Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Diane Provencher is active.

Publication


Featured researches published by Diane Provencher.


The New England Journal of Medicine | 2010

ARID1A Mutations in Endometriosis-Associated Ovarian Carcinomas

Kimberly C. Wiegand; Sohrab P. Shah; Osama M. Al-Agha; Yongjun Zhao; Kane Tse; Thomas Zeng; Janine Senz; Melissa K. McConechy; Michael S. Anglesio; Steve E. Kalloger; Winnie Yang; Alireza Heravi-Moussavi; Ryan Giuliany; Christine Chow; John Fee; Abdalnasser Zayed; Leah M Prentice; Nataliya Melnyk; Gulisa Turashvili; Allen Delaney; Jason Madore; Stephen Yip; Andrew McPherson; Gavin Ha; Lynda Bell; Sian Fereday; Angela Tam; Laura Galletta; Patricia N. Tonin; Diane Provencher

BACKGROUND Ovarian clear-cell and endometrioid carcinomas may arise from endometriosis, but the molecular events involved in this transformation have not been described. METHODS We sequenced the whole transcriptomes of 18 ovarian clear-cell carcinomas and 1 ovarian clear-cell carcinoma cell line and found somatic mutations in ARID1A (the AT-rich interactive domain 1A [SWI-like] gene) in 6 of the samples. ARID1A encodes BAF250a, a key component of the SWI–SNF chromatin remodeling complex. We sequenced ARID1A in an additional 210 ovarian carcinomas and a second ovarian clear-cell carcinoma cell line and measured BAF250a expression by means of immunohistochemical analysis in an additional 455 ovarian carcinomas. RESULTS ARID1A mutations were seen in 55 of 119 ovarian clear-cell carcinomas (46%), 10 of 33 endometrioid carcinomas (30%), and none of the 76 high-grade serous ovarian carcinomas. Seventeen carcinomas had two somatic mutations each. Loss of the BAF250a protein correlated strongly with the ovarian clear-cell carcinoma and endometrioid carcinoma subtypes and the presence of ARID1A mutations. In two patients, ARID1A mutations and loss of BAF250a expression were evident in the tumor and contiguous atypical endometriosis but not in distant endometriotic lesions. CONCLUSIONS These data implicate ARID1A as a tumor-suppressor gene frequently disrupted in ovarian clear-cell and endometrioid carcinomas. Since ARID1A mutation and loss of BAF250a can be seen in the preneoplastic lesions, we speculate that this is an early event in the transformation of endometriosis into cancer. (Funded by the British Columbia Cancer Foundation and the Vancouver General Hospital–University of British Columbia Hospital Foundation.).


The New England Journal of Medicine | 1998

Oral Contraceptives and the Risk of Hereditary Ovarian Cancer

Steven A. Narod; Harvey A. Risch; Roxana Moslehi; Anne Dørum; Susan L. Neuhausen; Håkan Olsson; Diane Provencher; Paolo Radice; Gareth Evans; Susan Bishop; Jean Sébastien Brunet; Bruce A.J. Ponder; J.G.M. Klijn

Background Women with mutations in either the BRCA1 or the BRCA2 gene have a high lifetime risk of ovarian cancer. Oral contraceptives protect against ovarian cancer in general, but it is not known whether they also protect against hereditary forms of ovarian cancer. Methods We enrolled 207 women with hereditary ovarian cancer and 161 of their sisters as controls in a case–control study. All the patients carried a pathogenic mutation in either BRCA1 (179 women) or BRCA2 (28 women). The control women were enrolled regardless of whether or not they had either mutation. Lifetime histories of oral-contraceptive use were obtained by interview or by written questionnaire and were compared between patients and control women, after adjustment for year of birth and parity. Results The adjusted odds ratio for ovarian cancer associated with any past use of oral contraceptives was 0.5 (95 percent confidence interval, 0.3 to 0.8). The risk decreased with increasing duration of use (P for trend, <0.001); use for six or...


