) have played an increasing role in preventing reflux of
particles.
C-arm cone-beam CT is now routinely used intraprocedurally in
DEVICES addition to digital subtraction angiography to determine vascular
There are two types of 90Y microspheres commercially available. supply to the tumor and detect potential extrahepatic vessels (78–82).
TheraSphere (BTG International, Ottawa, Ontario, Canada) consists It allows for visualization of tumors as well as intraprocedural
of 20–30-mm-diameter glass microspheres with 90Y integrated into the guidance to position microcatheters in the appropriate vessels for
glass structure. This device received US Food and Drug Administra- targeting (54). This intraprocedural imaging technique is strongly
tion (FDA) approval in 1999 under a Humanitarian Device Exemption recommended and may become the standard of care, but is currently
as radiation treatment or as a neoadjuvant therapy to surgery or not mandated as a result of the lack of universal availability.
transplantation in patients with unresectable HCC who can have
placement of appropriately positioned hepatic arterial catheters, or in
patients with HCC with partial or branch portal vein thrombosis/ 99m
Imaging with Tc MAA
occlusion, when clinical evaluation warrants the treatment. In the Technetium-99m MAA scintigraphy is performed at the time of the
European Union and Canada, it is approved for the treatment of pretreatment angiography to quantify the LSF and identify nontarget
hepatic neoplasia in patients who have appropriately positioned flow. It is important to recognize that the gold standard for assessment
arterial catheters. SIR-Spheres (Sirtex, North Sydney, Australia) are of extrahepatic arterial flow is angiography (with or without cone-beam
biodegradable resin 20–60-mm-diameter microspheres onto which 90Y CT) and not 99mTc-MAA scintigraphy. However, improved imaging
is adsorbed to the surface. This device received FDA approval in 2002 protocols and implementation of single-photon emission CT (SPECT)
for the treatment of metastatic colorectal cancer with concomitant use will continue to play a role in confirming the lack of extrahepatic
of intraarterial floxuridine. In the European Union, SIR-Spheres are deposition of 99mTc-MAA (83).
indicated for the treatment of patients with inoperable liver tumors.
Despite the relatively narrow FDA-approved indications for both
devices, both are often used on an off-label basis based on several
medical practice guidelines (62,63). There are other radiopharmaceut- Choosing Hepatic Target Volume
ical agents (ie, radionuclide/carrier combinations) available, but there The decision to use whole-liver, lobar, or selective (ie, two or fewer
are limited data available on their safety and efficacy (64–67). hepatic segments perfused) approaches is dependent on the patient’s
baseline laboratory values, clinical characteristics, tumor distribution,
and vascular anatomy. In general, patients with better performance
TECHNIQUE status, adequate liver function, and lack of previous treatment tend to
Multiple factors may influence technique of radioembolization, such as tolerate treatment of larger hepatic target volumes. Targeting is
tumor distribution, choice of 90Y device, and goals of therapy. principally accomplished with angiography, use of c-arm cone-beam
CT, and correlation with preprocedural imaging. This allows an
appropriate level of vessel selection.
Preprocedure Angiography Lobar infusion is the most common approach for multifocal
Pretreatment (ie, planning) angiography is an integral step before disease. For whole-liver therapy, sequential lobar injections (typically
radioembolization (68–72). Meticulous mapping of the vascular anat- separated by 4–8 wk) are the most common method. In select
omy allows the interventional radiologist to (i) identify the vascular situations, especially in the setting of metastatic liver tumors in which
supply of the liver and the tumor, (ii) identify and embolize vessels that no underlying liver disease is present, whole-liver infusion can be
may lead to nontarget deposition of the radioembolic material, and (iii) performed (via separate lobar injections in a single session). Selective
to quantify LSF. The choice to perform prophylactic embolization of multivessel infusions can potentially avoid nontarget deposition in
extrahepatic vessels (eg, gastroduodenal artery, right gastric artery) is proximal extrahepatic vessels (eg, cystic artery, supraduodenal artery).
largely operator-dependent and varies considerably across institutions. Segmental radioembolization, whereby one or two hepatic segments
It is recommended that new users have a low threshold to perform are treated, can be considered in patients with isolated tumor burden,
prophylactic embolization of extrahepatic vessels before radioemboli- and may allow higher administered dose because most segments of the
zation. However, many experienced, high-volume centers have moved liver would remain unaffected by treatment (15,16,84).
Volume 28 ’ Number 1 ’ January ’ 2017 7

DOSIMETRY the same imaging modality and temporal follow-up; if possible,


Standard dosimetric formulas are presented as follows: standardization is recommended.
Radiologic response has been shown to correlate with survival
(94). Tumor response can be measured strictly based on size by using
Dose Calculation for Glass Microspheres the World Health Organization criteria and Response Evaluation
Dose calculations require the use of 3D-reconstructed cross-sectional Criteria In Solid Tumors (RECIST). In cases of enhancing tumors
imaging to calculate the volume of the targeted liver tissue to be (ie, HCC), the European Association for Study of the Liver (EASL)
infused. The volume (in milliliters) is then used to calculate the mass (in and modified RECIST (mRECIST) guidelines (measuring change in
grams) by multiplying it by a factor of 1.03 (conversion factor that the amount of enhancing, ie, viable, tumor only) are commonly used
represents liver parenchymal density). The activity (A in Equation 1) in (95,96). The use of EASL guidelines and mRECIST for HCC has
gigabecquerels administered to the target area of the liver, assuming numerous advantages over conventional size measurements, and has
uniform distribution of microspheres, is calculated using the following been shown in multiple studies to be a better predictor of progression
the Medical Internal Radiation Dose formula as follows: and survival (97–99). Combining size and enhancement criteria has
A¼D  m=50 [Eq 1] been shown to improve the prediction of complete pathologic necrosis
at explant (100). Recently, use of the “primary index tumor” to assess
Where D is the dose administered in Grays and m is the mass in response following locoregional therapy in HCC has been reported,
kilograms of the perfused tumor-bearing liver tissue. The dose delivered thereby simplifying and standardizing response evaluation
to the treated mass also depends on the percent residual activity (R in methodology in HCC (101). The use of quantitative 3D imaging
Eq 2) in the vial and tubing after treatment and the LSF, which is assessment is a developing tool that may prove to have better accuracy
calculated beforehand by using 99mTc-MAA scintigraphy. These and correlation with outcomes (102,103).
factors are accounted for in the following formula: Fluorodeoxyglucose (FDG) positron emission tomography is a
D¼A  50  ð1LSFÞ  ð1RÞ=m [Eq 2] metabolic imaging technique that has a role in response assessment
following radioembolization of some metastatic hepatic tumors, but is
Although it is noted that this dosimetry model is independent of rarely used in the setting of HCC because HCC is not typically FDG-
tumor burden, for a given dose, different number of microspheres can avid (104–108).
be administered to account for larger tumor size.
Changes in Tumor Markers
Dose Calculation for Resin Microspheres Posttreatment changes in tumor markers, such as α-fetoprotein (AFP)
The model of dosimetry for SIR-Spheres is based on whole-liver in HCC and carcinoembryonic antigen in colorectal metastases,
infusion. The calculated activity of the whole liver is multiplied by correlate to response to treatment. Recent studies support the use of
the percentage of the target site as a proportion of the whole liver. The tumor markers following locoregional therapies (92). In many cases in
formula for dosimetry of SIR-Spheres is as follows: which response for HCC is challenging to assess with imaging, the use
 of AFP trends can help direct future therapy. In cases of solitary HCC
A¼body surface area–0:2 þ % tumor burden=100
and elevated baseline AFP level (4 200 ng/mL), AFP assessment may
Where A is the activity in gigabecquerels, body surface area is outperform imaging in predicting long-term outcomes (109).
measured in meters squared, and “% tumor burden” is the percentage
of the liver that is involved by tumor. For treatment of less than the Tumor Progression
whole liver, the dose is adjusted according to the proportion of liver It is important to be able to recognize progression following treatment.
treated. A lower number of microspheres per activity can now be RECIST, mRECIST, World Health Organization, and EASL guide-
administered when infused 1 day early relative to its calibration date. lines define progression distinctly. Progression should incorporate
Several drawbacks to both dosimetry models exist, leading many development of new tumor, development or expansion of vascular
experienced users to modify their approach to dosimetry with the intent invasion, and development of extrahepatic metastases. As staged
of maximizing tumor response and minimizing liver toxicity (85). For therapy has become more accepted for liver malignancy, the differ-
example, many centers have adopted partition modeling for resin entiation of local tumor progression and overall tumor progression is
microsphere dosimetry, which involves calculating tumor versus vital in defining treatment success. In addition, the pattern of extra-
nontumor distribution of dose (86,87). Other centers have adopted hepatic disease should be well-understood, particularly in HCC, to
quantitative SPECT/CT after 99mTc-MAA injection to adjust dosim- avoid premature discontinuation of effective treatment (110).
etry for glass microspheres (88–90). This area is still under
investigation.
Patient Mortality
Patient mortality unrelated to the intervention is expected as a result of
(i) cancer, (ii) progression of underlying liver disease (particularly in the
FOLLOW-UP AND OUTCOME ASSESSMENT
case of HCC), and (iii) other comorbidities. Mortality rates should not
The response to radioembolization may be evaluated by using (i)
be confused with survival. Mortality rates are represented by the
imaging findings, (ii) changes in tumor markers, and (iii) pathologic
percentage of patients who have died at a specific time, whereas
findings (91–93). The clinical impact should be assessed as well, such as
survival data represent extrapolated medians calculated by using the
change in energy levels and pain palliation. A clinic visit is recom-
Kaplan–Meier method.
mended with laboratory workup (including liver function tests) within
1 month of treatment. Radiologic evaluation is recommended at 1–3
months following treatment and at 3–6-month intervals thereafter.
ADVERSE EVENTS
Although early follow-up imaging at 1 month is often performed, one
Published rates for individual types of adverse events (Appendix B) are
should interpret tumor response with caution at such an early time
highly dependent on patient selection and may be based on series
period, as the effects of radioembolization occur over several months.
comprising several hundred patients, which is a volume larger than
most individual practitioners are likely to treat. Generally, the adverse
Radiologic Response event–specific thresholds should therefore be set higher than the
The imaging modality (ie, CT vs magnetic resonance imaging) should adverse event–specific reported rates listed here earlier. It is also
be consistent throughout the patient’s care to best assess response to recognized that a single adverse event can cause a rate to cross
therapy. It may be difficult in some situations to have patients receive above an adverse event–specific threshold when the adverse event
8 ’ Quality Improvement Guidelines for Hepatic Radioembolization Padia et al ’ JVIR

