scholarly journals Combination of gene set signatures correlates with response to nivolumab in platinum-resistant ovarian cancer

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Ryusuke Murakami ◽  
Junzo Hamanishi ◽  
J. B. Brown ◽  
Kaoru Abiko ◽  
Koji Yamanoi ◽  
...  

AbstractBased on our previous phase II clinical trial of anti-programmed death-1 (PD-1) antibody nivolumab for platinum-resistant ovarian cancer (n = 19, UMIN000005714), we aimed to identify the biomarkers predictive of response. Tumor gene expression was evaluated by proliferative, mesenchymal, differentiated, and immunoreactive gene signatures derived from high-grade serous carcinomas and a signature established prior for ovarian clear cell carcinoma. Resulting signature scores were statistically assessed with both univariate and multivariate approaches for correlation to clinical response. Analyses were performed to identify pathways differentially expressed by either the complete response (CR) or progressive disease (PD) patient groups. The clear cell gene signature was scored significantly higher in the CR group, and the proliferative gene signature had significantly higher scores in the PD group where nivolumab was not effective (respective p values 0.005 and 0.026). Combinations of gene signatures improved correlation with response, where a visual projection of immunoreactive, proliferative, and clear cell signatures differentiated clinical response. An applicable clinical response prediction formula was derived. Ovarian cancer-specific gene signatures and related pathway scores provide a robust preliminary indicator for ovarian cancer patients prior to anti-PD-1 therapy decisions.

2021 ◽  
Author(s):  
Ryusuke Murakami ◽  
Junzo Hamanishi ◽  
J. B. Brown ◽  
Kaoru Abiko ◽  
Koji Yamanoi ◽  
...  

Abstract Background Based on our previous phase II clinical trial of anti-programmed death-1 (PD-1) antibody nivolumab for platinum-resistant ovarian cancer (n=19, UMIN000005714), we aimed to identify the therapeutic response biomarkers to nivolumab in ovarian cancer. Methods Tumor gene expressions were evaluated by proliferative, mesenchymal, differentiated, and immunoreactive gene signatures derived from high-grade serous carcinomas in The Cancer Genome Atlas and a signature established prior to ovarian clear cell carcinoma. Gene sets were scored using the single-sample gene set enrichment analysis, and resulting scores were used to assess the correlation between each gene set and the clinical response to nivolumab therapy. Statistical analyses were performed to identify pathways differentially expressed by either the complete response (CR) or progressive disease (PD) groups. Results The clear cell gene signature significantly had higher score in the CR group, and the proliferative gene signature had significantly higher score in the PD group where nivolumab was not effective (respective p-values 0.005 and 0.026). Combinations of gene signatures improved correlation with response, where a projection of immunoreactive, proliferative, and clear cell signatures differentiated clinical response. Conclusion Ovarian cancer-specific gene signature and related pathway scores provide a preliminary indicator for ovarian cancer prior to receiving anti-PD-1 antibody therapy.


Blood ◽  
2011 ◽  
Vol 118 (21) ◽  
pp. 805-805
Author(s):  
Carolina Terragna ◽  
Daniel Remondini ◽  
Sandra Durante ◽  
Marina Martello ◽  
Francesca Patriarca ◽  
...  

