Skip to main content
Journal of Clinical Oncology logoLink to Journal of Clinical Oncology
. 2011 May 2;29(16):2259–2265. doi: 10.1200/JCO.2010.32.6397

Phase II Trial of Bevacizumab in Recurrent or Persistent Endometrial Cancer: A Gynecologic Oncology Group Study

Carol Aghajanian 1,, Michael W Sill 1, Kathleen M Darcy 1, Benjamin Greer 1, D Scott McMeekin 1, Peter G Rose 1, Jacob Rotmensch 1, Mack N Barnes 1, Parviz Hanjani 1, Kimberly K Leslie 1
PMCID: PMC3107744  PMID: 21537039

Abstract

Purpose

Bevacizumab, a recombinant humanized monoclonal antibody against vascular endothelial growth factor-A (VEGF-A), has clinical activity in multiple tumor types. We conducted a phase II trial to assess the activity and tolerability of single-agent bevacizumab in recurrent or persistent endometrial cancer (EMC).

Patients and Methods

Eligible patients had persistent or recurrent EMC after receiving one to two prior cytotoxic regimens, measurable disease, and Gynecologic Oncology Group performance status of ≤ 2. Treatment consisted of bevacizumab 15 mg/kg intravenously every 3 weeks until disease progression or prohibitive toxicity. VEGF-A was assessed by immunohistochemistry in archival tumor and by enzyme-linked immunosorbent assay in pretreatment plasma. Primary end points were progression-free survival (PFS) at 6 months and overall response rate.

Results

Fifty-six patients were enrolled. Fifty-two patients were eligible and evaluable. Median age was 62 years, and prior treatment consisted of one or two regimens in 33 (63.5%) and 19 (36.5%) patients, respectively. Twenty-nine patients (55.8%) received prior radiation. Adverse events were consistent with those expected with bevacizumab treatment. No GI perforations or fistulae were seen. Seven patients (13.5%) experienced clinical responses (one complete response and six partial responses; median response duration, 6.0 months), and 21 patients (40.4%) survived progression free for at least 6 months. Median PFS and overall survival times were 4.2 and 10.5 months, respectively. Suggested associations were observed between high VEGF-A and adjusted hazard of death or tumor response when evaluated in tumor/plasma or plasma, respectively.

Conclusion

Bevacizumab is well tolerated and active based on PFS at 6 months in recurrent or persistent EMC and warrants further investigation.

INTRODUCTION

Endometrial cancer affects an estimated 40,000 women in the United States every year, and long-term outcomes for patients with advanced-stage or recurrent disease are poor.1 Investigations focusing on new approaches to improve outcomes in this patient population are warranted.

There have been several randomized studies addressing the issue of optimal therapy for this group of patients. The most recently reported study randomly assigned 263 patients to doxorubicin and cisplatin (AP) versus paclitaxel, doxorubicin, and cisplatin (TAP). TAP was superior to AP in terms of overall response rate (ORR; 57% v 34%, respectively; P < .01), median progression-free survival (PFS; 8.3 v 5.3 months, respectively; P < .01), and median overall survival (15.3 v 12.3 months, respectively; P = .037).2 This improved efficacy came at the cost of increased toxicity. In an attempt to address this issue, a study of TAP compared with paclitaxel and carboplatin has completed accrual and is in follow-up (ClinicalTrials.gov identifier: NCT00063999). Once this initial therapy has been delivered, there are limited treatment options. Hormonal therapies, when given to chemotherapy-naive patients, can result in responses, but these responses are of short duration.37 Targeted therapies, other than hormonal therapies, have yet to be implemented in clinical practice. Completed studies have evaluated agents that target mammalian target of rapamycin (temsirolimus,8,9 everolimus,10 and ridaforolimus11), human epidermal growth factor receptor 2 (trastuzumab12), epidermal growth factor receptor (erlotinib,13 gefitinib,14 and cetuximab15), and vascular endothelial growth factor (VEGF; sunitinib,16 sorafenib,17 and thalidomide18), showing none to modest activity.

Bevacizumab monotherapy has been evaluated in persistent or recurrent ovarian and cervical cancers with positive results. A phase II trial of bevacizumab in patients with ovarian cancer (one to two prior chemotherapy regimens) demonstrated a single-agent response rate of 21%, with 40.3% of patients surviving progression free for at least 6 months.19 A second phase II trial in patients with platinum-resistant ovarian cancer (one to three prior chemotherapy regimens) demonstrated a response rate of 15.9%.20 In patients with cervical cancer (one or two prior chemotherapy regimens), a response rate of 10.9% was observed, with 23.9% of patients surviving progression free for at least 6 months.21

A phase II trial of single-agent bevacizumab was conducted in patients with recurrent or persistent EMC. The primary objective was to evaluate efficacy in terms of both the probability of surviving progression free for at least 6 months (PFS at 6 months) and clinical response.

PATIENTS AND METHODS

Patient Selection

Eligible patients met the following criteria: histologic confirmation of the primary tumor by central pathology review by the Gynecologic Oncology Group (GOG) Pathology Committee; GOG performance status of 0 to 2; measurable disease by Response Evaluation Criteria in Solid Tumors ( RECIST)22; one or two prior cytotoxic regimens; discontinuation of prior chemotherapy at least 3 weeks before registration and hormonal therapy at least 1 week before registration; recovery of the effects of recent surgery, radiotherapy, or chemotherapy; freedom from active infection requiring antibiotics; adequate hematologic (absolute neutrophil count ≥ 1,000/μL and platelets ≥ 100,000/μL), renal (serum creatinine ≤ 1.5× the institutional upper limit of normal [ULN] and urine protein-to-creatinine ratio < 1), hepatic (serum bilirubin ≤ 1.5× ULN and AST and alkaline phosphatase ≤ 2.5× ULN), and coagulation (prothrombin time such that international normalized ratio ≤ 1.5 or between 2 and 3 for patients receiving stable doses of therapeutic warfarin, and partial thromboplastin time ≤ 1.5× ULN) laboratory values; left ventricular ejection fraction ≥ 50% (for patients who received prior anthracycline); negative serum pregnancy test before study entry and agreement to practice an effective form of contraception in patients of childbearing potential; a signed approved informed consent in accordance with federal, state, and local requirements; and authorization permitting release of personal health information.

