Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Oct 1.
Published in final edited form as: Semin Radiat Oncol. 2011 Oct;21(4):294–302. doi: 10.1016/j.semradonc.2011.05.004

Radioembolization for primary and metastatic liver cancer

Khairuddin Memon 1, Robert J Lewandowski 1, Laura Kulik 2, Ahsun Riaz 1, Mary F Mulcahy 3, Riad Salem 1,3
PMCID: PMC3221012  NIHMSID: NIHMS314673  PMID: 21939859

Abstract

The incidence of hepatocellular carcinoma is increasing. Most patients present beyond potentially curative options and are usually affected by underlying cirrhosis. In this scenario, trans-arterial therapies, such as radioembolization, are rapidly gaining acceptance as a potential therapy for hepatocellular carcinoma and liver metastases. Radioembolization is a catheter-based liver-directed therapy that involves injection of micron-sized embolic particles loaded with a radioisotope by use of percutaneous transarterial techniques. Cancer cells are preferentially supplied by arterial blood and normal hepatocytes by portal venous blood; radioembolization therefore specifically targets tumor cells with a high dose of lethal radiation and spares healthy hepatocytes.

The antitumor effect mostly comes from radiation rather than embolization. The most commonly used radioisotope is Yttrium-90. The commercially available devices are TheraSphere® (glass-based) and SIR-Sphere® (resin-based). The procedure is performed on outpatient basis. The incidence of complications is generally less than other locoregional therapies and may include nausea, fatigue, abdominal pain, hepatic dysfunction, biliary injury, fibrosis, radiation pneumonitis, gastrointestinal ulcers and vascular injury. However, these can be avoided by meticulous pretreatment assessment, careful patient selection and adequate dosimetry. This article focuses on both the technical and clinical aspects of radioembolization with emphasis on patient selection, uses and complications.

2. Introduction

Hepatocellular carcinoma (HCC) is the sixth most common malignancy worldwide and the third most common cause of cancer-related mortality 1,2. The incidence of HCC has tripled in the USA from 1975-2005 3. Metastatic disease to the liver is the most common form of hepatic malignancy 4. Common tumors that metastasize to liver include colorectal carcinoma, breast carcinoma and neuroendocrine tumors. Surgical interventions (resection/liver transplantation) provide curative options for selected patients, but many patients are precluded from these treatment 5,6.

Given this background, novel liver-directed loco-regional therapies have been investigated for the treatment of primary and metastastic liver cancers. Among these are image-guided endovascular therapies including chemoembolization and radioembolization. Radioembolization is defined as the injection of micron-sized embolic particles loaded with a radioisotope by use of percutaneous transarterial techniques in order to deliver high focal doses of radiation to cancers. In HCC, chemoembolization and radioembolization can be used to delay disease progression, downstage or bridge HCC in order to permit liver transplantation 7, or as palliative therapies. Chemoembolization is considered as a standard of care in HCC; radioembolization with Yttrium-90 has regulatory approval for HCC. Randomized controlled trials comparing chemoembolization and radioembolization are underway. Recently, a comparative effectiveness study of radioembolization versus chemoembolization in a cohort of 123 patients (without portal venous thrombosis and/or extrahepatic metastases at baseline) treated with radioembolization and 122 with chemoembolization was published 8. The investigators concluded that radioembolization leads to longer time-to-progression and reduced toxicity with similar survival outcomes when compared with chemoembolization.

Herein, we discuss radioembolization as a potential therapy for hepatocellular carcinoma and liver metastases. Our discussion incorporates both clinical and technical aspects of this therapy with emphasis on appropriate patient selection, indications, clinical efficacy and complications.

3. Radioembolization: Technical Aspects

3.1 Vascular anatomy of the Liver

Given that radioembolization is hepatic arterial directed therapy, special attention to vascular anatomy, reviewed here, is required. The liver has a dual blood supply (portal vein and hepatic artery). The portal vein, which carries venous blood from the gastrointestinal tract to the liver is the predominant source of vascular supply for hepatocytes. The hepatic artery proper is a branch of the common hepatic artery which itself arises from the celiac trunk. This branch divides into the right and left which supply their corresponding hepatic lobes. The right hepatic artery also gives off the cystic artery supplying the gall bladder. Interestingly, normal hepatocytes are primarily supplied by portal blood whereas cancer cells are supplied primarily by the hepatic artery 9-12. This variable blood supply is exploited by intra-arterial therapies and forms the basis for prioritizing the delivery of cytotoxic therapy to the capillary bed of tumor cells via the hepatic arterial route while limiting the exposure to normal hepatocytes. Given this fact, very high doses of radiation can be selectively administered to hepatic tumors, with little dose to the surrounding liver.13

3.2 Available Radioisotopes

3.2.1 Yttrium-90

90Y is the most commonly used radioisotope used for radioembolization. It is a pure beta emitter with a half-life of 64.2 hours. It decays into the stable element Zirconium-90. The range of tissue penetration of the emissions is 2.5 to 11 mm. It is the most commonly used radionuclide for the treatment of both primary and secondary liver malignancies. There are two forms available (TheraSphere and SIR-Spheres).

TheraSphere® (MDS Nordion, Ottawa, Canada) consists of nonbiodegradable glass microspheres that have a diameter between 20 and 30 microns. It was approved by the FDA in 1999 as a bridge to transplantation 14 and recently has been approved for use in HCC patients with PVT. Six activity vials are available differing from each other only in the number of spheres per vial e.g. 1.2 million microspheres are present in a vial with an activity of 3 Gigabecquerel (GBq). Each microsphere has an activity of 2500 Becquerel at the time of calibration. The activity of the vial varies inversely with the time elapsed after calibration.

SIR-Spheres® (Sirtex, Lane Cove, Australia) consist of biodegradable resin microspheres. The spheres are slightly larger and less dense than TheraSphere® and hence associated with more embolic effect. One vial of SIR-Spheres® of 3 GBq is available and contains 40-80 million microspheres ranging from 20 to 60 microns. SIR-Spheres® was approved by the FDA for metastatic colorectal cancer to the liver. Each microsphere has a specific activity of 50 Bq at the time of calibration.

3.2.2 Iodine-131 Labeled Iodized Oil (I-131 Lipiodol)

I-131 is a beta and gamma emitter. The iodine moiety of lipiodol can be substituted for the radionuclide, 131I. It is used exclusively for the treatment of HCC. It has a half-life of 8 days. The thyroid gland, due to its use of iodine to make thyroid hormones, is susceptible to the toxicity of 131I. Therefore, the thyroid gland is blocked before and after the treatment to minimize toxicity. The radionuclide is excreted in urine and thus the patient has to be hospitalized for approximately six days after treatment as a radioactivity safety measure. Patients are told to refrain from possible conception until six months after treatment. The use of 131I-Lipiodol has been studied in a prospective randomized trial in 142 patients where it was compared to chemoembolization. Overall survival rates at 6 months, 1, 2, 3, and 4 years were 69%, 38%, 22%, 14%, and 10%, and 66%, 42%, 22%, 3%, and 0% in the I-131 labeled Lipiodol and chemoembolization groups, respectively. Complete response in 1 and 0 patients and partial response in 15 and 16 patients were noted in the I-131 labeled Lipiodol and chemoembolization groups, respectively. The incidence of adverse effects was low in I-131 group versus chemoembolization group (3 severe side effects vs. 29, P<0.001; 1 arterial thrombosis vs. 10, P<0.01) 15.