Abstract Abstract 805FN2 Background. Achievement of CR is generally associated with improved clinical outcomes for patients (pts) with MM and represents a primary endpoint of current clinical trials. The GIMEMA Italian Myeloma Network designed a phase 3 study to demonstrate that the triplet VTD regimen was superior over a doublet such as thalidomide-dexamethasone (TD) as induction therapy prior to double ASCT for newly diagnosed MM. On an intention-to-treat basis, the rate of complete or near complete response (CR/nCR) was 31% for the 236 pts on VTD induction therapy, while it was 11% (p<0.0001) for the 238 pts on TD induction therapy. Since enhanced rates of CR/nCR affected by VTD incorporated into ASCT resulted in extended progression-free survival, prediction of CR by pharmacogenomic tools is likely to be an important goal to prospectively select those pts who are more likely to benefit from a given therapy. Methods. For this purpose, in a molecular substudy to the main clinical study we assessed the ability of gene expression profile (GEP) to predict attainment of CR/nCR in 122 pts enrolled in the VTD arm of the study. Their characteristics at baseline, including cytogenetic abnormalities, were comparable with those of the whole population of 236 pts. Highly purified CD138+ plasma cells were obtained at diagnosis from each of these pts and were profiled for gene expression using the Affymetrix U133 Plus2.0 platform. In order to build a low-dimensional signature with optimal performance, genomic data were analyzed with an original algorithm that exploits quadratic discriminant analysis with a bottom-up approach that builds N-gene signatures starting from two-dimensional signatures. Gene models were applied to test datasets to predict achievement of either CR/nCR or less than nCR, and classification performances were validated by a leave-one-out crossvalidation procedure. Results. Thirty four pts out of the 122 (28%) who were included in the present analysis achieved a CR/nCR, while the remaining 88 patients failed this objective. The molecular approach described above allowed to identify several gene signatures among which we choose a 163-gene signature that provided a predictive capability of 79% sensitivity, 87% specificity, 71% positive predictive value (PPV) and 92% negative predictive value (NPV). These expression values were used in an unsupervised hierarchical clustering to stratify the population of 122 profilated pts into 3 well defined subgroups. Seventy nine pts were included in subgroup A, while the remaining 43 pts were included in either subgroup B (n=22) or subgroup C (n=21). Notably, 19 out the 34 CR/nCR pts (56%) clustered in subgroup B, whereas the remaining 15 pts were randomly distributed within subgroup A. Analysis of demographic and disease characteristics of the pts belonging to the 3 major subgroups, revealed that in subgroup B the frequencies of pts carrying del(13q) (78%) or del(17p) (22%) or with an IgA isotype (54%) were significantly higher in comparison with the corresponding values found in subgroup A (47%, 4%, and 10%, respectively) and subgroup C (38%, 10%, and 5%, respectively). In order to obtain a more feasible set of genes predictive of CR/nCR, several smaller signatures originating from the 163-gene signature were further analyzed by means of the same algorithm described above. The best predictive capability was obtained with a 41-gene signature that provided 88% sensitivity, 97% specificity, 91% PPV and 95% NPV. A GeneGo ® network analysis of genes included in the signatures showed that the most relevant network nodes included tumour suppressor genes (FBXW7 and MAD), genes involved in inflammatory response (TREM1 and TLR4) and genes involved in B cell development (IKZF1, IL10 and NFAM1). Genes included in the signatures do not gather in specific chromosomes, thus confirming the absence of bias on selection of signatures genes, potentially due to prevalence of MM typical chromosomal aberrations. Conclusions. GEP analysis of a subgroup of pts who received VTD induction therapy allowed to provide a 41-gene signature that was able to predict attainment of CR/nCR and, conversely, failure to achieve at least nCR in 91% and 95% of cases, respectively. These favorable results might represent a first step towards the possible application of a tailored therapy based on the single patient's genetic background. Supported by: Fondazione Del Monte di Bologna e Ravenna, Ateneo RFO grants (M.C.) BolognAIL. Disclosures: Bringhen: Celgene: Honoraria; Janssen-Cilag: Honoraria; Novartis: Honoraria; Merck Sharp & Dhome: Membership on an entity's Board of Directors or advisory committees. Offidani:Janssen: Honoraria; Celgene: Honoraria.


1993 ◽  
Vol 11 (10) ◽  
pp. 1952-1956 ◽  
Author(s):  
V Lorusso ◽  
A Catino ◽  
B Leone ◽  
M Rabinovich ◽  
G Gargano ◽  
...  

PURPOSE This study aimed to evaluate the activity and toxicity of carboplatin (PPL) and ifosfamide (IFO) in patients with epithelial ovarian cancer previously treated with cisplatin (CDDP)-containing regimens. PATIENTS AND METHODS From July 1989 to December 1991, 35 patients with epithelial ovarian cancer relapsed or refractory to CDDP as first-line chemotherapy were treated. PPL was administered at a dose of 300 mg/m2 intravenously (IV) on day 1 and IFO at a dose of 1,500 mg/m2 IV on days 1 to 3 every 3 to 4 weeks. Criteria for evaluating previous response to CDDP were strictly defined. RESULTS The overall response rate was 43% (complete response [CR], 6%; partial response [PR], 37%) and the median duration of response was 7 months (range, 3 to 16). In potentially platinum-sensitive (PPS; relapsed) patients, the overall response rate was 56%. None of the primary platinum-resistant (PPR) patients obtained a clinical response to PPL plus IFO, whereas one of five secondary platinum-resistant (SPR) patients obtained a PR. The regimen was easily manageable. CONCLUSION PPL plus IFO is useful and well-tolerated combination in salvage treatment of patients with advanced ovarian cancer. However, clear synergism between PPL and IFO that could overcome intrinsic or acquired CDDP resistance was not observed. The advantage of PPL plus IFO as compared with CDDP-containing regimens is represented by the increased tolerability and the reduced neurotoxicity, nephrotoxicity, and ototoxicity as compared with CDDP-containing regimens. It is essential that the patient population be defined according to their previous response to platinum therapy in trials involving second-line therapy of ovarian cancer.