The New England Journal of Medicine | 2009

Mutation of FOXL2 in granulosa-cell tumors of the ovary

Sohrab P. Shah; Martin Köbel; Janine Senz; Ryan D. Morin; Blaise Clarke; Kimberly C. Wiegand; Gillian Leung; Abdalnasser Zayed; Erika Mehl; Steve E. Kalloger; Mark Sun; Ryan Giuliany; Erika Yorida; Steven J.M. Jones; Richard Varhol; Kenneth D. Swenerton; Dianne Miller; Philip B. Clement; Colleen Crane; Jason Madore; Diane Provencher; Peter C. K. Leung; Anna deFazio; Jaswinder Khattra; Gulisa Turashvili; Yongjun Zhao; Thomas Zeng; J.N. Mark Glover; Barbara C. Vanderhyden; Chengquan Zhao

BACKGROUND Granulosa-cell tumors (GCTs) are the most common type of malignant ovarian sex cord-stromal tumor (SCST). The pathogenesis of these tumors is unknown. Moreover, their histopathological diagnosis can be challenging, and there is no curative treatment beyond surgery. METHODS We analyzed four adult-type GCTs using whole-transcriptome paired-end RNA sequencing. We identified putative GCT-specific mutations that were present in at least three of these samples but were absent from the transcriptomes of 11 epithelial ovarian tumors, published human genomes, and databases of single-nucleotide polymorphisms. We confirmed these variants by direct sequencing of complementary DNA and genomic DNA. We then analyzed additional tumors and matched normal genomic DNA, using a combination of direct sequencing, analyses of restriction-fragment-length polymorphisms, and TaqMan assays. RESULTS All four index GCTs had a missense point mutation, 402C-->G (C134W), in FOXL2, a gene encoding a transcription factor known to be critical for granulosa-cell development. The FOXL2 mutation was present in 86 of 89 additional adult-type GCTs (97%), in 3 of 14 thecomas (21%), and in 1 of 10 juvenile-type GCTs (10%). The mutation was absent in 49 SCSTs of other types and in 329 unrelated ovarian or breast tumors. CONCLUSIONS Whole-transcriptome sequencing of four GCTs identified a single, recurrent somatic mutation (402C-->G) in FOXL2 that was present in almost all morphologically identified adult-type GCTs. Mutant FOXL2 is a potential driver in the pathogenesis of adult-type GCTs.


Journal of Clinical Oncology | 2011

Phase II Study of Temsirolimus in Women With Recurrent or Metastatic Endometrial Cancer: A Trial of the NCIC Clinical Trials Group

Amit M. Oza; Laurie Elit; Ming-Sound Tsao; Suzanne Kamel-Reid; J. J. Biagi; Diane Provencher; Walter H. Gotlieb; Paul Hoskins; Prafull Ghatage; Katia Tonkin; Helen Mackay; John Mazurka; Joana Sederias; Percy Ivy; Janet Dancey; Elizabeth Eisenhauer

PURPOSE Phosphatase and tensin homolog (PTEN) is a tumor suppressor gene, and loss of function mutations are common and appear to be important in the pathogenesis of endometrial carcinomas. Loss of PTEN causes deregulated phosphatidylinositol-3 kinase/serine-threonine kinase/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling which may provide neoplastic cells with a selective survival advantage by enhancing angiogenesis, protein translation, and cell cycle progression. Temsirolimus, an ester derivative of rapamycin that inhibits mTOR, was evaluated in this setting. PATIENTS AND METHODS Sequential phase II studies evaluated single-agent activity of temsirolimus in women with recurrent or metastatic chemotherapy-naive or chemotherapy-treated endometrial cancer. Temsirolimus 25 mg intravenously was administered weekly in 4-week cycles. RESULTS In the chemotherapy-naive group, 33 patients received a median of four cycles (range, one to 23 cycles). Of the 29 patients evaluable for response, four (14%) had an independently confirmed partial response and 20 (69%) had stable disease as best response, with a median duration of 5.1 months (range, 3.7 to 18.4 months) and 9.7 months (range, 2.1 to 14.6 months). Only five patients (18%) had progressive disease. In the chemotherapy-treated group, 27 patients received a median of three cycles (range, one to six cycles). Of the 25 patients evaluable for response, one (4%) had an independently confirmed partial response, and 12 patients (48%) had stable disease, with a median duration of 4.3 months (range, 3.6 to 4.9 months) and 3.7 months (range, 2.4 to 23.2 months). PTEN loss (immunohistochemistry and mutational analysis) and molecular markers of PI3K/Akt/mTOR pathway did not correlate with the clinical outcome. CONCLUSION mTOR inhibition with temsirolimus has encouraging single-agent activity in endometrial cancer which is higher in chemotherapy-naive patients than in chemotherapy-treated patients and is independent of PTEN status. The difference in activity according to prior therapy should be factored into future clinical trial designs.


