A Randomised, Multi-centre Phase II Trial of Weekly Paclitaxel and Vistusertib in Platinum-Resistant Ovarian High-Grade Serous Carcinoma: OCTOPUS Arm 1 ======================================================================================================================================================= * S. Banerjee * G. Giannone * A. Clamp * D. Ennis * R. Glasspool * R. Herbertson * J. Krell * R. Riisnaes * H.B. Mirza * Z. Cheng * J. McDermott * C. Green * R.S. Kristeleit * A. George * C. Gourley * L-A Lewsley * D. Rai * U. Banerji * S. Hinsley * I.A. McNeish ## Abstract **Background** Preclinical studies support targeting PI3K/AKT/mTOR signalling in platinum-resistant ovarian cancer (PROC). A phase I study of the dual mTORC1/mTORC2 inhibitor vistusertib with weekly paclitaxel (wP) showed activity. We report the results of Arm 1 of OCTOPUS, the first randomised trial of weekly paclitaxel and dual mTORC1/2 inhibition in ovarian cancer. **Methods** Patients with platinum-resistant or refractory high grade serous carcinoma were randomised (1:1) to wP (80mg/m2 D1,8,15 of 28 day cycle) plus oral vistusertib (50mg BD) or placebo (P). The primary endpoint was progression-free survival (PFS). Secondary endpoints included response rate (RR) and overall survival (OS). **Results** 140 patients (median age 63, range: 36-86; 18% platinum-refractory; 54% ≥3 prior therapies) were randomised. There was no difference in PFS (median 4.5 vs 4.1m (HR 0.84; 80% CI (0.67, 1.07); 1-sided p=0.18), OS (median 9.7 vs 11.1m (HR 1.21; 80% CI (0.91, 1.60); 1-sided p=0.80) or RR (odds ratio 0.86; 80% CI (0.55, 1.36); 1-sided p=0.66). Grade 3/4 adverse events were 41.2% (wP+V) vs 36.7% (wP+P). Low tumour PTEN expression was associated with longer PFS in the wP+V arm (9.4 vs 4.1m p=0.003) but not in the wP arm (4.8 vs 4.2m p=0.60). Tumour genome-wide copy number (CN) analysis suggested that high CN signature 4 was associated with worse outcome in the wP+P arm (2.3 vs 4.6m p=0.018) but not the wP+V arm (5.4 vs 3.3m). **Conclusions** Vistusertib did not improve clinical activity of wP in PROC. However, low tumour cell PTEN expression may be a predictive biomarker for vistusertib activity. **Translational Relevance** Preclinical studies suggest that activation of the PI3K/AKT/mTOR signalling pathway contributes to platinum-resistance in ovarian high grade serous carcinoma (HGSC). Based on activity in a phase I study, we evaluated the clinical efficacy of the dual mTORC1/mTORC2 inhibitor vistusertib in combination with weekly paclitaxel in the OCTOPUS study - a multi-centre, randomised, placebo-controlled, phase II trial in platinum-resistant ovarian (HGSC). In the first randomised trial of weekly paclitaxel and dual mTORC1/2 inhibition in ovarian cancer, vistusertib did not improve clinical activity of weekly paclitaxel. However, translational analyses indicated that low tumour cell PTEN expression may be a predictive biomarker for vistusertib activity. We also showed genome-wide copy number (CN) analysis, in particular high exposure to CN signature 4, may also allow identification of patients with greater chance of benefit from dual mTORC inhibition. Potential predictive biomarkers identified in our study should be evaluated in ongoing/future studies. Keywords * Weekly paclitaxel * platinum-resistant * ovarian cancer * mTORC ## Introduction Despite improvements in first line treatment for advanced ovarian cancer, approximately 85% of patients recur and eventually develop fatal chemotherapy resistance. The duration of response following platinum therapy (platinum-free interval) remains an important consideration when selecting treatment options for recurrent ovarian cancer. For patients whose disease has relapsed during (platinum-refractory) or within 6 months of completing platinum-based therapy (‘platinum-resistant’), treatment options are limited. Weekly paclitaxel has activity in platinum-resistant ovarian cancer, with response rates of approximately 30%. However, the median PFS is generally 3-4 months and overall survival approximately 12 months (1). One potential mechanism of resistance to both platinum and taxane chemotherapy is activation of the PI3K/AKT/mTOR signalling pathway (2,3). Raised p-S6K expression in malignant cells isolated from ascites in ovarian cancer patients has been associated with chemoresistance and poor clinical outcome (4). However, response rates in single arm studies of first generation mTOR inhibitors such as temsirolimus were poor (5), which prompted the development of dual mTORC1/2 inhibitors that could prevent the AKT-mediated feedback observed with these rapalogues. Following encouraging preclinical data (6), a phase I study of weekly paclitaxel in combination with the dual mTORC1/2 inhibitor vistusertib was conducted with a dose expansion for patients with ovarian high grade serous carcinoma (HGSC). The recommended phase II dose (RP2D) was established as 80 mg/m2 paclitaxel with 50mg vistusertib bd 3 days per week. The RECIST response rate in the HGSC cohort was 52% (13/25), with median PFS of 5.8 months (95% CI: 3.3–18.5) (7). Here, we evaluated the clinical efficacy of vistusertib in combination with weekly paclitaxel in the OCTOPUS study - a multi-centre, randomised, placebo-controlled, phase II trial in platinum-resistant ovarian high grade serous carcinoma. ## Methods ### Study design OCTOPUS was a phase 2, randomised, double-blind, placebo-controlled, multicentre study evaluating the efficacy and safety of vistusertib in combination with paclitaxel compared to paclitaxel