1
|
Apelian S, Martincuks A, Whittum M, Yasukawa M, Nguy L, Mathyk B, Andikyan V, Anderson ML, Rutherford T, Cristea M, Stewart D, Kohut A. PARP Inhibitors in Ovarian Cancer: Resistance Mechanisms, Clinical Evidence, and Evolving Strategies. Biomedicines 2025; 13:1126. [PMID: 40426953 PMCID: PMC12108591 DOI: 10.3390/biomedicines13051126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/24/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
The introduction of poly (ADP-ribose) polymerase inhibitors (PARPi) into the management of ovarian cancer has transformed the treatment landscape for patients affected by this malignancy. However, as the use of PARPi expands into both frontline maintenance and recurrence settings, the emergence of drug resistance has become a significant clinical challenge in the treatment of these patients. Although platinum-based chemotherapy (PBC) and PARPi act through different mechanisms-PBC causes DNA damage while PARPi blocks its repair-both depend on the integrity of DNA damage repair (DDR) pathways, leading to overlapping mechanisms of resistance. Here, we review the key resistance mechanisms shared by PARPi and PBC, and then we discuss their clinical implications in the management of patients with ovarian cancer. We also examine clinical rationale supporting the hypothesis that prior PARPi exposure may reduce the efficacy of subsequent PBC in patients experiencing a disease recurrence. Furthermore, we review preliminary clinical data assessing the potential role of PARPi retreatment in patients who have previously progressed on PARPis.
Collapse
Affiliation(s)
- Shant Apelian
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of South Florida, Tampa, FL 33620, USA; (M.W.); (M.Y.); (L.N.); (B.M.); (V.A.); (M.L.A.); (T.R.); (A.K.)
- Division of Gynecologic Oncology, Tampa General Hospital Cancer Institute, Tampa, FL 33620, USA
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510, USA
| | - Antons Martincuks
- Department of Immuno-Oncology, City of Hope National Medicinal Center, Duarte, CA 91010, USA;
| | - Michelle Whittum
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of South Florida, Tampa, FL 33620, USA; (M.W.); (M.Y.); (L.N.); (B.M.); (V.A.); (M.L.A.); (T.R.); (A.K.)
- Division of Gynecologic Oncology, Tampa General Hospital Cancer Institute, Tampa, FL 33620, USA
| | - Maya Yasukawa
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of South Florida, Tampa, FL 33620, USA; (M.W.); (M.Y.); (L.N.); (B.M.); (V.A.); (M.L.A.); (T.R.); (A.K.)
- Division of Gynecologic Oncology, Tampa General Hospital Cancer Institute, Tampa, FL 33620, USA
| | - Lindsey Nguy
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of South Florida, Tampa, FL 33620, USA; (M.W.); (M.Y.); (L.N.); (B.M.); (V.A.); (M.L.A.); (T.R.); (A.K.)
- Division of Gynecologic Oncology, Tampa General Hospital Cancer Institute, Tampa, FL 33620, USA
| | - Begum Mathyk
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of South Florida, Tampa, FL 33620, USA; (M.W.); (M.Y.); (L.N.); (B.M.); (V.A.); (M.L.A.); (T.R.); (A.K.)
| | - Vaagn Andikyan
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of South Florida, Tampa, FL 33620, USA; (M.W.); (M.Y.); (L.N.); (B.M.); (V.A.); (M.L.A.); (T.R.); (A.K.)
- Division of Gynecologic Oncology, Tampa General Hospital Cancer Institute, Tampa, FL 33620, USA
| | - Matthew L. Anderson
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of South Florida, Tampa, FL 33620, USA; (M.W.); (M.Y.); (L.N.); (B.M.); (V.A.); (M.L.A.); (T.R.); (A.K.)