Journal of Clinical Oncology | 2010

Randomized, Double-Blind, Placebo-Controlled Phase II Study of AMG 386 Combined With Weekly Paclitaxel in Patients With Recurrent Ovarian Cancer

Beth Y. Karlan; Amit M. Oza; Gary Richardson; Diane Provencher; Vincent L. Hansen; Martin Buck; Setsuko K. Chambers; Prafull Ghatage; Charles H. Pippitt; John V. Brown; Allan Covens; Raj V. Nagarkar; Margaret Davy; Charles A. Leath; Hoa Nguyen; Daniel E. Stepan; David M. Weinreich; Marjan Tassoudji; Yu Nien Sun; Ignace Vergote

PURPOSE To estimate the efficacy and toxicity of AMG 386, an investigational peptide-Fc fusion protein that neutralizes the interaction between the Tie2 receptor and angiopoietin-1/2, plus weekly paclitaxel in patients with recurrent ovarian cancer. PATIENTS AND METHODS Patients with recurrent epithelial ovarian, fallopian tube, or primary peritoneal cancer were randomly assigned 1:1:1 to receive paclitaxel (80 mg/m(2) once weekly [QW], 3 weeks on/1 week off) plus intravenous AMG 386 10 mg/kg QW (arm A), AMG 386 3 mg/kg QW (arm B), or placebo QW (arm C). The primary end point was progression-free survival (PFS). Secondary end points included overall survival, objective response, CA-125 response, safety, and pharmacokinetics. RESULTS One hundred sixty-one patients were randomly assigned. Median PFS was 7.2 months (95% CI, 5.3 to 8.1 months) in arm A, 5.7 months (95% CI, 4.6 to 8.0 months) in arm B, and 4.6 months (95% CI, 1.9 to 6.7 months) in arm C. The hazard ratio for arms A and B combined versus arm C was 0.76 (95% CI, 0.52 to 1.12; P = .165). Further analyses suggested an exploratory dose-response effect for PFS across arms (Tarones test, P = .037). Objective response rates for arms A, B, and C were 37%, 19%, and 27%, respectively. The incidence of grade ≥ 3 adverse events (AEs) in arms A, B, and C was 65%, 55%, and 64%, respectively. Frequent AEs included hypertension (8%, 6%, and 5% in arms A, B, and C, respectively), peripheral edema (71%, 51%, and 22% in arms A, B, and C, respectively), and hypokalemia (21%, 15%, and 5% in arms A, B, and C, respectively). AMG 386 exhibited linear pharmacokinetic properties at the tested doses. CONCLUSION AMG 386 combined with weekly paclitaxel was tolerable, with a manageable and distinct toxicity profile. The data suggest evidence of antitumor activity and a dose-response effect, warranting further studies in ovarian cancer.


Lancet Oncology | 2014

Anti-angiopoietin therapy with trebananib for recurrent ovarian cancer (TRINOVA-1): a randomised, multicentre, double-blind, placebo-controlled phase 3 trial.

