Radioembolization as Salvage Therapy for Hepatic Metastasis of Uveal Melanoma: A Single-Institution Experience
Abstract
OBJECTIVE. The purpose of this study was to assess the safety and efficacy of radioembolization in the management of hepatic metastasis of uveal melanoma after failure of immunoembolization or chemoembolization.
MATERIALS AND METHODS. From January 2007 through April 2009, 32 patients underwent radioembolization therapy for hepatic metastasis of uveal melanoma. Pretreatment tumor burdens were divided into three categories: less than 25% (n = 25), 25–50% (n = 5), and greater than 50% (n = 2). Toxicity, extrahepatic disease, and hepatic tumor response were assessed 1 month and then every 3 months after treatment. Best radiographic response of hepatic metastasis was determined with the Response Evaluation Criteria in Solid Tumors criteria. Overall survival and progression-free survival of hepatic metastasis were estimated by Kaplan-Meier analysis. Differences in survival between subgroups were evaluated by log-rank test in univariate analysis.
RESULTS. The clinical follow-up period ranged from 1.0 to 29.0 months (median, 10.0 months). The median overall survival was 10.0 months, and the progression-free survival of hepatic metastasis, 4.7 months. Twenty-two patients died 1.0–29.0 months (median, 5.8 months) after treatment owing to progression of liver disease (n = 13), extrahepatic disease (n = 4), or both (n = 5). Patients who had a pretreatment tumor burden less than 25% had longer median overall survival (10.5 vs 3.9 months, p = 0.0003) and progression-free survival (6.4 vs 3.0 months, p = 0.03) than patients who had a pretreatment tumor burden of 25% or greater. Patients who had a complete response (n = 1), partial response (n = 1), or stable disease (n = 18) had longer median overall survival (14.7 vs 4.9 months, p = 0.0006) and progression-free survival of hepatic metastasis (7.9 vs 3.1 months, p < 0.0001) than patients with tumor progression (n = 12). Self-limiting grade 1–2 systemic toxicity included tiredness (n = 9), indigestion (n = 2), and abdominal discomfort (n = 5). Grade 3–4 hepatic toxicity was attributed to tumor progression.
CONCLUSION. Radioembolization is safe and effective salvage therapy for limited metastasis of uveal melanoma.
Introduction
Uveal melanoma is the most common primary intraocular malignant tumor among adults [1, 2]. The incidence of uveal melanoma is 4.3 cases per million population with a slightly higher rate among men (4.9 cases per million than women (3.7 cases per million) [3]. Uveal melanoma is more common in older age. The progressively increasing age-specific incidence peaks at age 70 years (24.5 cases per million among men, 17.8 cases per million among women) [3].
As many as 50% of patients have systemic metastasis after the initial diagnosis and treatment of primary uveal melanoma. Clinically evident metastatic disease at presentation is uncommon, however, indicating that early subclinical metastasis occurs in most cases [4, 5]. The liver is the predominant organ of involvement in 70–90% of patients with metastatic uveal melanoma and tends to be the first manifestation of metastatic disease [6–9]. Approximately 90% of patients with metastatic uveal melanoma die with hepatic metastasis [7]. In general, without treatment, survival after the development of hepatic metastasis of uveal melanoma is poor, the median survival period being 2–7 months [5, 6].
Treatment options for patients with metastatic uveal melanoma are limited. Only 9% of patients with hepatic metastasis are eligible for resection, predominantly because of the multiplicity of hepatic tumors when the patient comes to medical attention [9]. Furthermore, the standard systemic chemotherapies used for metastatic cutaneous melanoma have not been efficacious in the management of metastatic uveal melanoma [10]. However, survival has been lengthened with transarterial catheter-directed liver therapies, such as hepatic arterial chemoinfusion, transarterial chemoembolization (TACE), immunoembolization, and isolated hepatic perfusion [10–21]. Despite the development of various transarterial catheter-directed treatments, most patients with hepatic metastasis of uveal melanoma eventually progress through these first-line approaches. Therefore, subsequent salvage therapy is mandatory to lengthen the survival of patients with hepatic metastasis of uveal melanoma.