occurs within a small patient volume, (eg, early in a QI program). In phosphatase and bilirubin levels), ascites, and fatigue in the absence of
this situation, the overall procedure threshold is more appropriate for tumor progression. Although the incidence of REILD is relatively low
use in a QI program. All values in Table 3 were supported by the (typically less than 4%), it can progress to liver failure and death (114).
weight of literature evidence and panel consensus. Previous radioembolization, multiple lines of chemotherapy, abnormal
Adverse events following radioembolization may occur as a result baseline liver function, single-session whole-liver therapy, and empiric
of (i) a toxic dose to normal hepatic parenchyma, (ii) a toxic dose to model dosimetry with resin microspheres have been identified as risk
extrahepatic tissue, (iii) adverse events of angiography (eg, puncture factors for the development of REILD (55,114).
site adverse events, vessel dissection), and (iv) systemic side effects. It is Biliary adverse events from radioembolization have been
recommend to actively track additional clinical and biochemical reported, but are relatively uncommon compared with other intra-
toxicities representing liver function abnormalities, such as bilirubin arterial therapies, likely because of the minimally embolic effect of
toxicity and ascites. radioembolization (112). Patients with biliary stents, obstructed bile
Adverse events secondary to treatment delivery should be defined ducts, or previous manipulation of the ampulla of Vater are at
by using the National Cancer Institute Common Terminology Criteria increased risk of cholangitis and bilomas. Such a history tends to be
for Adverse Events, version 4.03 (111). These criteria are designed to be more prevalent in the setting of intrahepatic cholangiocarcinoma, as
applied to all treatment modalities. many of these patients initially present with biliary obstruction.
However, these should not be considered an absolute contraindi-
cation to radioembolization. Instead, a thorough assessment of
Hepatic Adverse Events competing therapies and discussion with the patient regarding risks
Hepatic adverse events include liver failure, portal hypertension
should be undertaken. The use of prophylactic antibiotic agents, as per
(caused by hepatic fibrosis), liver abscess, intrahepatic bilomas, and
the SIR antibiotics guidelines (116), may lower the rate of infection and
liver infarction (112–117). Worsening ascites, jaundice, and laboratory
should be considered.
test results (eg, elevated total bilirubin) may indicate postprocedural
Hepatic abscess is a known adverse event associated with all
liver dysfunction.
intraarterial therapies, including radioembolization. Its incidence is
The incidence of hepatic adverse events in the literature is highly
relatively low, likely because of the minimally embolic effect of 90Y
variable as a result of diverse patient selection, duration of patient
microspheres. Management is usually conservative, but percutaneous
follow-up to assess for toxicities, and definitions for grading toxicities.
drainage may be required occasionally.
The presence of underlying cirrhosis (in the case of HCC) further
Hepatic fibrosis can be observed months to years after radio-
complicates this picture, as the natural history of cirrhosis progression
embolization, and may manifest clinically as portal hypertension.
may be confounded with liver toxicity from radioembolization. This is
Several case series demonstrating hepatic volume reduction and splenic
especially the case in patients with Child–Pugh class B and C disease,
enlargement after radioembolization have been published (118–120).
as well as patients with rapidly deteriorating liver function before
Repeat treatments have been associated with an increased incidence of
treatment.
fibrosis, and, in these cases, alternative therapies should again be
Multiple factors have been identified that place patients at
evaluated. Refinements in dosimetry may help to reduce the incidence
increased risk of hepatic dysfunction, especially the presence of
in the future, as the fibrosis likely occurs from microsphere deposition
preexisting liver disease. For this reason, patients with Child–Pugh
into the normal hepatic parenchyma. In some scenarios, the develop-
class C disease are at markedly increased risk of further hepatic
ment of fibrosis in a treated lobe may be beneficial, as contralateral
toxicity, and radioembolization should be performed with extreme
lobar hypertrophy may occur, facilitating an eventual surgical resection
caution. Patients with elevated baseline bilirubin levels, low albumin
(119).
levels, or the presence of ascites are not ideal candidates for radio-
embolization. Patients with metastatic liver tumors should have normal
background hepatic parenchyma, and therefore any hepatic laboratory
abnormality should raise concern. This often occurs when patients have Extrahepatic Adverse Events
received numerous lines of systemic chemotherapy, some of which may Extrahepatic adverse events can be separated into adverse events
be hepatotoxic. Although several guidelines on absolute values of resulting from systemic effects of radioembolization (eg, lymphopenia)
acceptable liver function are available, one must also assess trends in a and extrahepatic deposition of injected material (ie, nontarget embo-
patient’s liver function tests over time to assess their candidacy for lization). The latter includes radiation pneumonitis and radiation-
radioembolization. induced pulmonary fibrosis, radiation cholecystitis, and gastrointestinal
REILD can present at any time after radioembolization, but ulcers. When these adverse events occur, reevaluation of relevant
usually occurs at least 1 month after treatment. REILD is characterized details of the vascular anatomy from angiography and 99mTc-MAA
by abnormal liver function test results (including elevated alkaline is recommended for QI purposes.

Table 3 . Adverse Events

Adverse Event Reported Rate (%) Suggested Threshold (%)


Radiation-induced liver disease 1–4 5
Biloma requiring percutaneous drainage o1 2
Abscess with functional sphincter of Oddi o1 2
Postembolization syndrome requiring extended stay or readmission 1–2 4
GI ulceration 0–5 3
Radiation-induced skin injury o1 4
Radiation-induced cholecystitis 2 5
Radiation pneumonitis o1 1
Radiation-induced pulmonary fibrosis o1 1
Iatrogenic dissection preventing treatment 1 5
Death within 30 d 0–2 4
Volume 28 ’ Number 1 ’ January ’ 2017 9

