Abstract
Objectives.
To determine the efficacy and tolerability of sunitinib in recurrent or persistent clear cell ovarian cancer patients.
Methods.
All patients had one or two prior regimens with measurable disease. Tumors were at least 50% clear cell histomorphology and negative for WT-1 antigen and estrogen receptor expression by immunohistochemistry. Sunitinib 50 mg per day for 4 weeks was administered in repeated 6-week cycles until disease progression or prohibitive toxicity. Primary end points were progression-free survival (PFS) at 6 months and clinical response. The study was designed to determine if the drug had a response rate of at least 20% or 6-month PFS of at least 25%.
Results.
Of 35 patients enrolled, 30 were treated and eligible (median age: 51, range: 27–73). Twenty-five (83%) were White, 4 (13%) Asian, and 1 (3%) unknown. The majority 28 (83%) patients, underwent ≤3 but 2 (7%) had 16 courses of study therapy. Five (16.7%) patients had PFS ≥6 months (90% CI: 6.8%−31.9%). Two (6.7%) patients had a partial or complete response (90% CI: 1.2%–19.5%). The median PFS was 2.7 months. The median overall survival was 12.8 months. The most common grade 3 adverse events were fatigue (4), hypertension (4), neutropenia (4), anemia (3), abdominal pain (3), and leukopenia (3). Grade 4–5 adverse events included: thrombocytopenia (5), anemia (2), acute kidney Injury (1), stroke (1), and allergic reaction (1).
Conclusion.
Sunitinib demonstrated minimal activity in the second- and third-line treatment of persistent or recurrent clear cell ovarian carcinoma.
Keywords: Sunitinib, Progression-free survival, Persistent or recurrent clear cell ovarian, carcinoma
1. Introduction
Accounting for 3–12% of all epithelial ovarian cancers, patients with clear cell carcinomas have a poorer prognosis compared to those with serous cancers [1–6]. Clinical and translational studies have shown that the biology and clinical behavior of clear cell carcinoma is distinct compared to other epithelial cell types [2,7–9].
Clear cell cancers of the kidney, ovary, and uterus have similar genomic profiles [10]. Renal and ovarian clear cell carcinomas have frequent mutational inactivation of the Von Hippel-Lindau (VHL) pathway [11]. Similar to renal cell cancer, angiogenesis also plays a central role in ovarian cancer progression [12,13]. Targeting angiogenesis in ovarian cancer resulted in the approval of bevacizumab for recurrent disease [14,15]. Given these similarities between renal and ovarian clear cell cancers, we hypothesized that biologic agents that are active in metastatic renal cell cancer may have activity in ovarian clear cell cancers.
Sunitinib (SU11248) is a highly potent, selective inhibitor of protein tyrosine kinases, including vascular endothelial growth factor receptor (VEGF-R) and platelet derived growth factor receptor (PDGF-R) [16–20]. In second-line treatment of metastatic renal cell cancer, a setting where no effective standard therapy, sunitinib therapy resulted in a response rate of 34% [20]. In 2006, the FDA approved sunitinib for the treatment of advanced renal cell carcinoma. Sunitinib has been shown to have modest activity in epithelial ovarian cancers based on three phase II trials from Canada, Europe and United States [21–23]. However, all of these trials included patients with various epithelial cell types with distinct molecular profiles. In fact, clear cell cancers comprised of <10% of these clinical trial patients. Furthermore, there was no defined criterion for clear cell histology with central pathology review. Since epithelial ovarian cancers are heterogeneous cancers, it is important to study clear cell ovarian cancer in a multi-center, cooperative group trial with central pathology review and standardized treatments. As such, we proposed to evaluate the anti-tumor activity and toxicity of sunitinib in persistent or recurrent clear cell ovarian cancer patients.
2. Methods
2.1. Patient eligibility and exclusions
Patients had either recurrent or persistent clear cell ovarian cancer. They must have had one prior platinum-based chemotherapeutic regimen for management of primary disease containing carboplatin, cisplatin, or another organo platinum compound. Patients were allowed, but not required, to receive one additional cytotoxic regimen for management of recurrent or persistent disease. Patients must have had measurable disease with at least one target lesion to be used to assess response.
Patients were excluded if they have received any non-cytotoxic therapy for management of recurrent or persistent disease such as VEGF inhibitors including bevacizumab. All patients were at least 18 years old with a Gynecologic Oncology Group (GOG) performance status score of 0 (fully active) to 2 (ambulatory and capable of self-care but unable to work; up and about >50% of waking hours). All chemotherapy was discontinued at least three weeks before registration. All patients had adequate bone marrow, renal, hepatic, and neurologic function.