- Division of Gynecologic Oncology, Tampa General Hospital Cancer Institute, Tampa, FL 33620, USA
| | - Thomas Rutherford
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of South Florida, Tampa, FL 33620, USA; (M.W.); (M.Y.); (L.N.); (B.M.); (V.A.); (M.L.A.); (T.R.); (A.K.)
- Division of Gynecologic Oncology, Tampa General Hospital Cancer Institute, Tampa, FL 33620, USA
| | | | - Daphne Stewart
- Department of Medicine, Division of Medical Oncology, USC Norris Comprehensive Cancer Center and Keck School of Medicine, Los Angeles, CA 90089, USA;
| | - Adrian Kohut
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, University of South Florida, Tampa, FL 33620, USA; (M.W.); (M.Y.); (L.N.); (B.M.); (V.A.); (M.L.A.); (T.R.); (A.K.)
- Division of Gynecologic Oncology, Tampa General Hospital Cancer Institute, Tampa, FL 33620, USA
| |
Collapse
|
2
|
Obasi J, Sharma K, De Sarkar N, Antonarakis ES, Kilari D. Platinum Chemotherapy After PARP Inhibition in HRR-Deficient Metastatic Castration-Resistant Prostate Cancer. Clin Genitourin Cancer 2024; 22:102187. [PMID: 39241311 DOI: 10.1016/j.clgc.2024.102187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 09/09/2024]
Affiliation(s)
- Jennifer Obasi
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI; School of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Komal Sharma
- Medical College of Wisconsin Cancer Center, Milwaukee, WI; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI; Data Science Institute, School of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Navonil De Sarkar
- Medical College of Wisconsin Cancer Center, Milwaukee, WI; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI; Data Science Institute, School of Medicine, Medical College of Wisconsin, Milwaukee, WI
| | - Emmanuel S Antonarakis
- Division of Hematology, Oncology, and Transplantation, Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Deepak Kilari
- Division of Hematology and Oncology, Medical College of Wisconsin, Milwaukee, WI.
| |
Collapse
|
3
|
Telli ML, Litton JK, Beck JT, Jones JM, Andersen J, Mina LA, Brig R, Danso M, Yuan Y, Symmans WF, Hopkins JF, Albacker LA, Abbattista A, Noonan K, Mata M, Laird AD, Blum JL. Neoadjuvant talazoparib in patients with germline BRCA1/2 mutation-positive, early-stage triple-negative breast cancer: exploration of tumor BRCA mutational status. Breast Cancer 2024; 31:886-897. [PMID: 38869771 PMCID: PMC11341741 DOI: 10.1007/s12282-024-01603-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 06/02/2024] [Indexed: 06/14/2024]
Abstract
BACKGROUND Talazoparib monotherapy in patients with germline BRCA-mutated, early-stage triple-negative breast cancer (TNBC) showed activity in the neoadjuvant setting in the phase II NEOTALA study (NCT03499353). These biomarker analyses further assessed the mutational landscape of the patients enrolled in the NEOTALA study. METHODS Baseline tumor tissue from the NEOTALA study was tested retrospectively using FoundationOne®CDx. To further hypothesis-driven correlative analyses, agnostic heat-map visualizations of the FoundationOne®CDx tumor dataset were used to assess overall mutational landscape and identify additional candidate predictive biomarkers of response. RESULTS All patients enrolled (N = 61) had TNBC. In the biomarker analysis population, 75.0% (39/52) and 25.0% (13/52) of patients exhibited BRCA1 and BRCA2 mutations, respectively. Strong concordance (97.8%) was observed between tumor BRCA and germline BRCA mutations, and 90.5% (38/42) of patients with tumor BRCA mutations evaluable for somatic-germline-zygosity were predicted to exhibit BRCA loss of heterozygosity (LOH). No patients had non-BRCA germline DNA damage response (DDR) gene variants with known/likely pathogenicity, based on a panel of 14 non-BRCA DDR genes. Ninety-eight percent of patients had TP53 mutations. Genomic LOH, assessed continuously or categorically, was not associated with response. CONCLUSION The results from this exploratory biomarker analysis support the central role of BRCA and TP53 mutations in tumor pathobiology. Furthermore, these data support assessing germline BRCA mutational status for molecular eligibility for talazoparib in patients with TNBC.