Bradley J. Monk; Andres Poveda; Ignace Vergote; Francesco Raspagliesi; Keiichi Fujiwara; Duk Soo Bae; Isabelle Ray-Coquard; Diane Provencher; Beth Y. Karlan; Catherine Lhommé; Gary Richardson; Dolores Gallardo Rincon; Robert L. Coleman; Thomas J. Herzog; Christian Marth; Arija Brize; Michel Fabbro; Andrés Redondo; Aristotelis Bamias; Marjan Tassoudji; Lynn Navale; Douglas Warner; Amit M. Oza

BACKGROUND Angiogenesis is a valid target in the treatment of epithelial ovarian cancer. Trebananib inhibits the binding of angiopoietins 1 and 2 to the Tie2 receptor, and thereby inhibits angiogenesis. We aimed to assess whether the addition of trebananib to single-agent weekly paclitaxel in patients with recurrent epithelial ovarian cancer improved progression-free survival. METHODS For this randomised, double-blind phase 3 study undertaken between Nov 10, 2010, and Nov 19, 2012, we enrolled women with recurrent epithelial ovarian cancer from 32 countries. Patient eligibility criteria included having been treated with three or fewer previous regimens, and a platinum-free interval of less than 12 months. We enrolled patients with a computerised interactive voice response system, and patients were randomly assigned using a permuted block method (block size of four) in a 1:1 ratio to receive weekly intravenous paclitaxel (80 mg/m(2)) plus either weekly masked intravenous placebo or trebananib (15 mg/kg). Patients were stratified on the basis of platinum-free interval (≥0 and ≤6 months vs >6 and ≤12 months), presence or absence of measurable disease, and region (North America, western Europe and Australia, or rest of world). The sponsor, investigators, site staff, and patients were masked to the treatment assignment. The primary endpoint was progression-free survival assessed in the intention-to-treat population. The trial is registered with ClinicalTrials.gov, NCT01204749, and is no longer accruing patients. FINDINGS 919 patients were enrolled, of whom 461 were randomly assigned to the trebananib group and 458 to the placebo group. Median progression-free survival was significantly longer in the trebananib group than in the placebo group (7·2 months [5·8-7·4] vs 5·4 months [95% CI 4·3-5·5], respectively, hazard ratio 0·66, 95% CI 0·57-0·77, p<0·0001). Incidence of grade 3 or higher adverse events was similar between treatment groups (244 [54%] of 452 patients in the placebo group vs 258 [56%] of 461 patients in the trebananib group). Trebananib was associated with more adverse event-related treatment discontinuations than was placebo (77 [17%] patients vs 27 [6%], respectively) and higher incidences of oedema (294 [64%] patients had any-grade oedema in the trebananib group vs 127 [28%] patients in the placebo group). Grade 3 or higher adverse events included ascites (34 [8%] in the placebo group vs 52 [11%] in the trebananib group), neutropenia (40 [9%] vs 26 [6%]), and abdominal pain (21 [5%] vs 22 [5%]). We recorded serious adverse events in 125 (28%) patients in the placebo group and 159 (34%) patients in the trebananib group. There was a difference of 2% or less in class-specific adverse events associated with anti-VEGF therapy (hypertension, proteinuria, wound-healing complications, thrombotic events, gastrointestinal perforations), except bleeding, which was more common in the placebo group than in the trebananib group (75 [17%] vs 46 [10%]). INTERPRETATION Inhibition of angiopoietins 1 and 2 with trebananib provided a clinically meaningful prolongation in progression-free survival. This non-VEGF anti-angiogenesis option for women with recurrent epithelial ovarian cancer should be investigated in other settings and in combination with additional agents. Although oedema was increased, typical anti-VEGF associated adverse events were not prominent. FUNDING Amgen.


American Journal of Human Genetics | 1998

Founder BRCA1 and BRCA2 mutations in French Canadian breast and ovarian cancer families

Patricia N. Tonin; Anne-Marie Mes-Masson; P. Andrew Futreal; Kenneth Morgan; Michelle Mahon; William D. Foulkes; David E. C. Cole; Diane Provencher; Parviz Ghadirian; Steven A. Narod