2.2. Pathology screening
Primary tumors had to have at least 50% clear cell histomorphology to be eligible or have a documented recurrence with at least 50% clear cell histomorphology and negative for expression of WT-1 antigen and estrogen receptor (ER) by immunohistochemistry. The trial was designed such that if the primary tumor did not have at least 50% clear cell histomorphology, a biopsy of the recurrent or persistent tumor was required. In this study, all patients met the initial histologic criteria and did not require a subsequent biopsy of the recurrent tumor. Appropriate tissue and immune-histochemical stained slides for WT-1 antigen and ER were available for histologic evaluation for central pathology review by NRG Oncology/Gynecologic Oncology Group.
2.3. Treatment plan
Sunitinib 50 mg per day was orally administered in repeated six-week cycles of daily therapy for four weeks, followed by two weeks off. Dose reduction for grade 3 to 4 toxicity was allowed to 37.5 mg per day and then to 25 mg per day. This six-week cycle was repeated until evidence of disease progression or unacceptable toxicity.
2.4. Efficacy and toxicity assessment
Clinical examination with evaluation of tumor burden was performed at baseline and before each cycle. Disease status was also assessed radiographically at baseline, before each odd cycle, and at the end of treatment. Response and progression were evaluated in this study using the new international criteria proposed by the revised Response Evaluation Criteria in Solid Tumors (RECIST) [24]. Using the National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE version 3.0), adverse events were assessed on day one of each cycle and were graded. Investigator-determined best overall response was defined by using RECIST criteria in patients with measurable tumors.
2.5. Objectives
The primary objectives were to determine the efficacy of sunitinib as estimated from the probability of surviving progression free for at least six months. Progression-free survival (PFS) was defined as the duration of time from start of treatment to time of progression or death, whichever occurs first. Response and progression were evaluated using RECIST [24]. Overall survival (OS) was defined as the duration of time from start of treatment to time of death or date of last contact.
2.6. Study oversight
The NRG Oncology/Gynecologic Oncology Group and GOG designed and conducted this study. The study was approved by the research ethics board at each participating center or by a central institutional review board and all patients provided written informed consent. With reviews by the data and safety monitoring committee, the data were collected, held, and analyzed by the statistical group. The first author (study chair) vouches for the integrity of the data and analyses reported and for the fidelity of the trial to the protocol. Representatives from the sponsors (the Cancer Therapy Evaluation Program of the National Cancer Institute and Pfizer) had no role in the design, accrual, management or analysis of the data. The drafting and content of the manuscript and the decision to publish was undertaken by the first author with input from all the coauthors.
2.7. Statistical analysis
This was a single arm, phase II clinical trial that used a flexible, bivariate two-stage design [25]. The primary hypothesis of this study tested the proportion of patients with objective tumor response (complete or partial) and the proportion of those surviving progression-free for at least six months. The null proportions were 10% for response rate and 15% for PFS at six months. The targeted accrual for the first stage was 19 eligible and evaluable patients (range: 15 to 22) and the cumulative targeted accrual for the second stage was 31 patients. The study was designed to determine if the drug had a response rate of at least 20% or a six-month PFS of 25%.
3. Results
From 2010 to 2014, 35 patients were enrolled and 30 were treated and eligible (median age: 51, range: 27–73). Five patients were not included: two never treated, and three ineligible. Twenty-five (83%) were White, 4 (13%) Asian, and 1 (3%) unknown. Performance status of 0,1, and 2 comprised of 18 (60%), 10 (33.3%), and 2 (6.7%) patients. Twenty (67%), 9 (30%) and 1 (3%) patients had 1, 2, and 3 cycles of prior chemotherapy. (Table 1) Two (6.7%) patients had a partial or complete response (90% CI: 1.2%–19.5%). The median PFS was 2.7 months. Eight (27%), 16 (53%), 1 (3%), 1 (3%), 1 (3%), 1 (3%), and 2 (7%) patients had 1, 2, 3, 4, 9, 11 and 16 courses of study therapy, respectively. The median OS was 12.8 months. Five (16.7%) patients had PFS ≥6 months (90% CI: 6.8%–31.9%). (Table 2 and Fig. 1).
Table 1.
Patient characteristics.
Number of patients | Percent of patients | |
---|---|---|
Age (y) | ||
20–29 | 2 | 6.7 |
30–39 | 1 | 3.3 |
40–49 | 10 | 33.3 |
50–59 | 9 | 30.0 |
60–69 | 7 | 23.3 |
70–79 | 1 | 3.3 |
Race | ||
White | 25 | 83.3 |
Asian | 4 | 13.3 |
Unknown | 1 | 3.3 |
Performance status | ||
0 | 18 | 60.0 |
1 | 10 | 33.3 |
2 | 2 | 6.7 |
Number of prior chemotherapies | ||
1 | 20 | 67 |
2 | 9 | 30 |
3 | 1 | 3 |
Table 2.