Gastrointestinal ulceration remains one of the most concerning fractions exceeding 10% as recommend by the manufacturer. Interest-
sequela of extrahepatic deposition of 90Y microspheres. This occurs ingly, a recent study of resin microspheres for HCC (21) reported two
from unintentional 90Y embolization to arteries supplying the stomach deaths (among a total of 63 patients) secondary to radiation-induced
or duodenum, such as the gastroduodenal artery, right gastric artery, lung injury. In both these cases, large tumors with arterial–venous
supraduodenal artery, left gastric artery, or accessory left gastric artery. fistulae were prophylactically embolized to lower the shunt fractions to
The consequences of gastrointestinal ulceration are potentially severe. less than 20%, but were hypothesized to reform fistulae by the time of
In the setting of metastatic disease, the use of biologic agents is not treatment. Although chemotherapy was not administered in these two
recommended if gastrointestinal ulceration occurs. In addition, it may cases, systemic chemotherapy may also play a role in predisposing
be necessary to withhold systemic chemotherapy agents to allow for patients to radiation pneumonitis (127).
healing. Because patients may have difficulty eating, weight loss and
poor nutritional status become a major concern, which may lead to
fatigue and decrease in performance status. Many patients may require
Vascular Adverse Events
Systemic chemotherapeutic agents may render blood vessels fragile.
long-term proton pump inhibitors, intravenous nutrition, and narcotic
Hence, vascular adverse events may occur more often following
pain control. Overall quality of life is markedly affected in patients with
systemic chemotherapy for secondary liver tumors. Biologic agents
gastrointestinal ulceration, and symptoms may last for years.
have been widely known to cause vascular adverse events during
In some cases, it may not be technically possible to safely
arteriography. The most common agent associated with vascular
administer 90Y without a significant risk of nontarget embolization to
adverse events is bevacizumab, a monoclonal antibody to vascular
the bowel. In such cases, alternative therapies (eg, systemic chemo-
endothelial growth factor commonly used in metastatic colon and
therapy, chemoembolization, bland particle embolization) should be
breast cancer. Arterial dissection, vasoconstriction, and poor vascular
considered.
flow have been reported (128,129). Therefore, it is recommended that
A recent series (121) has shown that stasis during injection of resin
radioembolization proceed at least 4–6 weeks after the last dose of
microspheres, a proximal injection location, and a distal origin of the
bevacizumab. Careful selection and manipulation of microcatheters
gastroduodenal artery (even if coil embolization has been performed)
and wires are mandatory in these situations.
are the greatest risk factors for ulceration. Infusion should never be
performed from the region of the common or proper hepatic artery,
and, at the very least, lobar (eg, right hepatic artery) injections should Systemic Side Effects
be performed. In the case of whole-liver infusions, a multivessel 90Y Systemic side effects are frequent consequences of radioembolization,
approach to cover the entire liver should be chosen instead of infusion but rarely result in substantial morbidity. The most common side effect
from a proximal position. is a postradioembolization syndrome that consists of a combination of
Radiation-induced skin injury has been reported from micro- fatigue and anorexia, occasionally with nausea, vomiting, fever, and
sphere deposition in the falciform artery (122,123). Severe cases can abdominal discomfort. Analgesic agents with or without oral steroids
manifest in dermatitis or even skin ulceration. Many patients report may be considered. Patients may be given antiemetic agents such as
symptoms of superficial abdominal pain, which is thought to result ondansetron on the day of treatment. Gastrointestinal prophylaxis with
from falciform artery deposition. Because of the exceedingly low H2 receptor blockers or proton pump inhibitors and may be consid-
incidence of severe adverse events from falciform artery uptake of ered. Postradioembolization syndrome is less severe than that observed
90
Y, it may not routinely require prophylactic embolization. In many after other embolic therapies (such as chemoembolization) in which
cases, the origin of the falciform artery is in a distal second- or third- fatigue and constitutional symptoms predominate (39,130,131).
order left hepatic artery branch, and attempted catheterization may Mild abdominal pain may be experienced following radioembo-
cause spasm or dissection of the left hepatic artery. Therefore, risks of lization. Rare cases of significant pain during infusion have been
coil embolization in these instances may exceed the potential benefits. reported. This is often accompanied by nausea and diaphoresis,
Inclusion of the falciform artery in the treated distribution is not an occasionally with alterations in blood pressure and heart rate. It has
absolute contraindication to radioembolization. been attributed to an idiosyncratic reaction, often to resin microspheres
Radiation-induced cholecystitis is a well-known adverse event (39).
when infusion is performed proximal to the cystic artery, and occurs in
as many as 2% of cases. Not uncommonly, the gallbladder wall may
enhance at follow-up imaging as a result of exposure to 90Y. This FUTURE DIRECTIONS
finding is most commonly clinically innocuous. Several studies have Future developments in radioembolization will continue to refine the
assessed the impact of prophylactic embolization (eg, gelatin, coils) of therapy, with the goal of expanding appropriate indications, improving
the cystic artery, but a decrease in the rate of cholecystitis has not been tumor response (and hence survival), and minimizing adverse events.
observed with these maneuvers (124). It is likely that cases of It is imperative that robust data be obtained for new indications
cholecystitis after prophylactic embolization may be ischemic in of radioembolization. For example, further research is needed to
nature. Most instances of radiation-induced cholecystitis can be appropriately define its role in the treatment of hepatic metastases
managed conservatively with hydration, pain control, and antibiotic from uveal melanoma, sarcoma, renal-cell carcinoma, lung cancer,
therapy. In rare cases, cholecystectomy or percutaneous cholecystos- pancreatic cancer, and prostate cancer. The challenge in obtaining data
tomy may be required; however, these measures should be reserved as a is the relatively low incidence of liver-only or liver-dominant metastases
last option (125). Prevention by administration of microspheres distal in these situations. For example, patients with liver metastases from
to the cystic artery should be attempted whenever feasible. renal-cell carcinoma often present with concurrent pulmonary and
Radiation pneumonitis is rare and likely occurs from hepatic osseous metastases. Although radioembolization is not commonly
arterial–hepatic venous shunting, resulting in particle deposition to the recommended in this setting, there may be instances in which extra-
lungs. Its incidence can be controlled by appropriate dose reduction in hepatic metastases are being well-controlled by systemic chemotherapy.
cases of elevated LSF. Elevated lung shunting is often seen with This is especially true in breast cancer. In this scenario, radioemboliza-
infiltrative disease, high tumor burden, and portal vein invasion (126). tion may result in improved time to progression and patient survival. It
However, the presence of these factors should not preclude mapping is widely believed that hepatic tumor progression in many of these
angiography, as many tumors with these features do not exhibit a high cancers represents the limiting factor in patients’ survival.
degree of lung shunting. For glass microspheres, dose should be Comparative data of radioembolization versus other locoregional
reduced to keep a single-session pulmonary dose under 30 Gy (in a therapies are important to collect, especially for primary liver cancer.
patient with normal pulmonary function). Resin microspheres may be Several studies comparing chemoembolization and radioembolization
dose-modified in the same manner, or can be reduced based on shunt have shown no significant superiority of any one therapy in overall
10 ’ Quality Improvement Guidelines for Hepatic Radioembolization Padia et al ’ JVIR