Patients were ineligible if they met any of the following criteria: prior treatment with bevacizumab or other VEGF pathway–targeted therapy; prior treatment with any noncytotoxic therapy (other than hormonal therapy); other malignancies (except nonmelanomatous skin cancer) evident within 5 years or prior cancer treatment that contradicts eligibility; nonhealing wound, ulcer, or bone fracture; abdominal fistula, GI perforation, or intra-abdominal abscess within 28 days; active bleeding or pathologic condition that carries high risk of bleeding; known CNS disease; clinically significant cardiovascular disease; and major surgical procedure within 28 days or anticipated on study.

Treatment

Enrolled patients were to receive bevacizumab 15 mg/kg intravenously every 21 days with no dose modification except for at least a 10% change in body weight. Treatment was planned until disease progression or adverse events prohibited further therapy. Toxicity was monitored with history, physical examination, and laboratory assessment before each treatment cycle, with adverse events defined and graded according to Common Terminology Criteria for Adverse Events (version 3.0). Bevacizumab was held for grade 3 nonhematologic toxicity for a maximum of 4 weeks to allow recovery to ≤ grade 1. Bevacizumab was discontinued for grade 4 allergic reactions; grade 2 (new or worsened) and 3 to 4 arterial thrombosis; symptomatic grade 4 venous thrombosis; grade 4 hypertension; grade 4 (or nephrotic syndrome) proteinuria; grade 4 hemorrhage; GI perforation, GI leak, or fistula; and reversible posterior leukoencephalopathy syndrome. Specific guidelines were implemented for modifying the treatment regimen in the event of hypertension, venous thrombosis, proteinuria, and hemorrhage.

Evaluation Criteria

Activity of bevacizumab was assessed according to RECIST16 either by palpation before each cycle or by computed tomography or magnetic resonance imaging at baseline, every other cycle for the first 6 months, and every four cycles thereafter.

VEGF-A Assessments

VEGF-A expression was detected by immunohistochemistry in archival formalin-fixed, paraffin-embedded primary, metastatic, or recurrent tumor using antigen retrieval (30 minutes in 1 mmol/L EDTA in a Nordicware pressure cooker) and the Ventana NexES automated immunohistochemistry staining system with the SC-152 (A-20) primary antibody against VEGF-A (Santa Cruz Biotechnology, Santa Cruz, CA). Controls for the immunohistochemistry assay included formalin-fixed, paraffin-embedded MCF7-WT human breast cancer cells and poorly differentiated endometrial cancer cells for high VEGF-A; doxorubicin-resistant MCF7-40F human breast cancer cells and well-differentiated endometrial cancer cells for low VEGF-A; and invasive breast carcinomas that do not express VEGF-A. Cytoplasmic VEGF-A staining was categorized by intensity (0 to 3+ relative to the negative controls and controls for high VEGF-A) and percent positive tumor cells (0% to 100%). VEGF-A concentration was quantified in picograms per milliliter by enzyme-linked immunosorbent assay (ELISA) using the DVE00 quantikine human VEGF immunoassay (R&D Systems, Minneapolis, MN) and recombinant human VEGF as instructed and previously described9 in pretreatment plasma prepared from blood drawn in a purple-top tube with EDTA. Assays for VEGF-A were performed by the GOG Core Laboratory for Targets and Receptors under the direction and supervision of the director (K.K.L.).

Statistics

The primary objective evaluated the efficacy of bevacizumab through the frequency of patients with objective tumor responses and the frequency of patients who survived progression free for at least 6 months. The null hypothesis relating to uninteresting levels of activity was determined from an analysis of a historical data set based on a similar population of patients where the levels of activity were believed to be inactive to modestly active (Table 1). The null hypothesis jointly specified the probability of a patient experiencing a tumor response to within ≤ 10% and the probability of a patient being progression free at 6 months to within ≤ 15%. Clinically significant differences were 20% increases in the probability of either event (ie, probability of 30% or 35%, respectively). Using a method by Sill and Yothers,32 a two-stage design was used with a goal of limiting patient exposure to inactive agents while restricting the probabilities of type I and type II errors to approximately 10%. Nineteen patients were targeted for accrual to the first stage, but the number was allowed to deviate for administrative flexibility. If the regimen demonstrated sufficient activity (> two or three patients with responses or PFS at 6 months, respectively), then the study targeted 42 patients (cumulatively) in stage 2. If more than seven or 10 patients had responses or were alive and progression free at 6 months, respectively, the regimen was deemed worthy of further study.

Table 1.

Historical Control Characteristics

Protocol Agent No. of Evaluable Patients Probability PFS at 6 Months
Response
Product Limit Estimate SE No. of Patients %
GOG 129-B23 Etoposide 25 0.08 0.05 0 0
GOG 129-C24 Paclitaxel 48 0.21 0.06 12 25
GOG 129-E25 Dactinomycin 27 0.04 0.04 3 11
GOG 129-H26 Liposomal doxorubicin 43 0.23 0.06 4 9
GOG 129-I27 Pyrazoloacridine 25 0.16 0.07 1 4
GOG 129-J28 Topotecan 28 0.25 0.08 2 7
GOG 129-K29 Oxaliplatin 52 0.27 0.06 7 13
GOG 129-L30 Irofulven 25 0.28 0.09 1 4
GOG 129-M31 Flavopiridol 21 0 0 0 0
GOG 229-B18 Thalidomide 24 0.08 0.06 3 12
GOG 229-E Bevacizumab 52 0.40 0.07 7 13.5

Abbreviations: PFS, progression-free survival; GOG, Gynecologic Oncology Group.