alone in patients with high grade serous ovarian carcinoma (HGSC) progressed during (platinum-refractory) or within 6 months of completing last platinum-based therapy (platinum-resistant). The study was conducted in accordance with the Research Governance Framework for Health and Community Care (Second edition; 2006) and the Medicines for Human Use (Clinical Trials) Regulations, 2004 SI 2004:1031 (as amended) and World Medical Association Declaration of Helsinki Ethical Principles for Medical Research Involving Human Subjects 1964 (as amended) and was co-sponsored by University of Glasgow and NHS Greater Glasgow and Clyde. Ethical approval was obtained from London–Brighton and Sussex Research Ethics Committee (reference 15/LO/1302) and all patients provided written informed consent. ### Patient population Patients (≥18 years) with histologically confirmed high grade serous carcinoma of ovarian, fallopian tube or primary peritoneal origin, with relapse in the platinum-resistant timeframe, were enrolled. Platinum resistant/refractory status was defined as either radiological progression (based on RECISTv.1.1) or CA125 rise (according to GCIG CA125 criteria) plus symptoms indicative of progression either during (platinum-refractory) or within 6 months of completing (platinum-resistant) prior platinum therapy. Measurable or evaluable disease according to RECISTv.1.1 and/or GCIG CA125 criteria was required (8). Patients who received weekly paclitaxel in combination with platinum as part of first line treatment were eligible if the interval since the last dose of weekly paclitaxel was >6 months at the time of randomisation. Patients who received prior weekly paclitaxel for platinum-resistant disease were excluded. A biopsy was mandatory at study entry if deemed technically feasible. There were no restrictions on number of lines of prior therapy (see supplementary for full inclusion/exclusion criteria) and the most recent chemotherapy did not have to be platinum-based. ### Study Design, Treatment and Conduct 140 participants were randomised (1:1) to receive treatment as follows: * wP+V: paclitaxel 80mg/m2 IV D1, 8, 15 plus Vistusertib 50mg bd days 1-3, 8-10 and 15-17 of a 28 day cycle (experimental arm) * wP+P: paclitaxel 80mg/m2 IV D1, 8, 15 plus placebo bd days 1-3, 8-10 and 15-17 of a 28 day cycle (control arm) Patients received 6 cycles (24 weeks) of combination treatment. Thereafter, patients who did not have progressive disease and had completed at least 4 cycles of combination treatment could continue on vistusertib (50mg bd)/placebo alone as continuous maintenance therapy. Patients could continue beyond 6 cycles of paclitaxel and vistusertib/placebo before receiving maintenance therapy, at the discretion of the Investigator and Chief Investigator, provided the patient had not progressed. After informed consent and eligibility confirmation, participants were allocated to treatment using minimisation with a random element, and the following minimisation factors: treatment centre, measurable disease (yes vs. no), platinum refractory vs. resistant, and taxane-free interval (<6 months vs. ≥6 months vs. no prior taxane) (1). ### Assessments Imaging assessments (CT chest, abdomen and pelvis or MRI) were performed at baseline and then 8 weekly until progression. CA125 was measured 4 weekly. Adverse events were graded according to National Cancer Institute Common Terminology Criteria for Adverse Events (NCI-CTCAE) v4.3. Quality of life (QoL) was assessed using EQ-5D at baseline, prior to each cycle of treatment, end of treatment and follow up. ### Statistical Considerations The primary end point was progression-free survival (PFS) in the intention-to-treat (ITT) population, defined as the time from randomisation until the first appearance of confirmed progressive disease as defined by a combined RECIST v1.1 and GCIG CA125 criteria (8) or death from any cause. Patients still alive and without progression at the time of analysis were censored at the date last known to be alive and progression-free. Secondary endpoints were Response rate (RR) (best recorded response according to combined RECISTv1.1 and GCIG CA125 criteria), Overall Survival (OS), defined as the time from randomisation until death from any cause, safety and tolerability (according to NCI CTCAE Version 4) and Quality of Life as measured by EQ-5D. The trial followed a three-outcome design (9): a PFS difference in favour of wP+V that was statistically significant at 10% was a clear signal that a subsequent phase III study is warranted. A result statistically significant at the 20% level (but not 10%) would require supportive data with improved response rate before a subsequent phase III would be considered. A total of 122 PFS events were required to detect a 50% improvement in median PFS from 3.7 months with wP+P to 5.55 months with wP+V with 90% power, at the 20% 1-sided level of statistical significance (or equivalently with 80% power at the 10% level of statistical significance). This required 140 patients (70 patients per arm) recruited over 16.5 months with 8 months subsequent follow-up. This incorporated an interim analysis for futility, after 40 PFS events had been observed, using a Lan-DeMets spending function (10) and Pocock type boundary (11). All analyses were pre-planned, unless specified, and performed using SAS Enterprise Guide 5.1. Efficacy analyses were undertaken on the ITT population, including all patients randomised into the study. Safety and tolerability analyses were undertaken on the safety