Clinical response to treatment.
Endpoint | Number of patients | Percent of cases | |
---|---|---|---|
PFS > 6 months | Yes | 5 | 17% |
No | 24 | 80% | |
Indeterminate | 1 | 3% | |
Clinical response | Partial response | 2 | 7% |
Stable disease | 4 | 13% | |
Progressive disease | 20 | 67% | |
Indeterminate | 4 | 13% |
Fig. 1.
Progression-free and Overall Survival.
The most common grade 3 adverse events were fatigue (n = 4), hypertension (n = 4), neutropenia (n = 4), anemia (n = 3), abdominal pain (n = 3), and leukopenia (n = 3). Grade 4–5 adverse events included: thrombocytopenia (n = 5), anemia (n = 2), acute kidney Injury (n =1), stroke (n = 1), and allergic reaction (n = 1). Three grade 5 events were reported. One patient died from a stroke which was attributed as possibly related to treatment. Two patients died due to disease. (Table 3).
Table 3.
Adverse events.
Grade |
|||||
---|---|---|---|---|---|
1 | 2 | 3 | 4 | 5 | |
Blood/Lymphatics | |||||
Anemia | 6 | 12 | 3 | 2 | 0 |
White Blood Cell Decreased | 6 | 9 | 3 | 0 | 0 |
Lymphocyte count decreased | 2 | 1 | 2 | 0 | 0 |
Neutrophil count decreased | 8 | 4 | 4 | 0 | 0 |
Platelet count decreased | 8 | 3 | 1 | 5 | 0 |
Cardiovascular | |||||
Hypertension | 1 | 5 | 4 | 0 | 0 |
Hypotension | 0 | 1 | 1 | 0 | 0 |
Thromboembolic Event | 0 | 1 | 2 | 0 | 0 |
Gastrointestinal | |||||
Nausea | 15 | 1 | 2 | 0 | 0 |
Vomiting | 7 | 2 | 2 | 0 | 0 |
Abdominal pain | 5 | 4 | 3 | 0 | 0 |
Rectal hemorrhage | |||||
Nervous system | |||||
Headache | 6 | 2 | 2 | 0 | 0 |
Stroke | 0 | 0 | 0 | 0 | 1 |
Renal | |||||
Creatinine increased | 0 | 0 | 1 | 2 | 0 |
Urinary tract infection | 1 | 0 | 1 | 0 | 0 |
Acute kidney injury | 0 | 0 | 1 | 1 | 0 |
Respiratory | |||||
Dyspnea | 0 | 3 | 2 | 0 | 0 |
Pleural Effusion | 0 | 0 | 1 | 0 | 0 |
Metabolism/nutrition | |||||
Hypokalemia | 1 | 0 | 2 | 0 | 0 |
Hypoalbuminemia | 1 | 2 | 2 | 0 | 0 |
General/Skin | |||||
Fatigue | 9 | 7 | 4 | 0 | 0 |
Pain | 1 | 2 | 2 | 0 | 0 |
Palmar-Plantar Erythrodysesthesia Syndrome | 2 | 0 | 1 | 0 | 0 |
4. Discussion
Compared to other epithelial cell types, clear cell ovarian carcinomas have a poorer prognosis [1–6]. Given that the molecular characteristic of clear cell ovarian cancers is similar to that of clear cell renal carcinoma, we hypothesized that effective and approved novel targeting agents for renal clear cell cancers may also be active in ovarian clear cell cancers. VHL-associated tumors, including most renal and clear cell ovarian cancers, produce high levels of vascular endothelial growth factor (VEGF) and platelet-derived growth factor receptor (PDGFR) alpha expression [26–31]. These investigations resulted in the development of biologic agents targeting the VHL-HIF pathway and angiogenesis in renal cell cancer, including tyrosine kinase and mTOR inhibitors such as sunitinib, sorafenib, and temsirolimus [32]. Sunitinib is a highly potent, selective inhibitor of protein tyrosine kinases, including VEGF-R and PDGF-R [16–20].