Biomarkers were screened for associations with demographic characteristics and clinical outcomes in an exploratory fashion to yield hypotheses for further testing. Characteristics of interest included age, performance status, prior treatment, cell grade, response, PFS at 6 months, and the hazards of progression and death. Where appropriate, biomarkers were dichotomized into high versus low values using the observed median as a threshold. Some characteristics were examined as ordinal categorical variables (eg, response with progressive disease < stable disease < response). Associations were examined with correlations (Kendall's τ-b and Spearman rank correlation coefficient), χ2 tests, and the Cox proportional hazards model. If P < .05, an association was designated as suggested. Given the small sample sizes, a lack of a suggested association should not be interpreted as definitive.

RESULTS

Patient Characteristics

From March 2006 to October 2007, GOG member institutions enrolled 56 patients onto this trial. Four patients were deemed ineligible because of second primary cancers (n = 2), inadequate pathology for central review (n = 1), and wrong primary cancer (n = 1); the remaining 52 patients were assessable for toxicity and response. Patient characteristics are listed in Table 2. A total of 368 cycles were administered, and a median of five cycles were given (range, one to 27 cycles). Twenty patients (38.5%) received eight or more cycles.

Table 2.

Patient Characteristics and Activity of Bevacizumab

Characteristics No. of Patients (N = 52) %
Age, years
    Median 62
    Range 32-84
Performance status
    0 34 65.4
    1 17 32.7
    2 1 1.9
Histology
    Endometrioid 26 50
    Serous 14 26.9
    Mixed epithelial 5 9.6
    Clear cell 4 7.7
    Adenocarcinoma, unspecified 1 1.9
    Mucinous 1 1.9
    Undifferentiated 1 1.9
Tumor grade
    1 3 5.8
    2 12 23.1
    3 37 71.2
No. of prior regimens
    1 33 63.5
    2 19 36.5
Prior radiation therapy 29 55.8
Response
    Complete response 1 1.9
    Partial response 6 11.5
    Stable disease 26 50.0
    Increasing disease 17 32.7
    Indeterminate 2 3.8
PFS ≥ 6 months
    No 31 59.6
    Yes 21 40.4
No. of treatment cycles
    1 5 9.6
    2 15 28.8
    3 2 3.8
    4 2 3.8
    5 3 5.8
    6 5 9.6
    7 0 0
    8+ 20 38.5

Abbreviation: PFS, progression-free survival.

Adverse Events

As shown in Table 3, safety of bevacizumab in all 52 patients was analyzed descriptively. No GI perforations or fistulae were reported. No treatment-related deaths were reported. One patient had a grade 4 hemorrhage of the stomach, and another patient had a grade 3 hemorrhage of the rectum. Two patients had grade 3 or 4 thrombosis/embolism. One patient had an asymptomatic pulmonary embolus noted on routine computed tomography scan after cycle 2 and was then removed from study after cycle 3 for a new deep venous thrombosis that developed on warfarin. The second patient developed a deep venous thrombosis after cycle 2 and continued study treatment once anticoagulation was instituted. Two patients had grade 3 or 4 proteinuria, and four patients had grade 3 hypertension. One episode of grade 3 hypotension was reported. One patient had grade 2 left ventricular systolic function decrease (comorbidities included prior anthracycline, coronary artery disease, elevated cholesterol, hypertension, and diabetes). The treating investigator listed this adverse event as possibly related to bevacizumab and possibly related to the comorbidities. One patient had a small bowel obstruction. Three patients were removed from study as a result of toxicity (one patient each for recurrent thrombosis, hemoptysis [grade 1, lung metastases], and proteinuria).

Table 3.

Adverse Events (N = 52)

Adverse Event Toxicity Grade* (No. of patients)
0 1 2 3 4 5
Leukopenia 48 4 0 0 0 0
Thrombocytopenia 46 6 0 0 0 0
Neutropenia 50 2 0 0 0 0
Anemia 36 9 6 1 0 0
Hypersensitivity 50 1 1 0 0 0
Rhinitis 50 1 1 0 0 0
Hypertension 43 2 3 4 0 0
Hypotension 50 1 0 1 0 0
Other cardiac 48 3 1 0 0 0
Constitutional 17 23 10 2 0 0
Dermatologic 47 5 0 0 0 0
Nausea 44 6 2 0 0 0
Vomiting 46 4 2 0 0 0
GI 28 21 2 1 0 0
Hemorrhage 40 9 1 1 1 0
Hepatobiliary 51 0 0 1 0 0
Infection 47 0 5 0 0 0
Edema (limb) 48 3 0 1 0 0
Metabolic 30 16 3 1 2 0
Musculoskeletal 48 1 0 3 0 0
Neurosensory 48 4 0 0 0 0
Other neurologic 47 4 0 1 0 0
Ocular/visual 49 3 0 0 0 0
Pain 33 11 4 4 0 0
*

The maximum severity of each adverse event per patient, graded according to Common Terminology Criteria for Adverse Events (version 3.0).

Activity of Bevacizumab

The activity of bevacizumab was analyzed in 52 patients (Table 2). One complete response and six partial responses were observed, for an ORR of 13.5% (90% CI for the true response rate, 6.5% to 27%), with median response duration of 6 months. The histologic subtypes of the observed responses were high-grade serous carcinoma for the complete response and three high-grade serous carcinomas, one endometrioid carcinoma, one clear cell carcinoma, and one adenocarcinoma unspecified for the partial responses. Twenty-one (40.4%; 90% CI for the true proportion, 29% to 53%) of 52 patients were progression free for at least 6 months. The percentage of patients surviving progression free for at least 6 months did not vary significantly across histologic subtype (35% v 36% for endometrioid and high-grade serous carcinomas, respectively). Figure 1 indicates that the median PFS in the study population was 4.17 months. The median overall survival was 10.55 months.