We evaluated our single-institution experience using radioembolization as salvage therapy in the care of patients with hepatic metastasis of uveal melanoma. Tumor response and related toxicity after radioembolization were assessed. The results were compared with those of other transarterial catheter-directed therapies to increase understanding of the efficacy of radioembolization in the management of hepatic metastasis of uveal melanoma.
Materials and Methods
From January 2007 through April 2009, 32 patients (14 men, 18 women; median age, 61 years; range, 29–89 years) with hepatic metastasis of uveal melanoma underwent radioembolization. Patient eligibility included tumor progression after treatment with immunoembolization or TACE, no extensive extrahepatic metastasis limiting life expectancy less than 3 months, Eastern Cooperative Oncology Group 0–2 performance status, and adequate liver function (bilirubin concentration < 1.8 mg/dL, albumin concentration > 3.0 g/dL, no ascites) and renal function (creatinine concentration ≤ 2.0 mg/dL). Exclusion criteria were uncorrectable arterial blood flow to the gastrointestinal tract diagnosed at arteriography or 99mTc–macroaggregated albumin scintigraphy, lung shunting greater than 20%, portal vein thrombosis, biliary obstruction, and previous radiation therapy in which the liver was included in the radiation field. Before treatment, all patients underwent PET, contrast-enhanced abdominal MRI, and CT of the chest, abdomen, and pelvis. Pretreatment tumor burden was divided into the three following categories: less than 25% (n = 25), 25–50% (n = 5), and greater than 50% (n = 2). Tumor burden percentages were calculated with MRI and the following formula: volume of tumor / (volume of tumor + volume of normal liver). Pretreatment tumor burden was determined at radioactive microsphere dosimetry.
Preparatory Arteriography and Radioembolization Treatments
The preparatory arteriograms and radioembolization treatments were performed by one of three interventional radiologists with certificates of added qualification. All patients underwent abdominal visceral angiography before treatment for definition of the intrahepatic and extrahepatic arterial anatomy. After prophylactic coil embolization of nontarget extrahepatic vessels (i.e., right gastric and gastroduodenal arteries), 99mTc–macroaggregated albumin was injected into the hepatic arteries as deemed necessary by the interventional radiologist performing the procedure. SPECT was performed to assess for unintentional delivery of radioactive microspheres to extrahepatic structures and to estimate the percentage of shunting to the lungs.
Radioembolization was performed with 90Y resin microspheres (SIR-Spheres, Sirtex Medical). The activity of 90Y microspheres was calculated by an experienced radiation oncologist using the following formula: 90Y microspheres = body surface area – 0.2 + (percentage tumor involvement / 100), where microspheres are measured in gigabecquerels and body surface area is measured in square meters [22]. Because radioembolization was being used as salvage therapy after failure of previous transarterial catheter-directed therapies, a dose reduction of 25% was applied to each patient. For each patient, the details of radioembolization treatments were determined on the basis of the location of the hepatic metastatic lesions, angiographic findings, and history of surgery. Procedure-related complications were classified according to the Society of Interventional Radiology classification system of complications by outcome [23].
Patient Assessment and Treatment Response
One month after completing therapy, patients were evaluated for acute toxicity and progression of extrahepatic metastasis. They then were evaluated every 3 months for assessment of clinical status, including radiographic response of hepatic metastasis. History interviews and physical examinations were performed in our multidisciplinary metastatic uveal melanoma clinic by both a medical and a radiation oncologist. Laboratory blood tests, including complete blood cell count, total bilirubin, aspartate aminotransferase, alanine aminotransferase, lactate dehydrogenase, alkaline phosphatase, albumin, and creatinine, were performed every week for 1 month and then every 2 weeks for 2 months and every month thereafter. The degree of toxicity was determined with the Common Terminology Criteria for Adverse Events, version 3.