We have identified four mutations in each of the breast cancer-susceptibility genes, BRCA1 and BRCA2, in French Canadian breast cancer and breast/ovarian cancer families from Quebec. To identify founder effects, we examined independently ascertained French Canadian cancer families for the distribution of these eight mutations. Mutations were found in 41 of 97 families. Six of eight mutations were observed at least twice. The BRCA1 C4446T mutation was the most common mutation found, followed by the BRCA2 8765delAG mutation. Together, these mutations were found in 28 of 41 families identified to have a mutation. The odds of detection of any of the four BRCA1 mutations was 18.7x greater if one or more cases of ovarian cancer were also present in the family. The odds of detection of any of the four BRCA2 mutations was 5.3x greater if there were at least five cases of breast cancer in the family. Interestingly, the presence of a breast cancer case <36 years of age was strongly predictive of the presence of any of the eight mutations screened. Carriers of the same mutation, from different families, shared similar haplotypes, indicating that the mutant alleles were likely to be identical by descent for a mutation in the founder population. The identification of common BRCA1 and BRCA2 mutations will facilitate carrier detection in French Canadian breast cancer and breast/ovarian cancer families.


In Vitro Cellular & Developmental Biology – Animal | 2000

CHARACTERIZATION OF FOUR NOVEL EPITHELIAL OVARIAN CANCER CELL LINES

Diane Provencher; H. Lounis; L. Champoux; M. Tétrault; E. N. Manderson; J. C. Wang; P. Eydoux; R. Savoie; Patricia N. Tonin; Anne-Marie Mes-Masson