Fig 1.

Fig 1.

Kaplan-Meier plots demonstrating overall survival (OS) and progression-free survival (PFS) for the 52 patients in the study population.

VEGF-A

Immunohistochemical expression of VEGF-A was examined in 40 primary, two metastatic, and two recurrent tumors from 44 of 52 patients. VEGF-A staining intensity was low in 34% (four were negative, and 11 had 1+ staining) and high in 66% (17 exhibited 2+ staining, and 12 displayed 3+ staining) of tumors. The percentage of VEGF-A–positive tumor cells varied from 0% to 100% and was categorized at the median (35%). VEGF-A concentration, quantified by ELISA in pretreatment plasma from 34 of 52 patients, ranged from less than the lower limit of detection (5 pg/mL) to 856 pg/mL and was categorized at the median as low (< 76.9 pg/mL) versus high (≥ 76.9 pg/mL). Of the various associations examined in Appendix Tables A1 and A2 (online only) and Figure 2, a suggested association was observed between high VEGF-A staining intensity (categorized at the median as high or low) and a reduced risk of death (adjusted hazard ratio [HR], 0.350; 95% CI, 0.153 to 0.797), high pretreatment plasma VEGF-A and lack of tumor response (r = −0.382; Fig 2A), and high plasma VEGF-A (categorized at the median as high or low) and worse survival (unadjusted HR, 2.719; 95% CI, 1.160 to 6.374; adjusted HR, 5.298; 95% CI, 2.002 to 14.022; Fig 2C).

Fig 2.

Fig 2.

(A) Scatter plot for vascular endothelial growth factor-A (VEGF-A) concentration in pretreatment plasma in picograms per milliliter by clinical tumor response classified as progressive disease (PD), stable disease (SD), and partial response (PR)/complete response (CR). (B) Kaplan-Meier estimates of progression-free survival by pretreatment plasma VEGF-A categorized at the median as low (< 76.9 pg/mL) versus high (≥ 76.9 pg/mL). (C) Kaplan-Meier estimates of overall survival by pretreatment plasma VEGF-A categorized at the median as low (< 76.9 pg/mL) versus high (≥ 76.9 pg/mL).

DISCUSSION

On the basis of historical controls for inactive cytotoxic and targeted agents investigated in previous GOG phase II trials, bevacizumab is worthy of further investigation based on PFS, with 21 patients (40.4%) surviving progression free for at least 6 months. In addition, seven patients (13.5%) experienced an objective response. This trial, as is the case for most previously completed trials of targeted therapy in recurrent/persistent endometrial cancer, entered all patients without regard to histologic or genomic type.

Endometrial cancers are heterogeneous and can be classified into at least two major types. Type I endometrial cancers, those with endometrioid histology, are the most common and are associated with unopposed estrogen exposure. Type II endometrial cancers have nonendometrioid histology (usually papillary serous or clear cell). Inactivation of the PTEN tumor-suppressor gene is the most common genetic defect in endometrial cancers and is seen in up to 83% of endometrioid tumors.3336 PIK3CA mutation, seen in 36% of endometrial carcinomas, is most frequent in tumors that also have PTEN mutation.37

GOG endometrial cancer trials are subject to central pathology review and categorization of histologic type. It is of great interest as to whether histologic type can be used as a biomarker of response to targeted therapies as a result of the previously mentioned known genetic alterations. In the current study, responses were seen across histologic type (although interestingly, the one patient with a complete response and three of six patients with a partial response had serous histology), and the percentage of patients alive and progression free at 6 months was similar for serous and endometrioid histologies. Patient numbers are too small to formally evaluate the role of histologic subtype and response to bevacizumab in this study, but it is worthy of further study.

Three mammalian target of rapamycin inhibitors, temsirolimus, everolimus, and ridaforolimus, are in clinical trials in endometrial cancer. Preliminary results of a phase II trial of temsirolimus in recurrent or metastatic endometrial cancer (chemotherapy naive) demonstrated encouraging results, with five confirmed partial responses (26%) in 19 evaluable patients.8 Evaluation of a second cohort, women who must have had treatment with one prior regimen of cytotoxic chemotherapy, revealed an ORR of 7% (two of 27 patients).9 A phase II trial of everolimus, in patients with one to two prior chemotherapy regimens, reported no responses.10 Entry was limited to patients with endometrioid histology. A phase II trial of intravenous ridaforolimus in recurrent or metastatic endometrial cancer and carcinosarcoma of the uterus (up to two prior cytotoxic regimens) revealed a response rate of 9% (four of 45 patients).11

Epidermal growth factor receptor as a therapeutic target has also been evaluated in endometrial cancer. Erlotinib was evaluated in chemotherapy-naive patients, showing an ORR of 12.5% (four of 32 patients).13 Gefitinib failed to meet criteria for further evaluation in patients with one to two prior chemotherapy regimens (ORR, 3.8% [one of 26 patients]; 6-month PFS, 8.3% [four of 26 patients]).14 One (5%) of 20 evaluable patients (one to four prior chemotherapy regimens) treated with cetuximab experienced a partial response.15 A phase II trial of trastuzumab did select patients based on human epidermal growth factor receptor 2 positivity (either by overexpression or amplification), although it failed to showed responses.12

Prior attempts to target VEGF have shown modest activity. Treatment of patients with recurrent/persistent endometrial cancer (one to two prior regimens) with thalidomide yielded an ORR of 12.5% (three of 24 patients), but with only 8.3% of patients (two of 24 patients) surviving progression free for at least 6 months.16 On preliminary report, the oral tyrosine kinase inhibitors sunitinib and sorafenib have resulted in minimal activity, with ORRs of 15% (three of 20 patients) and 5% (two of 39 patients), respectively.17

