Skip to main content

OPINION

Oncol Rev., 20 September 2023

Rationale for the Use of Homologous Recombination Proficient Molecular Profile as a Biomarker for Therapeutic Targeting in Ovarian Cancer

John Nemunaitis
John Nemunaitis1*Laura StanberyLaura Stanbery1Adam Walter,Adam Walter1,2Rodney RocconiRodney Rocconi3Philip Stephens,Philip Stephens1,4
  • 1Gradalis, Inc, Dallas, TX, United States
  • 2ProMedica, Toledo, OH, United States
  • 3Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Mobile, AL, United States
  • 4Naveris, Waltham, MA, United States

Introduction

Therapeutic options for advanced-stage ovarian cancer patients are limited in those subjects with homologous recombination proficient molecular profiles. A recent review of the existing literature demonstrates evidence of enhanced relapse-free survival and overall survival associated with treatment with Vigil in the Phase 2b trial in the HRP population. Homologous recombination (HR) is a genetic rearrangement in which molecular information is exchanged between two similar molecules of double-stranded or single-stranded nucleic acids [1]. The purpose of HR is to maintain genome stability by performing high-fidelity repair of complex DNA damage such as DNA double-strand breaks and interstrand crosslinks [24].

Homologous recombination is responsible for double-stranded DNA breaks and interstrand crosslink damage repair through the use of sister chromatids as a repair template. BRCA1/2 are critically important proteins in this pathway. HR deficiency (D) is the result of germline or somatic genetic alterations in HR genes (i.e., BRCA 1 or 2) [5]. Dysfunctional HR genes cause genome-wide errors and can lead to tumorigenesis [6, 7]. Tumors that are not HRD are considered HR proficient (P) and contain no functional genetic alterations in HR pathway genes, like BRCA1/2, resulting in faithful DNA repair, thereby reducing the mutation burden. While the HR pathway is responsible for repairing double-stranded breaks, the base excision repair pathway repairs single-stranded DNA breaks. Poly (ADP-ribose) polymerase proteins (PARPs) are essential proteins in this pathway. When PARPs are inhibited, single-stranded breaks are converted to double-stranded breaks during DNA replication. Synthetic lethality occurs in cells treated with a PARP inhibitor that have a BRCA mutation or are HRD.

Alterations in HR pathway genes, especially mutations in BRCA1/2, can be germline and confer familial risk for breast, ovarian, prostate, and pancreatic cancer [8] or somatic. For patients who demonstrate negative germline testing, somatic HR molecular status is assessed by NGS and is most commonly evaluated by Myriad’s MyChoice CDx-testing. This involves the analysis of BRCA 1 and 2 gene mutation status, loss of heterozygosity (LOH), telomeric allelic imbalance (TAI), and large-scale state transition score (LST) to determine a genomic instability score (GIS) [9]. Each is weighted and scored using a proprietary algorithm to determine the level of genomic instability. A GIS ≥42 in BRCA 1 or 2 negative patients defines HRD status. A GIS score <42 defines HRP status [10]. BRCA 1 or 2 mutations or HRD molecular profile tumors are a sensitive ovarian cancer population to PARP inhibitor therapy [1014] and are associated with a better prognosis in patients receiving platinum-based chemotherapy and/or bevacizumab [15]. However, ovarian cancer patients with HRP molecular status have a worse prognosis with standard-of-care therapy involving PARPIs, bevacizumab, and platinum-based chemotherapy [11, 1619]. This is related to the ability of HRP molecular status tumors to perform DNA repair, resulting in decreased DNA damage induced by these therapeutic options. Regardless of mutation status, all tumors demonstrate benefit from frontline maintenance treatment with PARP inhibitors, although the magnitude of benefit is greater in patients with a BRCA mutation or HRD, as demonstrated in multiple clinical trials [11, 12]. However, multiple resistance mechanisms have been demonstrated. Additionally, PARP inhibitors in the recurrent setting have recently been shown to be detrimental to OS, and the FDA has removed them from this setting in patients without a BRCA mutation1 [20, 21]. Additional studies are underway investigating PARP inhibitor combination therapy to overcome the limitations of PARP inhibitor therapy [22]. Results are expected soon from several large Phase 3 clinical trials evaluating combination PARP inhibitor and checkpoint inhibitor therapy. Previous studies evaluating checkpoint inhibitors in ovarian cancer have been largely negative [23, 24].