survival (26–28,132). However, many of these studies had broad APPENDIX A. CONSENSUS METHODOLOGY
inclusion criteria and included patients across the spectrum of Barce- Reported adverse event–specific rates in some cases reflect the aggre-
lona Clinic Liver Cancer staging. In several studies, even within the gate of adverse events of varying severities. Thresholds are derived
same tumor stage, radioembolization was often offered to patients who from critical evaluation of the literature, evaluation of empirical data
were not candidates for chemoembolization. Further work is needed to from Standards of Practice Committee members, and, when available,
compare these therapies based on tumor stage. For example, compar- the National Benchmarks from the National Quality Registry for
ing chemoembolization and radioembolization when performed in a Interventional Radiology. Modified Delphi technique may be used to
segmental fashion would be a fair comparison. Finally, many of the enhance effective decision-making (136,137).
previously published studies compared radioembolization versus che-
moembolization when the authors were experienced with chemoembo-
lization but were early in their experience with radioembolization. As
the radioembolization technique continues to be refined and improved, APPENDIX B. ADVERSE EVENT CLASSIFICATION
comparative studies may indeed reveal differences in the future. Adverse Event Description
The combination of systemic chemotherapy and radioemboliza- A. Description narrative of adverse event (including sedation and
tion continues to be debated for primary and metastatic disease. In the anesthesia)
case of HCC, patients with certain features that portend a high B. Adverse event severity assessment*: escalation of level of care.
likelihood of future metastatic disease (eg, high AFP level, vascular
invasion) may benefit from the addition of a systemic agent in addition 1. Mild adverse event: No therapy or nominal (non-substantial)
to local arterial-based therapy. In the case of metastatic disease, the therapy (post-procedural imaging performed and fails to show
safety of systemic agents may be potentially compromised by the manifestation of adverse event); near miss (e.g., wrong site of
addition of radioembolization. Future studies should continue to focus patient prepped, recognized and corrected prior to procedure,
on the safety and efficacy of combined strategies, as this will likely wrong patient information entered for procedure, etc.);
become the new paradigm for liver cancer treatment. 2. Moderate adverse event: moderate escalation of care, requiring
With further evolution and refinement of technology and techni- substantial treatment, e.g., intervention (description of interven-
ques, the efficacy of radioembolization will continue to improve for tion and result of intervention) under conscious sedation, blood
primary and secondary hepatic malignancy. A wide range of tumor product administration, extremely prolonged outpatient obser-
response rates following radioembolization has been published. Further vation or overnight admission post outpatient procedure not
research is needed to identify factors that could result in higher tumor typical for the procedure (excludes admission or hospital days
response rates without raising rates of hepatic toxicity. For example, unrelated to adverse event);
there has been significant work in improving dosimetry models for resin 3. Severe adverse event: marked escalation of care, i.e. hospital
and glass microspheres, acknowledging the limitations of the currently admission or prolongation of existing hospital admission for >
accepted dosimetry formulas. Several publications on non–FDG 24 h hospital admission that is atypical for the procedure,
positron emission tomography–based dosimetry and quantified single inpatient transfer from regular floor/telemetry to ICU or com-
photon-emission CT dosimetry may help to create threshold doses to plex intervention performed requiring general anesthesia in
achieve higher tumor response rates (133,134). previously non-intubated patient (generally excludes pediatrics
Further studies are needed to assess factors that result in hepatic or in circumstances where GA would primarily be used in lieu of
toxicity, especially radiation-induced liver disease. Refinements in conscious sedation, e.g., in mentally challenged or severely
dosimetry can play a large role in reducing hepatic toxicity. In many uncooperative patients);
cases, this is unavoidable, especially when multifocal tumor is present. 4. Life-threatening or disabling event, e.g. cardiopulmonary arrest,
However, refinements in dosimetry, use of segmental infusions, and use shock, organ failure, unanticipated dialysis, paralysis, loss of
of ancillary devices may help to improve rates of hepatic toxicity. limb or organ;
Several series have shown extraordinarily low rates of hepatic toxicity
5. Patient death or unexpected pregnancy abortion
from segmental radioembolization (15,16), as most of the hepatic
n
parenchyma is spared exposure to radiation. Future directions in The SIR Adverse event Severity Scale is intended to approximate
dosimetric quantification from the mapping 99mTc-MAA study may the surgical Clavien-Dingo scale and the NCI CTCAE scale. The SIR
further help to refine the dosimetry model. scale is tailored towards the procedures and adverse events encountered
Nontarget embolization of microspheres to the gastrointestinal in IR practices. The grading of interventional oncology adverse events
tract must be minimized, as radiation-induced ulcers represent a can selectively incorporate relevant adverse event grading definitions
significant cause of patient morbidity and can severely limit quality published in the current CTCAE for oncological interventions, which
of life. Future studies looking at various techniques to minimize may be particularly relevant in the context of research publications. All
nontarget deposition are needed. adverse events occurring within 30 days of a procedure should be
Finally, cost–benefit analyses are lacking in this arena. This is included in the adverse event description and analysis, regardless of
becoming increasingly important as the trend to reduce overall health causality, in the interest of objectivity. The adverse event scale itself
care expenditures continues. Although many believe radioembolization does not assess operator performance.
can be a costly procedure, cost–benefit comparisons of radioemboliza-
tion versus other techniques must take into account costs over specified Modifier:
time periods (ie, cost per patient year of survival) as well as quality of M = multiple adverse events, each of which is counted and
life (eg, number of days of hospitalization, days of work lost) (135). evaluated separately if possible;
The preceding part refers to adverse event description and severity
characterization. It is suitable for scientific use (presentations, publica-
tions etc.) as well as for adverse event reviews within a practice,
practice group, facility or specialty.
CONCLUSIONS