2007 ◽  
Vol 25 (18_suppl) ◽  
pp. 16076-16076
Author(s):  
M. Marinaccio ◽  
E. De Marino ◽  
E. Mele ◽  
R. Catacchio ◽  
D. Meo ◽  
...  

16076 Background: The prognosis of patients with recurrent epithelial ovarian cancer is extremely poor after several lines of chemotherapy; this situation becomes more and more difficult to manage in the presence of a platinum-resistance condition. Topotecan 1.5mg/m2 on days 1 through 5 of a 21-day cycle is often employed as a second-line chemotherapy also for platinum-resistant epithelial ovarian cancer. In this phase II study we investigated the safety as well response rate of weekly topotecan as a salvage therapy in heavily pretreated patients with recurrent platinum-resistant epithelial ovarian cancer (RprEOC). Methods: To be elegible for the study patients had to be considered resistant to platinum and paclitaxel pretreated. Eleven patients (median age 51 yrs, range 45 - 70) with performance status 0–2 affected by RprEOC with measurable disease, were planned to receive weekly topotecan. They had received at least 3 prior lines of chemotherapy. Topotecan was administered at the dose of 2.0 mg/m2 via a 30-minute i.v. infusion once every week until disease progression or unacceptable toxicity. Results: All patients were evaluable for toxicity and clinical response. All the 11 pts enrolled had stage III-IV disease. Median number of chemotherapy cycles was 5 (range 3 - 8). A total of 62 cycles were administered. Dose reduction was necessary for 12% of the cycles. Main toxicities included anemia (12%), leucopenia (18%), thrombocytopenia (15%) and asthenia (20%). No deaths were attributable to therapy. No one showed complete response, while two partial response (18.2%) and four stable disease (36.4%) were observed. Five pts (45.4%) progressed on therapy. The median progression-free interval was 13 weeks. Conclusion: Salvage therapy for patients with ovarian cancer who failed several platinum and paclitaxel treatments remains a therapeutic challenge. Topotecan administered at low weekly dosage (2.0 mg/m2) is an active option in the subset of heavily RprEOC and it seems also to be a more tolerable regimen compared to the classical 5-day schedule. No significant financial relationships to disclose.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 5082-5082
Author(s):  
Ron E. Swensen ◽  
Barbara Ann Goff ◽  
Jennifer Childs ◽  
Doreen Higgins ◽  
Theodore Gooley ◽  
...  

5082^ Background: Ovarian cancer patients with an anti-tumor immune response have a prolonged survival, suggesting that augmenting anti-tumor immunity may be therapeutic. We hypothesized that weekly nab paclitaxel (nabP) followed by GM-CSF may enhance anti tumor immunity and prolong time to progression (TTP). Methods: Eligible subjects had platinum resistant ovarian, primary peritoneal or fallopian tube cancer, and an elevated CA125. Conditional power estimate after 11 subjects showed 22 subjects had 80% power to show a response rate (RR) >21% if the true study RR is 35%. Study end points were RR and TTP. Progression (DP) was doubling of CA125 above the nadir. Complete response (CR) was a decline of CA125 below institutional normal. Partial response (PR) was a decline of >50% from baseline. Stable disease (SD) was all other scenarios. Subjects received nabP, 100mg/m2 days 1,8,15 followed by GM-CSF 250mcg days 16-26 of a 28 day cycle until progression or 6 cycles were complete. Responding subjects received up to 6 more cycles of GM-CSF on days 14-28. Results: 21 subjects received at least one dose of study medications. Median age was 61 (30-91) and had a median of 3 (1-13) prior regimens. Among those completing the study, the median TTP was 132 days vs. 272 days on the prior platinum regimen. 9/21 (43%) had a PR and 4/21 (19%) had a CR. Subjects with a response had a median TTP of 140 days. Assay of serial T-lymphocyte counts against CEA, MUC1, CA125 and tt and influenza controls are planned. Conclusions: Weekly nabP with GM-CSF had manageable toxicity and induced a high response rate (62% by CA125) in patients with platinum resistant ovarian cancer, however this regimen did not prolong the TTP beyond the TTP observed in the prior platinum regimen.


2021 ◽  
Vol 39 (15_suppl) ◽  
pp. 5538-5538
Author(s):  
Erika P. Hamilton ◽  
Camille Catherine Jackson ◽  
Ramez Nassef Eskander ◽  
Floor Jenniskens Backes ◽  
Vicky Makker ◽  
...  