Vigil

Vigil is a novel, triple-function, cell-based immunotherapy recently cleared by the FDA for the initiation of a phase 3 registration trial in newly diagnosed stage IIIb/IV HRP-positive ovarian cancer patients. Vigil expresses GMCSF, an immunostimulatory cytokine, and a bifunctional short-hairpin RNA to knockdown furin. Furin is the critical convertase responsible for the cleavage of TGFβ1 and 2. Finally, Vigil provides the full complement of personal neoantigens relevant to the patient’s cancer, allowing for T cell education and priming. Data supporting the efficacy of Vigil in the ovarian cancer population include results from phase 1, 2a, and 2b trials [2531].

A phase 2b, double-blind placebo-controlled trial [25] recently evaluated 91 newly diagnosed stage IIIb-IV ovarian cancer patients randomized to Vigil vs. placebo at maintenance following debulking surgery and combination cisplatin/taxane induction chemotherapy. The molecular profiles of the 91 patients enrolled in the study included BRCA-mutant, HRD, and HRP patients. At the time of trial initiation and accrual, the use of somatic HRD/HRP testing was not part of clinical practice, so somatic testing was not done prospectively. A post hoc analysis demonstrated improved clinical benefit correlated with increased DNA repair capacity (HRP) with Vigil. A trend toward clinical benefit in RFS was observed in all patients (HR 0.688, p = 0.078) [25]. However, a statistically significant survival benefit was observed in the non-germline mutated population (combination of HRD and HRP) in both RFS and OS (HR 0.514, p = 0.020; HR 0.493, p = 0.049, respectively). The HRD/HRP subgroups were evaluated using Myriad’s MyChoice CDx. The greatest benefit following Vigil treatment was observed in those patients with the highest capacity for DNA repair and those with the HRP molecular profile, in both RFS and OS (HR 0.386, p = 0.007 and HR 0.342, p = 0.019, respectively) [26, 27]. This effect was durable and continued at a long-term follow-up of 3 years [27].

Discussion

Recent literature, both preclinical and clinical, has convincingly demonstrated the role of clonal neoantigen burden in correlating OS improvement with checkpoint inhibitor therapy in advanced cancer patients [3234]. CD8+ lymphocytes reactive to clonal neoantigens have been identified in multiple studies [3236]. Durable clinical benefit has been correlated with the identification of T cells recognizing clonal neoantigens (not subclonal neoantigens) [3236]. Cytotoxic chemotherapy has been shown to increase the proportion of subclonal neoantigens and reduce the expression and visibility of clonal neoantigens, thereby reducing the responsiveness of malignant cells to immunotherapy [32, 34, 35]. The inconsistency of PD-L1 expression and checkpoint inhibitor response appears to be related to the proportion of clonal neoantigens targeting effector cells. We hypothesized that Vigil activity would be more likely to provide clinical benefit in patient tumors with higher expression of clonal neoantigens (present on all tumor cells) as opposed to subclonal neoantigens (only present on newly mutated subpopulations). It is our premise that Vigil construction involving autologous tumors and ex vivo transfection with a dual plasmid containing bi-shRNAi furin/GMCSF wild-type DNA would generate a more active clonal neoantigen profile to induce an effector cell response in malignant cells containing the HRP molecular profile [3740]. In this scenario, the clonally matched targets are more highly visible and associated with greater effector cell responsiveness as compared to effector cells impacting subclonal neoantigens that are selectively on tumor cells (Figure 1). This effect has been demonstrated in the BRCA-wt HRP population clinical benefit to Vigil.