The most striking translational finding was the relationship of high circulating VEGF-A levels with poor outcome in this study. This raises the following important question: What is the source of circulating VEGF-A? VEGF-A is produced by many cells in the body, including the vascular endothelium, and our finding that archival VEGF-A levels from tissues obtained remote from bevacizumab treatment did not correlate with pretreatment circulating levels suggests that sources other than the tumor contribute to plasma VEGF-A levels. Indeed, although high plasma VEGF-A concentrations assessed by ELISA were associated with lack of tumor response and an increased risk of death, high VEGF-A staining intensity in archival tumor was associated with a reduced risk of death. It is possible that VEGF-A staining in remote archival tumor does not reflect the state of the tumor immediately before treatment when the plasma pretreatment VEGF-A levels were assessed. Pretreatment biopsies of recurrent or persistent lesions were not performed in this study. It is also possible that because of the small patient numbers in this study, this result is spurious. Regardless, these data imply that relying on the tumor phenotype when assessed from archival tissues remote from treatment to predict clinical response may have limitations. Despite the limitations in sample size and exploratory nature of the studies, angiogenic markers in tumor and serum may provide prognostic value in recurrent/persistent endometrial cancer and are being prospectively evaluated in the GOG randomized phase II trial of paclitaxel, carboplatin, and bevacizumab; paclitaxel, carboplatin, and temsirolimus; and ixabepilone, carboplatin, and bevacizumab.

Appendix

The following Gynecologic Oncology Group institutions participated in this study: University of Alabama at Birmingham, Duke University Medical Center, Abington Memorial Hospital, Walter Reed Army Medical Center, University of Washington, University of North Carolina School of Medicine, University of Iowa Hospitals and Clinics, Wake Forest University School of Medicine, Rush-Presbyterian-St Luke's Medical Center, State University of New York Downstate Medical Center, University of New Mexico Health Sciences Center, The Cleveland Clinic Foundation, Washington University School of Medicine, Memorial Sloan-Kettering Cancer Center, Columbus Cancer Council, University of Oklahoma, Gynecologic Oncology Network/Brody School of Medicine, and The Hospital of Central Connecticut at New Britain General Hospital and Community Clinical Oncology Program.

Table A1.

Associations Between VEGF-A Assessments* and Clinical Features Including Patient Characteristics, Tumor Characteristics, Prior Treatment, Tumor Response, and PFS ≥ 6 Months

Tumor VEGF-A Staining
Plasma VEGF-A
Intensity
Percent Positive Tumor Cells
Concentration
Eligible/Evaluable Patients
Patients With IHC Results
Low
High
Low
High
Patients With ELISA Results
Low
High
Clinical Feature No. % No. % No. % No. % No. % No. % No. % No. % No. %
Total No. of patients 52 44 15 34.1 29 65.9 22 50.0 22 50.0 34 17 50.0 17 50.0
Age, years
    40-59 18 34.6 14 31.8 6 40.0 8 27.6 6 27.3 8 36.4 11 32.4 5 29.4 6 35.3
    60-69 22 42.3 18 40.9 5 33.3 13 44.8 8 36.4 10 45.5 14 41.2 10 58.8 4 23.5
    ≥ 70 12 23.1 12 27.3 4 26.7 8 27.6 8 36.4 4 18.2 9 26.5 2 11.8 7 42.2
Race
    White 44 84.6 37 84.1 12 80.0 25 86.2 19 86.4 18 81.8 28 82.4 13 76.5 15 88.2
    Hispanic 3 5.8 3 6.8 0 0 3 10.3 1 4.5 2 9.1 3 8.8 3 17.6 0 0
    African American 2 3.8 2 4.5 2 13.3 0 0 2 9.1 0 0 2 5.9 0 0 2 11.8
    Other 3 5.8 2 4.5 1 6.7 1 0.4 0 0 2 9.1 1 2.9 1 0.9 0 0
GOG performance status
    0 34 65.4 30 68.2 11 73.3 19 65.5 14 63.6 16 72.7 20 58.8 8 47.1 12 70.6
    1 18 34.6 14 31.8 4 26.7 10 34.5 8 36.4 6 27.3 14 41.2 9 52.9 5 29.4
Cell type
    Endometrioid 26 50.0 19 43.2 9 60.0 10 34.5 10 45.5 9 40.9 16 47.1 7 41.2 9 52.9
    Serous 14 26.9 14 31.8 4 26.7 10 34.5 5 22.7 9 40.9 9 26.5 4 23.5 5 29.4
    Other 12 23.1 11 25.0 2 13.3 9 31.0 7 31.8 4 18.2 9 26.5 6 35.3 3 17.6
Tumor grade
    1 or 2 15 28.8 11 25.0 4 26.7 7 24.1 4 18.2 7 31.8 10 29.4 6 35.3 4 23.5
    3 or not specified 37 71.2 33 75.0 11 73.3 22 75.9 18 81.8 15 68.2 24 70.6 11 64.7 13 76.5
Prior radiation
    No 23 44.2 19 43.2 4 26.7 15 51.7 9 40.9 10 45.5 17 50.0 11 64.7 6 35.3
    Yes 29 55.8 25 56.8 11 73.3 14 48.3 13 59.1 12 54.5 17 50.0 6 35.3 11 64.7
Prior chemotherapy
    No 33 63.5 28 63.6 8 53.3 20 69.0 13 59.1 15 68.2 22 64.7 12 70.6 10 58.8
    Yes 19 36.5 16 36.4 7 46.7 9 31.0 9 40.9 7 31.8 12 35.3 5 29.4 7 41.2
Tumor response
    Response 7 12.4 6 13.6 2 13.3 4 13.8 2 9.1 4 18.2 4 11.7 4 23.5 0 0
    Stable disease 26 50.0 21 47.7 7 46.7 14 48.3 13 59.1 8 36.4 17 50.0 9 52.9 8 47.1
    Progressive disease 17 32.7 15 34.1 5 33.3 10 34.5 5 22.7 10 45.5 13 38.2 4 23.5 9 52.9
    Indeterminate 2 3.8 2 4.5 1 6.7 1 3.4 2 9.1 0 0 0 0 0 0 0 0
PFS ≥ 6 months
    No 31 59.6 25 56.8 9 60.0 16 55.2 12 54.5 13 59.1 22 64.7 10 58.8 12 70.6
    Yes 21 40.6 19 43.2 6 40.0 13 44.8 10 45.5 9 40.9 12 35.3 7 41.2 5 29.4

Abbreviations: VEGF-A, vascular endothelial growth factor-A; PFS, progression-free survival; IHC, immunohistochemistry; ELISA, enzyme-linked immunosorbent assay; GOG, Gynecologic Oncology Group.

*

VEGF-A was categorized by staining intensity as low (negative or 1+ staining) versus high (2+ or 3+ staining), by median percent positive tumor cells as low (35%) versus high (≥ 35%), or by median plasma concentration as low (< 76.9 pg/mL) versus high (≥ 76.9 pg/mL).

A suggestive correlation was observed between plasma VEGF-A concentration and tumor response (Spearman correlation coefficient, −0.382). Correlation of VEGF-A concentration categorized at the median and tumor response was −0.373 (Kendall's correlation).

Table A2.

Associations Between VEGF-A Assessments* and Clinical Outcome

VEGF-A Assessments Progression-Free Survival
Overall Survival
Unadjusted Cox Model
Unadjusted Cox Model
Adjusted Cox Model
HR 95% CI HR 95% CI HR 95% CI
Tumor VEGF-A staining
    Intensity
        High v low 0.703 0.352 to 1.403 0.541 0.250 to 1.172 0.350 0.153 to 0.797
    Percent positive
        High v low 1.464 0.768 to 2.789 0.817 0.387 to 1.725 1.246 0.524 to 2.965
Plasma VEGF-A concentration (pg/mL)
    High v low 1.539 0.746 to 3.171 2.719 1.160 to 6.374 5.298 2.002 to 14.022

Abbreviations: VEGF-A, vascular endothelial growth factor-A; HR, hazard ratio.

*

VEGF-A was categorized by staining intensity as low (negative or 1+ staining) versus high (2+ or 3+ staining), by median percent positive tumor cells as low (35%) versus high (≥ 35%), or by median plasma concentration as low (< 76.9 pg/mL) versus high (≥ 76.9 pg/mL).

Cox regression modeling adjusted for Gynecologic Oncology Group performance status and tumor grade.

Suggested associations were observed between categorized VEGF-A intensity and adjusted overall survival and between categorized plasma VEGF-A concentration and unadjusted and adjusted overall survival.

Footnotes

Supported by the Cory Beach Family Fund for Endometrial Cancer Research (K.K.L.) and National Cancer Institute Grant No. U10CA27469 to the Gynecologic Oncology Group (GOG) Administrative Office, Tissue Bank, and Receptor Core; Grant No. U24CA114793 to the GOG Tissue Bank; Grant No. U10CA37517 to the GOG Statistical and Data Center; and Grant No. R01CA99908 (K.K.L.).

Presented in part at the 45th Annual Meeting of the American Society of Clinical Oncology, May 29-June 2, 2009, Orlando, FL.

Authors' disclosures of potential conflicts of interest and author contributions are found at the end of this article.

Clinical trial information can be found for the following: NCT00301964.

AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

Although all authors completed the disclosure declaration, the following author(s) indicated a financial or other interest that is relevant to the subject matter under consideration in this article. Certain relationships marked with a “U” are those for which no compensation was received; those relationships marked with a “C” were compensated. For a detailed description of the disclosure categories, or for more information about ASCO's conflict of interest policy, please refer to the Author Disclosure Declaration and the Disclosures of Potential Conflicts of Interest section in Information for Contributors.

Employment or Leadership Position: None Consultant or Advisory Role: None Stock Ownership: None Honoraria: None Research Funding: Kathleen M. Darcy, Genentech; Mack N. Barnes, Genentech, Daiichi Sankyo; Parviz Hanjani, Genentech Expert Testimony: None Other Remuneration: None

AUTHOR CONTRIBUTIONS

Conception and design: Carol Aghajanian, Michael W. Sill, Kathleen M. Darcy, Kimberly K. Leslie

Administrative support: Kathleen M. Darcy

Provision of study materials or patients: Carol Aghajanian, Benjamin Greer, D. Scott McMeekin, Peter G. Rose, Jacob Rotmensch, Mack N. Barnes, Parviz Hanjani

Collection and assembly of data: Michael W. Sill, Kathleen M. Darcy, D. Scott McMeekin, Jacob Rotmensch

Data analysis and interpretation: Carol Aghajanian, Michael W. Sill, Kathleen M. Darcy, Benjamin Greer, D. Scott McMeekin, Peter G. Rose, Mack N. Barnes, Parviz Hanjani