Collapse
Affiliation(s)
- Melinda L Telli
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| | - Jennifer K Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - J Thaddeus Beck
- Department of Medical Oncology and Hematology, Highlands Oncology, Springdale, AR, USA
| | - Jason M Jones
- Avera Medical Group Oncology & Hematology, Avera Cancer Institute, Sioux Falls, SD, USA
| | - Jay Andersen
- Medical Oncology, Compass Oncology, West Cancer Center, US Oncology Network, Tigard, OR, USA
| | - Lida A Mina
- Hematology Oncology Department, Banner MD Anderson Cancer Center, Gilbert, AZ, USA
| | - Raymond Brig
- Medical Oncology, Brig Center for Cancer Care and Survivorship, Knoxville, TN, USA
| | - Michael Danso
- Medical Oncology, Virginia Oncology Associates, Norfolk, VA, USA
| | - Yuan Yuan
- Department of Medical Oncology & Therapeutics Research, Cedars-Sinai Cancer Center, West Hollywood, CA, USA
| | - William F Symmans
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | - Kay Noonan
- Clinical Oncology, Pfizer Inc., Groton, CT, USA
| | | | | | - Joanne L Blum
- Department of Oncology, Texas Oncology-Baylor Charles A. Sammons Cancer Center, US Oncology Network, Dallas, TX, USA
| |
Collapse
|
4
|
Hecht F, Zocchi M, Alimohammadi F, Harris IS. Regulation of antioxidants in cancer. Mol Cell 2024; 84:23-33. [PMID: 38029751 PMCID: PMC10843710 DOI: 10.1016/j.molcel.2023.11.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/19/2023] [Accepted: 11/01/2023] [Indexed: 12/01/2023]
Abstract
Scientists in this field often joke, "If you don't have a mechanism, say it's ROS." Seemingly connected to every biological process ever described, reactive oxygen species (ROS) have numerous pleiotropic roles in physiology and disease. In some contexts, ROS act as secondary messengers, controlling a variety of signaling cascades. In other scenarios, they initiate damage to macromolecules. Finally, in their worst form, ROS are deadly to cells and surrounding tissues. A set of molecules with detoxifying abilities, termed antioxidants, is the direct counterpart to ROS. Notably, antioxidants exist in the public domain, touted as a "cure-all" for diseases. Research has disproved many of these claims and, in some cases, shown the opposite. Of all the diseases, cancer stands out in its paradoxical relationship with antioxidants. Although the field has made numerous strides in understanding the roles of antioxidants in cancer, many questions remain.