Dear Editor: Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy. EOCs originate from either the surface epithelium itself or from the crypts or inclusion cysts on the surface epithelium of the ovary (Whitman et al., 1993). EOCs are designated according to their cell type: serous, mucinous, endometrioid, clear cell, Brenner, or undifferentiated (Serov et al., 1973). They are graded according to degree of differentiation: borderline or low malignant potential represent minimal deviation from their benign counterpart while well differentiated tumors are grade 1, moderately differentiated are grade 2, and poorly differentiated are grade 3 carcinomas. At surgery EOCs are also classified according to stage, grade and amount of residual disease based on criteria established by the International Federation of Gynecology and Obstetrics (FIGO). A common behavior of EOC (in one-third of all cases) is seeding of the peritoneal fluid leading to subsequent implantation over peritoneal surfaces with ascites formation (Morrow and Curtin, 1998). EOCs can arise sporadically and in rarer instances in association with familial cancer syndromes (Easton et al., 1993). It is presently unclear whether the underlying molecular events in the development of all EOCs are similar or whether distinct events are associated with particular subtypes. We have previously described an efficient and rapid technique for the establishment of primary cultures from EOC (Lounis et al., 1994). While these primary cultures are ideally suited to certain studies, the major drawback is the inability to maintain these cultures over extended periods for long-term experiments and for tumor assays. Most human ovarian cancer cell lines described were derived from ascites or pleural effusion (DiSaia et al., 1975; Fogh and Trcmpe, 1975; Sinna et al., 1979; Bast et al,, 1981; Hamilton et al., 1983; Langdon et al., 1988; Golombick et al., 1990; Wong et al., 1990; Golombick and Bezwda, 1991; Yamada et al., 1991; Grunt et al., 1993; Provencher et al., 1993; Hirte et al., i994; Buller et al., 1995; Alama et al., 1996), with only a few cell lines derived from primary ovarian solid tumors (Woods et al., 1979; Langdon et al., 1988; Crickard et al., 1989) or metastases (Buick et al., 1985). In addition, most cell lines originate from tumor material obtained following adjuvant therapy such as chemotherapy or radiation therapy, which could introduce confounding genetic events. Following the derivation of primary EOC cultures (Lounis et al., 1994), we were able to establish four independent spontaneously immortalized epithelial ovarian cancer cell lines from patients who were never exposed to chemotherapy or radiation therapy. The cell lines were derived from ovarian malignant tumors (TOV-21G, TOV-81D, and TOV-112D) and from an ovarian malignant ascites (OV-90). The ovarian tumor cell lines are derived from different histopathologies of ovarian tumors: a clear cell carcinoma (TOV-21G), a papillary serous adenocarcinoma (TOV-81D), an endometrioid carcinoma (TOV-112D), and an adenocarcinoma (OV-90). The clinical information is summarized in Table 1. All patients were diagnosed with advanced disease. Three tumors were grade 3 while one, TOV-81D, was classified as grade 1-2. In Canada, the average age at diagnosis is 54 yr (NCIC, 1999) and therefore TOV112D was derived from a patient with early age of onset ovarian cancer (42 yr at the time of diagnosis). This patient survived less then 3 mo despite an optimal cytoreduetive surgical procedure and platinol-based chemotherapy. TOV-81D is particularly interesting since it was derived from a patient with a familial history of breast and ovarian cancer. In this patient the disease was rather indolent with relapse occurring greater than 5 yr later. Since all the cell lines described here were derived from women of French-Canadian descent they were screened for mutations found to occur in this population (Tonin et al., 1998). Through these studies it was possible to identify a germline BRCA2 mutation, a nucleotide 8765delAG mutation in exon 20, in TOV-81D. All cell lines expressed BRCA1 and BRCA2 as detected by reverse transcriptasepolymerase chain reaction (RT-PCR). Three of the cell lines grow as monolayer cultures on a solid surface (TOV-21G, TOV-81D, and OV-90) while the TOV-112D cell line is loosely adherent and the cells have a tendency to compact and form foci. TOV-81D cells have a very flat morphology and are large surface cells with abundant cytoplasm (Fig. 1D). The epithelial morphology is highly similar to the morphology of cell cultures derived from normal ovarian epithelium (Lounis et al., 1994) and resembles the morphology of a previously described ovarian cell line SKOV-3 (Fogh and Trempe, 1975) which was established from the ascites of a patient with an ovarian adenocarcinoma (Fig. 1E). In contrast, the cellular morphology of the TOV-21G (Fig. 1A), TOVl12D (Fig. 1B), and OV-90 (Fig. 1C) differ from SKOV-3, with ceils being generally smaller and more refractile (Fig. 1). In particular, the OV-90 cells maintain a classic morphology, characterized by ruffled membranes (Lounis et al., 1994), which have been observed in primary cultures from ovarian ascites. The expression of epithelial specific keratins was verified (Table 2). The TOV-21G, OV-90, and TOV-112D cell lines show strong immunofluorescence against the keratin specific CAM5.2 antibody while the TOV-81D cell line reacted weakly with the CAM5.2 antibody. Staining was as reported previously for OV and TOV cultures (Lounis et al., 1994) and was consistent with a staining pattern associated with transformed calls. In addition, we have assessed the expression of antigens MH99 (Mattes et al., 1983, 1987) and B72.3 whose expression correlates with epithelial ovarian carcinomas (Thor et al.~ 1986). All cell lines showed strong positive immunofluorescent staining for both MH99 and B72.3, and this staining appeared stronger than the staining pattern observed for SKOV-3 (Table 2). Staining for CAM5.2, MH99, and B72.1 were negative in the NIH3T3 fibroblast control cell line (Table 2). Finally, the expression of the HER2/NEU oncogene, has been associated with poor prognosis in ovarian cancer (Slamon et al., 1989), was determined. All four ovarian cell lines express neu to some extent


Journal of Clinical Oncology | 2008

Phase II Study of Erlotinib in Recurrent or Metastatic Endometrial Cancer: NCIC IND-148

Amit M. Oza; Elizabeth Eisenhauer; Laurie Elit; Jean-Claude Cutz; Akira Sakurada; Ming S. Tsao; Paul Hoskins; J. J. Biagi; Prafull Ghatage; John Mazurka; Diane Provencher; Naomi Dore; Janet Dancey; Anthony Fyles