Sunitinib is an active FDA-approved agent against solid tumors including renal cell cancer and gastrointestinal stromal tumors. The safety and efficacy of sunitinib has been evaluated in three phase II trials in ovarian cancers. The National Cancer Institute of Canada Clinical Trials Group treated 30 recurrent platinum-sensitive ovarian cancer patients with sunitinib; of which 20 (67%) had serous and 3 (6%) had clear cell cancers. The response rate was approximately 3% with a median PFS of four months [21]. These authors concluded that sunitinib has modest activity in recurrent platinum sensitive disease. In a European study, the AGO investigators performed a randomized multicenter phase II trial in 73 platinum-resistant ovarian cancer patients and showed a 16.7% response with a median PFS and OS of 4.8 and 13.6 months, respectively [22]. In another phase II study from United States, Campos et al. treated 36 recurrent and refractory epithelial ovarian cancer patients, of which five cases were clear cell, and found a response rate of 8.3% with a 6-month PFS of 36% [23].
Given the molecular similarities between renal and ovarian clear cell cancers and the activity of sunitinib in epithelial ovarian and renal cell cancers in prior clinical trials, we anticipated that sunitinib may have significant activity in clear cell ovarian cancer. The FDA approved sunitinib for renal cell cancer based on a randomized trial of treatment-naïve metastatic renal cell cancer patients and showed an objective response rate of 27.5% in the sunitinib arm compared to only 5.3% in the interferon-α arm [33]. In addition to the molecular similarities to renal cell cancers, clear cell ovarian tumors have frequent mutational inactivation of the Von Hippel-Lindau (VHL) pathway associated angiogenesis and disease progression, we anticipated that the anti-VEGF activity of sunitinib would be effective in the treatment of clear cell ovarian cancers patients [11,12,13]. However, we found minimal activity with a response rate of only 6.7% in recurrent clear cell ovarian cancer patients in this current study. These unanticipated findings may be explained by the differences in the biology of clear cell cancers and selection of patients.
In light of the recent discoveries on the pathogenesis of clear cell cancers since the conception of this current study, our results were not completely unexpected. While we utilized histology to select patients who may benefit from sunitinib, the current data indicate that directing targeted therapy based on molecular tumor aberrations may be a more rational approach for identifying promising agents in a target group of patients. Recent studies have shown that clear cell ovarian cancers may arise from malignant transformation of endometriosis with a shared genetic lineage [34–35]. AR1D1A, a tumor suppressor gene that encodes BAF250a chromatin remodeling protein, is mutated in nearly half of endometriosis-associated clear cell cancers [36]. Colony stimulating factor-1 receptor is a lethal target of AR1D1A-deficient endometriosis associated clear cell ovarian carcinoma [37]. In vitro inhibition of CSF1 selectively inhibited the proliferation of AR1D1A-deficient endometriosis associated clear cell ovarian cancer cell lines. Given these findings, the NRG (GOG283) is currently evaluating dasatinib (NSC #732517 IND #120636) in women with clear cell ovarian cancer with retention or loss BAF250a tumor expression to enhance the selection of patients and clinical activity of these targeted therapies.
Clinical trials evaluating single-agent biologics in clear cell ovarian cancer have shown minimal to modest activity [39–41]. (Table 4) In a recent trial of platinum-resistant ovarian cancer patients, AUREL1A investigators showed that the combination of chemotherapy with biologic agents was superior to chemotherapy alone [14]. In a subsequent retrospective study from our research group, we also found that patients who had chemotherapy combined with bevacizumab had better outcomes compared those with bevacizumab alone [38]. Therefore, it is possible that combination treatment rather than single-agent biologic or chemotherapy alone may be required to obtain sufficient activity in recurrent and resistant clear cell ovarian cancer.
Table 4.
Clinical trials for clear cell ovarian cancer.
Agent | Year open | Author, organization | Number of patients | Results/status |
---|---|---|---|---|
Chemotherapy | ||||
Docetaxel-irinotecan [45] | 2013 | Ueda, Osaka University | 62 | RR = 53% Disease control rate = 67% |
Paclitaxel + carboplatin vs. irinotecan + cisplatin [46] | 2010 | Takakura, JGOG | 99 | PFS tended to be longer with irinotecan + cisplatin in small volume disease |
Paclitaxel + carboplatin vs. Irinotecan + Cisplatin 47] | 2016 | Sugiyama, JGOG/GCIG | 667 | No significant survival benefit was found for Irinotecan + Cisplatin |
Biologic | ||||
Sunitinib | 2010 | Chan, NRG | 35 | RR = 20%, 6 mo PFS17% |
Paclitaxel-carboplatin-temsirolimus | 2010 | Farley, NRG | 90 | Completed |
Dasatinib | 2014 | Hyman, NRG | 62 | Suspended |
Nintedanib | 2015 | Glasspool, NHS | 120 | Recruiting |
Cabozantanib (XL184) | 2015 | Farley, NRG | NA | Closed after first stage |
ENMD-2076 | 2013 | Oza, University Health | 36 | Recruiting |
Everolimus or temsirolimus | NA | Vicus, NRG | NA | Under development |
Pazopanib | NA | Glen, NRG | NA | Under development |
RR = response rate; PFS = progression-free survival.
This current study included only clear cell ovarian cancer patients with recurrent, persistent, and most with platinum-resistant disease. We selected for all primary tumors with at least 50% clear cell histomorphology and negative for expression ofWT-1 antigen and estrogen receptor based on immunohistochemistry. All patients entered on trial had a primary tumor with at least 50% clear cell histomorphology and thus did not require a biopsy of recurrent tumor. Nevertheless, it may be more informative to obtain biopsies of recurrent disease to assure the selection of recurrent clear cell cancers based on histomorphologic criteria and genomic characteristics. Unlike renal cell cancers, recurrent ovarian clear cell cancers may be more heterogeneous and require molecular characterization of relapsed tumors to individualize effective treatments.
In clear cell ovarian cancer, numerous trials have evaluated chemotherapeutic and biologic agents with modest results [39]. 1mmune modulators targeting the microenvironment of clear cell cancers may have promise. In the CheckMate 025 trial with advanced recurrent renal cell cancer, nivolumab, a programmed cell death receptor 1 inhibitor, improved the median overall survival by 5.4 months compared to everolimus [40]. Further, the combination of nivolumab plus ipilimumab, a cytotoxic T lymphocyte-associated antigen 4 inhibitor, improved the overall survival compared with sunitinib, resulting in an update of the European Association of Urology Guidelines Recommendations [41]. In recurrent platinum-resistant ovarian cancer, Hamanishi et al. treated twenty patients with nivolumab and found an overall response rate of 15%. Of the two patients with a durable complete response, one had clear cell carcinoma [42]. Pembrolizumab, a programmed cell death receptor 1 inhibitor, was recently approved for the treatment of unresectable or metastatic, microsatellite instability high or mismatch repair deficient solid tumors, regardless of tumor site or histology [43]. In a case report of chemotherapy and radiation-resistant ovarian cancer, pembrolizumab led to an exceptional response with tumor harboring a PD-L1 gene structural variations causing aberrant PD-L1 expression [44].
This is the first study designed to treat patients with clear cell ovarian cancer. All tumors underwent central path review with histomorphologic validation, standardized treatment, and surgical staging by gynecologic oncologists. There are limitations that may have contributed to the minimal activity demonstrated with this agent during this trial. It is possible that the immune and microenvironment of tumors may be different and thus the same agent may not be as effective across organs. Moreover, the intra-tumoral heterogeneity of clear cell ovarian cancers may require personalized molecular therapy. Further, adaptive clinical trials may be necessary to validate tumor and serum biomarkers to advance novel treatments for clear cell ovarian cancer patients.
HIGHLIGHTS.
Efficacy of sunitinib in recurrent or persistent clear cell ovarian cancer
16.7% of patients had PFS N6months and 6.7% had responses with PFS of 2.7 months.
Common adverse events were fatigue, hypertension, neutropenia, and anemia.
Sunitinib was tolerable but had minimal activity.
Acknowledgements
This study was supported by National Cancer Institute grants to the Gynecologic Oncology Group (GOG) Administrative Office (CA 27469), the Gynecologic Oncology Group Statistical Office (CA 37517), NRG Oncology (1 U10 CA180822) and NRG Operations (U10CA180868), and Pfizer. The following Gynecologic Oncology institutions participated in this study: MD Anderson Cancer Center, UCSF-Mount Zion, Washington University School of Medicine, Duke University Medical Center, Abington Memorial Hospital, Cleveland Clinic Foundation, Seoul National University Hospital, University of Alabama at Birmingham, University of Colorado Cancer Center – Anschutz Cancer Pavilion, University of California Medical Center at Irvine-Orange Campus, Rush University Medical Center, University of Oklahoma Health Sciences Center, Case Western Reserve University, Carolinas Medical Center/Levine Cancer Institute, University of Iowa Hospitals and Clinics and Women and Infants Hospital. We would like to acknowledge The Fisher Family Foundation, Denise Cobb Hale, and Dr. John A. Kerner for their generous administrative research support.
Footnotes
ClinicalTrials.gov number, NCT00979992.