Manuscript writing: All authors

Final approval of manuscript: All authors

REFERENCES

  • 1.Jemal A, Siegel R, Xu J, et al. Cancer Statistics, 2010. CA Cancer J Clin. 2010;60:277–300. doi: 10.3322/caac.20073. [DOI] [PubMed] [Google Scholar]
  • 2.Fleming GF, Brunetto VL, Cella D, et al. Phase III trial of doxorubicin plus cisplatin with or without paclitaxel plus filgrastim in advanced endometrial carcinoma: A Gynecologic Oncology Group study. J Clin Oncol. 2004;22:2159–2166. doi: 10.1200/JCO.2004.07.184. [DOI] [PubMed] [Google Scholar]
  • 3.Fiorica M, Brunetto VL, Hanjani P, et al. Phase II trial of alternating course of megestrol acetate and tamoxifen in advanced endometrial carcinoma: A Gynecologic Oncology Group study. Gynecol Oncol. 2004;92:10–14. doi: 10.1016/j.ygyno.2003.11.008. [DOI] [PubMed] [Google Scholar]
  • 4.Whitney CW, Brunetto VL, Zaino RJ, et al. Phase II study of medroxyprogesterone acetate plus tamoxifen in advanced endometrial carcinoma: A Gynecologic Oncology Group study. Gynecol Oncol. 2004;92:4–9. doi: 10.1016/j.ygyno.2003.09.018. [DOI] [PubMed] [Google Scholar]
  • 5.Lentz SS, Brady MF, Major FJ, et al. High-dose megestrol acetate in advanced or recurrent endometrial carcinoma: A Gynecologic Oncology Group study. J Clin Oncol. 1996;14:357–361. doi: 10.1200/JCO.1996.14.2.357. [DOI] [PubMed] [Google Scholar]
  • 6.Thigpen JT, Brady MF, Alvarez RD, et al. Oral medroxyprogesterone acetate in the treatment of advanced or recurrent endometrial carcinoma: A dose-response study by the Gynecologic Oncology Group. J Clin Oncol. 1999;17:1736–1744. doi: 10.1200/JCO.1999.17.6.1736. [DOI] [PubMed] [Google Scholar]
  • 7.Thigpen T, Brady MF, Homesley HD, et al. Tamoxifen in the treatment of advanced or recurrent endometrial carcinoma: A Gynecologic Oncology Group study. J Clin Oncol. 2001;19:364–367. doi: 10.1200/JCO.2001.19.2.364. [DOI] [PubMed] [Google Scholar]
  • 8.Oza AM, Elit L, Biagi J, et al. Molecular correlates associated with a phase II study of temsirolimus (CCI-77) in patients with metastatic or recurrent endometrial cancer-NCIC IND 160. J Clin Oncol. 2006;24(suppl):121s. abstr 3003. [Google Scholar]
  • 9.Oza AM, Elit L, Provencher J, et al. NCIC CTG IND 160b: Phase II study of CCI-779 (temsirolimus) in patients with metastatic and/or locally advanced recurrent endometrial cancer. J Clin Oncol. 2008;26(suppl):296s. abstr 5516. [Google Scholar]
  • 10.Slomovitz BM, Lu KH, Johnston T, et al. A phase II study of the oral mammalian target of rapamycin, everolimus, in patients with recurrent endometrial carcinoma. Cancer. 2010;116:5415–5419. doi: 10.1002/cncr.25515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Colombo N, McMeekin S, Schwartz P, et al. A phase II trial of the mTOR inhibitor AP23573 as a single agent in advanced endometrial cancer. J Clin Oncol. 2007; 25(suppl):278s. abstr 5516. [Google Scholar]
  • 12.Fleming GF, Sill MW, Darcy KM, et al. Phase II trial of trastuzumab in women with advanced or recurrent HER2-positive endometrial carcinoma: A Gynecologic Oncology Group study. Gynecol Oncol. 2010;116:15–20. doi: 10.1016/j.ygyno.2009.09.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Oza AM, Eisenhauer EA, Elit L, et al. Phase II study of erlotinib in recurrent or metastatic endometrial cancer: NCIC IND-148. J Clin Oncol. 2008;26:4319–4325. doi: 10.1200/JCO.2007.15.8808. [DOI] [PubMed] [Google Scholar]
  • 14.Leslie KK, Sill MW, Darcy KM, et al. Efficacy and safety of gefitinib (Iressa, ZD1839) and potential prognostic value of soluble EGFR, EGFR mutations and tumor markers in a Gynecologic Oncology Group phase II trial of persistent or recurrent endometrial cancer. J Clin Oncol. 2009;(suppl):27. abstr e16542. [Google Scholar]
  • 15.Slomovitz B, Schmeler K, Miller D, et al. Phase II study of cetuximab (Erbitux) in patients with progressive or recurrent endometrial cancer. Gynecol Oncol. 2010; 116(suppl 1):13. doi: 10.1136/ijgc-2020-001859. abstr. [DOI] [PubMed] [Google Scholar]
  • 16.Correa R, Mackay H, Hirte HW, et al. A phase II study of sunitinib in recurrent or metastatic endometrial carcinoma: A trial of the Princess Margaret Hospital, The University of Chicago, and California Cancer Phase II Consortia. J Clin Oncol. 2010; 28(suppl):399s. abstr 5038. [Google Scholar]
  • 17.Nimeiri HS, Oza AM, Morgan RJ, et al. Sorafenib (SOR) in patients (pts) with advanced/recurrent uterine carcinoma (UCA) or carcinosarcoma (CS): A phase II trial of the University of Chicago, PMH, and California Phase II Consortia. J Clin Oncol. 2008; 26(suppl):313s. abstr 5585. [Google Scholar]
  • 18.McMeekin DS, Sill MW, Benbrook D, et al. A phase II trial of thalidomide in patients with refractory endometrial cancer and correlation with angiogenesis biomarkers: A Gynecologic Oncology Group study. Gynecol Oncol. 2007;105:508–516. doi: 10.1016/j.ygyno.2007.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Burger RA, Sill MW, Monk BJ, et al. Phase II trial of bevacizumab in persistent or recurrent epithelial ovarian cancer or primary peritoneal cancer: A Gynecologic Oncology Group study. J Clin Oncol. 2007;25:5165–5171. doi: 10.1200/JCO.2007.11.5345. [DOI] [PubMed] [Google Scholar]