Contrast-enhanced CT scans of the chest, abdomen, and pelvis were obtained 1 month and every 3 months after treatment for evaluation of extrahepatic disease and progression. In addition, gadolinium-enhanced abdominal MRI and whole-body PET were performed every 3 months after treatment for evaluation of tumor response in the liver. MRI of the abdomen was performed sooner than the scheduled 3-month follow-up evaluation if there was concern about intrahepatic disease progression or radiation-induced hepatic disease. Best radiographic response of hepatic metastasis was determined with the Response Evaluation Criteria in Solid Tumors, version 1.0 [24].
Statistical Analysis
Institutional review board approval was obtained for retrospective analysis of the records of patients who underwent radioembolization. The overall survival period was measured from initial administration of radioactive microspheres to death. The period of progression-free survival of hepatic metastasis was measured from initial administration of radioactive microspheres to confirmation of progression of hepatic metastasis at diagnostic imaging or death of the patient. Overall survival and progression-free survival of hepatic metastasis were estimated with Kaplan-Meier analysis. The log-rank test was used to conduct univariate analysis of survival differences between subgroups determined with pretreatment tumor burdens and radiographic response. A value of p < 0.05 was considered significant. Statistical analyses were conducted with the GraphPad Prism program (GraphPad Software).
Results
Thirty-two patients were treated with radioembolization as salvage therapy after failure of immunoembolization (median, 4 procedures; range 1–14, procedures) or TACE (median, 3 procedures; range, 1–14 procedures). The total number of procedures (immunoembolization and/or TACE) per patient ranged from 1 to 23 (median, 9). Thirty-one patients underwent radioembolization treatment without procedure-related complications. Common hepatic artery dissection occurred in one patient during preparatory arteriography. The patient received one fractionated whole-liver radioembolization treatment, but subsequent occlusion of the common hepatic artery precluded further treatment.
Radioembolization treatments were administered as either unfractionated whole-liver (eight patients), fractionated whole-liver (two patients), or consecutive right and left lobe (19 patients) treatments separated by 3–5 weeks. Three patients underwent single-lobe treatments, two of whom could not undergo the second radioembolization treatment because of deterioration of hepatic function related to progression of disease. One patient underwent only one radioembolization treatment of tumor isolated to the right lobe. Lobar treatment was generally preferred. Whole-liver treatment was performed in accordance with the arterial anatomy at the discretion of the treating interventional radiologist. Three of the patients who received unfractionated whole-liver treatments underwent left lobectomy for tumor resection before initiation of transarterial catheter-directed therapy. The median dose delivered was 1.08 GBq (29.28 mCi) per patient (range, 0.63–1.86 GBq [16.77–50.10 mCi]).
There were no procedure- or treatment-related deaths. Before radioembolization, 13 patients had preexisting liver dysfunction from previous transarterial catheter-directed therapy or intrahepatic tumor burden (Table 1). Hepatic toxicity grades after the procedure are shown in Table 1. Grade 3 and 4 liver toxicity (n = 4) 1 month after radioembolization was caused by tumor progression confirmed with either CT or MRI and was not related to the treatment itself. In the four patients with grade 3 or 4 toxicity after treatment, grades 1 (n = 1), 2 (n = 2), and 3 (n = 1) hepatic dysfunction was present before radioembolization. Only one patient with pretreatment grade 1 hepatic dysfunction had a two-level increase in hepatic dysfunction (grade 3), which was due to marked progression of liver metastasis after radioembolization. Systemic nonhepatic toxicity after radioembolization was mainly grade 1 or 2 mild tiredness (n = 9) and gastrointestinal symptoms such as indigestion (n = 2) and mild abdominal discomfort (n = 5), all of which were self-limiting.