FIGURE 1
www.frontiersin.org

FIGURE 1. Homologous Recombination Deficient (HRD) and Proficient (HRP) OvCs: neoantigen fraction (clonal vs. subclonal) affects immunogenicity. The clinical benefit of Vigil is improved with increased clonal neoantigen display.

In conclusion, the achievement of high clonal neoantigen targeting capacity provides an enhancement in immunotherapeutic proficiency and is likely induced by Vigil treatment and optimized by HRP molecular profile capacity.

Author Contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Conflict of Interest

JN reports employment by, and ownership of stocks in Gradalis, Inc. AW and LS report employment by Gradalis, Inc. RR reports sponsored research by Gradalis, Inc. PS reports consulting fees paid by Gradalis, Inc and is employed by Naveris.

The authors declare this study received funding from Gradalis, Inc. The funder had the following involvement with the study: manuscript preparation and the decision to submit for publication.

Acknowledgments

The authors would like to thank Brenda Marr for her competent and knowledgeable assistance in the preparation of the manuscript.

Footnotes

1Clovis Oncology. Rubraca Rucaparib for Treatment of BRCA-Mutated Ovarian Cancer after 2 or More Chemotherapies Is Voluntarily Withdrawn in the U.S. Health Care Provider Letter (2022).

References

1. Piazza, A, and Heyer, WD. Homologous Recombination and the Formation of Complex Genomic Rearrangements. Trends Cel Biol (2019) 29(2):135–49. doi:10.1016/j.tcb.2018.10.006

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Li, X, and Heyer, WD. Homologous Recombination in DNA Repair and DNA Damage Tolerance. Cell Res (2008) 18(1):99–113. doi:10.1038/cr.2008.1

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Paques, F, and Haber, JE. Multiple Pathways of Recombination Induced by Double-Strand Breaks in Saccharomyces cerevisiae. Microbiol Mol Biol Rev (1999) 63(2):349–404. doi:10.1128/mmbr.63.2.349-404.1999

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Hunter, N. Meiotic Recombination. In: A Aguilera, and R Rothstein, editors. Homologous Recombination. Berlin-Heidelberg: Springer-Verlag (2007).

Google Scholar

5. da Cunha Colombo Bonadio, RR, Fogace, RN, Miranda, VC, and Diz, MPE. Homologous Recombination Deficiency in Ovarian Cancer: A Review of its Epidemiology and Management. Clinics (Sao Paulo) (2018) 73(1):e450s. doi:10.6061/clinics/2018/e450s

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Hoeijmakers, JH. Genome Maintenance Mechanisms for Preventing Cancer. Nature (2001) 411(6835):366–74. doi:10.1038/35077232

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Petrucelli, N, Daly, MB, and Feldman, GL. Hereditary Breast and Ovarian Cancer Due to Mutations in BRCA1 and BRCA2. Genet Med (2010) 12(5):245–59. doi:10.1097/gim.0b013e3181d38f2f

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Heeke, A, Pishvaian, MJ, Lynce, F, Xiu, J, Brody, JR, Chen, WJ, et al. Prevalence of Homologous Recombination-Related Gene Mutations across Multiple Cancer Types. JCO Precis Oncol (2018) 2018(10). doi:10.1200/PO.17.00286

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Miller, RE, Leary, A, Scott, C, Serra, V, Lord, C, Bowtell, D, et al. ESMO Recommendations on Predictive Biomarker Testing for Homologous Recombination Deficiency and PARP Inhibitor Benefit in Ovarian Cancer. Ann Oncol (2020) 31(12):1606–22. doi:10.1016/j.annonc.2020.08.2102

PubMed Abstract | CrossRef Full Text | Google Scholar

10. 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

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Ray-Coquard, I, Pautier, P, Pignata, S, Pérol, D, González-Martín, 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

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Gonzalez-Martin, A, Pothuri, B, Vergote, I, DePont Christensen, R, Graybill, W, Mirza, MR, et al. Niraparib in Patients with Newly Diagnosed Advanced Ovarian Cancer. N Engl J Med (2019) 381(25):2391–402. doi:10.1056/nejmoa1910962