population, defined as participant who received at least one dose of study treatment. PFS and OS were analysed using Cox regression via a model incorporating the blinded study arm and the factors used in the minimisation algorithm. A test of the proportional hazards (PH) assumption was performed by testing the significance of a time dependent covariate in the Cox model, as well as being assessed graphically. Response rates (complete and partial combined) were compared between the blinded study arms using logistic regression via a model incorporating study arm and the factors used in the minimization algorithm. QoL was analysed using mixed effects and repeated measures models, adjusting for minimisation factors, as well as time-point and interaction terms (a change from the SAP). Both complete case analyses (CC; ignoring missing data), and analyses accounting for missing values using multiple imputation (MI) with 40 imputations were undertaken (12). #### Translational analyses Details of the translational samples and analysis are given in Supplementary Methods. ## Results ### Patient Demographics A total of 140 patients were randomised (n=70 vistusertib, n=70 placebo) from 20 UK sites between January 2016 and March 2018 and included in the ITT population (Figure 1). Baseline characteristics are summarised in Table 1. The median age was 65 years (IQR 58–70) in the wP+V arm and 61 years (58-66) in the wP+P arm. 51% and 55% respectively had ≥3 prior treatment lines; 9% and 6% respectively had a taxane-free interval of <6 months whilst 29 patients (20.7%) had received prior weekly paclitaxel in combination with carboplatin, two of whom had received it twice. 139/140 patients had high grade serous histology (1 patient deemed ineligible after starting trial treatment was confirmed to have low grade serous carcinoma) and 17.9% were platinum-refractory. View this table: [Table 1.](http://medrxiv.org/content/early/2022/05/27/2022.05.26.22275618/T1) Table 1. Baseline characteristics ![Figure 1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/05/27/2022.05.26.22275618/F1.medium.gif) [Figure 1.](http://medrxiv.org/content/early/2022/05/27/2022.05.26.22275618/F1) Figure 1. CONSORT diagram ### Efficacy Median PFS was 4.5 (wP+V) vs 4.1 months (wP+P) (adjusted hazard ratio (HR) 0.84; 80% CI (0.67, 1.07); 1-sided p=0.18) (Figure 2A). Median OS was 9.7 vs 11.1 months (adjusted HR 1.21; 80% CI (0.91, 1.60); 2-sided p=0.80) (Figure 2B). Testing of the proportional hazards assumption confirmed that the above hazard ratios from the Cox proportional hazards model are appropriate. ![Figure 2A.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/05/27/2022.05.26.22275618/F2.medium.gif) [Figure 2A.](http://medrxiv.org/content/early/2022/05/27/2022.05.26.22275618/F2) Figure 2A. Progression-free survival: Kaplan-Meier curve ![Figure 2B.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/05/27/2022.05.26.22275618/F3.medium.gif) [Figure 2B.](http://medrxiv.org/content/early/2022/05/27/2022.05.26.22275618/F3) Figure 2B. Overall survival: Kaplan-Meier curve The RECIST 1.1 response rates were 29% vs 30% for wP+V and wP+P respectively and GCIG combined RECIST 1.1/CA125 criteria response rates were 53% and 54% respectively. There was no difference in response rate by combined GCIG RECIST1.1/CA125 criteria (adjusted odds ratio 0.86; 80% CI (0.55, 1.36); 1-sided p=0.66). Although PFS was significant at the 20% level, there was no improvement in response rate and therefore the primary endpoint was not met as per statistical considerations. The Forest plot in Figure 2C indicates the estimated hazard ratios for clinical minimisation factors with p-values for interaction test. There was no difference in PFS or OS according to measurable disease status or platinum status (refractory/resistant). Although there appears to be an interaction with taxane-free interval, a further analysis splitting taxane-free interval into approximate quartiles (to account for bias due the small number of patients in the <6 months group) showed no evidence of an interaction (p=0.83). ![Figure 2C.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/05/27/2022.05.26.22275618/F4.medium.gif) [Figure 2C.](http://medrxiv.org/content/early/2022/05/27/2022.05.26.22275618/F4) Figure 2C. Forest plot for clinical minimisation factors with p-values for interaction test ### Tolerability 136 participants were included in the safety population (n=68 in each arm). Table S1 summarises adverse events that occurred during combination therapy at grade 2 or above in >10% of patients in either arm or that are different significantly between the arms. Grade 3/4 adverse events were reported in 41.2% (wP+V) vs 36.7% (wP+P). Grade 1/2 adverse events were reported in 58.9% vs 63.2%. The most frequent grade 3/4 toxicities were lymphopenia (13% wP+V vs 9% wP+P) and fatigue (9% vs 4%). There was significantly more gastro-oesophageal reflux (grade 1/2 10% v 0%), rash (grade 2/3 9% v 0%) and lymphopenia (grade 2/3/4 47 v 31%) with wP+V compared to wP+P. There were no grade 4 or 5 events. View this table: [Table S1.](http://medrxiv.org/content/early/2022/05/27/2022.05.26.22275618/T2) Table S1. Adverse events that occurred during combination therapy at grade 2 or above in >10% of patients in either arm or that are different significantly between the arms ### Treatment delivery Table S2 summarises the treatment delivery of weekly paclitaxel and vistusertib/placebo. Dose intensity of weekly paclitaxel was similar between the wP+V and wP+P arms. Dose reductions and missed doses were greater for vistusertib than placebo (>1 dose reduction 19% vs 6%). The rate of discontinuation of vistusertib or placebo due to toxicity was 5% vs 0%. View this table: [Table S2.](http://medrxiv.org/content/early/2022/05/27/2022.05.26.22275618/T3) Table S2. Treatment delivery of weekly paclitaxel and vistusertib/placebo ### Quality of Life 126 patients were included in the QoL analysis. HRQoL questionnaires were completed at baseline (94.3%), weekly during treatment and 8-weekly post treatment. Overall QoL completion at time-points used in the analysis (up to and including week 28) was 79.3%. Figure S1 shows the EQ5D scores by time point. There were no statistically significant differences (at the 10% or 20% level) between the two treatment arms for EQ5D index or EQ5D VAS (p>0.25 in all analyses). ![Figure S1.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/05/27/2022.05.26.22275618/F7.medium.gif) [Figure S1.](http://medrxiv.org/content/early/2022/05/27/2022.05.26.22275618/F7) Figure S1. EQ5D scores by time-point. ### Biomarker Analysis Digital quantification of PTEN status showed a high correlation between QuPath and pathologist scores (r=0.94, *p*<0.0001 for tumour: r=0.70, *p*=0.009 for non-tumour – Figure S2, S3). Expression of PTEN was more variable in tumour cells (H-score range 23-258) than in non-tumour cells (range 44-177). Using pre-defined criteria (see Supplementary Methods), 13 (19.1%) cases were defined as PTEN low and 55 (80.9%) as PTEN high. There was a significant interaction between the randomisation arm and PTEN status (*p*=0.015) for PFS. Patients with PTEN low tumours showed a longer PFS compared with PTEN high patients in the experimental arm (median 9.4 (95%CI 2.8-16) vs 4.1 (95%CI 1.1-7) months respectively *p*=0.009; HR =0.14 (95%CI 0.03-0.62) but not in the control arm (4.8 (95%CI 2.5-7.0) vs 4.2 (95%CI 2.5-5.9) months respectively *p*=0.60) (Figure 3). No difference in OS was recorded although there was a trend toward a longer OS for PTEN low patients in the experimental arm (p=0.076). ![Figure S2](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/05/27/2022.05.26.22275618/F8.medium.gif) [Figure S2](http://medrxiv.org/content/early/2022/05/27/2022.05.26.22275618/F8) Figure S2 Hierarchical pathway on QuPath for PTEN Scoring. ![Figure 3.](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/05/27/2022.05.26.22275618/F5.medium.gif) [Figure 3.](http://medrxiv.org/content/early/2022/05/27/2022.05.26.22275618/F5) Figure 3. Progression-free survival according to PTEN IHC status A: Placebo; median PFS 4.4m (PTEN high) vs 4.7m (PTEN low); p=0.6077 B: Vistusertib: median PFS 4.0m (PTEN high) vs 9.3m (PTEN low); p=0.0031 HR=0.31 (95%CI 0.16–0.63) We used sWGS to analyse genome-wide absolute CN in 78 samples from 66 patients (with 12 matched study entry and archival pairs). There were no differences in ploidy, rates of focal somatic CN alterations or CN signature exposure between diagnosis and study entry (Figure S4), allowing us to combine archival and study entry samples as a single cohort for survival analyses. No CN signature had a statistically significant predictive value although a trend toward an interaction between the treatment and exposure to normalized signature 4 (*p*=0.071) was recorded for PFS. Therefore, we divided our cohort in two groups according to a pre-defined cut-off, the mean normalized signature 4 exposure. 40 (60.6%) patients had high exposure (defined as ≥mean) and 26 (39.4%) low (defined as 20% were marked up and sections were cut in the Experimental Cancer Medicine Centre (ECMC)/Imperial Cancer Biomarker Resource Centre (ICBRC) laboratory. DNA was thereafter extracted from 5–10 consecutive 10 μm sections using QIAmp DNA FFPE Tissue Kit (Qiagen, UK) according to the manufacturer’s protocol. 50-200ng was sheared with a Covaris LE220 focused ultrasonicator (Covaris, Woburn, MA) to produce 100-200bp fragments. Quantity was assessed using Qbit and they were immediately frozen in an ultra-low temperature freezer with a remote alarm system. Thereafter libraries were prepared in batches. ### Shallow Whole Genome Sequencing sWGS was executed using a HiSeq4000 system (Illumina Cambridge, UK), with paired-end 150 bp protocols. The input DNA was 250 ng according to the manufacturer’s instructions. Assuming a mean ploidy of 3 and 30kb bin size, we aimed to get 10 million reads per sample. This should allow detection of a CN change of 1 (tumour purity 0.6) – 2 (tumour purity 0.3) at 80% power, alpha=0.01 [https://gmacintyre.shinyapps.io/sWGS_power/](https://gmacintyre.shinyapps.io/sWGS_power/). Five 150PE HiSeq4000 lanes allowed us to sequence 146 DNA samples. The minimum number of reads per sample was set at 5–10 million (mean coverage of 0.1×). ### Absolute copy number and copy number signature calling sWGS reads were aligned to reference human genome hg19 using the BWA-MEM (3). Relative copy numbers (CN) were obtained for predefined 30kb bins using a modified version of the QDNASeq package (4). We obtained absolute CN using ACE (5). For each sample, we looked at top 10 best fits for purity and ploidy from ACE and chose the best fit that closely matched the *TP53* Variant Allele Frequency, as determined using targeted next-generation sequencing (Illumina AmpliSeq). In most cases, the ACE top fit was found to be the closest to *TP53* VAF. We defined a CN<2 as loss, 2-2.5 as normal, CN ≥2.5 but <5.0 as gain and ≥5 as amplification. CN signatures were called as previously described (6) and compared between cohorts using Kruskal Wallis test with Dunn’s multiple comparisons test on pairwise analysis of signatures-by-cohort. ### Statistics and analyses CN signature data are compositional (i.e. they sum to 1 in each sample). Thus, we selected one normalizing signature to perform a COX regression analysis. Signature 5 was chosen as it had the lowest variability across our two cohorts (archival and study entry). To avoid division errors, all signature exposures equal to 0 were converted to 0.01. The normalization of the remaining signature exposures was calculated using the log ratio of their exposure to the exposure of signature 5. A p value of 0.05 or lower was considered statistically significant. Unless otherwise specified, statistical analyses were performed using IBM SPSS Statistics (v27). ## Acknowledgements We thank all the patients who agreed to participate in this study; all participating sites, principal investigators and site staff; Cancer Research UK Glasgow Clinical Trials Unit, AstraZeneca, NIHR CRN Cancer Alliance, NHS Greater Glasgow and Clyde Sponsor Pharmacy Team, Translational Pharmacology Laboratory, WWRC, University of Glasgow, Lady Garden Foundation, European Society of Medical Oncology (GG recipient of translational research fellowship). the NIHR Biomedical Research Centres at The Royal Marsden NHS Foundation Trust/Institute of Cancer Research and Imperial, the CRUK/NIHR Experimental Cancer Medicine Centres at Imperial and The Institute of Cancer Research. ## Footnotes * **Disclosures/Conflicts of Interest** * **S Banerjee** Research Grants: Astrazeneca, GlaxoSmithKline; Advisory Boards: Amgen, Astrazeneca, Genmabs, GlaxoSmithKline Immunogen, Merck Sharpe Dohme, Merck Sereno, Mersana, Oncxerna, Seagen, Shattuck Labs; Honoraria for lectures: Amgen, Astrazeneca, Clovis, GlaxoSmithKine, Immunogen, Merck Sharpe Dohme, Mersana, Pfizer, Roche, Takeda. Clinical Advisory Board (uncompensated) Epsilogen * **G Giannone** Payment for educational events from Mylan, outside the submitted manuscript. * **A Clamp** Research grant- AstraZeneca, Advisory Board- GSK, MSD, Honoraria- Clovis Oncology, AstraZeneca * **R Glasspool** Rosalind Glasspool is Principal Investigator for trials sponsored by AstraZeneca, Clovis, GlaxoSmithKline/Tesaro, Immunogen, AbbVie, Pfizer, Lilly, and Novartis, and reports research funding from Clovis, Boehringer Ingelheim, and Lilly/Ignyta; institution research funding from GlaxoSmithKline/Tesaro; personal speaker and/or consultancy fees from AstraZeneca, MSD, Clovis, Tesaro, GlaxoSmithKline, Immunogen, and Sotio; and institutional consultancy fees from Novartis * **R Kristeleit** Grants: Clovis, MSD Advisory Boards/Consulting: GSK, Eisai, Astra Zeneca, Clovis, MSD, Basilea, Shattuck Labs, Zydus Cadila, iTEOS, InCyte, Regeneron. * **U Banerji** Honoraria from Astellas, Phoenix Solutions, Eli Lilly, Janssen, Boehringer Ingelheim Grant/Research support from BTG international, Carrick Therapeutics, Chugai, Verastem Oncology * **I A McNeish** Advisory boards and speaker fees for AZ, Clovis Oncology, GSK/Tesaro, Roche, OncoC4, Epsila Bio, Theolytics and Duke St Bio, all outside the scope of OCTOPUS. Institutional grant income from AZ also outside the scope of OCTOPUS. * Received May 26, 2022. * Revision received May 27, 2022. * Accepted May 27, 2022. * © 2022, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NoDerivs 4.0 International), CC BY-ND 4.0, as described at [http://creativecommons.org/licenses/by-nd/4.0/](http://creativecommons.org/licenses/by-nd/4.0/) ## References 1. 1.McNeish IA, Ledermann JA, Webber L, James L, Kaye SB, Hall M, et al. A randomised placebo-controlled trial of weekly paclitaxel and saracatinib (AZD0530) in platinum-resistant ovarian, fallopian tube or primary peritoneal cancer. Ann Oncol 2014;25(10):1988–95 doi 10.1093/annonc/mdu363. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/annonc/mdu363&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25070546&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) 2. 2.Peng DJ, Wang J, Zhou JY, Wu GS. Role of the Akt/mTOR survival pathway in cisplatin resistance in ovarian cancer cells. Biochemical and biophysical research communications 2010;394(3):600–5 doi 10.1016/j.bbrc.2010.03.029. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.bbrc.2010.03.029&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20214883&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) 3. 3.Choi HJ, Heo JH, Park JY, Jeong JY, Cho HJ, Park KS, et al. A novel PI3K/mTOR dual inhibitor, CMG002, overcomes the chemoresistance in ovarian cancer. Gynecol Oncol 2019;153(1):135–48 doi 10.1016/j.ygyno.2019.01.012. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ygyno.2019.01.012&link_type=DOI) 4. 4.Carden CP, Stewart A, Thavasu P, Kipps E, Pope L, Crespo M, et al. The association of PI3 kinase signaling and chemoresistance in advanced ovarian cancer. Mol Cancer Ther 2012;11(7):1609–17 doi 10.1158/1535-7163.Mct-11-0996. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6Im1vbGNhbnRoZXIiO3M6NToicmVzaWQiO3M6OToiMTEvNy8xNjA5IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDUvMjcvMjAyMi4wNS4yNi4yMjI3NTYxOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 5. 5.Behbakht K, Sill MW, Darcy KM, Rubin SC, Mannel RS, Waggoner S, et al. Phase II trial of the mTOR inhibitor, temsirolimus and evaluation of circulating tumor cells and tumor biomarkers in persistent and recurrent epithelial ovarian and primary peritoneal malignancies: a Gynecologic Oncology Group study. Gynecol Oncol 2011;123(1):19–26 doi 10.1016/j.ygyno.2011.06.022. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ygyno.2011.06.022&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21752435&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) 6. 6.Wong Te Fong AC, Thavasu P, Gagrica S, Swales KE, Leach MO, Cosulich SC, et al. Evaluation of the combination of the dual m-TORC1/2 inhibitor vistusertib (AZD2014) and paclitaxel in ovarian cancer models. Oncotarget 2017;8(69):113874–84 doi 10.18632/oncotarget.23022. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.18632/oncotarget.23022&link_type=DOI) 7. 7.Basu B, Krebs MG, Sundar R, Wilson RH, Spicer J, Jones R, et al. Vistusertib (dual m-TORC1/2 inhibitor) in combination with paclitaxel in patients with high-grade serous ovarian and squamous non-small-cell lung cancer. Ann Oncol 2018;29(9):1918–25 doi 10.1093/annonc/mdy245. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/annonc/mdy245&link_type=DOI) 8. 8.Rustin GJ, Vergote I, Eisenhauer E, Pujade-Lauraine E, Quinn M, Thigpen T, et al. Definitions for response and progression in ovarian cancer clinical trials incorporating RECIST 1.1 and CA 125 agreed by the Gynecological Cancer Intergroup (GCIG). Int J Gynecol Cancer 2011;21(2):419–23 doi 10.1097/IGC.0b013e3182070f17. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NDoiaWpnYyI7czo1OiJyZXNpZCI7czo4OiIyMS8yLzQxOSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIyLzA1LzI3LzIwMjIuMDUuMjYuMjIyNzU2MTguYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 9. 9.Hong S, Wang Y. A three-outcome design for randomized comparative phase II clinical trials. Statistics in medicine 2007;26(19):3525–34 doi 10.1002/sim.2824. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/sim.2824&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17262879&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) 10. 10.Lan KK, DeMets DL. Changing frequency of interim analysis in sequential monitoring. Biometrics 1989;45(3):1017–20. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2307/2531701&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=2790114&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=A1989AQ54000024&link_type=ISI) 11. 11.Pampallona S, Tsiatis AA, Kim K. Interim Monitoring of Group Sequential Trials Using Spending Functions for the Type I and Type II Error Probabilities. Drug Information Journal 2001;35(4):1113–21 doi 10.1177/009286150103500408. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/009286150103500408&link_type=DOI) 12. 12.Graham JW, Olchowski AE, Gilreath TD. How many imputations are really needed? Some practical clarifications of multiple imputation theory. Prev Sci 2007;8(3):206–13 doi 10.1007/s11121-007-0070-9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1007/s11121-007-0070-9&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17549635&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000249206400004&link_type=ISI) 13. 13.Lheureux S, Cristea MC, Bruce JP, Garg S, Cabanero M, Mantia-Smaldone G, et al. Adavosertib plus gemcitabine for platinum-resistant or platinum-refractory recurrent ovarian cancer: a double-blind, randomised, placebo-controlled, phase 2 trial. Lancet 2021;397(10271):281–92 doi 10.1016/s0140-6736(20)32554-x. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/s0140-6736(20)32554-x&link_type=DOI) 14. 14.Pujade-Lauraine E, Hilpert F, Weber B, Reuss A, Poveda A, Kristensen G, et al. Bevacizumab Combined With Chemotherapy for Platinum-Resistant Recurrent Ovarian Cancer: The AURELIA Open-Label Randomized Phase III Trial. J Clin Oncol 2014;32(13):1302–8 doi 10.1200/jco.2013.51.4489. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIzMi8xMy8xMzAyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDUvMjcvMjAyMi4wNS4yNi4yMjI3NTYxOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 15. 15.Poveda AM, Selle F, Hilpert F, Reuss A, Savarese A, Vergote I, et al. Bevacizumab Combined With Weekly Paclitaxel, Pegylated Liposomal Doxorubicin, or Topotecan in Platinum-Resistant Recurrent Ovarian Cancer: Analysis by Chemotherapy Cohort of the Randomized Phase III AURELIA Trial. J Clin Oncol 2015;33(32):3836–8 doi 10.1200/jco.2015.63.1408. [FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiRlVMTCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIzMy8zMi8zODM2IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDUvMjcvMjAyMi4wNS4yNi4yMjI3NTYxOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 16. 16.Oza A, Kaye S, Van Tornout J, Sessa C, Gore M, Naumann RW, et al. Phase 2 study evaluating intermittent and continuous linsitinib and weekly paclitaxel in patients with recurrent platinum resistant ovarian epithelial cancer. Gynecol Oncol 2018;149(2):275–82 doi 10.1016/j.ygyno.2018.01.019. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ygyno.2018.01.019&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) 17. 17.Kurzeder C, Bover I, Marmé F, Rau J, Pautier P, Colombo N, et al. Double-Blind, Placebo-Controlled, Randomized Phase III Trial Evaluating Pertuzumab Combined With Chemotherapy for Low Tumor Human Epidermal Growth Factor Receptor 3 mRNA-Expressing Platinum-Resistant Ovarian Cancer (PENELOPE). J Clin Oncol 2016;34(21):2516–25 doi 10.1200/jco.2015.66.0787. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIzNC8yMS8yNTE2IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDUvMjcvMjAyMi4wNS4yNi4yMjI3NTYxOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 18. 18.Grisham RN, Moore KN, Gordon MS, Harb W, Cody G, Halpenny DF, et al. Phase Ib Study of Binimetinib with Paclitaxel in Patients with Platinum-Resistant Ovarian Cancer: Final Results, Potential Biomarkers, and Extreme Responders. Clin Cancer Res 2018;24(22):5525–33 doi 10.1158/1078-0432.Ccr-18-0494. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6MTA6IjI0LzIyLzU1MjUiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMi8wNS8yNy8yMDIyLjA1LjI2LjIyMjc1NjE4LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 19. 19.Monk BJ, Kauderer JT, Moxley KM, Bonebrake AJ, Dewdney SB, Secord AA, et al. A phase II evaluation of elesclomol sodium and weekly paclitaxel in the treatment of recurrent or persistent platinum-resistant ovarian, fallopian tube or primary peritoneal cancer: An NRG oncology/gynecologic oncology group study. Gynecol Oncol 2018;151(3):422–7 doi 10.1016/j.ygyno.2018.10.001. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ygyno.2018.10.001&link_type=DOI) 20. 20.Ray-Coquard I, Pautier P, Pignata S, Perol D, Gonzalez-Martin A, Berger R, et al. Olaparib plus Bevacizumab as First-Line Maintenance in Ovarian Cancer. N Engl J Med 2019;381(25):2416–28 doi 10.1056/NEJMoa1911361. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1911361&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) 21. 21.Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, et al. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med 2011;365(26):2484–96 doi 10.1056/NEJMoa1103799 [doi]. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1103799&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22204725&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000298544600009&link_type=ISI) 22. 22.Burger RA, Brady MF, Bookman MA, Fleming GF, Monk BJ, Huang H, et al. Incorporation of bevacizumab in the primary treatment of ovarian cancer. N Engl J Med 2011;365(26):2473–83 doi 10.1056/NEJMoa1104390 [doi]. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1104390&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22204724&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000298544600008&link_type=ISI) 23. 23.Burger RA, Sill MW, Monk BJ, Greer BE, Sorosky JI. 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(33):5165–71. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyNS8zMy81MTY1IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDUvMjcvMjAyMi4wNS4yNi4yMjI3NTYxOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 24. 24.Cannistra SA, Matulonis UA, Penson RT, Hambleton J, Dupont J, Mackey H, et al. Phase II Study of Bevacizumab in Patients With Platinum-Resistant Ovarian Cancer or Peritoneal Serous Cancer. J Clin Oncol 2007;25(33):5180–6. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MzoiamNvIjtzOjU6InJlc2lkIjtzOjEwOiIyNS8zMy81MTgwIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDUvMjcvMjAyMi4wNS4yNi4yMjI3NTYxOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 25. 25.TCGA. Integrated genomic analyses of ovarian carcinoma. Nature 2011;474(7353):609–15. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature10166&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21720365&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000292204300032&link_type=ISI) 26. 26.Patch A-M, Christie EL, Etemadmoghadam D, Garsed DW, George J, Fereday S, et al. Whole–genome characterization of chemoresistant ovarian cancer. Nature 2015;521(7553):489–94 doi 10.1038/nature14410 [http://www.nature.com/nature/journal/v521/n7553/abs/nature14410.html#supplementary-information](http://www.nature.com/nature/journal/v521/n7553/abs/nature14410.html#supplementary-information). [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/nature14410&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26017449&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) 27. 27.LoRusso PM. Inhibition of the PI3K/AKT/mTOR Pathway in Solid Tumors. J Clin Oncol 2016;34(31):3803–15 doi 10.1200/jco.2014.59.0018. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1200/jco.2014.59.0018&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) 28. 28.Blagden SP, Hamilton AL, Mileshkin L, Wong S, Michael A, Hall M, et al. Phase IB Dose Escalation and Expansion Study of AKT Inhibitor Afuresertib with Carboplatin and Paclitaxel in Recurrent Platinum-resistant Ovarian Cancer. Clin Cancer Res 2019;25(5):1472–8 doi 10.1158/1078-0432.Ccr-18-2277. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTA6ImNsaW5jYW5yZXMiO3M6NToicmVzaWQiO3M6OToiMjUvNS8xNDcyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDUvMjcvMjAyMi4wNS4yNi4yMjI3NTYxOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 29. 29.Del Campo JM, Birrer M, Davis C, Fujiwara K, Gollerkeri A, Gore M, et al. A randomized phase II non-comparative study of PF-04691502 and gedatolisib (PF-05212384) in patients with recurrent endometrial cancer. Gynecol Oncol 2016;142(1):62–9 doi 10.1016/j.ygyno.2016.04.019. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ygyno.2016.04.019&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27103175&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) 30. 30.Wilson MK, Pujade-Lauraine E, Aoki D, Mirza MR, Lorusso D, Oza AM, et al. Fifth Ovarian Cancer Consensus Conference of the Gynecologic Cancer InterGroup: recurrent disease. Ann Oncol 2017;28(4):727–32 doi 10.1093/annonc/mdw663. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/annonc/mdw663&link_type=DOI) 31. 31.van der Ploeg P, Uittenboogaard A, Thijs AMJ, Westgeest HM, Boere IA, Lambrechts S, et al. The effectiveness of monotherapy with PI3K/AKT/mTOR pathway inhibitors in ovarian cancer: A meta-analysis. Gynecol Oncol 2021;163(2):433–44 doi 10.1016/j.ygyno.2021.07.008. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.ygyno.2021.07.008&link_type=DOI) 32. 