Liver Toxicity Grade | No. of Patients With Preexisting Liver Dysfunction | No. of Patients With Liver Toxicity 1 Month After Radioembolization |
---|---|---|
0 | 19 | 14 |
1 | 9 | 10 |
2 | 3 | 4 |
3 | 1 | 3 |
4 | 0 | 1 |
The clinical follow-up lasted 1.0–29.0 months (median, 10.0 months). At the end of the follow-up period (September 2009), 10 patients were alive 4.7–27.0 months (median, 9.4 months) after treatment. Twenty-two patients died 1.0–29.0 months (median, 5.8 months) after radioembolization owing to progression of liver disease (n = 13), extrahepatic disease (n = 4), or both (n = 5). Two patients underwent treatment to a single lobe then died 1.0 and 3.3 months after radioembolization before completing therapy to all hepatic metastatic lesions. The causes of death were progression of hepatic metastasis and extrahepatic disease (pulmonary metastasis). The causes of death of the two patients were determined with imaging (CT and MRI) and laboratory and clinical evaluation. The patient who had common hepatic artery dissection during preparatory arteriography underwent one fractionated whole-liver treatment and died 9.8 months after radioembolization owing to progression of liver disease. These three patients were included in the intent-to-treat analysis of overall survival. Therefore, overall survival of the entire patient sample (n = 32) ranged from 1.0 to 29.0 months (median, 10.0 months) (Fig. 1). Median overall survival was found to be significantly longer for patients with a pretreatment tumor burden less than 25% (10.5 months; range, 3.1–29.0 months) than for those with a pretreatment tumor burden of 25% or greater (3.9 months; range, 1.0–12.1 months) (p = 0.0003) (Fig. 2A).
With respect to best radiologic response of hepatic metastasis, one patient had a complete response, one patient had a partial response, 18 patients had stable disease, and 12 patients had disease progression. The median overall survival period among patients with complete response, partial response, or stable disease (14.7 months) was significantly longer than the median overall survival among patients with disease progression (4.9 months) (p = 0.0006) (Fig. 2B). The median progression-free survival of hepatic metastasis for the entire patient sample (n = 32) was 4.7 months (range, 1.0–26.5 months) (Fig. 1). Patients who had a pretreatment tumor burden less than 25% had significantly longer median progression-free survival of hepatic metastasis than did patients with a 25% or greater pretreatment tumor burden (6.4 versus 3.0 months) (p = 0.03) (Fig. 3A). The median period of progression-free survival of hepatic metastasis among patients who had a complete response, partial response, or stable disease also was significantly longer than that among patients who had disease progression after radioembolization (7.9 versus 3.1 months) (p < 0.0001) (Fig. 3B).
Discussion
Transarterial catheter-directed therapies for hepatic metastasis are most advantageous when systemic chemotherapy is not effective or beneficial, the liver is the only or predominant site of involvement, or there is urgent need to control hepatic progression for symptom control or palliation. These criteria are often applicable to patients with hepatic metastasis of uveal melanoma. Therefore, various transarterial catheter-directed therapies have been investigated for the management of these metastatic lesions, including hepatic arterial infusion, isolated hepatic perfusion, TACE, and immunoembolization. Results of clinical studies of transarterial catheter-directed liver treatments for hepatic metastasis of uveal melanoma are shown in Table 2. Although the number of patients in the individual studies is small, the reported overall survival among patients who received hepatic arterial infusion, TACE, immunoembolization, or isolated hepatic perfusion as first-line therapy for hepatic metastasis of uveal melanoma ranges from 6.0 to 21.0 months (median, ≈1 year) (Table 2).
Overall Survival (mo) | ||||||
---|---|---|---|---|---|---|
Source | Treatment | No. of Patients | Drug | Responders | Nonresponders) | Median |
Mavligit et al. [9] | TACE first-line | 30 | Cisplatin | 14 | 6 | 11 |
Bedikian et al. [10] | TACE first-line | 44 | Cisplatin | 14.5 | 5 | 6 |
Bedikian et al. [10] | TACE second-line | 20 | Cisplatin | < 6 | ||
Huppert et al. [16] | TACE first-line | 14 | Cisplatin, carboplatin | 14.5 | 10 | 11.5 |
Patel et al. [15] | TACE first-line | 24 | BCNU | 21.9 | 3.3 | 5.2 |
Dayani et al. [20] | TACE first-line | 21 | Mitomycin C, cisplatin, doxorubicin | 12.7 (mean) | 3.7 (mean) | 7.6 (mean) |
Vogl et al. [19] | TACE first-line | 12 | Mitomycin C | 21 | 16.5 | 21 |
Leyvraz et al. [11] | HAI first-line | 31 | Fotemustine | 14 | ||
Peters et al. [21] | HAI first-line | 101 | Fotemustine | 15 | ||
Sato [17] | Transarterial immunoembolization first-line | 34 | GM-CSF | 14.4 | ||
Alexander [13] | Isolated hepatic perfusion first-line | 29 | Melphalan | 12.1 |
Note—TACE = transarterial chemoembolization, HAI = hepatic arterial infusion, BCNU = 1,3-bis (2-chloroethyl)-1-nitrosourea, GM-CSF, granulocyte–macrophage colony-stimulating factor
Radioembolization with the pure β-emitting isotope 90Y has been used for several years to manage both primary and metastatic hepatic tumors [25–28]. The therapeutic advantage of radioembolization is based on two important principles: first, hepatic tumors receive 80–100% of their blood supply exclusively from the hepatic artery and, second, tumor neovascularity is denser than the vasculature supplying the normal hepatic parenchyma [29, 30]. Therefore, 90Y microspheres infused into the hepatic artery concentrate in much higher doses in hepatic tumors than in the normal surrounding liver [30]. Yttrium-90 microspheres, which have a half-life of 64.2 hours, are permanently embedded in hepatic tumors and have an average energy of 0.94 MeV [22, 30]. Most of the radiation dose is released over 14 days at approximately 50 cGy/min [31]. The mean tissue penetration of 90Y is 2.5 mm with a maximum penetration of approximately 10 mm [22, 31]. Most of the radiation dose is administered directly to the tumor with relative sparing of the normal liver parenchyma. Because radioactive plaque is one of the standards of care in the management of primary uveal melanoma, radioembolization is a reasonable approach to the management of hepatic metastasis of uveal melanoma.
Kennedy et al. [31] reported their experience treating a small number of patients with hepatic metastasis of uveal melanoma using 90Y radioactive microspheres. In that retrospective multicenter study, 11 patients underwent 12 radioembolization procedures with a median activity of 1.55 GBq delivered per treatment. Toxicity was minimal. Only one patient experienced a grade 3 event, a gastric ulcer that healed uneventfully within 6 weeks with supportive care. No radiation-induced liver disease was detected. CT and PET of hepatic metastatic lesions 3 months after treatment showed one complete response, six partial responses, one case of stable disease, and one case of disease progression in nine evaluable patients. Among the 10 patients with available clinical follow-up information, the 1-year survival rate was 80%. The relatively low response rate (one complete response, one partial response, and 18 cases of stable disease in 32 patients) in our study compared with that found by Kennedy et al. may be explained in part by the inclusion of previously treated patients with bulky progressive hepatic metastatic lesions. Pretreatment tumor burden has been found to be an important predictor of response after TACE. In our phase 2 trial of 1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU) for TACE [15], the overall survival period among patients with less than 20% pretreatment tumor burden was found to be significantly longer (median, 19.0 months; range, 3.8–27.6 months) than that among patients with 20–50% (median, 5.6 months; range, 0.1–14.0 months) or greater than 50% pretreatment tumor burden (median, 2.1 months; range, 0.6–7.5 months). Huppert et al. [16] reported similar results of TACE using cisplatin and carboplatin in the care of 14 patients with hepatic metastasis of uveal melanoma. Median survival was 17 and 11 months among patients with pretreatment tumor burdens of less than 25% (n = 7) and 25% or greater (n = 7) (p = 0.02). These results correlate well with those of a study by Dunfee et al. [32], who evaluated prognostic indicators associated with survival after radioembolization [32]. In that study, 130 patients underwent radioembolization therapy for either primary (n = 7) or secondary (n = 123) malignant disease of the liver after lack of response to standard-of-care therapy. In that patient sample, large pretreatment tumor burden (> 50%) was found to be a statistically significant negative prognostic indicator of survival. On the basis of those observations, radioembolization may have limited benefit in the care of patients with a large pretreatment tumor burden, and, therefore, exclusion from future clinical trials should be considered. Furthermore, the median 90Y dose delivered in our study was 1.08 GBq compared with 1.55 GBq in the report by Kennedy et al. [31]. The relatively low dose of 90Y, which was deemed necessary to avoid adverse events in our heavily pretreated population, may have limited the effectiveness of our radioembolization treatments and may explain the lower overall response rate among our patients.