5538 Background: The phase (Ph) 1b part of LIO-1 (NCT04042116; ENGOT-GYN3/AGO/LIO) assessed the oral antiangiogenic, multikinase inhibitor lucitanib + immune checkpoint inhibitor nivolumab, confirming the recommended Ph2 dose (RP2D) of lucitanib as 6 mg QD + nivolumab (480 mg IV every 28 days). To maximize lucitanib exposure and potential clinical benefit of the combination, individualized lucitanib dose titration is being explored in a Ph2 part, across 4 recurrent gynecologic malignancies (endometrial, cervical, ovarian, and ovarian/endometrial clear-cell cancers) using a Simon 2-stage design. We present updated Ph1b data and describe initial experience for the first 24 patients (pts) enrolled in the Ph2 ovarian cancer (OC) cohort. Methods: In Ph1b, pts with advanced, metastatic solid tumors received lucitanib at 6, 8, and 10 mg QD + nivolumab (in a 4+3 dose escalation). In the Ph2 OC cohort, pts with recurrent high-grade epithelial ovarian, fallopian tube, or primary peritoneal cancer (excluding clear-cell histology) with ≥2 prior chemotherapy regimens (including ≥1 platinum doublet) received the combination at RP2D; lucitanib dose was escalated from 6 mg to 8 mg and then to 10 mg QD for pts who met safety-based titration criteria. Visit cutoff was Feb 1, 2021. Results: In the Ph1b part (N = 17), median treatment duration was 109 days (range 14–505+). There has been 1 confirmed complete response (CR; anal cancer) and 1 confirmed partial response (PR; cervical cancer) per RECIST v1.1 with durations of 7.1 and 12.8 months, respectively. Ten pts had stable disease (SD), 3 had progressive disease, and 2 were nonevaluable; 3 pts remain on treatment. Overall disease control rate (CR + PR + SD ≥16 wk) was 47.1%. One dose-limiting toxicity (DLT; grade [G] 3 proteinuria) was observed in a pt receiving lucitanib 6 mg, leading to lucitanib discontinuation; no DLTs were seen at 8 or 10 mg. G ≥3 treatment-emergent adverse events (TEAEs) reported in ≥2 pts included hypertension (HTN; n = 4), fatigue (n = 2), and proteinuria (n = 2). Of the first 24 pts enrolled in the Ph2 OC cohort, 13 (54%) remain on treatment (median duration 59 [2–167+] days). Most frequent any-grade TEAEs were HTN (n = 10), fatigue (n = 8), nausea (n = 7), and proteinuria (n = 6). The only G ≥3 TEAE experienced in ≥2 pts was HTN (n = 4); 1 pt discontinued due to G4 HTN/G2 angina pectoris and 1 pt to G2 colonic perforation. To date, 21 pts have completed ≥1 cycle; 11 met safety-based dose-titration criteria, 10 of whom escalated to the 8 mg lucitanib dose. Of these, 5 pts subsequently escalated to 10 mg. One pt required dose reduction from 6 mg to 4 mg lucitanib. Conclusions: Ph1b data suggest that lucitanib + nivolumab has promising signs of antitumor activity. A safety-based dose-titration strategy appears feasible with manageable toxicity, based on experience from the Ph2 OC cohort to date; efficacy data from this cohort will also be presented. Clinical trial information: NCT04042116.


2010 ◽  
Vol 9 ◽  
pp. CIN.S2892 ◽  
Author(s):  
Yarong Yang ◽  
Eric J. Kort ◽  
Nader Ebrahimi ◽  
Zhongfa Zhang ◽  
Bin T. Teh

Background Gene set enrichment analysis (GSEA) is an analytic approach which simultaneously reduces the dimensionality of microarray data and enables ready inference of the biological meaning of observed gene expression patterns. Here we invert the GSEA process to identify class-specific gene signatures. Because our approach uses the Kolmogorov-Smirnov approach both to define class specific signatures and to classify samples using those signatures, we have termed this methodology “Dual-KS” (DKS). Results The optimum gene signature identified by the DKS algorithm was smaller than other methods to which it was compared in 5 out of 10 datasets. The estimated error rate of DKS using the optimum gene signature was smaller than the estimated error rate of the random forest method in 4 out of the 10 datasets, and was equivalent in two additional datasets. DKS performance relative to other benchmarked algorithms was similar to its performance relative to random forests. Conclusions DKS is an efficient analytic methodology that can identify highly parsimonious gene signatures useful for classification in the context of microarray studies. The algorithm is available as the dualKS package for R as part of the bioconductor project.


Sign in / Sign up

Export Citation Format

Share Document