Conflicts of interest
Dr. Monk discloses that St. Joseph’s Hospital institution has received research grants from Novartis, Amgen, Lilly Genentech, Janssen/Johnson & Johnson, Array, TESARO, and Morphotek. He has received honoraria for speaker bureaus from Roche/Genentech, AstraZeneca, Janssen/Johnson & Johnson, Myriad, TESARO, and Clovis. Additionally, Dr. Monk has been a consultant for Roche/Genentech, Merck, TESARO, AstraZeneca, Gradalis, Advaxis, Cerulean, Amgen, Vemillion, ImmunoGen, Pfizer, Bayer, NuCana, Insys, GlaxoSmithKline, Verastem, Mateon (formally OxiGENE), PPD, Clovis, Precision Oncology, Biodesix, Perthera, ImmunoGen and Cue. Dr. Chan disclose that he has received honoraria for speaker bureaus and/or consultation fees from Roche/Genentech, AstraZeneca, Tesaro, and Clovis.
References
- [1].Sugiyama T, Kamura T, Kigawa J, et al. , Clinical characteristics of clear cell carcinoma of the ovary: a distinct histologic type with poor prognosis and resistance to platinum-based chemotherapy, Cancer 88 (2000) 2584–2589. [PubMed] [Google Scholar]
- [2].Montag AG, Jenison EL, Griffiths CT, et al. , Ovarian clear cell carcinoma. A clinico-pathologic analysis of 44 cases, Int. J. Gynecol. Pathol 8 (1989) 85–96. [DOI] [PubMed] [Google Scholar]
- [3].Chan JK, Teoh D, Hu JM, et al. , Do clear cell ovarian carcinomas have poorer prognosis compared to other epithelial cell types? A study of 1411 clear cell ovarian cancers, Gynecol. Oncol 109 (2008) 370–376. [DOI] [PubMed] [Google Scholar]
- [4].Chan JK, Tian C, Fleming GF, et al. , The potential benefit of 6 vs. 3 cycles of chemo-therapy in subsets of women with early-stage high-risk epithelial ovarian cancer: an exploratory analysis of a gynecologic oncology group study, Gynecol. Oncol 116 (2010) 301–306. [DOI] [PubMed] [Google Scholar]
- [5].Chan JK, Tian C, Monk BJ, et al. , Prognostic factors for high-risk early-stage epithetlial ovarian cancer: a gynecologic oncology group study, Cancer 112 (2008) 2202–2210. [DOI] [PubMed] [Google Scholar]
- [6].Chan JK, Cheung MK, Husain A, et al. , Patterns and progress in ovarian cancer over 14 years, Obstet. Gynecol 108 (2006) 521–528. [DOI] [PubMed] [Google Scholar]
- [7].Jenison EL, Montag AG, Griffiths CT, et al. , Clear cell adenocarcinoma of the ovary: a clinical analysis and comparison with serous carcinoma, Gynecol. Oncol. 32 (1989) 65–71. [DOI] [PubMed] [Google Scholar]
- [8].Goff BA, Sainz De La Cuesta R, Muntz HG, et al. , Clear cell carcinoma of the ovary: a distinct histologic type with poor prognosis and resistance to platinum-based chemotherapy in stage III disease, Gynecol. Oncol 60 (1996) 412–417. [DOI] [PubMed] [Google Scholar]
- [9].Behbakht K, Randall TC, Benjamin I, et al. , Clinical characteristics of clear cell carcinoma of the ovary, Gynecol. Oncol 70 (1998) 255–258. [DOI] [PubMed] [Google Scholar]
- [10].Zorn KK, Bonome T, Gangi L, et al. , Gene expression profiles of serous, endometrioid, and clear cell subtypes of ovarian and endometrial cancer, Clin. Cancer Res 11 (2005) 6422–6430. [DOI] [PubMed] [Google Scholar]
- [11].Simsir A, Palacios D, Linehan WM, et al. , Detection of loss of heterozygosity at chromosome 3p25-26 in primary and metastatic ovarian clear-cell carcinoma: utilization of microdissection and polymerase chain reaction in archival tissues, Diagn. Cytopathol 24 (2001) 328–332. [DOI] [PubMed] [Google Scholar]
- [12].Yoneda J, Kuniyasu H, Crispens MA, et al. , Expression of angiogenesis-related genes and progression of human ovarian carcinomas in nude mice, J. Natl. Cancer Inst 90(1998)447–454. [DOI] [PubMed] [Google Scholar]
- [13].Nakanishi Y, Kodama J, Yoshinouchi M, et al. , The expression of vascular endothelial growth factor and transforming growth factor-beta associates with angiogenesis in epithelial ovarian cancer, Int. J. Gynecol. Pathol. 16 (1997) 256–262. [DOI] [PubMed] [Google Scholar]