As summarized in Table 2, most studies show significantly longer overall survival among treatment responders than among non-responders. For example, in the previously discussed TACE trial of BCNU in the treatment of 30 patients with hepatic metastasis of uveal melanoma [15], median overall survival was significantly longer for patients with complete response and partial response (21.9 months; range, 7.4–27.6 months) than for patients with disease progression (3.3 months; range, 1.6–5.6 months). In our phase 1 clinical trial [17, 18] of immunoembolization with granulocyte–macrophage colony-stimulating factor (GM-CSF) in the management of hepatic metastasis of uveal melanoma in 34 patients with less than 50% tumor burden, overall survival for patients who achieved complete or partial response (33.7 months) was significantly longer than that among patients with stable disease or disease progression (12.4 months) (p = 0.0043). Furthermore, in the study by Dunfee et al. [32], response to radioembolization was found to be a statistically significant positive prognostic indicator of survival and correlated well with the findings of our study.
Information on the benefit of salvage transarterial catheter-directed therapy for hepatic metastasis of uveal melanoma is limited. In 1995, Bedikian et al. [10] reported their results in the care of 20 patients with metastatic uveal melanoma treated with TACE with a cisplatin-based regimen after failure of previous first-line therapy. Five of 20 patients treated with salvage TACE had a partial response, and two patients had stable disease. However, the overall survival of this patient sample was only 5 months, inferior to the 10.0-month median overall survival of our patient sample who underwent radioembolization salvage therapy.
To better understand the efficacy of radioembolization as salvage therapy for hepatic metastasis of uveal melanoma, we compared the results of the current study with those of previous clinical trials performed at our institution: immunoembolization with GM-CSF and TACE with BCNU [15, 17, 18]. For patients with limited (< 50%) pretreatment tumor burden, the overall survival and progression-free survival of hepatic metastasis after first-line immunoembolization and TACE were similar to the results of radioembolization as salvage therapy (Table 3). Therefore, our institutional data indicate that radioembolization may be beneficial when previous immunoembolization or TACE has failed. Furthermore, because of its safety profile and potential efficacy, the addition of radioembolization to our treatment armamentarium affords a useful means of further lengthening the survival of patients with metastatic uveal melanoma.
Therapy | No. of Patients | Median Overall Survival Period (mo) | Median Period of Progression-Free Survival of Hepatic Metastasis (mo) |
---|---|---|---|
Salvage radioembolization | 30 | 10.0 | 4.7 |
First-line transarterial immunoembolization with granulocyte–macrophage colony-stimulating factor [17, 18] | 34 | 14.4 | 4.8 |
First-line transarterial chemoembolization with 1,3-bis (2-chloroethyl)-1-nitrosourea [15] | 19 | 9.8 | 6.4 |
It can be argued that our results might have been better if radioembolization had been used as first-line instead of salvage therapy. Distal hepatic artery attenuation and obliteration of segmental hepatic artery branches have been reported after transarterial catheter-directed therapy [33]. Because our patients were heavily pretreated with either immunoembolization or TACE (median, 9 procedures), adequate delivery of radioactive microspheres to metastatic lesions of uveal melanoma might have been hampered, limiting the effectiveness of the radioembolization treatments. In this regard, we have started a new phase 2 clinical trial to prospectively investigate the safety and efficacy of radioembolization with 90Y resin microspheres as first-line therapy for hepatic metastasis of uveal melanoma. In that clinical trial, patient stratification will be based on history of transarterial catheter-directed therapy (first-line vs salvage). The safety and efficacy of treatments will be separately analyzed in these two patient cohorts. Furthermore, we will investigate whether the molecular and genetic characteristics of hepatic metastasis of uveal melanoma correlate with the outcome of radioembolization [34]. Biopsy specimens of hepatic metastatic lesions obtained before radioembolization may provide insight on the biologic activity of these tumors and thereby aid in prediction of treatment response. Because of the retrospective nature of this study, genetic analysis of hepatic metastatic lesions of uveal melanoma before radioembolization treatments was not performed, and that is a limitation.