Radioembolization has an established role in the management of The following part pertains to adverse event analysis. It is designed to
hepatic malignancies. Ongoing research will help to refine the technol- enable a confidential and constructive review of any adverse event
ogy and techniques, further validate indications, and help determine its within an IR practice or practice group. Applicability for scientific
exact place among other therapies for primary and secondary liver publications is limited and there is none for other public use. The
cancer. following content is meant to provide a strictly confidential, legally
Volume 28 ’ Number 1 ’ January ’ 2017 11

non-discoverable, non-punitive, objective, consistent and clinically ACKNOWLEDGMENTS


constructive analytic guide that may result in quality improvement
measures to advance the quality of patient care in interventional Siddharth A. Padia, MD, authored the first draft of this document and
radiology. served as topic leader during the subsequent revisions of the draft.
T. Gregory Walker, MD, and James E. Silberzweig, MD, are co-chairs
of the SIR Standards of Practice Committee. Boris Nikolic, MD,
Adverse Event Analysis MBA, is Councilor of the SIR Standards Division. All other authors
A. Causality are listed alphabetically. Other members of the Standards of Practice
Category 1. Adverse event not caused by the procedure Committee and SIR who participated in the development of this
Category 2. Unknown whether adverse event was caused by the guideline are (listed alphabetically): J. Fritz Angle, MD, Bulent Arslan,
procedure MD, Stephen Balter, PhD, Kevin Baskin, MD, Olga Brook, MD,
Category 3. Adverse event caused by the procedure Drew Caplin, MD, Michael Censullo, MD, Abbas Chamsuddin, MD,
B. Patient and procedural risk modifier: Christine Chao, MD, Marco Cura, MD, Mandeep S. Dagli, MD, Sean
R, Dariushnia, MD, Jon Davidson, MD, A. Michael Devane, MD,
Risk modifier: Eduardo Eyheremendy, MD, Florian Fintelmann, MD, Suvranu
Category 1. High risk patient AND technically challenging Ganguli, MD, Joseph J. Gemmete, MD, Vyacheslav Gendel, MD,
procedure Kirk Giesbrandt, MD, Jennifer Gould, MD, Tara Graham, MD, John
Category 2. High risk patient (e.g. ASA 4, uncorrectable coagul- Hancock, MD, Mark Hogan, MD, Bertrand Janne d’Othee, MD,
opathy, poor functional status (ECOG 3 & 4), poly- MPH, Ahmed Kamel Abdel Aal, MD, MSc, PhD, Sanjeeva Kalva,
pharmacy/polyintravenous therapy and transfusion, MD, Baljendra Kapoor, MD, Maureen P. Kohi, MD, Naganathan B.
septicemia, hemodynamic instability, recent cata- Mani, MD, Gloria Martinez‑Salazar, MD, Mehran Midia, MD,
strophic event/ICU admission/major surgery or inter- Donald L. Miller, MD, John Moriarty, MD, Christopher Morris,
ventions) etc. OR low risk patient and technically MD, Waleska Pabon‑Ramos, MD, MPH, Indravadan Patel, MD, Anil
challenging procedure (e.g. TIPS with occluded portal Pillai, MD, Uei Pua, MD, Ellen Redstone, MD, Anne C. Roberts,
vein, percutaneous biliary drain placement in non- MD, Tarun Sabharwal, MD, Cindy K. Saiter, NP, Marc Sapoval,
dilated biliary system, etc.) MD, PhD, Brian J. Schiro, MD, Samir Shah, MD, Paul Shyn, MD,
Category 3. No modifier Nasir Siddiqi, MD, LeAnn Stokes, MD, Rajeev Suri, MD, Timothy L.
C. Adverse event preventability Swan, MD, Naciye Turan, MD, Ulku Turba, MD, Aradhana
Category 1: Rarely preventable: i.e. well described and “typical” for Venkatesan, MD, Jeffrey L. Weinstein, MD, and Joan C. Wojak, MD.
the procedure and occurring despite adequate precau-
tionary and preventive measures
Category 2: Potentially preventable
Category 3: Consistently preventable: e.g. inappropriateness of pro- REFERENCES
cedural indication (may use checklist, see appendixn)
D. Adverse event Management 1. Salem R, Lewandowski RJ, Gates VL, et al. Research reporting stand-
Category 1: Most operators would have handled the adverse event ards for radioembolization of hepatic malignancies. J Vasc Interv Radiol
similarly; 2011; 22:265–278.
Category 2: Some operators would have handled the adverse event 2. Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global
differently; cancer statistics, 2012. CA Cancer J Clin 2015; 65:87–108.
3. El-Serag HB, Kanwal F. Epidemiology of hepatocellular carcinoma in the
Category 3: Most operators would have handled the adverse event
United States: where are we? Where do we go? Hepatology 2014; 60:
differently. 1767–1775.
4. El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carci-
noma. Gastroenterology 2012; 142:1264–1273.
Consistently Preventable Events 5. Mazzaferro V, Regalia E, Doci R, et al. Liver transplantation for the
treatment of small hepatocellular carcinomas in patients with cirrhosis. N
 Wrong patient
Engl J Med 1996; 334:693–699.
 Absolute contraindication for procedure 6. Lu DS, Yu NC, Raman SS, et al. Radiofrequency ablation of hepatocel-
 Wrong side for procedure lular carcinoma: treatment success as defined by histologic examination
of the explanted liver. Radiology 2005; 234:954–960.
 Wrong procedure
7. Llovet JM, Ricci S, Mazzaferro V, et al. Sorafenib in advanced hepato-
 Wrong medication/contrast agent/blood product (dose/ cellular carcinoma. N Engl J Med 2008; 359:378–390.
administration route) 8. Harding JJ, Abou-Alfa GK. Treating advanced hepatocellular carcinoma:
 Exposure to known allergens how to get out of first gear. Cancer 2014; 120:3122–3130.
9. Llovet J, Real MI, Montana X, et al. Arterial embolisation or chemo-
 Intraarterial placement of catheter meant to be intravenous or
embolisation versus symptomatic treatment in patients with unresect-
nonvenous placement of inferior vena cava filter able hepatocellular carcinoma: a randomised controlled trial. Lancet 2002;
 Ferromagnetic devices contraindicating performance of mag- 359:1734–1739.
netic resonance imaging 10. Salem R, Lewandowski RJ, Mulcahy MF, et al. Radioembolization for
hepatocellular carcinoma using yttrium-90 microspheres: a comprehen-
 Failure to follow up or communicate laboratory, pathology, or
sive report of long-term outcomes. Gastroenterology 2010; 138:52–64.
radiology results
11. Benson AB 3rd, D’Angelica MI, Abrams TA, et al. Hepatobiliary cancers,
 Use of known malfunctioning equipment or patient version 2.2014. J Natl Compr Canc Netw 2014; 12:1152–1182.
monitor system 12. Kulik LM, Carr BI, Mulcahy MF, et al. Safety and efficacy of 90Y
 Lack or inappropriate use of monitoring equipment during radiotherapy for hepatocellular carcinoma with and without portal vein
thrombosis. Hepatology 2007; 47:71–81.
sedation
13. Carr BI. Hepatic arterial 90yttrium glass microspheres (Therasphere) for
unresectable hepatocellular carcinoma: interim safety and survival data
on 65 patients. Liver Transpl 2004; 10:S107–S110.
Billing, Coding, and Reimbursement 14. Lewandowski RJ, Kulik LM, Riaz A, et al. A comparative analysis of
The most current coding updates are available in the member area of transarterial downstaging for hepatocellular carcinoma: chemoemboliza-
the SIR Web site at www.sirweb.org. tion versus radioembolization. Am J Transplant 2009; 9:1920–1928.
12 ’ Quality Improvement Guidelines for Hepatic Radioembolization Padia et al ’ JVIR

15. Riaz A, Gates VL, Atassi B, et al. Radiation segmentectomy: a novel fluorouracil/leucovorin chemotherapy alone in advanced colorectal can-
approach to increase safety and efficacy of radioembolization. Int J Radiat cer. J Surg Oncol 2004; 88:78–85.
Oncol Biol Phys 2011; 79:163–171. 36. Hendlisz A, Eynde MVD, Peeters M, et al. Phase III trial comparing
16. Padia SA, Kwan SW, Roudsari B, Monsky WL, Coveler A, Harris protracted intravenous fluorouracil infusion alone or with yttrium-90 resin
WP. Superselective yttrium-90 radioembolization for hepatocellular microspheres radioembolization for liver-limited metastatic colorectal
carcinoma yields high response rates with minimal toxicity. J Vasc Interv cancer refractory to standard chemotherapy. J Clin Oncol 2010; 28:
Radiol 2014; 25:1067–1073. 3687–3694.
17. Vouche M, Lewandowski RJ, Atassi R, et al. Radiation lobectomy: 37. van Hazel GA, Heinemann V, Sharma NK, et al. SIRFLOX: randomized
Time-dependent analysis of future liver remnant volume in unresectable phase III trial comparing first-line mFOLFOX6 (plus or minus bevacizu-
liver cancer as a bridge to resection. J Hepatol 2013; 59:1029–1036. mab) versus mFOLFOX6 (plus or minus bevacizumab) plus selective
18. Saxena A, Bester L, Chua TC, Chu FC, Morris DL. Yttrium-90 radio- internal radiation therapy in patients with metastatic colorectal cancer. J
therapy for unresectable intrahepatic cholangiocarcinoma: a preliminary Clin Oncol 2016; 34:1723–1731.
assessment of this novel treatment option. Ann Surg Oncol 2010; 17: 38. Gonsalves CF, Eschelman DJ, Sullivan KL, Anne PR, Doyle L, Sato
484–491. T. Radioembolization as salvage therapy for hepatic metastasis of uveal
19. Hoffmann RT, Paprottka PM, Schön A, et al. Transarterial hepatic melanoma: a single-institution experience. AJR Am J Roentgenol 2011;
yttrium-90 radioembolization in patients with unresectable intrahepatic 196:468–473.
cholangiocarcinoma: factors associated with prolonged survival. Cardio- 39. Murthy R, Xiong H, Nunez R, et al. Yttrium 90 resin microspheres for
vasc Intervent Radiol 2012; 35:105–116. the treatment of unresectable colorectal hepatic metastases after failure
20. Mouli S, Memon K, Baker T, et al. Yttrium-90 radioembolization for of multiple chemotherapy regimens: preliminary results. J Vasc Interv
intrahepatic cholangiocarcinoma: safety, response, and survival analysis. Radiol 2005; 16:937–945.
J Vasc Interv Radiol 2013; 24:1227–1234. 40. Abbott AM, Kim R, Hoffe SE, et al. Outcomes of therasphere radio-
21. Gramenzi A, Golfieri R, Mosconi C, et al. Yttrium-90 radioembolization embolization for colorectal metastases. Clin Colorectal Cancer 2015; 14:
vs sorafenib for intermediate-locally advanced hepatocellular carcinoma: 146–153.
a cohort study with propensity score analysis. Liver Int 2015; 35: 41. Kalva SP, Rana RS, Liu R, et al. Yttrium-90 radioembolization as salvage
1036–1047. therapy for liver metastases from colorectal cancer. Am J Clin Oncol
22. Mazzaferro V, Sposito C, Bhoori S, et al. Yttrium-90 radioembolization 2014.
for intermediate-advanced hepatocellular carcinoma: a phase 2 study. 42. Saxena A, Meteling B, Kapoor J, Golani S, Morris DL, Bester L. Is
Hepatology 2013; 57:1826–1837. yttrium-90 radioembolization a viable treatment option for unresectable,
23. Sangro B, Carpanese L, Cianni R, et al. Survival after yttrium-90 resin chemorefractory colorectal cancer liver metastases? A large single-center
microsphere radioembolization of hepatocellular carcinoma across Bar- experience of 302 patients. Ann Surg Oncol 2015; 22:794–802.
celona clinic liver cancer stages: a European evaluation. Hepatology 43. Kennedy AS, Ball D, Cohen SJ, et al. Multicenter evaluation of the
2011; 54:868–878. safety and efficacy of radioembolization in patients with unresectable
24. Kokabi N, Camacho JC, Xing M, et al. Open-label prospective study of colorectal liver metastases selected as candidates for (90)Y resin micro-
the safety and efficacy of glass-based yttrium 90 radioembolization for spheres. J Gastrointest Oncol 2015; 6:134–142.
infiltrative hepatocellular carcinoma with portal vein thrombosis. Cancer 44. Lewandowski RJ, Memon K, Mulcahy MF, et al. Twelve-year experi-
2015; 121:2164–2174. ence of radioembolization for colorectal hepatic metastases in 214
25. Saxena A, Meteling B, Kapoor J, et al. Yttrium-90 radioembolization is a patients: survival by era and chemotherapy. Eur J Nucl Med Mol Imaging
safe and effective treatment for unresectable hepatocellular carcinoma: a 2014; 41:1861–1869.
single centre experience of 45 consecutive patients. Int J Surg 2014; 12: 45. Rhee TK, Lewandowski RJ, Liu DM, et al. 90Y radioembolization for
1403–1408. metastatic neuroendocrine liver tumors. Ann Surg 2008; 247:1029–1035.
26. Kooby DA, Egnatashvili V, Srinivasan S, et al. Comparison of yttrium-90 46. King J, Quinn R, Glenn DM, et al. Radioembolization with selective
radioembolization and transcatheter arterial chemoembolization for the internal radiation microspheres for neuroendocrine liver metastases.
treatment of unresectable hepatocellular carcinoma. J Vasc Interv Radiol Cancer 2008; 113:921–929.
2010; 21:224–230. 47. Saxena A, Chua TC, Bester L, Kokandi A, Morris DL. Factors predicting
27. Salem R, Lewandowski RJ, Kulik L, et al. Radioembolization results in response and survival after yttrium-90 radioembolization of unresectable
longer time-to-progression and reduced toxicity compared with chemo- neuroendocrine tumor liver metastases: a critical appraisal of 48 cases.
embolization in patients with hepatocellular carcinoma. Gastroenterology Ann Surg 2010; 251:910–916.
2011; 140:497–507. 48. Gordon AC, Gradishar WJ, Kaklamani VG, et al. Yttrium-90 radioembo-
28. Lance C, McLennan G, Obuchowski N, et al. Comparative analysis of lization stops progression of targeted breast cancer liver metastases after
the safety and efficacy of transcatheter arterial chemoembolization and failed chemotherapy. J Vasc Interv Radiol 2014; 25:1523–1532.
yttrium-90 radioembolization in patients with unresectable hepatocellular 49. Saxena A, Kapoor J, Meteling B, Morris DL, Bester L. Yttrium-90
carcinoma. J Vasc Interv Radiol 2011; 22:1697–1705. radioembolization for unresectable, chemoresistant breast cancer liver
29. Salem R, Gilbertsen M, Butt Z, et al. Increased quality of life among metastases: a large single-center experience of 40 patients. Ann Surg
hepatocellular carcinoma patients treated with radioembolization, com- Oncol 2014; 21:1296–1303.
pared with chemoembolization. Clin Gastroenterol Hepatol 2013; 11: 50. Klingenstein A, Haug AR, Zech CJ, Schaller UC. Radioembolization as
1358–1365. locoregional therapy of hepatic metastases in uveal melanoma patients.
30. Valle J, Wasan H, Palmer DH, et al. Cisplatin plus gemcitabine versus Cardiovasc Intervent Radiol 2013; 36:158–165.
gemcitabine for biliary tract cancer. N Engl J Med 2010; 362:1273–1281. 51. Yang TX, Chua TC, Morris DL. Radioembolization and chemoemboliza-
31. Gray B, Van Hazel G, Hope M, et al. Randomised trial of SIR-Spheres tion for unresectable neuroendocrine liver metastases - a systematic
plus chemotherapy vs. chemotherapy alone for treating patients with review. Surg Oncol 2012; 21:299–308.
liver metastases from primary large bowel cancer. Ann Oncol 2001; 12: 52. Transue DL, Hackworth J, Johnson MS, et al. Multi-institutional expe-
1711–1720. rience treating metastatic soft tissue sarcomas using yttrium-90 micro-
32. Kennedy AS, Dezarn WA, McNeillie P, et al. Radioembolization for spheres. J Vasc Interv Radiol 2015; 26:S133.
unresectable neuroendocrine hepatic metastases using resin 90Y-micro- 53. Abdelmaksoud MHK, Louie JD, Hwang GL, Kothary N, Minor DR, Sze
spheres: early results in 148 patients. Am J Clin Oncol 2008; 31:271–279. DY. Yttrium-90 radioembolization of renal cell carcinoma metastatic to
33. Sharma RA, Van Hazel GA, Morgan B, et al. Radioembolization of liver the liver. J Vasc Interv Radiol 2012; 23:323–330.
metastases from colorectal cancer using yttrium-90 microspheres with 54. Tacher V, Radaelli A, Lin M, Geschwind JF. How I do it: cone-beam CT
concomitant systemic oxaliplatin, fluorouracil, and leucovorin chemo- during transarterial chemoembolization for liver cancer. Radiology 2015;
therapy. J Clin Oncol 2007; 25:1099–1106. 274:320–334.
34. Mulcahy MF, Lewandowski RJ, Ibrahim SM, et al. Radioembolization of 55. Gil-Alzugaray B, Chopitea A, Iñarrairaegui M, et al. Prognostic factors
colorectal hepatic metastases using yttrium-90 microspheres. Cancer and prevention of radioembolization-induced liver disease. Hepatology
2009; 115:1849–1858. 2013; 57:1078–1087.
35. Van Hazel G, Blackwell A, Anderson J, et al. Randomised phase 2 trial 56. Hickey R, Mulcahy MF, Lewandowski RJ, et al. Chemoradiation of
of SIR-Spheres plus fluorouracil/leucovorin chemotherapy versus hepatic malignancies: prospective, phase 1 study of full-dose
Volume 28 ’ Number 1 ’ January ’ 2017 13

capecitabine with escalating doses of yttrium-90 radioembolization. Int J radioembolization in the treatment of liver tumors. Cardiovasc Intervent
Radiat Oncol Biol Phys 2014; 88:1025–1031. Radiol 2010; 33:523–531.
57. Seo YS, Kim YJ, Um SH, et al. Evaluation of the prognostic powers of 78. Orth RC, Wallace MJ, Kuo MD. Technology assessment committee of
various tumor status grading scales in patients with hepatocellular the society of interventional radiology. C-arm cone-beam CT: general
carcinoma. J Gastroenterol Hepatol 2008; 23:1267–1275. principles and technical considerations for use in interventional radiology.
58. Kinoshita A, Onoda H, Fushiya N, Koike K, Nishino H, Tajiri H. Staging J Vasc Interv Radiol 2008; 19:814–820.
systems for hepatocellular carcinoma: current status and future perspec- 79. Louie JD, Kothary N, Kuo WT, et al. Incorporating cone-beam CT into
tives. World J Hepatol 2015; 7:406–424. the treatment planning for yttrium-90 radioembolization. J Vasc Interv
59. Memon K, Kulik LM, Lewandowski RJ, et al. Comparative study of Radiol 2009; 20:606–613.
staging systems for hepatocellular carcinoma in 428 patients treated with 80. Wallace MJ, Murthy R, Kamat PP, et al. Impact of C-arm CT on hepatic
radioembolization. J Vasc Interv Radiol 2014; 25:1056–1066. arterial interventions for hepatic malignancies. J Vasc Interv Radiol 2007;
60. Johnson PJ, Berhane S, Kagebayashi C, et al. Assessment of liver 18:1500–1507.
function in patients with hepatocellular carcinoma: a new evidence-based 81. Miyayama S, Yamashiro M, Okuda M, et al. Usefulness of cone-beam
approach-the ALBI grade. J Clin Oncol 2015; 33:550–558. computed tomography during ultraselective transcatheter arterial chemo-
61. Cholapranee A, van Houten D, Deitrick G, et al. Risk of liver abscess embolization for small hepatocellular carcinomas that cannot be demon-
formation in patients with prior biliary intervention following yttrium-90 strated on angiography. Cardiovasc Intervent Radiol 2009; 32:255–264.
radioembolization. Cardiovasc Intervent Radiol 2015; 38:397–400. 82. Meyer BC, Frericks BB, Voges M, et al. Visualization of hypervascular
62. Kennedy A, Nag S, Salem R, et al. Recommendations for radioembo- liver lesions during TACE: comparison of angiographic C-arm CT and
lization of hepatic malignancies using yttrium-90 microsphere brachy- MDCT. AJR Am J Roentgenol 2008; 190:W263–W269.
therapy: a consensus panel report from the radioembolization 83. Lenoir L, Edeline J, Rolland Y, et al. Usefulness and pitfalls of MAA
brachytherapy oncology consortium. Int J Radiat Oncol Biol Phys 2007; SPECT/CT in identifying digestive extrahepatic uptake when planning
68:13–23. liver radioembolization. Eur J Nucl Med Mol Imaging 2012; 39:872–880.
63. Giammarile F, Bodei L, Chiesa C, et al. EANM procedure guideline for 84. Vouche M, Habib A, Ward TJ, et al. Unresectable solitary hepatocellular
the treatment of liver cancer and liver metastases with intra-arterial carcinoma not amenable to radiofrequency ablation: multicenter radiol-
radioactive compounds. Eur J Nucl Med Mol Imaging 2011; 38: ogy-pathology correlation and survival of radiation segmentectomy.
1393–1406. Hepatology 2014; 60:192–201.
64. Raoul JL, Guyader D, Bretagne JF, et al. Prospective randomized trial of 85. Lau WY, Kennedy AS, Kim YH, et al. Patient selection and activity
chemoembolization versus intra-arterial injection of 131I-labeled-iodized planning guide for selective internal radiotherapy with yttrium-90 resin
oil in the treatment of hepatocellular carcinoma. Hepatology 1997; 26: microspheres. Int J Radiat Oncol Biol Phys 2012; 82:401–407.
1156–1161. 86. Kao YH, Tan EH, Ng CE, Goh SW. Clinical implications of the body
65. Kumar A, Srivastava DN, Chau TTM, et al. Inoperable hepatocellular surface area method versus partition model dosimetry for yttrium-90
carcinoma: transarterial 188Re HDD-labeled iodized oil for treatment– radioembolization using resin microspheres: a technical review. Ann Nucl
prospective multicenter clinical trial. Radiology 2007; 243:509–519. Med 2011; 25:455–461.
66. Wong JY, Somlo G, Odom-Maryon T, et al. Initial clinical experience 87. Lam MG, Louie JD, Abdelmaksoud MHK, Fisher GA, Cho-Phan CD, Sze
evaluating yttrium-90-chimeric T84.66 anticarcinoembryonic antigen anti- DY. Limitations of body surface area-based activity calculation for
body and autologous hematopoietic stem cell support in patients with radioembolization of hepatic metastases in colorectal cancer. J Vasc
carcinoembryonic antigen-producing metastatic breast cancer. Clin Can- Interv Radiol 2014; 25:1085–1093.
cer Res 1999; 5:3224s–3231s. 88. Lam MG, Banerjee A, Goris ML, et al. Fusion dual-tracer SPECT-based
67. Kim JK, Han KH, Lee JT, et al. Long-term clinical outcome of phase IIb hepatic dosimetry predicts outcome after radioembolization for a wide
clinical trial of percutaneous injection with holmium-166/chitosan com- range of tumour cell types. Eur J Nucl Med Mol Imaging 2015; 42:
plex (Milican) for the treatment of small hepatocellular carcinoma. Clin 1192–1201.
Cancer Res 2006; 12:543–548. 89. Garin E, Lenoir L, Edeline J, et al. Boosted selective internal radiation
68. Salem R, Thurston KG. Radioembolization with 90yttrium micro- therapy with (90)Y-loaded glass microspheres (B-SIRT) for hepatocellular
spheres: a state-of-the-art brachytherapy treatment for primary and carcinoma patients: a new personalized promising concept. Eur J Nucl
secondary liver malignancies. part 2: special topics. J Vasc Interv Radiol Med Mol Imaging 2013; 40:1057–1068.
2006; 17:1425–1439. 90. Garin E, Lenoir L, Rolland Y, et al. Dosimetry based on 99mTc-macro-
69. Salem R, Thurston KG. Radioembolization with 90yttrium micro- aggregated albumin SPECT/CT accurately predicts tumor response and
spheres: a state-of-the-art brachytherapy treatment for primary and survival in hepatocellular carcinoma patients treated with 90Y-loaded
secondary liver malignancies. part 1: technical and methodologic consid- glass microspheres: preliminary results. J Nucl Med 2012; 53:255–263.
erations. J Vasc Interv Radiol 2006; 17:1251–1278. 91. Ibrahim SM, Nikolaidis P, Miller FH, et al. Radiologic findings following
70. Salem R, Lewandowski RJ, Sato KT, et al. Technical Aspects of Y90 radioembolization for primary liver malignancies. Abdom Imaging
Radioembolization with 90Y Microspheres. Tech Vasc Interv Radiol 2009; 34:566–581.
2007; 10:12–29. 92. Riaz A, Ryu RK, Kulik LM, et al. Alpha-fetoprotein response after
71. Liu DM, Salem R, Bui JT, et al. Angiographic considerations in patients locoregional therapy for hepatocellular carcinoma: oncologic marker of
undergoing liver-directed therapy. J Vasc Interv Radiol 2005; 16:911–935. radiologic response, progression, and survival. J Clin Oncol 2009; 27:
72. Lewandowski RJ, Sato KT, Atassi B, et al. Radioembolization with 90Y 5734–5742.
microspheres: angiographic and technical considerations. Cardiovasc 93. Riaz A, Kulik L, Lewandowski RJ, et al. Radiologic-pathologic correlation
Intervent Radiol 2007; 30:571–592. of hepatocellular carcinoma treated with internal radiation using yttrium-
73. Hamoui N, Minocha J, Memon K, et al. Prophylactic embolization of the 90 microspheres. Hepatology 2009; 49:1185–1193.
gastroduodenal and right gastric arteries is not routinely necessary before 94. Memon K, Kulik L, Lewandowski RJ, et al. Radiographic response to
radioembolization with glass microspheres. J Vasc Interv Radiol 2013; 24: locoregional therapy in hepatocellular carcinoma predicts patient survival
1743–1745. times. Gastroenterology 2011; 141:526–535.
74. Petroziello MF, McCann JW, Gonsalves CF, et al. Side-branch emboli- 95. Bruix J, Sherman M, Llovet JM, et al. Clinical management of
zation before 90Y radioembolization: rate of recanalization and new hepatocellular carcinoma. Conclusions of the Barcelona-2000 EASL
collateral development. AJR Am J Roentgenol 2011; 197:W169–W174. conference. European Association for the Study of the Liver. J Hepatol
75. Abdelmaksoud MHK, Hwang GL, Louie JD, et al. Development of new 2001; 35:421–430.
hepaticoenteric collateral pathways after hepatic arterial skeletonization 96. Lencioni R, Llovet J. Modified RECIST (mRECIST) assessment for
in preparation for yttrium-90 radioembolization. J Vasc Interv Radiol 2010; hepatocellular carcinoma. Semin Liver Dis 2010; 30:52–60.
21:1385–1395. 97. Edeline J, Boucher E, Rolland Y, et al. Comparison of tumor response
76. Abdelmaksoud MHK, Louie JD, Kothary N, et al. Consolidation of by Response Evaluation Criteria in Solid Tumors (RECIST) and modified
hepatic arterial inflow by embolization of variant hepatic arteries in RECIST in patients treated with sorafenib for hepatocellular carcinoma.
preparation for yttrium-90 radioembolization. J Vasc Interv Radiol 2011; Cancer 2012; 118:147–156.
22:1364–1371. 98. Forner A, Ayuso C, Varela M, et al. Evaluation of tumor response after
77. Bilbao JI, Garrastachu P, Herráiz MJ, et al. Safety and efficacy assess- locoregional therapies in hepatocellular carcinoma. Cancer 2009; 115:
ment of flow redistribution by occlusion of intrahepatic vessels prior to 616–623.
14 ’ Quality Improvement Guidelines for Hepatic Radioembolization Padia et al ’ JVIR

99. Seyal AR, Gonzalez-Guindalini FD, Arslanoglu A, et al. Reproducibility 118. Nosher JL, Ohman-Strickland PA, Jabbour S, Narra V, Nosher B.
of mRECIST in assessing response to transarterial radioembolization Changes in liver and spleen volumes and liver function after radio-
therapy in hepatocellular carcinoma. Hepatology 2015; 62:1111–1121. embolization with yttrium-90 resin microspheres. J Vasc Interv Radiol
100. Riaz A, Memon K, Miller FH, et al. Role of the EASL, RECIST, and 2011; 22:1706–1713.
WHO response guidelines alone or in combination for hepatocellular 119. Gaba RC, Lewandowski RJ, Kulik LM, et al. Radiation lobectomy:
carcinoma: radiologic-pathologic correlation. J Hepatol 2011; 54: preliminary findings of hepatic volumetric response to lobar yttrium-90
695–704. radioembolization. Ann Surg Oncol 2009; 16:1587–1596.
101. Riaz A, Miller FH, Kulik LM, et al. Imaging response in the primary 120. Theysohn JM, Ertle J, Müller S, et al. Hepatic volume changes after
index lesion and clinical outcomes following transarterial locoregional lobar selective internal radiation therapy (SIRT) of hepatocellular carci-
therapy for hepatocellular carcinoma. JAMA 2010; 303:1062–1069. noma. Clin Radiol 2014; 69:172–178.
102. Tacher V, Lin M, Duran R, et al. Comparison of existing response 121. Lam MG, Banerjee S, Louie JD, et al. Root cause analysis of gastro-
criteria in patients with hepatocellular carcinoma treated with trans- duodenal ulceration after yttrium-90 radioembolization. Cardiovasc
arterial chemoembolization using a 3D quantitative approach. Radiology Intervent Radiol 2013; 36:1536–1547.
2015.
122. Leong QM, Lai HK, Lo RG, Teo TK, Goh A, Chow PK. Radiation
103. Chapiro J, Duran R, Lin M, et al. Early survival prediction after intra-
dermatitis following radioembolization for hepatocellular carcinoma: a
arterial therapies: a 3D quantitative MRI assessment of tumour
case for prophylactic embolization of a patent falciform artery. J Vasc
response after TACE or radioembolization of colorectal cancer meta-
Interv Radiol 2009; 20:833–836.
stases to the liver. Eur Radiol 2015; 25:1993–2003.
123. Riaz A, Lewandowski RJ, Kulik LM, et al. Complications following
104. Lin M, Shon IH, Wilson R, D’Amours SK, Schlaphoff G, Lin P. Treat-
radioembolization with yttrium-90 microspheres: a comprehensive
ment response in liver metastases following 90Y SIR-spheres: an
literature review. J Vasc Interv Radiol 2009; 20:1121–1130.
evaluation with PET. Hepatogastroenterology 2007; 54:910–912.
124. Powerski M, Busse A, Seidensticker M, et al. Prophylactic emboliza-
105. Fendler WP, Philippe Tiega DB, Ilhan H, et al. Validation of several
SUV-based parameters derived from 18F-FDG PET for prediction of tion of the cystic artery prior to radioembolization of liver malignancies–
survival after SIRT of hepatic metastases from colorectal cancer. J Nucl an evaluation of necessity. Cardiovasc Intervent Radiol 2015; 38:
Med 2013; 54:1202–1208. 678–684.
106. Zerizer I, Al-Nahhas A, Towey D, et al. The role of early 18F-FDG PET/ 125. Hickey R, Lewandowski RJ. Hepatic radioembolization complicated by
CT in prediction of progression-free survival after 90Y radioembolization: radiation cholecystitis. Semin intervent Radiol 2011; 28:230–233.
comparison with RECIST and tumour density criteria. Eur J Nucl Med 126. Gaba RC, Zivin SP, Dikopf MS, et al. Characteristics of primary and
Mol Imaging 2012; 39:1391–1399. secondary hepatic malignancies associated with hepatopulmonary
107. Piduru SM, Schuster DM, Barron BJ, Dhanasekaran R, Lawson DH, Kim shunting. Radiology 2014; 271:602–612.
HS. Prognostic value of 18f-fluorodeoxyglucose positron emission 127. Wright CL, Werner JD, Tran JM, et al. Radiation pneumonitis following
tomography-computed tomography in predicting survival in patients yttrium-90 radioembolization: case report and literature review. J Vasc
with unresectable metastatic melanoma to the liver undergoing yttrium- Interv Radiol 2012; 23:669–674.
90 radioembolization. J Vasc Interv Radiol 2012; 23:943–948. 128. Wiggins E, Ibrahim SM, Lewandowski RJ, Sato KT, Omary RA, Salem
108. Haug AR, Tiega Donfack BP, Trumm C, et al. 18F-FDG PET/CT R. Effect of chemotherapy on hepatic vasculature in patients under-
predicts survival after radioembolization of hepatic metastases from going Y-90 radioembolization for metastatic disease. J Vasc Interv
breast cancer. J Nucl Med 2012; 53:371–377. Radiol 2008; 19:S48–S49.
109. Memon K, Kulik L, Lewandowski RJ, et al. Alpha-fetoprotein response 129. Murthy R, Eng C, Krishnan S, et al. Hepatic yttrium-90 radioembolo-
correlates with EASL response and survival in solitary hepatocellular therapy in metastatic colorectal cancer treated with cetuximab or
carcinoma treatedwith transarterial therapies: a subgroup analysis. J bevacizumab. J Vasc Interv Radiol 2007; 18:1588–1591.
Hepatol 2012; 56:1112–1120. 130. Salem R, Lewandowski RJ, Atassi B, et al. Treatment of unresectable
110. Senthilnathan S, Memon K, Lewandowski RJ, et al. Extrahepatic hepatocellular carcinoma with use of 90Y microspheres (TheraSphere):
metastases occur in a minority of hepatocellular carcinoma patients safety, tumor response, and survival. J Vasc Interv Radiol 2005; 16:
treated with locoregional therapies: analyzing patterns of progression in 1627–1639.
285 patients. Hepatology 2012; 55:1432–1442. 131. Kennedy AS, Coldwell D, Nutting C, et al. Resin 90Y-microsphere
111. National Cancer Institute. Common Terminology Criteria for Adverse brachytherapy for unresectable colorectal liver metastases: modern
Events (CTCAE) v4.03. NIH Publication No. 09-5410. 2010. Available at: USA experience. Int J Radiat Oncol Biol Phys 2006; 65:412–425.
https://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03_2010-06-14_QuickRe
132. Pitton MB, Kloeckner R, Ruckes C, et al. Randomized comparison of
ference_5x7.pdf. Accessed October 27, 2016.
selective internal radiotherapy (SIRT) versus drug-eluting bead trans-
112. Atassi B, Bangash AK, Lewandowski RJ, et al. Biliary sequelae
arterial chemoembolization (DEB-TACE) for the treatment of hepatocel-
following radioembolization with yttrium-90 microspheres. J Vasc Interv
lular carcinoma. Cardiovasc Intervent Radiol 2014; 38:352–360.
Radiol 2008; 19:691–697.
133. Padia SA, Alessio A, Kwan SW, Lewis DH, Vaidya S, Minoshima
113. Jakobs TF, Saleem S, Atassi B, et al. Fibrosis, portal hypertension, and
S. Comparison of positron emission tomography and bremsstrahlung
hepatic volume changes induced by intra-arterial radiotherapy with
imaging to detect particle distribution in patients undergoing yttrium-90
90yttrium microspheres. Dig Dis Sci 2008; 53:2556–2563.
radioembolization for large hepatocellular carcinomas or associated
114. Sangro B, Gil-Alzugaray B, Rodriguez J, et al. Liver disease induced by
radioembolization of liver tumors: description and possible risk factors. portal vein thrombosis. J Vasc Interv Radiol 2013; 24:1147–1153.
Cancer 2008; 112:1538–1546. 134. Martí-Climent JM, Prieto E, Elosúa C, et al. PET optimization for
115. Kennedy AS, McNeillie P, Dezarn WA, et al. Treatment parameters improved assessment and accurate quantification of 90Y-microsphere
and outcome in 680 treatments of internal radiation with resin 90Y- biodistribution after radioembolization. Med Phys 2014; 41:092503.
microspheres for unresectable hepatic tumors. Int J Radiat Oncol Biol 135. Ray CE, Battaglia C, Libby AM, Prochazka A, Xu S, Funaki B. Interven-
Phys 2009; 74:1494–1500. tional radiologic treatment of hepatocellular carcinoma-a cost analysis
116. Khan W, Sullivan KL, McCann JW, et al. Moxifloxacin prophylaxis for from the payer perspective. J Vasc Interv Radiol 2012; 23:306–314.
chemoembolization or embolization in patients with previous biliary 136. Fink A, Kosefcoff J, Chassin M, Brook RH. Consensus methods:
interventions: a pilot study. AJR Am J Roentgenol 2011; 197: characteristics and guidelines for use. Am J Public Health 1984; 74:
W343–W345. 979–983.
117. Lam MG, Banerjee A, Louie JD, Sze DY. Splenomegaly-associated 137. Leape LL, Hilborne LH, Park RE, et al. The appropriateness of use of
thrombocytopenia after hepatic yttrium-90 radioembolization. Cardio- coronary artery bypass graft surgery in New York State. JAMA 1993;
vasc Intervent Radiol 2014; 37:1009–1017. 269:753–760.
Volume 28 ’ Number 1 ’ January ’ 2017 15

SIR DISCLAIMER
The clinical guidelines of the Society of Interventional Radiology attempt to define principles that generally should
assist in producing high quality medical care. These guidelines are voluntary and are not rules. A physician may
deviate from these guidelines, as necessitated by the individual patient and available resources. These guidelines
should not be deemed inclusive of all proper methods of care or exclusive of other methods of care that are
reasonably directed towards the same result. Other sources of information may be used in conjunction with these
principles to produce a process leading to high quality medical care. The ultimate judgment regarding the conduct of
any specific procedure or course of management must be made by the physician, who should consider all
circumstances relevant to the individual clinical situation. Adherence to the SIR Quality Improvement Program will
not assure a successful outcome in every situation. It is prudent to document the rationale for any deviation from the
suggested guidelines in the department policies and procedure manual or in the patient’s medical record.

You might also like