- [14].Pujade-Lauraine E, Hilpert F, Weber B, et al. , Bevacizumab combined with chemotherapy for platinum-resistant recurrent ovarian cancer: the AURELIA open-label randomized phase III trial, J. Clin. Oncol 32 (2014) 1302–1308. [DOI] [PubMed] [Google Scholar]
- [15].Coleman RL, Brady MF, Herzog TJ, et al. Bevacizumab and paclitaxel-carboplatin chemotherapy and secondary cytoreduction in recurrent, platinum-sensitive ovarian cancer (NRG Oncology/Gynecologic Oncology Group study GOG-0213): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 18:779–791. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].O’Farrell AM, Abrams TJ, Yuen HA, et al. , SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo, Blood 101 (2003) 3597–3605. [DOI] [PubMed] [Google Scholar]
- [17].Schueneman AJ, Himmelfarb E, Geng L, et al. , SU11248 maintenance therapy prevents tumor regrowth after fractionated irradiation of murine tumor models, Cancer Res 63 (2003)4009–4016. [PubMed] [Google Scholar]
- [18].Abrams TJ, Murray LJ, Pesenti E, et al. ,Preclinical evaluation of the tyrosine kinase inhibitor SU11248 as a single agent and in combination with “standard of care” therapeutic agents for the treatment of breast cancer, Mol. Cancer Ther 2 (2003) 1011–1021. [PubMed] [Google Scholar]
- [19].Mendel DB, Laird AD, Xin X, et al. , In vivo antitumor activity of SU11248, a novel tyrosine kinase inhibitor targeting vascular endothelial growth factor and platelet-derived growth factor receptors: determination of a pharmacokinetic/pharmacodynamic relationship, Clin. Cancer Res 9 (2003) 327–337. [PubMed] [Google Scholar]
- [20].Motzer RJ, Michaelson MD, Redman BG, et al. , Activity of SU11248, a multitargeted inhibitor of vascular endothelial growth factor receptor and platelet-derived growth factor receptor, in patients with metastatic renal cell carcinoma, J. Clin. Oncol 24(2006) 16–24. [DOI] [PubMed] [Google Scholar]
- [21].Biagi JJ, Oza AM, Chalchal HI, et al. , A phase II study of sunitinib in patients with recurrent epithelial ovarian and primary peritoneal carcinoma: an NCIC clinical trials group study, Ann. Oncol 22 (2011) 335–340. [DOI] [PubMed] [Google Scholar]
- [22].Baumann KH, du Bois A, Meier W, et al. , A phase II trial (AGO 2.11) in platinum-resistant ovarian cancer: a randomized multicenter trial with sunitinib (SU11248) to evaluate dosage, schedule, tolerability, toxicity and effectiveness of a multitargeted receptor tyrosine kinase inhibitor monotherapy, Ann. Oncol 23 (2012) 2265–2271. [DOI] [PubMed] [Google Scholar]
- [23].Campos SM, Penson RT, Matulonis U, et al. , A phase II trial of Sunitinib malate in recurrent and refractory ovarian, fallopian tube and peritoneal carcinoma, Gynecol. Oncol 128 (2013) 215–220. [DOI] [PubMed] [Google Scholar]
- [24].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 92 (2000) 205–216. [DOI] [PubMed] [Google Scholar]
- [25].Sill MW, Rubinstein L, Litwin S, et al. , A method for utilizing co-primary efficacy outcome measures to screen regimens for activity in two-stage phase II clinical trials, Clin Trials 9 (2012) 385–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [26].Zbar B, Brauch H, Talmadge C, et al. , Loss of alleles of loci on the short arm of chromosome 3 in renal cell carcinoma, Nature 327 (1987) 721–724. [DOI] [PubMed] [Google Scholar]
- [27].Motzer RJ, Russo P, Systemic therapy for renal cell carcinoma,J. Urol. 163 (2000) 408–417. [PubMed] [Google Scholar]
- [28].Brown LF, Berse B, Tognazzi K, et al. , Vascular permeability factor mRNA and protein expression in human kidney, Kidney Int. 42 (1992) 1457–1461. [DOI] [PubMed] [Google Scholar]
- [29].Takahashi A, Sasaki H, Kim SJ, et al. , Markedly increased amounts of messenger RNAs for vascular endothelial growth factor and placenta growth factor in renal cell carcinoma associated with angiogenesis, Cancer Res. 54 (1994) 4233–4237. [PubMed] [Google Scholar]
- [30].Schraml P, Struckmann K, Hatz F, et al. , VHL mutations and their correlation with tumour cell proliferation, microvessel density, and patient prognosis in clear cell renal cell carcinoma, J. Pathol 196 (2002) 186–193. [DOI] [PubMed] [Google Scholar]