To our knowledge, the current study included the largest number of patients with metastatic uveal melanoma treated with radioembolization at a single institution. The results of this study indicate that radioembolization is safe and effective salvage therapy for metastatic uveal melanoma in patients with a limited pretreatment tumor burden. The addition of radioembolization to our treatment armamentarium constitutes a useful means of lengthening survival among patients with metastatic uveal melanoma after failure of immunoembolization and TACE. Further investigation is needed to determine whether radioembolization should be used as an alternative first-line therapy for hepatic metastasis of uveal melanoma.
Footnote
Address correspondence to C. F. Gonsalves ([email protected]).
References
1.
Strickland D. JA L. Melanomas of eye: stability of rates. Am J Epidemiol 1981; 113:700-702
2.
[No authors listed]. Accuracy of diagnosis of choroidal melanomas in the Collaborative Ocular Melanoma Study: COMS report no. 1 Arch Ophthalmol 1990; 108:1268-1273
3.
Singh AD. A T. Incidence of uveal melanoma in the United States: 1973-1997. Ophthalmology 2003; 110:956-961
4.
Kujala E, Makitie T, Kivela T. Very long-term prognosis of patients with malignant uveal melanoma. Invest Ophthalmol Vis Sci 2003; 44:4651-4659
5.
Singh AD, Topham A. Survival rates with uveal melanoma in the United States: 1973–1997. Ophthalmology 2003; 110:962-965
6.
Rajpal S, Moore R, Karakousis CP. Survival in metastatic ocular melanoma. Cancer 1983; 52:334-336
7.
Char DH. Metastatic choroidal melanoma. Am J Ophthalmol 1978; 86:76-80
8.
Einhorn LH, Burgess MA, Gottlieb JA. Metastatic patterns of choroidal melanoma. Cancer 1974; 34:1001-1004
9.
Mavligit GM, Charnsangavej C, Carrasco CH, Patt YZ, Benjamin RS, Wallace S. Regression of ocular melanoma metastatic to the liver after hepatic arterial chemoembolization with cisplatin and polyvinyl sponge. JAMA 1988; 260:974-976
10.
Bedikian AY, Legha SS, Mavligit G, et al. Treatment of uveal melanoma metastatic to the liver: a review of the MD Anderson Cancer Center experience and prognostic factors. Cancer 1995; 76:1665-1671
11.
Leyvraz S, Spataro V, Bauer J, et al. Treatment of ocular melanoma metastatic to the liver by hepatic arterial chemotherapy. J Clin Oncol 1997; 15:2589-2595
12.
Cantore M, Fiorentini G, Aitini E, et al. Intra-arterial hepatic carboplatin-based chemotherapy for ocular melanoma metastatic to the liver: report of a phase II study. Tumori 1994; 80:37-39
13.
Alexander HR, Libutti SK, Bartlett DL, Puhlmann M, Fraker DL, Bachenheimer LC. A phase I-II study of isolated hepatic perfusion using melphalan with or without tumor necrosis factor for patients with ocular melanoma metastatic to liver. Clin Cancer Res 2000; 6:3062-3070
14.
Farma JM, Pingpank JF, Libutti SK, et al. Percutaneous hepatic perfusion (PHP) with melphalan for patients with metastatic melanoma to the liver. Proceedings of the 2005 Gastrointestinal Cancers Symposium. Alexandria, VA: American Society of Clinical Oncology,2005
15.