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Kaufman, B, Shapira-Frommer, R, Schmutzler, RK, Audeh, MW, Friedlander, M, Balmaña, J, et al. Olaparib Monotherapy in Patients with Advanced Cancer and a Germline BRCA1/2 Mutation. J Clin Oncol (2015) 33(3):244–50. doi:10.1200/jco.2014.56.2728

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Miller, RE, Elyashiv, O, El-Shakankery, KH, and Ledermann, JA. Ovarian Cancer Therapy: Homologous Recombination Deficiency as a Predictive Biomarker of Response to PARP Inhibitors. OncoTargets Ther (2022) 15:1105–17. doi:10.2147/ott.s272199

CrossRef Full Text | Google Scholar

15. Tewari, KS, Burger, RA, Enserro, D, Norquist, BM, Swisher, EM, Brady, MF, et al. Final Overall Survival of a Randomized Trial of Bevacizumab for Primary Treatment of Ovarian Cancer. J Clin Oncol (2019) 37(26):2317–28. doi:10.1200/jco.19.01009

PubMed Abstract | CrossRef Full Text | Google Scholar

16. Ni, J, Guo, W, Zhao, Q, Cheng, X, Xu, X, Zhou, R, et al. Homologous Recombination Deficiency Associated with Response to Poly (ADP-Ribose) Polymerase Inhibitors in Ovarian Cancer Patients: The First Real-World Evidence from China. Front Oncol (2021) 11:746571. doi:10.3389/fonc.2021.746571

PubMed Abstract | CrossRef Full Text | Google Scholar

17. Creeden, JF, Nanavaty, NS, Einloth, KR, Gillman, CE, Stanbery, L, Hamouda, DM, et al. Homologous Recombination Proficiency in Ovarian and Breast Cancer Patients. BMC Cancer (2021) 21(1):1154. doi:10.1186/s12885-021-08863-9

PubMed Abstract | CrossRef Full Text | Google Scholar

18. Banerjee, S, Gonzalez-Martin, A, Harter, P, Lorusso, D, Moore, KN, Oaknin, A, et al. First-Line PARP Inhibitors in Ovarian Cancer: Summary of an ESMO Open - Cancer Horizons Round-Table Discussion. ESMO Open (2020) 5(6):e001110. doi:10.1136/esmoopen-2020-001110

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Swisher, EM, Kaufmann, S, Birrer, M, Levine, D, Moore, K, Fleming, G, et al. Exploring the Relationship Between Homologous Recombination Score and Progression-Free Survival in BRCA Wildtype Ovarian Carcinoma: Analysis of Veliparib Plus Carboplatin/paclitaxel in the Velia Study. Gynecol Oncol (2020) 159:51. doi:10.1016/j.ygyno.2020.06.107

CrossRef Full Text | Google Scholar

20. GlaxoSmithKline. Zejula (Niraparib) Important Prescribing Information for the Maintenance Treatment of Adult Patients with Non-gBRCAmut Recurrent Epithelial Ovarian, Fallopian Tube, or Primary Peritoneal Cancer Who Are in a Complete or Partial Response to Platinum-Based Chemotherapy in Second or Later Line Setting (2022). Available from: https://www.zejulahcp.com/content/dam/cf-pharma/hcp-zejulahcp-v2/en_US/pdf/ZEJULA%20(niraparib)%20Dear%20HCP%20Letter%20November%202022.pdf (Accessed March 17, 2023).

Google Scholar

21. GlaxoSmithKline. ZEJULA® Niraparib for the Treatment of Adult Patients with Advanced Ovarian, Fallopian Tube, or Primary Peritoneal Cancer Who Have Been Treated with 3 or More Prior Chemotherapy Regimens Is Voluntarily Withdrawn in the U.S. (2022). Available from: https://medinfo.gsk.com/5f95dbd7-245e-4e65-9f36-1a99e28e5bba/57e2a3fa-7b9b-432f-a220-5976a509b534/57e2a3fa-7b9b-432f-a220-5976a509b534_viewable_rendition__v.pdf (Accessed March 17, 2023).