- [31].Matei D, Emerson RE, Lai YC, et al. , Autocrine activation of PDGFRalpha promotes the progression of ovarian cancer, Oncogene 25 (2006) 2060–2069. [DOI] [PubMed] [Google Scholar]
- [32].Cowey CL, Rathmell WK, VHL gene mutations in renal cell carcinoma: role as a biomarker of disease outcome and drug efficacy, Curr. Oncol. Rep 11 (2009) 94–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [33].Motzer RJ, Hutson TE, Tomczak P, et al. , Sunitinib versus interferon alfa in meta-static renal-cell carcinoma, N. Engl. J. Med 356 (2007) 115–124. [DOI] [PubMed] [Google Scholar]
- [34].Ness RB, Endometriosis and ovarian cancer: thoughts on shared pathophysiology, Am. J. Obstet. Gynecol. 189 (2003) 280–294. [DOI] [PubMed] [Google Scholar]
- [35].Vigano P, Somigliana E, Chiodo I, et al. , Molecular mechanisms and biological plausibility underlying the malignant transformation of endometriosis: a critical analysis, Hum. Reprod. Update 12 (2006) 77–89. [DOI] [PubMed] [Google Scholar]
- [36].Wiegand KC, Shah SP, Al-Agha OM, et al. , ARID1A mutations in endometriosis-associated ovarian carcinomas, N. Engl. J. Med 363 (2010) 1532–1543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Mashkani B, Griffith R, Ashman LK, Colony stimulating factor-1 receptor as a target for small molecule inhibitors, Bioorg. Med. Chem. 18 (2010) 1789–1797. [DOI] [PubMed] [Google Scholar]
- [38].Fuh KC, Secord AA, Bevis KS, et al. , Comparison of bevacizumab alone or with chemotherapy in recurrent ovarian cancer patients, Gynecol. Oncol 139 (2015) 413–418. [DOI] [PubMed] [Google Scholar]
- [39].Chan JK, Kapp DS, Clear cell ovarian Cancer: optimum management and prognosis remain hazy, Gynecol. Oncol 147 (2017) 237–239. [DOI] [PubMed] [Google Scholar]
- [40].Motzer RJ, Escudier B, Mcdermott DF, et al. , Nivolumab versus everolimus in advanced renal-cell carcinoma, N. Engl. J. Med 373 (2015) 1803–1813. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Powles T, Albiges L, Staehler M, et al. , Updated European Association of Urology guidelines recommendations for the treatment of first-line metastatic clear cell renal cancer, Eur. Urol (2017) 10.1016/j.eururo.2017.11.016 pii: S0302-2838(17)31001-1. [Epub ahead ofprint]. [DOI] [PubMed] [Google Scholar]
- [42].Hamanishi J, Mandai M, Ikeda T, et al. , Safety and antitumor activity of anti-PD-1 antibody, Nivolumab, in patients with platinum-resistant ovarian Cancer, J. Clin. Oncol 33 (2015) 4015–4022. [DOI] [PubMed] [Google Scholar]
- [43].Le DT, Uram JN, Wang H, et al. , PD-1 blockade in tumors with mismatch-repair deficiency, N. Engl. J. Med. 372 (2015) 2509–2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [44].Bellone S, Buza N, Choi J, et al. , Exceptional response to pembrolizumab in a metastatic, chemotherapy/radiation resistant ovarian cancer patient harboring a CD274/ PD-L1-genetic rearrangement, Clin. Cancer Res (2018) 10.1158/1078-0432.CCR-17-1805 [Epub ahead ofprint] PubMed PMID: 29351920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [45].Ueda Y, Miyatake T, Nagamatsu M, et al. , A phase II study of combination chemotherapy using docetaxel and irinotecan for TC-refractory or TC-resistant ovarian carcinomas (GOGO-OV2 study) and for primary clear or mucinous ovarian carcinomas (GOGO-OV3 study), Eur.J. Obstet. Gynecol. Reprod. Biol 170 (2013) 259–263. [DOI] [PubMed] [Google Scholar]
- [46].Takakura S, Takano M, Takahashi F, et al. , Randomized phase II trial of paclitaxel plus carboplatin therapy versus irinotecan plus cisplatin therapy as first-line chemotherapy for clear cell adenocarcinoma of the ovary: a JGOG study, Int. J. Gynecol. Cancer 20 (2010) 240–247. [DOI] [PubMed] [Google Scholar]
- [47].Sugiyama T, Okamoto A, Enomoto T, et al. , Randomized phase III trial of irinotecan plus cisplatin compared with paclitaxel plus carboplatin as first-line chemotherapy for ovarian clear cell carcinoma: JGOG3017/GCIG trial, J. Clin. Oncol 34 (2016) 2881–2887. [DOI] [PubMed] [Google Scholar]