Patel K, Sullivan K, Berd D, et al. Chemoembolization of the hepatic artery with BCNU for metastatic uveal melanoma: results of a phase II study. Melanoma Res 2005; 15:297-304
16.
Huppert PE, Fierlbeck G, Pereira P, et al. Transarterial chemoembolization of liver metastases in patients with uveal melanoma. Eur J Radiol 2010; 74:e38-e44 [Epub 2009 May 21]
17.
Sato T, Eschelman DJ, Gonsalves CF, et al. Immunoembolization of malignant liver tumors, including uveal melanoma, using granulocyte-macrophage colony-stimulating factor. J Clin Oncol 2008; 26:5436-5442
18.
Yamamoto A, Chervoneva I, Sullivan KL, et al. High-dose immunoembolization: survival benefit in patients with hepatic metastases from uveal melanoma. Radiology 2009; 252:290-298
19.
Vogl T, Eichler K, Zangos S, et al. Preliminary experience with transarterial chemoembolization (TACE) in liver metastases of uveal malignant melanoma: local tumor control and survival. J Cancer Res Clin Oncol 2007; 133:177-184
20.
Dayani PN, Gould JE, Brown DB, Sharma KV, Linette GP, Harbour JW. Hepatic metastasis from uveal melanoma: angiographic pattern predictive of survival after hepatic artery chemoembolization. Arch Ophthalmol 2009; 127:628-632
21.
Peters S, Voelter V, Zografos L, et al. Intra-arterial hepatic perfusion for the treatment of liver metastases from uveal melanoma: experience in 101 patients. Ann Oncol 2006; 17:578-583
22.
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; 17:1251-1278
23.
Sacks D, McClenny TE, Cardella JF, et al. Society of Interventional Radiology clinical practice guidelines. J Vasc Interv Radiol 2003; 14:S199-S202
24.
Therasse P, Arbruck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst 2000; 92:205-216
25.
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; 12:1711-1720
26.
Kennedy AS, Coldwell D, Nutting C, et al. Resin 90Y-microspheres brachytherapy for unresectable colorectal liver metastases: modern UA experience. Int J Radiat Oncol Biol Phys 2006; 65:412-425
27.
Salem R, Lewandowski RJ, Atassi B, et al. Treatment of unresectable hepatocellular carcinoma with use of 90Y microspheres (Theraspheres): safety, tumor response, and survival. J Vasc Interv Radiol 2005; 16:1627-1639
28.
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; 31:271-279
29.
Breedis C, Young G. The blood supply of neoplasms in the liver. Am J Pathol 1954; 30:969-984
30.
Sato KT, Omary RA, Takehana C, et al. The role of vascularity in predicting survival after yttrium 90 radioembolization of liver metastases. J Vasc Interv Radiol 2009; 20:1564-1569
31.
Kennedy AS, Nutting C, Jakobs T, et al. A first report of radioembolization for hepatic metastases from ocular melanoma. Cancer Invest 2009; 27:682-690
32.
Dunfee BL, Riaz A, Lewandowski RJ, et al. Yttrium-90 radioembolization for liver malignancies: prognostic factors associated with survival. J Vasc Interv Radiol 2010; 21:90-95
33.
Chung JW, Kim HC, Yoon JH, et al. Transcatheter arterial chemoembolization of hepatocellular carcinoma: prevalence and causative factors of extrahepatic collateral arteries in 479 patients. Korean J Radiol 2006; 7:257-266
34.
Han F, Terai M, Yamamoto A, et al. A pilot study on genome-wide aberration analysis in metastatic uveal melanoma. (abstract). In: AACR Meeting Abstracts. Philadelphia, PA: American Association for Cancer Research,2008 :4517
Information & Authors
Information
Published In
Copyright
© American Roentgen Ray Society.
History
Submitted: April 26, 2010
Accepted: June 14, 2010
First published: November 23, 2012
Keywords
Authors
Metrics & Citations
Metrics
Citations
Export Citations
To download the citation to this article, select your reference manager software.