Google Scholar

22. Giannini, A, Di Dio, C, Di Donato, V, D’oria, O, Salerno, MG, Capalbo, G, et al. PARP Inhibitors in Newly Diagnosed and Recurrent Ovarian Cancer. Am J Clin Oncol (2023) 46(9):414–9. doi:10.1097/COC.0000000000001024

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Monk, BJ, Colombo, N, Oza, AM, Fujiwara, K, Birrer, MJ, Randall, L, et al. Chemotherapy with or without Avelumab Followed by Avelumab Maintenance Versus Chemotherapy Alone in Patients with Previously Untreated Epithelial Ovarian Cancer (JAVELIN Ovarian 100): An Open-Label, Randomised, Phase 3 Trial. Lancet Oncol (2021) 22(9):1275–89. doi:10.1016/s1470-2045(21)00342-9

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Moore, KN, Bookman, M, Sehouli, J, Miller, A, Anderson, C, Scambia, G, et al. Atezolizumab, Bevacizumab, and Chemotherapy for Newly Diagnosed Stage III or IV Ovarian Cancer: Placebo-Controlled Randomized Phase III Trial (IMagyn050/GOG 3015/ENGOT-OV39). J Clin Oncol (2021) 39(17):1842–55. doi:10.1200/jco.21.00306

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Rocconi, RP, Grosen, EA, Ghamande, SA, Chan, JK, Barve, MA, Oh, J, et al. Gemogenovatucel-T (Vigil) Immunotherapy as Maintenance in Frontline Stage III/IV Ovarian Cancer (VITAL): A Randomised, Double-Blind, Placebo-Controlled, Phase 2b Trial. Lancet Oncol (2020) 21(12):1661–72. doi:10.1016/s1470-2045(20)30533-7

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Rocconi, RP, Monk, BJ, Walter, A, Herzog, TJ, Galanis, E, Manning, L, et al. Gemogenovatucel-T (Vigil) Immunotherapy Demonstrates Clinical Benefit in Homologous Recombination Proficient (HRP) Ovarian Cancer. Gynecol Oncol (2021) 161(3):676–80. doi:10.1016/j.ygyno.2021.03.009

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Walter, A, Rocconi, RP, Monk, BJ, Herzog, TJ, Manning, L, Bognar, E, et al. Gemogenovatucel-T (Vigil) Maintenance Immunotherapy: 3-Year Survival Benefit in Homologous Recombination Proficient (HRP) Ovarian Cancer. Gynecol Oncol (2021) 163(3):459–64. doi:10.1016/j.ygyno.2021.10.004

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Senzer, N, Barve, M, Kuhn, J, Melnyk, A, Beitsch, P, Lazar, M, et al. Phase I Trial of "Bi-ShRNAi(furin)/GMCSF DNA/autologous Tumor Cell" Vaccine (FANG) in Advanced Cancer. Mol Ther (2012) 20(3):679–86. doi:10.1038/mt.2011.269

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Senzer, N, Barve, M, Nemunaitis, J, Kuhn, J, Melnyk, A, Beitsch, P, et al. Long Term Follow up: Phase I Trial of “Bi-ShRNA Furin/GMCSF DNA/Autologous Tumor Cell” Immunotherapy (FANG™) in Advanced Cancer. J Vaccin Vaccination (2013) 4(8):209.