Collapse
Affiliation(s)
- Fabio Hecht
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Marco Zocchi
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Fatemeh Alimohammadi
- Department of Pharmacology and Physiology, University of Rochester, Rochester, NY 14642, USA; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Isaac S Harris
- Department of Biomedical Genetics, University of Rochester Medical Center, Rochester, NY 14642, USA; Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
5
|
Brown TJ, Yablonovitch A, Till JE, Yen J, Kiedrowski LA, Hood R, O'Hara MH, Teitelbaum U, Karasic TB, Schneider C, Carpenter EL, Nathanson K, Domchek SM, Reiss KA. The Clinical Implications of Reversions in Patients with Advanced Pancreatic Cancer and Pathogenic Variants in BRCA1, BRCA2, or PALB2 after Progression on Rucaparib. Clin Cancer Res 2023; 29:5207-5216. [PMID: 37486343 PMCID: PMC10806928 DOI: 10.1158/1078-0432.ccr-23-1467] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/13/2023] [Accepted: 07/20/2023] [Indexed: 07/25/2023]
Abstract
PURPOSE PARP inhibitors (PARPi) provide an effective maintenance option for patients with BRCA- or PALB2-mutated pancreatic cancer. However, mechanisms of PARPi resistance and optimal post-PARPi therapeutic strategies are poorly characterized. EXPERIMENTAL DESIGN We collected paired cell-free DNA samples and post-PARPi clinical data on 42 patients with advanced, platinum-sensitive pancreatic cancer who were treated with maintenance rucaparib on NCT03140670, of whom 32 developed progressive disease. RESULTS Peripherally detected, acquired BRCA or PALB2 reversion variants were uncommon (5/30; 16.6%) in patients who progressed on rucaparib. Reversions were significantly associated with rapid resistance to PARPi treatment (median PFS, 3.7 vs. 12.5 months; P = 0.001) and poor overall survival (median OS, 6.2 vs. 23.0 months; P < 0.0001). All patients with reversions received rechallenge with platinum-based chemotherapy following PARPi progression and experienced faster progression on this therapy than those without reversion variants (real-world time-to-treatment discontinuation, 2.4 vs. 5.8 months; P = 0.004). Of the patients who progressed on PARPi and received further chemotherapy, the OS from initiation of second-line therapy was significantly lower in those with reversion variants than in those without (5.5 vs. 12.0 months, P = 0.002). Finally, high levels of tumor shedding were independently associated with poor outcomes in patients who received rucaparib. CONCLUSIONS Acquired reversion variants were uncommon but detrimental in a population of patients with advanced BRCA- or PALB2-related pancreatic ductal adenocarcinoma who received maintenance rucaparib. Reversion variants led to rapid progression on PARPi, rapid failure of subsequent platinum-based treatment, and poor OS of patients. The identification of such variants in the blood may have both predictive and prognostic value. See related commentary by Tsang and Gallinger, p. 5005.
Collapse
Affiliation(s)
- Timothy J Brown
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
- Penn Center for Cancer Care Innovation, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | - Jacob E Till
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Ryan Hood
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Mark H O'Hara
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ursina Teitelbaum
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Thomas B Karasic
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Charles Schneider
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Erica L Carpenter
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Katherine Nathanson
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Susan M Domchek
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kim A Reiss
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
6
|
Wei CH, Shehayeb S, Santiago NL, Kruper L, Han E, Wang E, Cristea M, Rodriguez-Rodriguez L, Yost SE, Stewart D. BRCA germline mutations in multiethnic gynecologic patients: A 10-year retrospective analysis from a single cancer institute. PLoS One 2023; 18:e0286998. [PMID: 37310942 PMCID: PMC10263324 DOI: 10.1371/journal.pone.0286998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 05/29/2023] [Indexed: 06/15/2023] Open
Abstract
Histologic and genetic mutation information from racially and ethnically diverse populations is warranted to better inform future cancer predisposition and promote health equity. A single institutional, retrospective capture of patients with gynecologic conditions and genetic susceptibilities to malignant neoplasms of the breast or ovaries was performed. This was achieved with manual curation of the electronic medical record (EMR) from 2010-2020 with the use of ICD-10 code searches. Among 8983 consecutive women identified with gynecologic conditions, 184 were diagnosed with pathogenic/likely pathogenic (P/LP) germline BRCA (gBRCA) mutations. Median age was 54 (22-90). Mutations included insertion/deletion (majority frameshift, 57.4%), substitution (32.4%), large structural rearrangement (5.4%), and alteration in splice site/intronic sequence (4.7%). A total of 48% were non-Hispanic White, 32% Hispanic or Latino, 13% Asian, 2% Black, and 5% Other. The most common pathology was high grade serous carcinoma (HGSC, 63%), followed by unclassified/high grade carcinoma (13%). Additional multigene panels led to the detection of 23 additional BRCA-positive patients with germline co-mutations and/or variants of uncertain significance in genes functionally involved in DNA repair mechanisms. Hispanic or Latino and Asian individuals comprised 45% of patients with concomitant gynecologic condition and gBRCA positivity in our cohort, confirming that germline mutations are represented across racial and ethnic groups. Insertion/deletion mutations, the majority of which led to a frameshift change, occurred in approximately half of our patient cohort, which may have prognostic implication for therapy resistance. Prospective studies are needed to unravel the significance of germline co-mutations in gynecologic patients.