PURPOSE Epidermal growth factor receptor (EGFR) overexpression is common in endometrial cancers and may have a major role in tumor growth and progression. Erlotinib is an orally active, selective inhibitor of EGFR tyrosine kinase activity. PATIENTS AND METHODS A multinomial design two-stage phase II study was performed to evaluate single-agent activity of erlotinib in women with advanced endometrial cancer with recurrent or metastatic disease who were chemotherapy naïve and had received up to one line of prior hormonal therapy. Erlotinib was administered at daily dose of 150 mg. Archival tumor tissue was analyzed for EGFR expression by immunohistochemistry (IHC) and gene amplification by fluorescent in situ hybridization (FISH). Mutational status of EGFR was determined in responders. RESULTS Thirty-two of 34 entered patients are assessable for response. Treatment was well tolerated and severe toxicity infrequent, with the only grade 4 toxicity being an elevation of transaminases (AST). There were four confirmed partial responses (PRs; 12.5%; 95% CI, 3.5% to 29%) lasting 2 to 36 months. Fifteen patients had stable disease (SD), with median duration of 3.7 months (range, 2 to 12 months). EGFR expression was analyzed in thirty patients; 19 were positive, nine were negative, and two were not assessable. Of the 19 patients who were EGFR positive, three had PR (16%), seven SD, and eight progressive disease, and one was not assessable. No mutations were identified in responders. FISH showed no correlation of response with gene amplification. CONCLUSION Erlotinib is well tolerated with an overall objective response rate of 12.5%. Molecular analysis did not identify EGFR mutations in responders or correlation of response with gene amplification.


Lancet Oncology | 2012

Intravenous aflibercept for treatment of recurrent symptomatic malignant ascites in patients with advanced ovarian cancer: a phase 2, randomised, double-blind, placebo-controlled study

Walter H. Gotlieb; Frédéric Amant; Advani Sh; Chanchal Goswami; Hal Hirte; Diane Provencher; Naresh Somani; S. Diane Yamada; Jean-Francois Tamby; Ignace Vergote

BACKGROUND Targeting of VEGF is a potential therapeutic option in patients with malignant ovarian ascites. We present the final results of a multicentre study of the efficacy and safety of aflibercept, a potent inhibitor of both VEGF and placental growth factor, in the treatment of malignant ascites. METHODS In this double-blind, placebo-controlled, parallel-group, phase 2 study, patients with advanced chemoresistant ovarian cancer and recurrent symptomatic malignant ascites were randomly assigned (1:1) via an interactive voice response system to either intravenous aflibercept (4 mg/kg every 2 weeks) or placebo, stratified by interval of time (≤ 2 weeks vs > 2 weeks) between the two most recent paracenteses before randomisation. Patients participated in the double-blind period (during which patients, investigators, and sponsor personnel were masked to treatment assignment) until they had a repeat paracentesis and for at least 60 days, and could also participate in an optional open-label period during which all patients received aflibercept. The primary efficacy endpoint was time to repeat paracentesis based on response during the double-blind period alone, and was analysed in the intention-to-treat population with censoring of patients who did not have a repeat paracentesis as of the last day of the double-blind period. Safety analyses included both double-blind and open-label periods. This study is registered at ClinicalTrials.gov, number NCT00327444. FINDINGS 55 patients with a median of four (range two to 11) previous lines of chemotherapy were randomly assigned to receive placebo (n=26) or aflibercept (n=29). Mean time to repeat paracentesis was significantly longer with aflibercept than with placebo (55·1 [SE 7·3] vs 23·3 [7·7] days; difference 31·8 days, 95% CI 10·6-53·1; p=0·0019). In the aflibercept group, two patients did not need a repeat paracentesis during 6 months of double-blind treatment. The most common grade 3 or 4 treatment-emergent adverse events were dyspnoea (six [20%] aflibercept vs two [8%] placebo), fatigue or asthenia (four [13%] vs 11 [44%]), and dehydration (three [10%] vs three [12%]). The frequency of fatal gastrointestinal events was higher with aflibercept (three intestinal perforations) than with placebo (one intestinal fistula leading to sepsis). INTERPRETATION This study shows the effectiveness of VEGF blockade in the reduction of malignant ascites, but confirms the significant clinical risk of fatal bowel perforation in this population of patients with very advanced cancer. VEGF blockade should be used with caution in advanced ovarian cancer with abdominal carcinomatosis, and the benefit-risk balance should be thoroughly discussed for each patient. FUNDING Sanofi Oncology.

Collaboration


Dive into the Diane Provencher's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amit M. Oza

Princess Margaret Cancer Centre

View shared research outputs
Top Co-Authors

Avatar

Suzanna L. Arcand

McGill University Health Centre

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ignace Vergote

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge