Article Text

other Versions

Download PDFPDF

New horizons for platinum-resistant ovarian cancer: insights from the 2023 American Society of Clinical Oncology (ASCO) and European Society for Medical Oncology (ESMO) Annual Meetings
Free
  1. Khalid El Bairi1,2,
  2. Ainhoa Madariaga3,
  3. Dario Trapani4,5,
  4. Ouissam Al Jarroudi1,6 and
  5. Said Afqir1,6
  1. 1Department of Medical Oncology, Mohammed VI University Hospital, Oujda, Morocco
  2. 2Faculty of Medical Sciences, University Mohammed 6 Polytechnic, Ben Guerir, Morocco
  3. 3Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
  4. 4European Institute of Oncology, IRCCS, Milan, Italy
  5. 5Department of Oncology and Hematology, University of Milan, Milan, Italy
  6. 6Faculty of Medicine and Pharmacy, Mohammed Ist University, Oujda, Morocco
  1. Correspondence to Mr Khalid El Bairi, Faculty of Medical Sciences, University Mohammed 6 Polytechnic, Ben Guerir, Morocco; k.elbairi{at}ump.ac.ma

Abstract

Platinum-resistant ovarian cancer is difficult to treat and has a poor prognosis. Patients with platinum-resistant ovarian cancer have limited treatment options and often have a limited benefit from existing chemotherapeutic agents. There is a lack of contemporary effective anticancer drugs and reliable predictive biomarkers for this aggressive cancer. Recent cutting-edge research presented at the 2023 American Society of Clinical Oncology (ASCO) and the European Society for Medical Oncology (ESMO) Annual Meetings has provided insights into several potential therapeutic targets, such as DNA damage repair proteins, cell-cycle regulators, and immune-modulating agents. In addition, antibody–drug conjugates have provided new practice-changing results in platinum-resistant ovarian cancer. Here, we review the results of research presented at this annual event, with a focus on clinical trials investigating novel treatment approaches for platinum-resistant ovarian cancer, in addition to predictive and prognostic biomarkers for optimal patient selection, and other topics, such as real-world evidence.

  • ovarian cancer

Statistics from Altmetric.com

Request Permissions

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

Introduction

Platinum-resistant epithelial ovarian, fallopian tube or primary peritoneal cancer denotes a treatment-resistant type of ovarian cancer associated with poor prognosis, and it poses a significant challenge in clinical practice. Patients with platinum-resistant ovarian cancer have restricted therapeutic options and commonly present limited benefit from available chemotherapy and targeted agents. Moreover, the development of new anticancer drugs and validated predictive biomarkers has been scarce in this space. Platinum resistance in ovarian cancer has traditionally been assessed using the platinum-free interval definition. Patients are classified as platinum-sensitive, platinum-resistant, or platinum-refractory based on the time elapsed between their last dose of platinum-based chemotherapy and relapse.1 2 Nevertheless, this approach faces challenges due to disease heterogeneity, evolving mechanisms of drug resistance, the imprecise estimation of a 6-month threshold, recent advances in predictive biomarkers within this context, and the emergence of non-platinum and targeted therapies. Consequently, these factors have prompted a reconsideration of patient stratification in contemporary phase III clinical trial design, moving beyond the platinum-free interval.3

To date, only three randomized controlled trials in platinum-resistant ovarian cancer have demonstrated a progression-free survival advantage. This includes two trials that investigated anti-angiogenic agents (bevacizumab and trebananib) alongside chemotherapy, but without improvements in overall survival.4 The lack of robust predictive biomarkers, insufficient preclinical and early clinical evidence, and inadequate methodology could explain these disappointing results. Promisingly, the recent MIRASOL trial that investigated mirvetuximab soravtansine was the first to demonstrate superior progression-free survival and overall survival for the first time in this setting.5 Recent research developments have provided valuable insights into this particular setting of ovarian cancer, which is known for its poor survival outcomes.6

Preclinical studies, early-phase trials, and randomized clinical trials exploring emerging therapeutic targets, including DNA damage repair proteins, such as ataxia telangiectasia and Rad3-related protein (ATR) and cell-cycle regulators like WEE1 and FAK kinases, as well as immune-modulating targets, including programmed cell death protein 1 (PD1)/programmed cell death ligand 1 (PD-L1) inhibitor combinations, have shown promising directions for future therapeutic approaches. Antibody drug conjugates have been broadly implemented in clinical research for multiple cancer types and have similarly dominated the scene for platinum-resistant ovarian cancer drug development recently.

The 2023 American Society of Clinical Oncology (ASCO) and European Society of Medical Oncology (ESMO) Annual Meetings (ASCO23, ESMO23) serve as a platform for oncology professionals worldwide to share and discuss the latest advances in cancer research and treatments. In this paper, we review cutting-edge research presented at these meetings, focusing on clinical trials investigating novel treatment approaches for patients with platinum-resistant ovarian cancer.

DEVELOPMENTAL THERAPEUTICS: MOLECULARLY TARGETED AGENTS

Phase III Trials: MIRASOL, OVAL, and ARTISTRY-7

MIRASOL

Several clinical trials investigating novel anticancer drugs such as antibody–drug conjugates and inducers of antitumor-directed immune response for the treatment of platinum-resistant ovarian cancer were presented at ASCO23, including 10 early-phase studies and three phase III trials encompassing one study protocol (Table 1).5 7–22 The advent of antibody–drug conjugates has yielded promising findings in some solid cancers, propelling their clinical development for platinum-resistant ovarian cancer.23 Antibody–drug conjugates possess a distinct advantage of using the principle of immune bioconjugation to selectively administer cytotoxic drugs to cancer cells that express multiple antigens based on specific monoclonal antibodies. This innovative therapeutic class has demonstrated encouraging preliminary evidence of clinical efficacy in platinum-resistant ovarian cancer,23 a cancer type commonly displaying chemotherapy resistance. Mirvetuximab soravtansine is an antibody–drug conjugate that targets folate receptor α (FR-α) and induces cell death in FR-α expressing tumors. Encouraging results were demonstrated in the multi-institutional single-arm SORAYA phase II study (NCT04296890) that investigated mirvetuximab soravtansine in an FR-α-high platinum-resistant ovarian cancer population.24 These findings prompted further exploration of the activity in the MIRASOL (GOG-3045; NCT04209855) phase III confirmatory trial, that randomized 453 patients with high-grade platinum-resistant ovarian cancer to receive mirvetuximab soravtansine or investigator’s choice of chemotherapy including topotecan, pegylated liposomal doxorubicin, or paclitaxel in a 1:1 fashion.5 Progression-free survival as assessed by investigators was the primary endpoint, and overall survival and objective response rate were secondary endpoints. Patient selection was accomplished based on biomarker immunohistochemical analysis for FR-α using FOLR1 assay: patients with platinum-resistant ovarian cancer expressed in at least 75% of viable tumor cells exhibiting ≥2+ level membrane staining intensity, including those with BRCA1/2 mutations. Patients were randomized to receive mirvetuximab soravtansine at a dose of 6 mg/kg on day 1 every 3 weeks (q3w) or investigator’s choice of chemotherapy. Of note, nearly half of enrolled patients (47%) had received three prior lines of therapies. Moreover, prior poly-ADP ribose polymerase (PARP) inhibitors and bevacizumab were allowed as maintenance therapies and had been received by 55% and 62% of enrolled patients, respectively. At the time of data cut-off and after a median follow-up of 13.1 months, progression-free survival was improved, reporting 5.62 months in the mirvetuximab soravtansine arm compared with 3.98 months in the control (HR 0.65, 95% CI 0.52 to 0.81, p<0.0001). In addition, a superior objective response rate was detected in the mirvetuximab soravtansine arm (42.3% vs 15.9%, p<0.0001), including 12 complete responses. In a pre-planned sensitivity analysis, these findings were consistent with those of a blinded independent central review. The interim analysis of overall survival showed a better outcome in patients treated with mirvetuximab soravtansine than those who received the investigator’s choice of chemotherapy (16.46 vs 12.75 months; HR 0.67, 95% CI 0.50 to 0.89, p=0.0046).5 Of note, this was the first study to report an improved overall survival in phase III trials for platinum-resistant ovarian cancer.

Table 1

Summary of clinical trials on developmental therapeutics

In an exploratory subgroup analysis, this clinical activity was more pronounced in bevacizumab-naive patients. The group of patients (n=172) who had no previous exposure to bevacizumab had progression-free survival superior by 34% as compared with the investigator’s choice of chemotherapy (HR 0.66, 95% CI 0.46 to 0.94, p=0.02). Similarly, this group had also a reduced risk of death by 49% (overall survival HR 0.51, 95% CI 0.31 to 0.86, p<0.01). Regarding patients who received prior treatments that included bevacizumab (n=281), progression-free survival only was improved in the mirvetuximab soravtansine subset compared with investigator’s choice of chemotherapy (HR 0.64, 95% CI 0.49 to 0.84, p<0.01) and overall survival was not significantly different between the two arms (HR 0.74, p=0.07).5 Regarding the toxicity profile specific to mirvetuximab soravtansine, grade ≥3 ocular events including 8% blurred vision and 9% keratopathy were remarkable in this trial, which may considerably impair patients’ quality of life (QoL). Ocular toxicities must be carefully screened, promptly diagnosed, and timely treated with this drug.

While the effectiveness of biomarker-driven mirvetuximab soravtansine has been confirmed in high-FRα-expressing patients in the MIRASOL trial, ocular toxicity is a worrying adverse event and needs a mitigation plan before infusion.25 The US Prescribing Information (USPI) alerted healthcare providers in a boxed warning about the potential risks associated with mirvetuximab soravtansine ocular toxicity that may lead to significant vision impairment and corneal disorders in addition to pneumonitis and peripheral neuropathy as other considerable safety risks.26 Ocular toxicities likely associated with off-target effects can go unnoticed without symptoms until significant damage occurs. Other important safety risks such as pneumonitis and peripheral neuropathy should also be considered and are included as a warning box for mirvetuximab soravtansine. Therefore, dose optimization strategies could be considered for mirvetuximab soravtansine to improve its safety profile, such as fractionated dosing regimens, as it could achieve a similar efficacy profile as the approved dose/schedule.26

Patients who had primary platinum-refractory disease were excluded from this trial. This also raises concerns about this patient population that has limited treatment options and does not benefit from the recent advances in treating this highly aggressive gynecological cancer, particularly on how the trial mirrors a real-life population. Primary patient-reported outcomes in MIRASOL were not disclosed yet and are awaited in the upcoming meetings. These outcomes are critical patient-centric endpoints to evaluate treatments and support patient-centered care more comprehensively. Globally, only a portion of patients with platinum-resistant ovarian cancer in real-life setting may have similar inclusion criteria than the one used in the MIRASOL study. The financial toxicity of antibody–drug conjugates in general, and particularly of mirvetuximab soravtansine,27 is also an important factor that will limit access to this innovative drug globally as it places a significant financial burden on patients and their families. It is crucial to acknowledge that recognizing these problems holds equal significance as celebrating these wins.

To date, mirvetuximab soravtansine received accelerated approval from the US Food and Drug Administration (FDA) on November 14, 2022, for treating patients with platinum-resistant ovarian cancer who have FR-α-positive tumors and received 1–3 prior lines of systemic treatments. The FDA has also approved the VENTANA FOLR1 (FOLR-2.1) RxDx Assay as a companion diagnostic to identify eligible patients for mirvetuximab soravtansine.26 Doublet-based combination approaches using mirvetuximab soravtansine and other drugs such as bevacizumab may provide improved response and meaningful clinical benefits in the platinum-resistant ovarian cancer population.28 Of course, preventing and optimizing the safety profile is now a priority.

OVAL

The OVAL (GOG-3018) phase III trial investigated ofranergene obadenovec in platinum-resistant ovarian cancer. Ofranergene obadenovec, known as VB-111, is a viral-mediated anti-angiogenic and vascular disrupting agent, based on a non-replicating adenovirus targeted against hypoxic tissue targets to exert endothelial cell killing to the neoangiogenic tumor vasculature.29 Mechanistically, the precise molecular mechanisms by which ofranergene obadenovec acts on ovarian cancer cells are not fully understood. Ofranergene obadenovec was engineered to induce the expression of the Fas-TNFR1 chimeric pro-apoptotic protein exclusively in angiogenic vessels. It is believed that its interaction with tumour necrosis factor α (TNF-α) in the tumor microenvironment of ovarian cancer leads to the apoptosis of endothelial cells.29

Previously, ofranergene obadenovec was studied in a dose-escalation phase I/II study that enrolled 21 patients with platinum-resistant ovarian cancer, including 50% with platinum-refractory disease, to receive ofranergene obadenovec in combination with weekly paclitaxel.29 At a therapeutic dose (1×1013 viral particles), ofranergene obadenovec showed superior overall survival as compared with the subtherapeutic dose (16.6 vs 5.8 months) and tolerable toxicities, mainly mild flu-like symptoms. More recently, the OVAL study was designed to confirm these preliminary signals of activity. Four hundred and nine patients with platinum-resistant ovarian cancer were randomly assigned in a 1:1 ratio to receive ofranergene obadenovec at a therapeutic dose every 8 weeks combined with paclitaxel at a dose of 80 mg/m2 or placebo with paclitaxel until disease progression, inacceptable toxicity, and per patient preference.7 Progression-free survival and overall survival by blinded independent central review were both co-primary endpoints. Topline mature data presented at ASCO23 showed that the results of the OVAL study were negative. In fact, median progression-free survival was not different between the two arms (5.29 vs 5.36 months, HR 1.03, 95% CI 0.83 to 1.29, p=0.7823). Similarly, median overall survival was not improved in the experimental arm (13.37 vs 13.14 months, HR 0.97, 95% CI 0.75 to 1.27, p=0.8440). The safety profile was similar to that reported in previous early-phase studies.7 These disappointing results were not in line with a futility analysis of this trial, that reported promising interim results.30 A pre-planned interim analysis suggested that the initial response rates based on cancer antigen 125 (CA125) in the OVAL trial were supportive of the study plan.30 Yet, the final OVAL findings raise questions regarding the value of using CA125 to predict response in this perspective, particularly in large phase III trials that enroll an important number of patients without sufficient quality evidence from early-phase studies or interim analyses. This is an important matter to protect patients by avoiding overtreatment in clinical trials with ineffective therapies. Evidence from the OVAL study should be used to pause and reflect on the way we track the disease trajectory in clinical trials to understand the benefit of treatments, and how they may not apply to all treatment types—in this case, with a biological agent.

ARTISTRY-7

ARTISTRY-7 (GOG-3063, NCT05092360) is an ongoing phase III trial that is currently recruiting patients with platinum-resistant ovarian cancer to be randomized to receive the engineered cytokine nemvaleukin alfa (ALKS 4230) and pembrolizumab, pembrolizumab monotherapy, nemvaleukin monotherapy, versus protocol-specific investigator’s choice chemotherapy.8 Nemvaleukin was designed to activate antitumor immunity mediated by CD8+T cells and natural killers by binding to interleukin 2 (IL-2) receptor.31 The protocol of this study presented at ASCO23 indicated a study population of 376 patients with platinum-resistant ovarian cancer who will be randomly assigned to one of four arms (3:1:1:3) including intravenous nemvaleukin at a dose of 6 µg/kg on days 1 to 5 and pembrolizumab at a dose of 200 mg as an infusion on day 1 of q3w cycle. The other groups will receive pembrolizumab, nemvaleukin as single agents or investigator’s choice chemotherapy (pegylated liposomal doxorubicin, weekly paclitaxel, topotecan, or gemcitabine). To be included, patients may have received up to five prior lines of systemic treatment in a platinum resistant setting and at least one line of therapy containing bevacizumab. Moreover, those with mucinous histology or primary platinum-refractory disease or with platinum-free interval of <3 months from first-platinum, will be excluded.8 Of note, this study will also enroll patients with clear cell ovarian cancer, a rare type that is frequently neglected in major clinical trials. A pre-planned stratification based on PD-L1 status, histology, and chemotherapy was intentional. Progression-free survival by the investigator is the primary endpoint and overall survival and other tumor measures are secondary endpoints.8 The existing data on this novel agent are scarce. In fact, the available conference proceedings showed that nemvaleukin combined with pembrolizumab in the ARTISTRY-1 phase I/II multicohort study (NCT02799095) was active in five subjects of total patients enrolled with heavily pre-treated platinum-resistant ovarian cancer.32 This included one confirmed complete response, two confirmed partial responses, and one unconfirmed partial response. In addition, one patient exhibited stable disease after being on the treatment for approximately 1.5 years.32 However, one can argue that conducting a phase III trial without clear positive results from powered early-phase studies is questionable, as efficacy in early-phase trials can anticipate the overall benefit in later stages of drug development. Moreover, progression-free survival should not be considered the best endpoint in this setting when investigating doublets. In platinum-resistant ovarian cancer, overall survival and QoL outcomes should be the gold standards to objectively assess the benefits of novel agents.33 In fact, progression-free survival as a surrogate endpoint to overall survival is unclear in trials assessing immunotherapy and in gynecological cancers.33

Phase I/II Trials

Luveltamab Tazevibulin

Luveltamab tazevibulin (STRO-002) is an FR-α-targeting antibody–drug conjugate with a drug-to-antibody ratio of 4 that was investigated in ovarian cancer settings with the advantages of no reported ocular toxicities.34–36 This antibody–drug conjugate uses the anti-tubulin payload 3-aminophenyl hemiasterlin to induce cytotoxic and immunologic cell death.37 Luveltamab tazevibulin received fast-track approval by the FDA in August 2021. An interim analysis of recent safety data of this agent used in patients at a higher dose (5.4 mg/kg once q3w) showed grade ≥3 neutropenia requiring pegfilgrastim treatment but without ocular adverse events.37 Of note, pegfilgrastim is a pegylated granulocyte-colony stimulating factor (G-CSF) used to enhance white blood cell production and is commonly used to prevent neutropenia in patients with cancer.38 A higher dose of luveltamab tazevibulin was associated with superior benefits including an objective response rate of 43.8%. Based on this, the phase II/III REFRaME trial (NCT05200364) will be initiated to investigate luveltamab tazevibulin in platinum-resistant ovarian cancer.35 At ASCO23, Oaknin et al reported recent updated data on luveltamab tazevibulin which was studied for relapsed patients with ovarian cancer in the global STRO-002-GM1 phase I trial.9 The study enrolled patients with platinum-sensitive and platinum-resistant ovarian cancer and this latter group had progressive disease after 1–3 prior lines of treatments. These patients were assigned to receive intravenous q3w luveltamab tazevibulin at doses of 4.3 or 5.2 mg/m2 (1:1 ratio) until disease progression. Biomarker analysis in this trial was not planned and it was provided as a post-hoc review of archived tissues. Objective response rate by Response Evaluation Criteria in Solid Tumors (RECIST, v1.1) was the primary endpoint, and progression-free survival and duration of response were secondary objectives. The majority of enrolled patients had prior exposure to bevacizumab and PARP inhibitors. The objective response rate was 31.7% in the overall population and 37.5% in FR-α >25%. In addition, the objective response rate was more pronounced (43.8%) in the higher starting dose in patients with FR-α >25% tumors. Yet, data regarding the impact of platinum sensitivity is lacking. The most frequent grade ≥3 adverse events included neutropenia, arthralgia, and anemia (70.5%, 18.2%, and 13.6%, respectively). These emerging adverse events were associated with a dose delay and reduction in 80% and 61% of the study participants, respectively, particularly in those who received a higher therapeutic dose (5.2 mg/kg).9 However, this novel antibody–drug conjugate may provide new therapeutic perspectives for platinum-resistant ovarian cancer if demonstrated in large trials, particularly given the early response seen in patients with a broader expression of FR-α. Currently, a phase I study (NCT05200364) evaluating the combination of luveltamab tazevibulin with bevacizumab is recruiting patients with advanced refractory or relapsed ovarian cancer after progression on available standard chemotherapies, and it is expected to release early findings in January 2024. Moreover, the REFRaME-O1 phase II/III trial investigating the efficacy and safety of luveltamab tazevibulin in patients with platinum-resistant ovarian cancer with FR-α >25% is currently open for enrollment (NCT05870748).

Fostamatinib

Fostamatinib is an orally available inhibitor of spleen tyrosine kinase (SYK). SYK has been found to be involved in conferring resistance to paclitaxel in ovarian cancer by modulating the stability of microtubules and has been correlated with poor overall survival in ovarian cancer patients.39 Preclinical models have shown that the use of fostamatinib sensitizes resistant cells to paclitaxel, thereby inducing cytotoxic effects.39 Based on these findings, the toxicity and early efficacy of fostamatinib were investigated in a recent phase I study presented at ASCO23.12 This trial enrolled patients with recurrent platinum-resistant ovarian cancer in a dose-escalation manner to determine the maximum tolerated dose and recommended phase II dose. Response rates were evaluated using RECIST v1.1 criteria. Eligible patients (n=27) received fostamatinib orally twice a day at three dose levels (100, 150, and 200 mg) in combination with intravenous paclitaxel (80 mg/m2, on days 1, 8, and 15, q4w). Adverse events attributed to the treatment included diarrhea, fatigue, anemia, neutropenia, nausea, dysgeusia, and hypertension. Grade 3 or 4 toxicities observed were neutropenia (37%), anemia (26%), and thromboembolic events (22%). The determined dose for the expansion cohort will be 200 mg when combined with paclitaxel.12 The efficacy data provided in the available abstract showed 39% of partial and complete responses; the study is still ongoing (NCT03246074).

SL-172154

SL-172154, also known as SIRPα-Fc-CD40L, is a fusion protein that incorporates the extracellular domains of signal regulatory protein α (SIRPα) and CD40L linked by an Fc domain.40 This complex molecule functions by blocking CD47, thereby increasing the capacity of macrophages to enhance phagocytosis and promoting CD40-based processing of antigens, leading to their presentation on antigen-presenting cells to CD8+T lymphocytes. A first-in-human phase I trial (NCT04406623) with a dose escalation design investigated the toxicity and pharmacokinetics of SL-172154 in patients with platinum-resistant ovarian cancer through intravenous administration of five different doses (0.1, 0.3, 1.0, 3.0, 10 mg/kg).13 The available results from a cohort of 27 patients with advanced platinum-resistant ovarian cancer showed one dose-limiting toxicity of grade 3 alanine aminotransferase increase. Grade 3/4 adverse events included aspartate aminotransferase elevation and lymphopenia (7% each). Infusion-related reaction rates increased with higher doses. The best response observed was stable disease in 22% (n=6) of evaluable patients.13 A maximum tolerated dose was not reached in this study. Combination therapies that target the CD47-SIRPα axis have shown higher response rates in solid tumors, as indicated by a recent meta-analysis.41

Debio-0123

Debio-0123 is a WEE1 inhibitor that has been studied in a phase I trial (Debio 0123–101; NCT03968653). WEE1 is a tyrosine kinase that regulates the cell cycle to prevent entry into the mitotic process.42 It has been identified as a prognostic biomarker of poor overall survival in patients with serous ovarian carcinomas treated with chemotherapy (reviewed elsewhere, see Schutte et al42). Targeting WEE1 by Debio-0123, which is an orally available, ATP-competitive WEE1 inhibitor, is being studied as a single agent or in combination with carboplatin in various solid tumors.17 Currently, no comprehensive data on this agent, including preclinical studies, are available to the public. During ASCO23, a poster presentation provided insights into the toxicity and preliminary activity of Debio-0123. The authors of a phase I trial (n=38) reported on the pharmacokinetics, pharmacodynamics, and anticancer activity of combining carboplatin with Debio-0123 in patients who had recurred or progressed following prior platinum-based therapy.17 The study followed a two-arm dose escalation design, administering Debio-0123 as a single agent and in combination with carboplatin. The maximum tolerated dose was found to be 520 mg, and the majority of adverse events were grade 1 or 2. In terms of clinical benefits, four out of the 12 enrolled patients with platinum-resistant ovarian cancer achieved partial responses.17

PRGN-3005 Autologous UltraCAR-T Cells

Chimeric antigen receptor (CAR)-T has recently attracted significant attention and has rapidly progressed to enhance the arsenal of cancer treatments, particularly in blood cancers, where it has shown remarkable results. CAR-T cell strategies involve targeting specific tumor cell antigens expressed in cancer. In ovarian cancer, proof of concept evidence on therapeutic targeting of various antigens, such as CA125 and mesothelin, has yielded promising T cell infiltration in both primary and recurrent disease.43 In mouse models, treatment using CAR-T cells was found to control the dissemination of ovarian cancer in vivo.44 Recently, an investigational phase I/Ib clinical trial (NCT03907527) evaluated PRGN-3005 UltraCAR-T cells, manufactured overnight and targeting CA125, in a population of patients with platinum-resistant ovarian cancer.14 In the phase I part of this trial, heavily pre-treated patients were infused intraperitoneally with PRGN-3005 without lymphodepletion or intravenously at three dose levels. Dose level 3 of intravenous injection was further assessed in patients who also received cyclophosphamide. PRGN-3005 was found to be well-tolerated at up to 65.5×105 cells/kg, and no dose-limiting toxicity was reported, including known adverse events of CAR-T cells such as grade ≥3 cytokine-release syndrome and on-target/off-target toxicities. Regarding clinical response, evaluated by RECIST v1.1, 20% of enrolled patients had a response to at least one target lesion. The group treated with intravenous doses had an 85.7% disease control rate, including a decrease in target tumor burden in four of the seven enrolled women.14 The phase Ib part of this trial is currently recruiting more patients, and final results are expected to be delivered in November 2028 (NCT03907527).

Raludotatug Deruxtecan

Raludotatug deruxtecan (DS-6000) is a novel antibody–drug conjugate containing a stable linker and a topoisomerase I inhibitor targeting cadherin 6 (CDH6). CDH6 is frequently expressed in primary and relapsed ovarian cancer of all histotypes, particularly in high-grade serous tumors.45 Early findings from an ongoing phase I trial (NCT04707248) demonstrated premature efficacy signals in heavily pretreated ovarian cancer patients, including those with platinum-resistant disease.22 In this study, patients who had received prior taxane and platinum treatments were enrolled to receive raludotatug deruxtecan as a single agent, without CDH6 preselection, at various dose-escalating levels. The maximum tolerated dose was determined to be 8 mg/kg. A subgroup analysis of this trial (n=40 with platinum-resistant ovarian cancer), presented at ESMO23, reported a 50% incidence of grade ≥3 adverse events, including nausea, fatigue, diarrhea, vomiting, anemia, and decreased neutrophil and platelet counts. These events were all manageable and consistent with previous reports on similar antibody–drug conjugates using the same payload.22 The assessment of response based on RECIST v1.1 revealed a 38% objective response rate at 4.8 mg/kg, 33% at 6.4 mg/kg, and 31% at 8 mg/kg. The encouraging activity observed in heavily pretreated platinum-resistant ovarian cancer supports the continuation of this ongoing phase I trial.

Cobimetinib

Cobimetinib is an orally available small molecule that inhibits mitogen-activated protein kinase (MEK) within the mitogen-activated protein kinase (MAPK)/ERK pathway.19 This signaling pathway is commonly over-activated in human tumors, including certain histotypes of ovarian cancer.46 The BOUQUET study (NCT04931342), presented at ESMO23, is a phase II trial evaluating various treatments in biomarker-selected patients with rare ovarian cancer who exhibit lower responsiveness to standard treatments.19 The interim analysis of this study, which investigated cobimetinib in women with BRAF/KRAS/NRAS or NF1 alterations compared with atezolizumab and bevacizumab in ineligible patients, reported an objective response rate of 16% in the cobimetinib arm and 14% in the combination arm. Notably, patients with low-grade serous tumors and mesonephric-like adenocarcinoma showed an improved response rate of 33%, supporting further enrollment of these patients with these rare histotypes.19

IN10018

IN10018 selectively inhibits focal adhesion kinase (FAK), a non-receptor tyrosine kinase with a primary role in regulating adhesion, migration, and cell survival.47 In ovarian cancer, FAK has been found to be overexpressed and associated with prognostic features such as stage and overall survival.47 Additionally, FAK orchestrates immunosuppression, lymphangiogenesis, and metastasis in ovarian cancer.48 49 The targeting of FAK is now fully explored as a therapeutic strategy in ovarian cancer, particularly in low-grade serous carcinomas.50 Recently, survival data from the phase Ib trial of IN10018 (n=61; NCT05551507) were presented at ESMO23.20 The study investigated the preliminary efficacy of IN10018 in combination with pegylated liposomal doxorubicin in patients with platinum-resistant ovarian cancer, focusing specifically on high-grade serous tumors. The primary endpoint, objective response rate, was 46.3%, and secondary objectives, including progression-free survival at 7.3 months and overall survival at 20.9 months, were achieved. The safety profile was manageable, primarily associated with known toxicities of pegylated liposomal doxorubicin.20 A phase II trial comparing this combination to a placebo is currently in the planning stages.

Pressurized Intraperitoneal Aerosolized Chemotherapy (PIPAC)

Treating recurrent ovarian cancer with peritoneal metastases poses a challenge as it is often associated with suboptimal debulking and resistance to intravenous platinum-based chemotherapy. Pressurized intraperitoneal aerosolized chemotherapy (PIPAC) was developed as an alternative method to deliver chemotherapy as aerosol compounds while maintaining high pressure in the peritoneum. Trials using various PIPAC procedures have shown encouraging objective response rates and improvements in QoL. Therefore, PIPAC has been proposed as a therapeutic option for patients with refractory peritoneal disease of various origins, including ovarian cancer. Previously, prospective case series and early evidence on the use of PIPAC in platinum-resistant ovarian cancer showed positive objective tumor response in a setting that poses important clinical challenges.51 52 A recent first-in-US phase I multicohort study communicated as a poster session investigated the role of PIPAC in seven pretreated patients with platinum-resistant ovarian cancer using a combination of cisplatin and doxorubicin.18 Eligible patients had recurrent ovarian cancer with peritoneal involvement and possible extraperitoneal metastases. Dose limiting toxicity was the primary endpoint, and progression-free survival and QoL were secondary objectives. A total of 71.4% of the participants in the study successfully completed PIPAC without encountering any surgical complications. The median decrease in peritoneal cancer index was −1.4% (from cycle 1 to cycle 2) and −2.8% (from cycle 1 to cycle 3). The clinical benefits of PIPAC were seen in a subset of patients with low-grade serous histology treated with six cycles and showed a partial response according to RECIST criteria.18

To date, this study is the only one to assess PIPAC efficacy as a palliative therapeutic option in selected platinum-resistant ovarian cancer patients, and merits further investigations compared with conventional chemotherapy. Of course, the evidence is insufficient to recommend any clinical implementation now. Other findings from negative and ongoing early-phase trials for patients with platinum-resistant ovarian cancer are summarized in Table 1.

ADVANCES IN BIOMARKERS, REAL-WORLD EVIDENCE, QUALITY OF LIFE, AND COST-EFFECTIVENESS ANALYSIS

Other presented studies at these events comprised biomarker/pharmacokinetic analyses (n=6), preclinical studies (n=2), real-world evidence (n=2), QoL (n=1), and cost-effectiveness analyses (n=1) (Table 2).53–64

Table 2

Summary of biomarker studies and other topics

Preclinical Studies

Oncolytic virotherapy, which involves the intratumor replication and spread of viral vectors to enhance the antitumor effects, represents a distinctive therapeutic approach for ovarian cancer. In platinum-resistant ovarian cancer, early clinical response to this therapeutic approach was seen in women who received intraperitoneal Olvi-Vec, a modified vaccinia virus, as monotherapy in a phase Ib trial.65 Mechanistically, the use of oncolytic viruses was found to induce immune activation by enhancing tumor infiltration of CD8+T lymphocytes and activation of tumor-specific T cells in circulation, in addition to natural killer (NK) cell cytotoxicity and macrophage phagocytosis.65 66 Krell et al demonstrated recently at the ASCO23 that THEO-260, which is an oncolytic virus-based agent targeting both cancer cells and cancer-associated fibroblasts, is active in treatment-naive and platinum-resistant patient derived samples.61 The authors of this abstract showed that THEO-260 enhances interferon γ-mediated T cell response. This agent was found to exert antitumor effects in in vivo murine models with PARP and platinum resistant tumors.37

Another preclinical study, presented at ESMO23, has provided novel insights into the molecular mechanisms of sensitivity to PARP inhibitors observed in a subgroup of patients with platinum-resistant ovarian cancer.67 The authors investigated CircIGF1R_0001, a circular endogenous non-coding RNA that regulates various cell functions.67 They demonstrated that its overexpression promotes resistance to platinum drugs while enhancing the response to PARP inhibitors by upregulating the PARylation activity of PARP1.67 This finding may open a new avenue of opportunity for the use of PARP inhibitors in selected patients with platinum-resistant ovarian cancer.

Biomarker/Pharmacokinetic Analyses

The ATR-CHK1-WEE1 axis contains several cell cycle checkpoint kinases that are upregulated in response to replication stress. Mechanistically, active proteins in this signaling cascade promote cell cycle arrest, replication fork stabilization, and DNA repair.68 Investigating this pathway in ovarian cancer has produced several clinical candidates for development in early phase studies.42 69 To enhance the response to platinum-based chemotherapy by sensitizing ovarian cancer, a WEE1 inhibitor, azenosertib, was developed and studied in a phase I trial (NCT04516447).53 This orally available agent was demonstrated to selectively inhibit DNA repair; it was assessed in a multicenter phase I trial in combination with paclitaxel, gemcitabine, carboplatin, or pegylated liposomal doxorubicin for platinum-resistant ovarian cancer patients who received one or two lines of platinum-based chemotherapy. Safety and recommended dose for phase II were primary objectives and clinical activity based on objective response rate and progression-free survival were secondary endpoints. At data cut-off, 94 of 103 patients were eligible for efficacy review. The highest efficacy was noticed for the combination of azenosertib and paclitaxel (objective response rate of 50%). In a biomarker analysis of a group of 80 patients, highly expressed cyclin E1 was shown to be correlated with superior outcomes (objective response rate 31.3% vs 7.7% and progression-free survival 10.35 vs 3.25 months).53

In another biomarker analysis of a proof-of-concept phase II study that investigated the checkpoint kinase 1 (CHK1) inhibitor prexasertib,70 innate immunity was found to modulate drug response.54 This study was a phase II trial that enrolled 49 patients with BRCA1/2 wild type platinum-resistant ovarian cancer to receive intravenous prexasertib every 2 weeks (q2w) in a 28-day cycle. In patients who had improved progression-free survival, flow cytometry analysis revealed a high rate of CD83 positive dendritic cells in the blood with a mature and functional phenotype (p<0.001 and p<0.001, respectively).54 Patients who did not benefit from prexasertib had an increase of immunosuppressive monocytic myeloid-derived suppressor cells (p<0.001) and classical monocytes (p=0.003).54 This suggests that response to this CHK1 inhibitor may be attributable to innate immunity. The study further confirmed the negative impact of these cells on immunosurveillance as modifiers of response to CHK1 inhibitors including prexasertib.71 According to the latest updates on the status of this trial based on ClinicalTrials.gov (NCT02203513), the study was prematurely terminated by the sponsor.

Dynamic changes in cancer immunity as a predictive biomarker are also being studied in other ongoing trials including its correlation with response to pembrolizumab and lenvatinib as monotherapies or in combination for patients with platinum-resistant ovarian cancer.59 An ongoing pilot phase II study (NCT05114421) in this setting will enroll 30 patients to assess the synergism between these two drugs and the effects of peritoneal tumor microenvironment based on multichannel flow cytometry. This includes proliferative and dysfunctional T cell populations in patients who received single agents or as doublets.59 This study is expected to release preliminary findings in early 2024. The impact of the immune component in conferring a chemotherapy resistance phenotype in ovarian cancer was also highlighted in another study presented at ESMO23.62 This was a longitudinal multicenter investigation that prospectively enrolled 159 patients, including 31 women with platinum-resistant ovarian cancer. A significantly lower activity of the JAK/STAT signaling pathway was observed in refractory ovarian cancer. This finding was further confirmed using an additional method that demonstrated a reduced expression of transcriptional target genes associated with the interferon α/β and γ pathways. Additionally, this group exhibited lower CD4+ T cell infiltration.62 Notably, mutational signatures related to homologous recombination repair were less common in samples derived from platinum-resistant ovarian cancer treated with neoadjuvant chemotherapy.62

Real-World Evidence: Does PARP Inhibition Increase the Risk of Platinum Resistance?

PARP inhibitors are approved as a maintenance therapy as they were demonstrated to enhance the duration of progression-free survival in women with newly diagnosed and recurrent platinum-sensitive ovarian cancer.72 73 To date, little is known about the evolution of disease behavior following treatment with these novel agents. Nonetheless, there is an increasing amount of evidence suggesting poor response to subsequent platinum-based chemotherapy after exposure to PARP inhibitors.74–76 An abstract of a retrospective real-world Canadian cohort (n=91) that included patients with recurrent high-grade epithelial ovarian cancer following niraparib or olaparib maintenance treatments showed that patients who experienced progression following PARP inhibitors had a reduced response to subsequent platinum-based chemotherapy.56 In fact, 59.3% of the study population had disease progression after initiating PARP inhibitors, suggesting a possible contribution of these agents in inducing platinum-resistance in recurrent disease.56 Similarly, another abstract by Guo and Zhang investigated the effects of PARP inhibitors used as maintenance treatment after first-line and second-line chemotherapy on response to platinum in a Chinese cohort of 172 patients with recurrent high-grade serous ovarian cancer.58 Patients who were exposed to PARP inhibitors were more likely to develop platinum resistance independently of the duration of PARP inhibitor use. These novel findings confirm the previous observations74–76 which demonstrated that patients receiving PARP inhibitors are less likely to respond to platinum-based subsequent chemotherapy.74–76 Therefore, impairment of platinum sensitivity after PARP inhibitors raises concerns for possible involvement of these new agents in conferring resistance to chemotherapy in this aggressive cancer where chemotherapy still plays a cornerstone role. Moreover, emerging hypotheses suggest that treatment of ovarian cancer using PARP inhibitors may also impact the response to non-platinum agents and surgical debulking rates.77 However, the association of PARP inhibition and sensitivity to treatments is not yet well established and more prospective research is needed to elucidate this phenomenon.

Quality of Life, Access, Policy, and Equity

Patients with platinum-resistant ovarian cancer face health challenges associated with extensive treatments involving multiple lines of therapies, in addition to cumulative burden of disease progressions. Notably, QoL in this setting predicts overall survival.78 A systematic review, presented at ESMO23, aimed to characterize the health-related QoL in this population by evaluating 39 studies.64 The findings showed a significant baseline burden of symptoms for platinum-resistant ovarian cancer patients, encompassing gastrointestinal concerns, fatigue, sleep disturbances, pain, anxiety, and depression. Across studies and therapeutic regimens, outcomes were diverse, with the majority of patients reporting either stable or worsening QoL.64 The limited benefits observed from existing treatments underscore the urgent need for interventions that enhance QoL, highlighting the need for tolerable treatments able to alleviate patients’ symptoms in this aggressive disease.

ASCO23 has been declined toward the themes of an improvement of access and sustainability of cancer care. A group of researchers from Korea conceptualized an innovative approach to PIPAC that is devoted to feasibility, implementation, and sustainability as a goal. One key issue with new cancer treatments, including complex and resource intensive approaches like PIPAC, is that it can commonly be provided in selected institutions, thus generating a pabulum of possible social exclusion and inequity for patients who cannot access or afford those interventions. Understanding how innovation can be implemented requires feasibility and economic evaluations, that are less often underlined during oncology meetings. At ASCO23, a group of researchers from the Seoul National University Hospital provided cost-effectiveness estimates for a new approach to delivering intraperitoneal chemotherapy. Such an approach consists of a device capable to deliver aerosolized chemotherapy agents, installed on an oscillating pendulum capable to angulate the jet of infusion, and more efficiently spread across the abdominal cavity: it is called rotational intraperitoneal pressurized aerosol chemotherapy (RIPAC). It is under development, and preclinical models have been previously developed.79 In the abstract presented at ASCO23, the authors developed a cost-utility analysis, embedding data on the effectiveness of RIPAC, as compared with standard of care chemotherapy agents including pegylated liposomal doxorubicin, gemcitabine, and topotecan in the setting of platinum-resistant ovarian cancer.57 The authors estimated that three cycles of palliative chemotherapy would result in direct medical costs of US$9000 to US$16 000. Intraperitoneal pressurized aerosol chemotherapy is not a standard procedure, and the cost for implementing the device was assumed from an ingredient-based micro-costing exercise to be around US$4000. Assuming a QoL gain that is similar to the standard chemotherapy at least, the authors concluded that intraperitoneal pressurized aerosol chemotherapy could be cost-effective from the Korean payers’ perspective. It is clear that one cannot deliver ultimate cost-effectiveness considerations without high-level quality data on how the health interventions perform and impact on patient outcomes, but it is remarkable that the authors wanted to frame cost assumptions as part of the transparent plan of development of a new therapeutic approach. In fact, while attitudes of exclusive profiting from healthcare are common with innovative cancer treatments, implementing innovation in oncology should be directed toward access and sustainability. At this point, improving outcomes solely in clinical trials is not sufficient anymore. Access to all is key.

CONCLUSIONS

Platinum resistance is a poor prognostic factor in ovarian cancer. This poses a significant challenge in the management of this disease. Results from studies presented at ASCO23 and ESMO23 provided a promise for contemporary approaches to enhance therapeutic effectiveness and overcome tumor resistance mechanisms. This encompassed antibody–drug conjugates for treating patients with platinum-resistant ovarian cancer, with mirvetuximab soravtansine showing promising results in biomarker-selected populations, although ocular toxicity was a notable concern that requires mitigation strategies. Full publications of these studies are awaited for a better appraisal of the current emerging evidence. Future large studies in this setting should be built on sufficient evidence from early phase clinical trials to limit the exposure of patients to non-active treatments.

Ethics statements

Patient consent for publication

References

Footnotes

  • Twitter @elbairikhalid19, @AinhoaMada

  • Correction notice This article has been corrected since it was first published. An inaccurate study completion date has been removed.

  • Contributors KE wrote the manuscript. AM, DT reviewed the manuscript. OA and SA supervised the review writing. All authors approved the final version of the manuscript.

  • Funding The authors have not declared a specific grant for this research from any funding agency in the public, commercial or not-for-profit sectors.

  • Competing interests KE reports receiving fees and honoraria from NCODA, Techspert, Elsevier and Springer and funding from the Cancer Research Institute (Morocco). DT, SA, and OA: nothing to report. AM: reports honoraria from AstraZeneca, GSK, MSD, PharmaMar.

  • Provenance and peer review Not commissioned; externally peer reviewed.