3.2.3 Rhenium-188 HDD Labeled Iodized Oil

Transarterial radionuclide therapy (TART) with Rhenium-188 (188Re) 4-hexadecyl-1, 2, 9, 9-tetramethyl-4, 7-diaza-1, 10-decanethiol (HDD)-labeled iodized oil has been recently studied for use in inoperable HCC. 188Re has a shorter half-life (16.9 hours), high beta energy (maximum: 2.1 MeV) and low gamma energy (155 keV) emissions, and is available through a Tungsten-188 (188W-188Re) generator 16. The quantity of 188Re HDD iodized oil (lipiodol) administered is based on the radiation absorbed dose (RAD) to critical organs, which is calculated after administration of test dose of the radioconjugate, transarterially 16. The incidence of serious adverse events is low.

3.2.4 Milican/Holmium-166 Microspheres (HoMS)

Holmium-166 is used in a complex with Chitosan, which is a unique substance derived from chitin. It has the ability to liquefy in an acidic environment and form a gel in basic environments. The gel has embolic effects. Holmium-166 has a half-life of 26.8 hours and emits both a beta particle for therapy and a gamma that can be used for imaging. The use of Holmium/chitosan complex (Milican, Dong Wha Pharmaceutical Co., Seoul, Korea) has been shown to be effective in treating small HCCs in a novel study based on 40 patients with single HCC <3cm in size from Korea17 with satisfactory response rates and survival rates of 87.2%, 71.8%, and 65.3% at 1,2 and 3 years respectively. Two patients had transient bone marrow depression requiring hospitalization.

Since 90Y is the most commonly used radioisotope, it will be the topic of discussion for the remaining portion of this manuscript.

3.3 Pretreatment Evaluation

All patients undergo pretreatment assessment consisting of history, laboratory and imaging work-up. Pretreatment cross sectional imaging is essential for treatment planning and post treatment response assessment. All patients undergo the following essential pretreatment evaluation procedures approximately 1 week before the planned first treatment. They are not usually repeated for any subsequent treatments.

3.3.1 Pretreatment Angiography

Given the high propensity for arterial variants and hepatic tumors to exhibit arteriovenous shunting, all patients being evaluated for 90Y must undergo pretreatment mesenteric angiography 10-12,14. This permits tailoring of the treatment plan according to each patient's individual anatomy and helps assess for the possibility of any inadvertent spread of the microspheres to non-target organs18. This can be mitigated by prophylactic embolization of aberrant vessels to non-hepatic targets.

An aortogram is necessary to assess the tortuosity and the presence of atherosclerosis in the aorta; it also permits optimal base catheter selection. The superior mesenteric and celiac trunk angiograms permit the interventional radiologists an opportunity to study the hepatic vascular anatomy. The patency of the portal vein and the presence of arterio-portal shunting are also assessed. Prophylactic embolization of the gastroduodenal artery and right gastric artery is recommended as a safe and efficacious mode of minimizing the risks of hepato-enteric flow since this can lead to inadvertent deposition of microspheres in the gastrointestinal tract causing severe ulcers that are highly symptomatic and difficult to manage 10,19,20. Other vessels that need to be interrogated and potentially embolized are the falciform, inferior esophageal, left inferior phrenic, accessory left gastric, supraduodenal and retroduodenal arteries.

Diagnostic angiography is essential to ensure that the blood supply to the tumor(s) has been adequately identified as incomplete identification of the blood supply to the tumor may lead to incomplete targeting and treatment. This facilitates accurate calculations of target volumes21.

3.3.2 Pulmonary Shunting and Technetium-99m labeled macroaggregated albumin (99mTc-MAA) scan

In contrast to metastatic tumours, one of the angiographic features of HCC is the direct arteriovenous shunting bypassing the capillary bed to the liver22; shunting of 90Y microspheres to lungs therefore becomes a concern as this could result in radiation pneumonitis23. Because the size of 99mTc-MAA particle closely mimics 90Y, it is assumed that the distribution of the two will be identical; this concept is utilized in assessing splanchnic and pulmonary shunting. It is important to correlate the findings of angiography to the findings of the 99mTc-MAA scan as the proximity of some portions of the gastrointestinal tract to the liver may confuse the findings of nuclear medicine scans. Lung shunt fraction (LSF) is used to calculate the dose delivered to the lungs and appropriate adjustment for this parameter minimizes the risk of radiation pneumonitis. If LSF is deemed to be high, appropriate reduction is made in the overall dose administered.

3.4 Dose Calculation

3.4.1 Dose Calculation for TheraSphere®

The volume of the liver to be infused in cubic centimeters is calculated using 3-dimensional reconstruction through commercially available software. This value is used to calculate the mass of infused liver tissue in grams by multiplying it by a factor of 1.03 mg/cc. The activity administered to the target volume of the liver (A) in GBq, assuming uniform distribution of microspheres, is calculated using the following formula:

A=D×m/50

Where D is the dose administered in Gray (Gy) and m is the mass in kilograms (kg).

Dose delivered to the treated mass also depends on the percent residual activity (R) in the vial after treatment and the LSF which is calculated beforehand using the 99mTc-MAA scan. These factors are accounted for in the following formula,

D=A×50×(1-LSF)×(1-R)/m

3.4.2 Dose Calculation for SIR-Spheres®

The dosimetry model for SIR-Spheres® is based on whole liver infusion. The calculated activity in GBq of the whole liver is multiplied by the ratio of the target volume to the whole liver volume. Three methods for dosimetry of SIR-Spheres® are recommended by the manufacturers. The partition method is seldom used as it is applicable only in special circumstances. The simple empiric method is based on percent tumor involvement of liver and recommends doses of 2 GBq for ≤25% involvement, 2.5 GBq for 25-50% involvement and 3 GBq for >50% involvement.

The most widely used body surface area (BSA) method is calculated as follows,

A=BSA0.2+(%tumor burden/100)

Where A is the activity in GBq, BSA is the body surface area in meters squared (m2), and % tumor burden is the percentage of the liver that is involved by tumor. There is often a 20% dose reduction factor that is applied in order to reduce the incidence of radiation pneumonitis. The issue of LSF is not commonly associated with metastatic disease and hence dose reduction is usually applied only for HCC. Moreover, the dose calculation is independent of tumor burden.

4. Radioembolization: Clinical Aspects

4.1 Patient Selection

All patients undergo clinical evaluation which includes history, physical examination, laboratory profile including liver function and detailed radiological imaging to establish the extent of disease. A multidisciplinary team consisting of professionals from interventional radiology, hepatology, medical, surgical and radiation oncology, transplant surgery and nuclear medicine is involved in selecting suitable patients for radioembolization. Patients are selected according to following criteria:

4.1.1 Inclusion criteria

(a) confirmed diagnosis of surgically unresectable HCC or intrahepatic cholangiocarcinoma (ICC) or metastatic disease, (b) age >18 years, (c) Eastern Cooperative Oncology Group (ECOG) performance status ≤ 2, (d) adequate pulmonary function test findings, (e) adequate hematologic parameters (granulocyte count >1.5 × 109/L, platelet count >50 × 109/L), renal function (serum creatinine level <2.0 mg/dL) and liver function (serum bilirubin level <3.0 mg/dL) and, (f) ability to undergo angiography and selective visceral catheterization 24,25. Most commonly, patients with Child-Pugh score ≤7 are eligible for radioembolization; however, Child-Pugh score > 7 is not an absolute contraindication.

4.1.2 Exclusion criteria (Contraindications)

(a) any other liver therapy planned for cancer treatment, (b) uncorrectable flow to the gastrointestinal tract, (c) estimated radiation doses to the lungs greater than 30 Gy in a single administration or 50 Gy in multiple administrations, and (d) significant extrahepatic disease representing imminent life-threatening outcome.

4.2 90Y Radioembolization Procedure

The procedure is carried out according to previously published guidelines21,26. The administrating device for the administration of 90Y is designed to minimize radiation exposure to persons involved in the procedure. The tumor is approached under fluoroscopic guidance; the activity vial is injected into the vessel feeding the tumor. Tumor distribution guides for the selectivity of the treatment i.e. to one or more lobes/segments as required. A physicist is present throughout the case to ensure that proper protocols are followed to minimize accidental radiation exposure. Radioembolization is performed as an outpatient procedure and patients are discharged 4-6 hours after treatment 14. Due to the possibility of GI ulcers, prophylactic proton pump inhibitors are advised for a period of 14 days.

4.3 Post-treatment Assessment

Clinical, laboratory and radiological follow-up is essential to monitor response to treatment and identify any toxicity. Regular laboratory follow-up includes the hepatic panel and tumor markers. Cross sectional imaging is performed one month post-treatment and then every three months to assess response to treatment or progression of disease.

4.4 Response Assessment Following Radioembolization

4.4.1 Imaging Following Radioembolization

Changes in tumor size using World Health Organization (WHO) and Response Evaluation Criteria in Solid Tumors (RECIST) guidelines, and amount of enhancing tissue by European Association for the Study of the Liver (EASL) guidelines are used to assess tumor response to therapy following radioembolization 27-29.

In a recent series of 245 patients treated with locoregional therapies, Riaz et al investigated the concept of the Index lesion and inter-method agreement between RECIST, WHO and EASL guidelines. They concluded that there is high agreement between RECIST and WHO guidelines but low between each of these and EASL. Moreover, the primary index lesion (the dominant lesion) can be used to measure response to therapy by applying the above mentioned guidelines30. In another series of 35 patients who underwent radioembolization, Riaz et al correlated post-treatment radiologic findings to pathologic data 31. They concluded that changes in amount of enhancing tissue and size of a tumor following therapy are predictors of complete pathologic necrosis. In a recent study, investigators compared various combinations of WHO, EASL and RECIST guidelines and devised various scoring systems based on the combinations of these guidelines. They concluded that EASL×WHO scoring system provides a simple and clinically acceptable method of response assessment and radiological-pathological correlation 32.

Rhee et al concluded that HCC tumor response using diffusion-weighted imaging changes after Yttrium-90 radioembolization (90Y) preceded anatomic size changes. Conventional anatomic imaging studies are not able to assess tumor response until six weeks have elapsed after treatment and functional MRI may have a role in earlier detection of tumor response 33.

In addition to conventional cross-sectional imaging such as CT and MRI, positron emission tomography (PET) has a role in assessing response to treatment for secondary liver tumors.

4.4.2 Using Tumor Markers as Biomarkers

Riaz et al investigated the use of AFP in locoregional therapies; they concluded that AFP response seen after transarterial locoregional therapy could be used as an ancillary method of assessing tumor response and potential surrogate for survival 34. Although utilized clinically, the benefit of other tumor markers such as carcinoembryonic agent and CA 19-9 in assessing response following radioembolization of secondary liver tumors have not been studied.

4.5 Indications for Radioembolization

4.5.1 Hepatocellular carcinoma (HCC)

The use of radioembolization has been shown to limit progression of the disease in HCC. This helps in bridging the patient to transplant as it allows the patient more time to wait for donor organs 35. Lewandowski et al compared chemoembolization to radioembolization in their retrospective analysis in patients with HCC beyond Milan criteria 36. Radioembolization was shown to be a better tool than chemoembolization for downstaging the disease to within transplant criteria. In a recent study analyzing the long-term outcomes of patients treated with 90Y for HCC, investigators concluded that patients with Child-Pugh A class disease, with or without PVT, benefited most from the treatment, and patients with Child-Pugh B class disease who had PVT had poor outcomes. Time-to-progression was higher for Child-Pugh A and B patients without PVT (15.5 and 13 months respectively) than those with PVT (5.6 and 5.9 months respectively). Overall survival for patients with Child–Pugh A and B disease was: CP-A, 17.2 months; CP-B, 7.7 months; P = .002) Time-to-progression and overall survival varied by patient stage at baseline 37.

Malignant vascular invasion in patients with advanced HCC is an exclusion criterion for transplantation. Embolic therapies are also relatively contraindicated as they may lead to further deterioration of blood supply to already compromised liver parenchyma. However, patient with vascular invasion have shown a survival benefit with 90Y as it is not a macro-embolic procedure 38.

Kooby et al have compared radioembolization to chemoembolization in a series of retrospectively studied patients and conclude that radioembolization and chemoembolization have similar effectiveness and safety profiles 39. In a similar analysis, Carr et al performed a retrospective study of North American patients who had unresectable HCC 40. They concluded that radioembolization and chemoembolization are equivalent locoregional therapies for patients with unresectable and non-metastatic HCC.

In a recent comprehensive comparison analysis, Salem et al compared 123 patients treated with radioembolization and 122 patients with chemoembolization. They concluded that radioembolization leads to longer time-to-progression and less toxicity than chemoembolization with similar survival times 8.

4.5.2. Intrahepatic Cholangiocarcinoma (ICC)

A pilot study analyzing the use of 90Y in 24 patients with biopsy proven ICC has shown a favorable response to treatment and survival outcomes 41. Patients with better performance status according to the Eastern Cooperation Oncology Group (ECOG) criteria had a significantly better survival in this study. In a recent series of 25 patients with unresectable intrahepatic cholangiocarcinoma, Saxena et al studied the safety and efficacy of 90Y radioembolization. They concluded that 90Y may be a safe and efficacious treatment for unresectable ICC with a median survival of 9.3 months and low incidence of grade 3 toxicities. Further investigations are underway 42.

4.5.3 Secondary liver tumors

Colorectal Carcinoma (CRC)

Radioembolization is employed to treat patients with unresectable hepatic CRC metastases. In a phase III randomized trial of 74 patients with CRC liver confined metastases, systemic chemotherapy alone was compared to the combination of radioembolization and systemic chemotherapy; the combination was shown to have significantly better tumor response (44% vs. 17.6%, P = 0.01), a longer time to progression (15.9 vs. 9.7, P = 0.001), similar survival outcome(72%, 39%, 17% and 3.5%, compared to 68%, 29%, 6.5% and 0% and 1,2,3 and 5 years respectively) and an acceptable safety profile 43. Studies have shown a better response with increasing doses; a study showed that doses >95 Gy (median) reduced mortality by 50% and were associated with odds of tumor response of 3.1 times greater than the odds of tumor response for doses ≤95 Gy. 44.

In a series of 72 patients with unresectable hepatic colorectal metastases, Mulcahy et al investigated the safety and efficacy of 90Y radioembolization. They observed that grade III and IV bilirubin toxicities were seen in 12.6% of patients. The CT response rate was 40.3%; PET response rate was 77%, and the median response duration and time to hepatic progression was 15 and 15.4 months respectively. Overall survival was 14.5 months from first treatment date. They concluded that 90Y is a safe and efficacious therapy for colorectal metastases 45.

In a recent multi-center phase II trial, Cosimelli et al found that radioembolization produced meaningful responses and disease stabilization in patients with advanced, unresectable and chemorefractory metastatic CRC. By RECIST criteria, complete response, partial response, stable disease and progressive disease were noted in 22%, 24%, 44% and 8% patients respectively. Median overall survival was 12.6 months. 46

Sharma et al performed a phase I study analyzing the combination of radioembolization with modified FOLFOX4 systemic chemotherapy in patients with unresectable CRC metastases in the liver in a series of 20 patients with the primary endpoint of toxicity 47. Five patients experienced grade 3 abdominal pain (two of whom had microsphere-induced gastric ulcers); grade 3 or 4 neutropenia was recorded in 12 patients; one episode of transient grade 3 hepatotoxicity was noted. Partial responses were demonstrated in 18 patients and stable disease in two patients. Median progression-free survival was 9.3 months, and median time-to-progression in the liver was 12.3 months. They concluded that this chemo-radiation regimen merits investigation in a phase II-III trial.

In a recent multicenter phase III randomized trial of 46 patients with unresectable liver-limited metastatic colorectal cancer, Hendlisz et al compared intravenous fluorouracil alone (arm A) with combination of radioembolization and intravenous fluorouracil (arm B). Median overall survival was 7.3 and 10.0 months in arms A and B respectively (P = 0.80). Median time to tumor progression (TTP) was 2.1 and 4.5 months, respectively (P = 0.03). Grade 3 or 4 toxicities were recorded in six patients after FU monotherapy and in one patient after radioembolization plus FU treatment (P = 0.10). They concluded that combination therapy is well tolerated and significantly improves time-to-progression compared with fluorouracil alone, and that this procedure is a valid therapeutic option for chemotherapy-refractory liver metastases from CRC 48. Due to low accrual and follow up longer than planned, the trial was closed earlier with 46 patients included in the study instead of the anticipated 58 patients required to obtain estimated 90% power.

Neuroendocrine Tumors

Patients with unresectable metastatic neuroendocrine tumors have also been treated with radioembolization, with limited toxicity and prolonged responses noted. A 148 patient analysis by Kennedy reported response rates of 63.2% with survival of 70 months 49. In a multicenter phase II study with 42 patients of hepatic neuroendocrine tumor metastases, Rhee et al investigated the efficacy and safety of 90Y radioembolization. They observed that 92% and 94% of patients treated with glass and resin microspheres respectively showed either partial response or stable disease at 6 months and median survival was 22 and 28 months respectively. Complete response was not observed in any patient. Grade 3 toxicities were recorded in 6 patients and included bilirubin toxicity (n=1), ALT toxicity (n=1), AST toxicity (n=1) and ALP toxicity (n=3). They concluded that 90Y is a viable therapy with acceptable toxicity for hepatic metastases of neuroendocrine tumors 50.

Other Metastases

90Yhas been used to treat patients with other primary cancers metastatic to liver with variable results. Since not established in these tumor types, we recommend that it be limited to use in patients unsuitable for standard systemic therapies or those who have become chemorefractory 51.

Breast cancer has a tendency to metastasize to the liver. However, a survival benefit from this treatment in this patient population has not been established. Bangash et al investigated 90Y radioembolization in 27 patients with progressing liver metastases of breast cancer on standard polychemotherapy. The response rate was 39.1%; stable and progressive disease was seen in 52.1% and 8.8% respectively; response on PET was noted in 63%. Median survival was 6.8 and 2.6 months in patients with ECOG 0 vs. 1, 2 and 3. 11% of patients showed grade III bilirubin toxicity. They concluded that 90Y may be a viable option for treating patients with breast cancer metastases to liver who have progressed on standard polychemotherapy 52. Coldwell et al investigated the use of 90Y microspheres in the treatment of unresectable chemorefractory liver metastases from breast cancer in 44 patients in a multi-institutional study. No treatment-related procedure deaths or liver toxicity was seen. Computed tomographic imaging partial response was 47% and positron emission tomographic response 95%. Patients without PET or CT response had a median survival of 3.6 months. Survival was longer for responders and patients with slowly progressing disease; however, median survival was not reached with a follow up of 14 months. 53.

4.6 Complications and Toxicities

The post-radioembolization syndrome (PRS) consists of the following clinical symptoms: fatigue, nausea, vomiting, anorexia, fever, abdominal discomfort, and cachexia. However, it occurs less commonly after radioembolization due to the small particle size and microembolic effect 54-56. Serious adverse events related to radioembolization are explained below:

4.6.1 Hepatobiliary dysfunction

Radiation induced liver disease (RILD) usually occurs between four to eight weeks after radioembolization; its incidence varies from 0%-4%. Classic and non-classic RILD may be seen following radioembolization. Ascites and jaundice are associated with an increased risk of ultimate liver failure 57. The presence of factors such as abnormal hepatic functions at baseline, increased age and activity delivered may influence the risk of patients to RILD. It has been shown that liver radiation dose to a maximum of 150 Gy for a single administration is associated with increased risk of liver toxicities. The biochemical toxicity rates following radioembolization have ranged between 15-20% 57,58.

The incidence of biliary sequelae after radioembolization is reported to be <10%. According to Atassi et al, less than 2% of patients required intervention for the biliary toxicity induced by radioembolization 33. These included drainage of three bilomas, one abscess and two cholecystectomies. Radiation-induced cholangitis has also been reported 33.

In the long-term, radioembolization has been shown to cause liver fibrosis, resulting in the contraction of the hepatic parenchyma and portal hypertension radiologically. Despite the imaging findings indicative of portal hypertension, the clinically significant occurrence of portal hypertension is low, 59 as clinically relevant manifestations such as reduced platelet counts (<100,000/dL) or variceal bleeding are rarely seen. It is recommended to observe for radiologic and clinical evidence of portal hypertension routinely, as this is not an acute process 60. 61. This finding is more commonly seen with bilobar treatment and its incidence is increased in patients who have chemotherapy associated steatohepatitis. The presence of preexisting cirrhosis leading to portal hypertension in most HCC patients makes them more susceptible to the aggravation of this complication.

4.6.2 Radiation Pneumonitis

The incidence of radiation pneumonitis is less than 1% if standard dosimetry protocols are followed 62. Caution has to be taken when the LSF is >13% and resin microspheres are used 63. A restrictive pulmonary dysfunction may be seen after radioembolization in a few cases with a predisposing high LSF. The LSF is used to calculate the dose that will be administered to the lung; patients with elevated LSF resulting in a delivery of >30 Gy in a single session or >50 Gy cumulatively over multiple sessions are contraindicated from receiving radioembolization64.

4.6.3 Gastrointestinal Complications

Gastrointestinal complications after radioembolization have been reported; the incidence is <5% when proper techniques are used i.e. slow and controlled injection of microspheres and prophylactic coil embolization of vessels to prevent deposition of microspheres in the GI tract.65. The inadvertent deposition of microspheres to the gastrointestinal tract may result in ulceration 65,66. The pathophysiology behind this complication is the ectopic distribution of radioembolic microspheres into the lining of the gastrointestinal tract. Severe epigastric pain after treatment should be aggressively managed; early intervention may prevent serious complications. As opposed to a normal ulcer that develops at the mucosal surface, 90Y-induced ulcers originate from the serosal surface.

4.6.4 Vascular Injury

Radioembolization is an invasive procedure. The incidence of vascular injury is very low and is mostly seen in patients previously exposed to systemic chemotherapy 67. This might cause increased fragility of the vessel wall leading to a susceptibility to injury (spasm, dissection). These adverse effects occur in <1% of patients and are related to technique/previous chemotherapy rather than the dose of microspheres.

5. Conclusion

Radioembolization is a novel tans-arterial locoregional therapy that is gaining recognition as a treatment option for primary and metastastic liver cancers. The incidence of complications is low and can be further reduced by rigorous pretreatment assessment and careful patient selection i.e. patients with preserved liver function without ascites or encephalopathy, Child-Pugh score ≤7, hypervascular tumors and low lung shunt fraction. Randomized comparative studies with other therapies are lacking; however a comparative effectiveness study with chemoembolization has been recently published, suggesting longer time-to-progression and reduced toxicities for patients treated with radioembolization (Table) 8. Additional studies will be required to evaluate the combination of radioembolization with other locoregional and systemic therapies in order to enhance the treatment options for patient with liver cancer further.

Table 1. Summary of few landmark studies.

Salem et al Hilgard et al Salem et al Kennedy et al Hendlisz et al
Purpose To assess clinical outcomes of HCC patients treated with 90Y To assess clinical outcomes of HCC patients treated with 90Y in Europe To compare the effectiveness of 90Y vs. TACE in HCC patients To assess 90Y as a salvage therapy for unresectable colorectal liver metastases Prospective randomized phase III trial comparing FU alone (arm A) with 90Y+ FU (arm B) for unresectable CRC metastases
Disease HCC HCC HCC CRC metastases CRC metastases
Patients 291 108 90Y: 123
TACE: 122
208 Arm A: 23
Arm B: 21
Response rate WHO: 42%
EASL: 57%
EASL: 40% 90Y: 49%
TACE: 36%
CT: 35%
PET: 91%
CAE: 70%
-
TTP (months) 7.9 10 90Y: 13.3
TACE: 8.4
- Arm A: 2.1
Arm B: 4.5
Survival (months) Child-Pugh A:17.2
Child-Pugh B:7.7
16.4 90Y:20.5
TACE: 17.4
Responders:10.5
Nonresponders: 4.5
Arm A: 7.3
Arm B: 10

Acknowledgments

Role of Funding: There was no funding provided for this monograph. Riad Salem is supported in part by NIH grant CA126809.

Footnotes

Conflict of Interest: Riad Salem is advisor to MDS Nordion. None of the other authors have any conflict of interest.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Parkin DM, Bray F, Ferlay J, Pisani P. Global cancer statistics, 2002. CA Cancer J Clin. 2005 Mar-Apr;55(2):74–108. doi: 10.3322/canjclin.55.2.74. [DOI] [PubMed] [Google Scholar]
  • 2.Bosch FX, Ribes J, Diaz M, Cleries R. Primary Liver cancer: worldwide incidence and trends. Gastroenterology. 2004;127(S5-S16) doi: 10.1053/j.gastro.2004.09.011. [DOI] [PubMed] [Google Scholar]
  • 3.Alterkruse SF, M K, Reichman ME. Hepatocellular carcinoma incidence, mortality, and survival benefits in the United States From 1975 to 2005. J Clin Oncol. 2009;27:1485–1491. doi: 10.1200/JCO.2008.20.7753. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Lewandowski RJ, Thurston KG, Goin JE, et al. 90Y microsphere (TheraSphere) treatment for unresectable colorectal cancer metastases of the liver: response to treatment at targeted doses of 135-150 Gy as measured by [18F]fluorodeoxyglucose positron emission tomography and computed tomographic imaging. J Vasc Interv Radiol. 2005 Dec;16(12):1641–1651. doi: 10.1097/01.RVI.0000179815.44868.66. [DOI] [PubMed] [Google Scholar]
  • 5.Yao FY, Bass NM, Nikolai B, et al. A follow-up analysis of the pattern and predictors of dropout from the waiting list for liver transplantation in patients with hepatocellular carcinoma: implications for the current organ allocation policy. Liver Transpl. 2003 Jul;9(7):684–692. doi: 10.1053/jlts.2003.50147. [DOI] [PubMed] [Google Scholar]
  • 6.Maddala YK, Stadheim L, Andrews JC, et al. Drop-out rates of patients with hepatocellular cancer listed for liver transplantation: outcome with chemoembolization. Liver Transpl. 2004 Mar;10(3):449–455. doi: 10.1002/lt.20099. [DOI] [PubMed] [Google Scholar]
  • 7.Mazzaferro V, Regalia E, Doci R, et al. Liver transplantation for the treatment of small hepatocellular carcinomas in patients with cirrhosis. N Engl J Med. 1996 Mar 14;334(11):693–699. doi: 10.1056/NEJM199603143341104. [DOI] [PubMed] [Google Scholar]
  • 8.Salem R, Lewandowski RJ, Kulik L, et al. Radioembolization Results in Longer Time-to-Progression and Reduced Toxicity Compared With Chemoembolization in Patients With Hepatocellular Carcinoma. Gastroenterology. 2011 Feb;140(2):497–507. e492. doi: 10.1053/j.gastro.2010.10.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Breedis C, Young G. The blood supply of neoplasms in the liver. Am J Pathol. 1954 Sep-Oct;30(5):969–977. [PMC free article] [PubMed] [Google Scholar]
  • 10.Liu DM, Salem R, Bui JT, et al. Angiographic considerations in patients undergoing liver-directed therapy. J Vasc Interv Radiol. 2005 Jul;16(7):911–935. doi: 10.1097/01.RVI.0000164324.79242.B2. [DOI] [PubMed] [Google Scholar]
  • 11.Lewandowski RJ, Sato KT, Atassi B, et al. Radioembolization with (90)y microspheres: angiographic and technical considerations. Cardiovasc Intervent Radiol. 2007 Jul-Aug;30(4):571–592. doi: 10.1007/s00270-007-9064-z. [DOI] [PubMed] [Google Scholar]
  • 12.Salem R, Lewandowski RJ, Sato KT, et al. Technical aspects of radioembolization with 90Y microspheres. Tech Vasc Interv Radiol. 2007 Mar;10(1):12–29. doi: 10.1053/j.tvir.2007.08.001. [DOI] [PubMed] [Google Scholar]
  • 13.Riaz A, Gates VL, Atassi B, et al. Radiation segmentectomy: a novel approach to increase safety and efficacy of radioembolization. Int J Radiat Oncol Biol Phys. 2011 Jan 1;79(1):163–171. doi: 10.1016/j.ijrobp.2009.10.062. [DOI] [PubMed] [Google Scholar]
  • 14.Salem R, Thurston KG, Carr BI, Goin JE, Geschwind JF. Yttrium-90 microspheres: radiation therapy for unresectable liver cancer. J Vasc Interv Radiol. 2002 Sep;13(9 Pt 2):S223–229. doi: 10.1016/s1051-0443(07)61790-4. [DOI] [PubMed] [Google Scholar]
  • 15.Raoul JL, Guyader D, Bretagne JF, et al. Prospective randomized trial of chemoembolization versus intra-arterial injection of 131I-labeled-iodized oil in the treatment of hepatocellular carcinoma. Hepatology. 1997 Nov;26(5):1156–1161. doi: 10.1002/hep.510260511. [DOI] [PubMed] [Google Scholar]
  • 16.Kumar A, Srivastava DN, Chau TT, et al. Inoperable hepatocellular carcinoma: transarterial 188Re HDD-labeled iodized oil for treatment--prospective multicenter clinical trial. Radiology. 2007 May;243(2):509–519. doi: 10.1148/radiol.2432051246. [DOI] [PubMed] [Google Scholar]
  • 17.Kim JK, Han KH, Lee JT, et al. Long-term clinical outcome of phase IIb clinical trial of percutaneous injection with holmium-166/chitosan complex (Milican) for the treatment of small hepatocellular carcinoma. Clin Cancer Res. 2006 Jan 15;12(2):543–548. doi: 10.1158/1078-0432.CCR-05-1730. [DOI] [PubMed] [Google Scholar]
  • 18.Covey AM, Brody LA, Maluccio MA, Getrajdman GI, Brown KT. Variant hepatic arterial anatomy revisited: digital subtraction angiography performed in 600 patients. Radiology. 2002 Aug;224(2):542–547. doi: 10.1148/radiol.2242011283. [DOI] [PubMed] [Google Scholar]
  • 19.Murthy R, Nunez R, Szklaruk J, et al. Yttrium-90 microsphere therapy for hepatic malignancy: devices, indications, technical considerations, and potential complications. Radiographics. 2005 Oct;25 1:S41–55. doi: 10.1148/rg.25si055515. [DOI] [PubMed] [Google Scholar]
  • 20.Cosin O, Bilbao JI, Alvarez S, de Luis E, Alonso A, Martinez-Cuesta A. Right gastric artery embolization prior to treatment with yttrium-90 microspheres. Cardiovasc Intervent Radiol. 2007 Jan-Feb;30(1):98–103. doi: 10.1007/s00270-006-0028-5. [DOI] [PubMed] [Google Scholar]
  • 21.Salem R, Thurston KG. Radioembolization with 90Yttrium Microspheres: A State-of-the-Art Brachytherapy Treatment for Primary and Secondary Liver Malignancies: Part 1: Technical and Methodologic Considerations. J Vasc Interv Radiol. 2006 Aug;17(8):1251–1278. doi: 10.1097/01.RVI.0000233785.75257.9A. [DOI] [PubMed] [Google Scholar]
  • 22.Chen JH, Chai JW, Huang CL, Hung HC, Shen WC, Lee SK. Proximal arterioportal shunting associated with hepatocellular carcinoma: features revealed by dynamic helical CT. AJR Am J Roentgenol. 1999 Feb;172(2):403–407. doi: 10.2214/ajr.172.2.9930792. [DOI] [PubMed] [Google Scholar]
  • 23.Ho S, Lau WY, Leung TW, et al. Partition model for estimating radiation doses from yttrium-90 microspheres in treating hepatic tumours. Eur J Nucl Med. 1996 Aug;23(8):947–952. doi: 10.1007/BF01084369. [DOI] [PubMed] [Google Scholar]
  • 24.Wong CY, Salem R, Raman S, Gates VL, Dworkin HJ. Evaluating 90Y-glass microsphere treatment response of unresectable colorectal liver metastases by [18F]FDG PET: a comparison with CT or MRI. Eur J Nucl Med Mol Imaging. 2002 Jun;29(6):815–820. doi: 10.1007/s00259-002-0787-4. [DOI] [PubMed] [Google Scholar]
  • 25.Wong CY, Salem R, Qing F, et al. Metabolic response after intraarterial 90Y-glass microsphere treatment for colorectal liver metastases: comparison of quantitative and visual analyses by 18F-FDG PET. J Nucl Med. 2004 Nov;45(11):1892–1897. [PubMed] [Google Scholar]
  • 26.Salem R, Lewandowski RJ, Gates VL, et al. Research reporting standards for radioembolization of hepatic malignancies. J Vasc Interv Radiol. 2011 Mar;22(3):265–278. doi: 10.1016/j.jvir.2010.10.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. European Organization for Research and Treatment of Cancer, National Cancer Institute of the United States, National Cancer Institute of Canada. J Natl Cancer Inst. 2000 Feb 2;92(3):205–216. doi: 10.1093/jnci/92.3.205. [DOI] [PubMed] [Google Scholar]
  • 28.Bruix J, Sherman M, Llovet JM, et al. Clinical management of hepatocellular carcinoma. Conclusions of the Barcelona-2000 EASL conference. European Association for the Study of the Liver. J Hepatol. 2001 Sep;35(3):421–430. doi: 10.1016/s0168-8278(01)00130-1. [DOI] [PubMed] [Google Scholar]
  • 29.Forner A, Ayuso C, Varela M, et al. Evaluation of tumor response after locoregional therapies in hepatocellular carcinoma: are response evaluation criteria in solid tumors reliable. Cancer. 2009 Feb 1;115(3):616–623. doi: 10.1002/cncr.24050. [DOI] [PubMed] [Google Scholar]
  • 30.Riaz A, Miller FH, Kulik LM, et al. Imaging response in the primary index lesion and clinical outcomes following transarteial locoregional theapy for hepatocellular carcinoma. JAMA. 2010;303(11):1062–1069. doi: 10.1001/jama.2010.262. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Riaz A, Kulik L, Lewandowski RJ, et al. Radiologic-pathologic correlation of hepatocellular carcinoma treated with internal radiation using yttrium-90 microspheres. Hepatology. 2009 Apr;49(4):1185–1193. doi: 10.1002/hep.22747. [DOI] [PubMed] [Google Scholar]
  • 32.Riaz A, Memon K, Miller FH, et al. Role of the EASL, RECIST, and WHO response guidelines alone or in combination for hepatocellular carcinoma: Radiologic–pathologic correlation. Journal of Hepatology. 2011;54(4):695–704. doi: 10.1016/j.jhep.2010.10.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Rhee TK, Naik NK, Deng J, et al. Tumor response after yttrium-90 radioembolization for hepatocellular carcinoma: comparison of diffusion-weighted functional MR imaging with anatomic MR imaging. J Vasc Interv Radiol. 2008 Aug;19(8):1180–1186. doi: 10.1016/j.jvir.2008.05.002. [DOI] [PubMed] [Google Scholar]
  • 34.Riaz A, Ryu RK, Kulik LM, et al. Alpha-fetoprotein response after locoregional therapy for hepatocellular carcinoma: oncologic marker of radiologic response, progression, and survival. J Clin Oncol. 2009 Dec 1;27(34):5734–5742. doi: 10.1200/JCO.2009.23.1282. [DOI] [PubMed] [Google Scholar]
  • 35.Kulik LM, Atassi B, van Holsbeeck L, et al. Yttrium-90 microspheres (TheraSphere(R)) treatment of unresectable hepatocellular carcinoma: Downstaging to resection, RFA and bridge to transplantation. J Surg Oncol. 2006 Oct 17;94(7):572–586. doi: 10.1002/jso.20609. [DOI] [PubMed] [Google Scholar]
  • 36.Lewandowski RJ, Kulik LM, Riaz A, et al. A comparative analysis of transarterial downstaging for hepatocellular carcinoma: chemoembolization versus radioembolization. Am J Transplant. 2009 Aug;9(8):1920–1928. doi: 10.1111/j.1600-6143.2009.02695.x. [DOI] [PubMed] [Google Scholar]
  • 37.Salem R, Lewandowski RJ, Mulcahy MF, et al. Radioembolization for hepatocellular carcinoma using Yttrium-90 microspheres: a comprehensive report of long-term outcomes. Gastroenterology. 2010 Jan;138(1):52–64. doi: 10.1053/j.gastro.2009.09.006. [DOI] [PubMed] [Google Scholar]
  • 38.Kulik LM, Carr BI, Mulcahy MF, et al. Safety and efficacy of (90)Y radiotherapy for hepatocellular carcinoma with and without portal vein thrombosis. Hepatology. 2007 Nov 20;47(1):71–81. doi: 10.1002/hep.21980. [DOI] [PubMed] [Google Scholar]
  • 39.Kooby DA, Egnatashvili V, Srinivasan S, et al. Comparison of yttrium-90 radioembolization and transcatheter arterial chemoembolization for the treatment of unresectable hepatocellular carcinoma. J Vasc Interv Radiol. Feb;21(2):224–230. doi: 10.1016/j.jvir.2009.10.013. [DOI] [PubMed] [Google Scholar]
  • 40.Carr BI, Kondragunta V, Buch SC, Branch RA. Therapeutic equivalence in survival for hepatic arterial chemoembolization and yttrium 90 microsphere treatments in unresectable hepatocellular carcinoma: a Two-Cohort Study. Cancer. Jan 11;116(5):1305–1314. doi: 10.1002/cncr.24884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ibrahim SM, Nikolaidis P, Miller FH, et al. Radiologic findings following Y90 radioembolization for primary liver malignancies. Abdom Imaging. 2009 Sep-Oct;34(5):566–581. doi: 10.1007/s00261-008-9454-y. [DOI] [PubMed] [Google Scholar]
  • 42.Saxena A, Bester L, Chua TC, Chu FC, Morris DL. Yttrium-90 Radiotherapy for Unresectable Intrahepatic Cholangiocarcinoma: A Preliminary Assessment of This Novel Treatment Option. Ann Surg Oncol. 2009 Oct 30; doi: 10.1245/s10434-009-0777-x. [DOI] [PubMed] [Google Scholar]
  • 43.Gray B, Van Hazel G, Hope M, et al. Randomised trial of SIR-Spheres plus chemotherapy vs. chemotherapy alone for treating patients with liver metastases from primary large bowel cancer. Ann Oncol. 2001 Dec;12(12):1711–1720. doi: 10.1023/a:1013569329846. [DOI] [PubMed] [Google Scholar]
  • 44.Goin JE, Dancey JE, Hermann GA, Sickles CJ, Roberts CA, MacDonald JS. Treatment of unresectable metastatic colorectal carcinoma to the liver with intrahepatic Y-90 microspheres: a dose-ranging study. World J of Nuc Med. 2003;2:216–225. [Google Scholar]
  • 45.Mulcahy MF, Lewandowski RJ, Ibrahim SM, et al. Radioembolization of colorectal hepatic metastases using yttrium-90 microspheres. Cancer. 2009 May 1;115(9):1849–1858. doi: 10.1002/cncr.24224. [DOI] [PubMed] [Google Scholar]
  • 46.Cosimelli M, Golfieri R, Cagol PP, et al. Multi-centre phase II clinical trial of yttrium-90 resin microspheres alone in unresectable, chemotherapy refractory colorectal liver metastases. Br J Cancer. 2010 Jul 27;103(3):324–331. doi: 10.1038/sj.bjc.6605770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Sharma RA, Van Hazel GA, Morgan B, et al. Radioembolization of liver metastases from colorectal cancer using yttrium-90 microspheres with concomitant systemic oxaliplatin, fluorouracil, and leucovorin chemotherapy. J Clin Oncol. 2007 Mar 20;25(9):1099–1106. doi: 10.1200/JCO.2006.08.7916. [DOI] [PubMed] [Google Scholar]
  • 48.Hendlisz A, Van den Eynde M, Peeters M, et al. Phase III trial comparing protracted intravenous fluorouracil infusion alone or with yttrium-90 resin microspheres radioembolization for liver-limited metastatic colorectal cancer refractory to standard chemotherapy. J Clin Oncol. 2010 Aug 10;28(23):3687–3694. doi: 10.1200/JCO.2010.28.5643. [DOI] [PubMed] [Google Scholar]
  • 49.Kennedy AS, Dezarn WA, McNeillie P, et al. Radioembolization for unresectable neuroendocrine hepatic metastases using resin 90Y-microspheres: early results in 148 patients. Am J Clin Oncol. 2008 Jun;31(3):271–279. doi: 10.1097/COC.0b013e31815e4557. [DOI] [PubMed] [Google Scholar]
  • 50.Rhee TK, Lewandowski RJ, Liu DM, et al. 90Y Radioembolization for metastatic neuroendocrine liver tumors: preliminary results from a multi-institutional experience. Ann Surg. 2008 Jun;247(6):1029–1035. doi: 10.1097/SLA.0b013e3181728a45. [DOI] [PubMed] [Google Scholar]
  • 51.Sato KT, Lewandowski RJ, Mulcahy MF, et al. Unresectable chemorefractory liver metastases: radioembolization with 90Y microspheres--safety, efficacy, and survival. Radiology. 2008 May;247(2):507–515. doi: 10.1148/radiol.2472062029. [DOI] [PubMed] [Google Scholar]
  • 52.Bangash AK, Atassi B, Kaklamani V, et al. 90Y Radioembolization of Metastatic Breast Cancer to the Liver: Toxicity, Imaging Response, Survival. J Vasc Interv Radiol. 2007 May;18(5):621–628. doi: 10.1016/j.jvir.2007.02.019. [DOI] [PubMed] [Google Scholar]
  • 53.Coldwell DM, Kennedy AS, Nutting CW. Use of Yttrium-90 Microspheres in the Treatment of Unresectable Hepatic Metastases from Breast Cancer. Int J Radiat Oncol Biol Phys. 2007 May 22; doi: 10.1016/j.ijrobp.2007.03.056. [DOI] [PubMed] [Google Scholar]
  • 54.Salem R, Lewandowski RJ, Atassi B, et al. Treatment of unresectable hepatocellular carcinoma with use of 90Y microspheres (TheraSphere): safety, tumor response, and survival. J Vasc Interv Radiol. 2005 Dec;16(12):1627–1639. doi: 10.1097/01.RVI.0000184594.01661.81. [DOI] [PubMed] [Google Scholar]
  • 55.Kennedy AS, Coldwell D, Nutting C, et al. Resin 90Y-microsphere brachytherapy for unresectable colorectal liver metastases: modern USA experience. Int J Radiat Oncol Biol Phys. 2006 Jun 1;65(2):412–425. doi: 10.1016/j.ijrobp.2005.12.051. [DOI] [PubMed] [Google Scholar]
  • 56.Murthy R, Xiong H, Nunez R, et al. Yttrium 90 resin microspheres for the treatment of unresectable colorectal hepatic metastases after failure of multiple chemotherapy regimens: preliminary results. J Vasc Interv Radiol. 2005 Jul;16(7):937–945. doi: 10.1097/01.RVI.0000161142.12822.66. [DOI] [PubMed] [Google Scholar]
  • 57.Sangro B, Gil-Alzugaray B, Rodriguez J, et al. Liver disease induced by radioembolization of liver tumors: description and possible risk factors. Cancer. 2008 Apr 1;112(7):1538–1546. doi: 10.1002/cncr.23339. [DOI] [PubMed] [Google Scholar]
  • 58.Young JY, Rhee TK, Atassi B, et al. Radiation Dose Limits and Liver Toxicities Resulting from Multiple Yttrium-90 Radioembolization Treatments for Hepatocellular Carcinoma. J Vasc Interv Radiol. 2007 Nov;18(11):1375–1382. doi: 10.1016/j.jvir.2007.07.016. [DOI] [PubMed] [Google Scholar]
  • 59.Jakobs TF, Saleem S, Atassi B, et al. Fibrosis, portal hypertension, and hepatic volume changes induced by intra-arterial radiotherapy with 90yttrium microspheres. Dig Dis Sci. 2008 Sep;53(9):2556–2563. doi: 10.1007/s10620-007-0148-z. [DOI] [PubMed] [Google Scholar]
  • 60.Gaba RC, Lewandowski RJ, Kulik LM, et al. Radiation lobectomy: preliminary findings of hepatic volumetric response to lobar yttrium-90 radioembolization. Ann Surg Oncol. 2009 Jun;16(6):1587–1596. doi: 10.1245/s10434-009-0454-0. [DOI] [PubMed] [Google Scholar]
  • 61.Jakobs TF, Saleem S, Atassi B, et al. Fibrosis, Portal Hypertension, and Hepatic Volume Changes Induced by Intra-arterial Radiotherapy with (90)Yttrium Microspheres. Dig Dis Sci. 2008 Sep;53(9):2556–2563. doi: 10.1007/s10620-007-0148-z. [DOI] [PubMed] [Google Scholar]
  • 62.Salem R, Parikh P, Atassi B, et al. Incidence of radiation pneumonitis after hepatic intra-arterial radiotherapy with yttrium-90 microspheres assuming uniform lung distribution. Am J Clin Oncol. 2008 Oct;31(5):431–438. doi: 10.1097/COC.0b013e318168ef65. [DOI] [PubMed] [Google Scholar]
  • 63.Leung TW, Lau WY, Ho SK, et al. Radiation pneumonitis after selective internal radiation treatment with intraarterial 90yttrium-microspheres for inoperable hepatic tumors. Int J Radiat Oncol Biol Phys. 1995 Nov 1;33(4):919–924. doi: 10.1016/0360-3016(95)00039-3. [DOI] [PubMed] [Google Scholar]
  • 64.TheraSphere Yttrium-90 microspheres package insert. MDS Nordion; Kanata, Canada: 2004. [Google Scholar]
  • 65.Murthy R, Brown DB, Salem R, et al. Gastrointestinal complications associated with hepatic arterial Yttrium-90 microsphere therapy. J Vasc Interv Radiol. 2007 Apr;18(4):553–561. doi: 10.1016/j.jvir.2007.02.002. quiz 562. [DOI] [PubMed] [Google Scholar]
  • 66.Carretero C, Munoz-Navas M, Betes M, et al. Gastroduodenal Injury After Radioembolization of Hepatic Tumors. Am J Gastroenterol. 2007 Mar 13; doi: 10.1111/j.1572-0241.2007.01172.x. [DOI] [PubMed] [Google Scholar]
  • 67.Murthy R, Eng C, Krishnan S, et al. Hepatic yttrium-90 radioembolotherapy in metastatic colorectal cancer treated with cetuximab or bevacizumab. J Vasc Interv Radiol. 2007 Dec;18(12):1588–1591. doi: 10.1016/j.jvir.2007.08.015. [DOI] [PubMed] [Google Scholar]

RESOURCES