32.Sieuwerts AM, Inda MA, Smid M, van Ooijen H, van de Stolpe A, Martens JWM, et al. ER and PI3K Pathway Activity in Primary ER Positive Breast Cancer Is Associated with Progression-Free Survival of Metastatic Patients under First-Line Tamoxifen. Cancers 2020;12(4) doi 10.3390/cancers12040802. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3390/cancers12040802&link_type=DOI) 33. 33.Mafi S, Mansoori B, Taeb S, Sadeghi H, Abbasi R, Cho WC, et al. mTOR-Mediated Regulation of Immune Responses in Cancer and Tumor Microenvironment. Frontiers in immunology 2021;12:774103 doi 10.3389/fimmu.2021.774103. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.3389/fimmu.2021.774103&link_type=DOI) 34. 34.Martins FC, Santiago I, Trinh A, Xian J, Guo A, Sayal K, et al. Combined image and genomic analysis of high-grade serous ovarian cancer reveals PTEN loss as a common driver event and prognostic classifier. Genome biology 2014;15(12):526 doi 10.1186/s13059-014-0526-8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/s13059-014-0526-8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25608477&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) 35. 35.Martins FC, Couturier DL, Paterson A, Karnezis AN, Chow C, Nazeran TM, et al. Clinical and pathological associations of PTEN expression in ovarian cancer: a multicentre study from the Ovarian Tumour Tissue Analysis Consortium. Br J Cancer 2020;123(5):793–802 doi 10.1038/s41416-020-0900-0. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41416-020-0900-0&link_type=DOI) 36. 36.Ciriello G, Miller ML, Aksoy BA, Senbabaoglu Y, Schultz N, Sander C. Emerging landscape of oncogenic signatures across human cancers. Nat Genet 2013;45(10):1127–33 doi 10.1038/ng.2762. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/ng.2762&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=24071851&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) 37. 37.Macintyre G, Goranova T, De Silva D, Ennis D, Piskorz AM, Eldridge M, et al. Copy-number signatures and mutational processes in ovarian carcinoma. Nat Genet 2018;50(9):1262–70 doi 10.1038/s41588-018-0179-8. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-018-0179-8&link_type=DOI) 38. 38.Cheng Z, Mirza H, Ennis DP, Smith P, Morrill Gavarró L, Sokota C, et al. The Genomic Landscape of Early-Stage Ovarian High-Grade Serous Carcinoma. Clin Cancer Res 2022:Of1–of12 doi 10.1158/1078-0432.Ccr-21-1643. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1158/1078-0432.Ccr-21-1643&link_type=DOI) 39. 39.Coleman RL, Oza AM, Lorusso D, Aghajanian C, Oaknin A, Dean A, et al. Rucaparib maintenance treatment for recurrent ovarian carcinoma after response to platinum therapy (ARIEL3): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390(10106):1949–61 doi 10.1016/S0140-6736(17)32440-6. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(17)32440-6&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) 40. 40.Mirza MR, Monk BJ, Herrstedt J, Oza AM, Mahner S, Redondo A, et al. Niraparib Maintenance Therapy in Platinum-Sensitive, Recurrent Ovarian Cancer. N Engl J Med 2016;375(22):2154–64 doi 10.1056/NEJMoa1611310. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa1611310&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27717299&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) ## References 1. 1.Bakkar RM, Xie SS, Urbauer DL, Djordjevic B, Vu K, Broaddus RR. Intact PTEN Expression by Immunohistochemistry is Associated With Decreased Survival in Advanced Stage Ovarian/Primary Peritoneal High-grade Serous Carcinoma. International journal of gynecological pathology : official journal of the International Society of Gynecological Pathologists. 2015;34(6):497–506. 2. 2.Martins FC, Couturier DL, Paterson A, Karnezis AN, Chow C, Nazeran TM, et al. Clinical and pathological associations of PTEN expression in ovarian cancer: a multicentre study from the Ovarian Tumour Tissue Analysis Consortium. Br J Cancer. 2020;123(5):793–802. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41416-020-0900-0&link_type=DOI) 3. 3.Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25(14):1754–60. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/bioinformatics/btp324&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19451168&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F05%2F27%2F2022.05.26.22275618.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000267665900006&link_type=ISI) 4. 4.Scheinin I, Sie D, Bengtsson H, van de Wiel MA, Olshen AB, van Thuijl HF, et al. DNA copy number analysis of fresh and formalin-fixed specimens by shallow whole-genome sequencing with identification and exclusion of problematic regions in the genome assembly. Genome research. 2014;24(12):2022–32. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6NjoiZ2Vub21lIjtzOjU6InJlc2lkIjtzOjEwOiIyNC8xMi8yMDIyIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDUvMjcvMjAyMi4wNS4yNi4yMjI3NTYxOC5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 5. 5.Poell JB, Mendeville M, Sie D, Brink A, Brakenhoff RH, Ylstra B. ACE: absolute copy number estimation from low-coverage whole-genome sequencing data. Bioinformatics (Oxford, England). 2019;35(16):2847–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1093/BIOINFORMATICS/BTY1055&link_type=DOI) 6. 6.Macintyre G, Goranova T, De Silva D, Ennis D, Piskorz AM, Eldridge M, et al. Copy-number signatures and mutational processes in ovarian carcinoma. Nat Genet. 2018;50(9):1262–70. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41588-018-0179-8&link_type=DOI)