Google Scholar

30. Oh, J, Barve, M, Matthews, CM, Koon, EC, Heffernan, TP, Fine, B, et al. Phase II Study of Vigil® DNA Engineered Immunotherapy as Maintenance in Advanced Stage Ovarian Cancer. Gynecol Oncol (2016) 143(3):504–10. doi:10.1016/j.ygyno.2016.09.018

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Oh, J, Barve, M, Senzer, N, Aaron, P, Manning, L, Wallraven, G, et al. Long-Term Follow-Up of Phase 2A Trial Results Involving Advanced Ovarian Cancer Patients Treated with Vigil® in Frontline Maintenance. Gynecol Oncol Rep (2020) 34:100648. doi:10.1016/j.gore.2020.100648

PubMed Abstract | CrossRef Full Text | Google Scholar

32. McGranahan, N, Furness, AJS, Rosenthal, R, Ramskov, S, Lyngaa, R, Saini, SK, et al. Clonal Neoantigens Elicit T Cell Immunoreactivity and Sensitivity to Immune Checkpoint Blockade. Science (2016) 351(6280):1463–9. doi:10.1126/science.aaf1490

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Rizvi, NA, Hellmann, MD, Snyder, A, Kvistborg, P, Makarov, V, Havel, JJ, et al. Mutational Landscape Determines Sensitivity to PD-1 Blockade in Non–Small Cell Lung Cancer. Science (2015) 348(6230):124–8. doi:10.1126/science.aaa1348

PubMed Abstract | CrossRef Full Text | Google Scholar

34. Snyder, A, Makarov, V, Merghoub, T, Yuan, J, Zaretsky, JM, Desrichard, A, et al. Genetic Basis for Clinical Response to CTLA-4 Blockade in Melanoma. N Engl J Med (2014) 371(23):2189–99. doi:10.1056/nejmoa1406498

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Johnson, BE, Mazor, T, Hong, C, Barnes, M, Aihara, K, McLean, CY, et al. Mutational Analysis Reveals the Origin and Therapy-Driven Evolution of Recurrent Glioma. Science (2014) 343(6167):189–93. doi:10.1126/science.1239947

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Hanada, KI, Zhao, C, Gil-Hoyos, R, Gartner, JJ, Chow-Parmer, C, Lowery, FJ, et al. A Phenotypic Signature that Identifies Neoantigen-Reactive T Cells in Fresh Human Lung Cancers. Cancer Cell (2022) 40(5):479–93 e6. doi:10.1016/j.ccell.2022.03.012

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Morand, S, Stanbery, L, Walter, A, Rocconi, RP, and Nemunaitis, J. BRCA1/2 Mutation Status Impact on Autophagy and Immune Response: Unheralded Target. JNCI Cancer Spectr (2020) 4(6):pkaa077. doi:10.1093/jncics/pkaa077

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Nemunaitis, J, Senzer, N, and Plato, L. Tumor Vaccines and Cellular Immunotherapies. Ann Transl Med (2016) 4(1):S24. doi:10.21037/atm.2016.10.54

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Yu, TW, Chueh, HY, Tsai, CC, Lin, CT, and Qiu, JT. Novel GM-CSF-Based Vaccines: One Small Step in GM-CSF Gene Optimization, One Giant Leap for Human Vaccines. Hum Vaccin Immunother (2016) 12(12):3020–8. doi:10.1080/21645515.2016.1221551

PubMed Abstract | CrossRef Full Text | Google Scholar

40. Fan, CA, Reader, J, and Roque, DM. Review of Immune Therapies Targeting Ovarian Cancer. Curr Treat Options Oncol (2018) 19(12):74. doi:10.1007/s11864-018-0584-3

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: homologous recombination, neoantigen, immune response, ovarian cancer, immunotherapy

Citation: Nemunaitis J, Stanbery L, Walter A, Rocconi R and Stephens P (2023) Rationale for the Use of Homologous Recombination Proficient Molecular Profile as a Biomarker for Therapeutic Targeting in Ovarian Cancer. Oncol. Rev. 17:11471. doi: 10.3389/or.2023.11471

Received: 12 April 2023; Accepted: 31 August 2023;
Published: 20 September 2023.

Edited by:

Angela Cappello, University of Rome Tor Vergata, Italy

Reviewed by:

Claudia Wiese, Colorado State University, United States
Camilla Didio, Sapienza University of Rome, Italy

Copyright © 2023 Nemunaitis, Stanbery, Walter, Rocconi and Stephens. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: John Nemunaitis, johnnemunaitis@gmail.com

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.