  • 20.Cannistra SA, Matulonis UA, Penson RT, et al. Phase II study of bevacizumab in patients with platinum-resistant ovarian cancer or peritoneal serous cancer. J Clin Oncol. 2007;25:5180–5186. doi: 10.1200/JCO.2007.12.0782. [DOI] [PubMed] [Google Scholar]
  • 21.Monk BJ, Sill MW, Burger RA, et al. Phase II trial of bevacizumab in the treatment of persistent or recurrent squamous cell carcinoma of the cervix: A Gynecologic Oncology Group study. J Clin Oncol. 2009;27:1069–1074. doi: 10.1200/JCO.2008.18.9043. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Therasse P, Arbuck SG, Eisenhauer EA, et al. New guidelines to evaluate the response to treatment in solid tumors. J Natl Cancer Inst. 2000;92:205–216. doi: 10.1093/jnci/92.3.205. [DOI] [PubMed] [Google Scholar]
  • 23.Rose PG, Blessing JA, Lewandowski GA, et al. A phase II trial of prolonged oral etoposide (VP-16) as second-line therapy for advanced or recurrent endometrial carcinoma: A Gynecologic Oncology Group study. Gynecol Oncol. 1996;63:101–104. doi: 10.1006/gyno.1996.0286. [DOI] [PubMed] [Google Scholar]
  • 24.Lincoln S, Blessing JA, Lee RB, et al. Activity of paclitaxel as second-line chemotherapy in endometrial carcinoma: A Gynecologic Oncology Group study. Gynecol Oncol. 2003;88:277–281. doi: 10.1016/s0090-8258(02)00068-9. [DOI] [PubMed] [Google Scholar]
  • 25.Moore DH, Blessing JA, Dunton C, et al. Evaluation of dactinomycin in the treatment of recurrent or persistent endometrial carcinoma: A phase II study of the Gynecologic Oncology Group. Gynecol Oncol. 1999;75:473–475. doi: 10.1006/gyno.1999.5652. [DOI] [PubMed] [Google Scholar]
  • 26.Muggia FM, Blessing JA, Sorosky J, et al. Phase II trial of the pegylated liposomal doxorubicin in previously treated metastatic endometrial cancer: A Gynecologic Oncology Group study. J Clin Oncol. 2002;20:2360–2364. doi: 10.1200/JCO.2002.08.171. [DOI] [PubMed] [Google Scholar]
  • 27.Plaxe S, Blessing JA, Husseindazeh N, et al. Evaluation of pyrazoloacridine (PZA) (NSC #366140) in the treatment of recurrent or persistent endometrial carcinoma: A Gynecologic Oncology Group study. Gynecol Oncol. 2002;84:241–244. [Google Scholar]
  • 28.Miller DS, Blessing JA, Lentz SS, et al. Evaluation of topotecan in the treatment of recurrent or persistent endometrial carcinoma: A Gynecologic Oncology Group study. Gynecol Oncol. 2002;87:247–251. doi: 10.1006/gyno.2002.6804. [DOI] [PubMed] [Google Scholar]
  • 29.Fracasso PM, Blessing JA, Molpus KL, et al. Phase II study of oxaliplatin as second-line chemotherapy in endometrial carcinoma: A Gynecologic Oncology Group study. Gynecol Oncol. 2006;103:523–526. doi: 10.1016/j.ygyno.2006.03.043. [DOI] [PubMed] [Google Scholar]
  • 30.Schilder R, Blessing JA, Pearl ML, et al. Evaluation of irofulven (MGI-114) in the treatment of recurrent or persistent endometrial carcinoma: A phase II study of the Gynecologic Oncology Group. Invest New Drugs. 2004;22:343–349. doi: 10.1023/B:DRUG.0000026262.77502.31. [DOI] [PubMed] [Google Scholar]
  • 31.Grendys E, Blessing J, Burger R, et al. A phase II evaluation of flavorpiridol as second-line chemotherapy of endometrial carcinoma: A Gynecologic Oncology Group study. Gynecol Oncol. 2005;98:249–253. doi: 10.1016/j.ygyno.2005.05.017. [DOI] [PubMed] [Google Scholar]
  • 32.Sill MW, Yothers G. A method for utilizing bivariate efficacy outcome measures to screen agents for activity in 2-stage phase II clinical trials: Technical Report 06-08, Department of Biostatistics, University at Buffalo. http://sphhp.buffalo.edu/biostat/research/techreports/UB_Biostatistics_TR0608.pdf.
  • 33.Hecht JL, Mutter GL. Molecular and pathologic aspects of endometrial carcinogenesis. J Clin Oncol. 2006;24:4783–4791. doi: 10.1200/JCO.2006.06.7173. [DOI] [PubMed] [Google Scholar]
  • 34.Mutter GL, Lin MC, Fitzgerald JT, et al. Altered PTEN expression as a diagnostic marker for the earliest endometrial precancers. J Natl Cancer Inst. 2000;92:924–930. doi: 10.1093/jnci/92.11.924. [DOI] [PubMed] [Google Scholar]
  • 35.Kurose K, Zhou XP, Araki T, et al. Frequent loss of PTEN expression is linked to elevated phosphorylated Akt levels, but not associated with p27 and cyclin D1 expression, in primary epithelial ovarian carcinomas. Am J Pathol. 2001;158:2097–2106. doi: 10.1016/S0002-9440(10)64681-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Zhu X, Kwon CH, Schlosshauer PW, et al. PTEN induces G1 cell cycle arrest and decreases cyclin D3 levels in endometrial carcinoma cells. Cancer Res. 2001;61:4569–4575. [PubMed] [Google Scholar]
  • 37.Oda K, Stokoe D, Takentani Y, et al. High frequency of coexistent mutations of PIK3CA and PTEN genes in endometrial carcinoma. Cancer Res. 2005;65:10669–10673. doi: 10.1158/0008-5472.CAN-05-2620. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Clinical Oncology are provided here courtesy of American Society of Clinical Oncology

RESOURCES