Collapse
Affiliation(s)
- Christina H. Wei
- Department of Pathology, City of Hope Comprehensive Cancer Institute, Duarte, California, United States of America
| | - Susan Shehayeb
- Department of Population Sciences, City of Hope Comprehensive Cancer Institute, Duarte, California, United States of America
| | - Nicole Lugo Santiago
- Department of Surgery, City of Hope Comprehensive Cancer Institute, Duarte, California, United States of America
| | - Laura Kruper
- Department of Surgery, City of Hope Comprehensive Cancer Institute, Duarte, California, United States of America
| | - Ernest Han
- Department of Surgery, City of Hope Comprehensive Cancer Institute, Duarte, California, United States of America
| | - Edward Wang
- Department or Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Institute, Duarte, CA, United States of America
| | - Mihaela Cristea
- Department or Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Institute, Duarte, CA, United States of America
| | - Lorna Rodriguez-Rodriguez
- Department of Surgery, City of Hope Comprehensive Cancer Institute, Duarte, California, United States of America
| | - Susan E. Yost
- Department or Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Institute, Duarte, CA, United States of America
| | - Daphne Stewart
- Department or Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Institute, Duarte, CA, United States of America
| |
Collapse
|
7
|
Keane F, Bajwa R, Selenica P, Park W, Roehrl MH, Reis-Filho JS, Mandelker D, O'Reilly EM. Dramatic, durable response to therapy in gBRCA2-mutated pancreas neuroendocrine carcinoma: opportunity and challenge. NPJ Precis Oncol 2023; 7:40. [PMID: 37087482 PMCID: PMC10122663 DOI: 10.1038/s41698-023-00376-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/30/2023] [Indexed: 04/24/2023] Open
Abstract
Poorly differentiated pancreatic neuroendocrine tumors (PDNEC), are a subtype of pancreatic cancer encompassing both small cell and large cell neuroendocrine carcinoma subtypes, and are characterized as distinct in terms of biology and prognosis compared to the more common pancreatic adenocarcinoma. Until recently, there has been a paucity of data on the genomic features of this cancer type. We describe a male patient diagnosed with PDNEC and extensive metastatic disease in the liver at diagnosis. Genomic analysis demonstrated a germline pathogenic variant in BRCA2 with somatic loss-of-heterozygosity of the BRCA2 wild-type allele. Following a favorable response to platinum-based chemotherapy (and the addition of immunotherapy), the patient received maintenance therapy with olaparib, which resulted in a further reduction on follow-up imaging (Fig. 1). After seventeen months of systemic control with olaparib, the patient developed symptomatic central nervous system metastases, which harboured a BRCA2 reversion mutation. No other sites of disease progression were observed. Herein, we report an exceptional outcome through the incorporation of a personalized management approach for a patient with a pancreatic PDNEC, guided by comprehensive genomic sequencing.
Collapse
Affiliation(s)
- Fergus Keane
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, New York, NY, USA
| | - Raazi Bajwa
- David M. Rubenstein Center for Pancreatic Cancer Research, New York, NY, USA
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pier Selenica
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wungki Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- David M. Rubenstein Center for Pancreatic Cancer Research, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Michael H Roehrl
- David M. Rubenstein Center for Pancreatic Cancer Research, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge S Reis-Filho
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Diana Mandelker
- David M. Rubenstein Center for Pancreatic Cancer Research, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Diagnostic Molecular Genetics Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- David M. Rubenstein Center for Pancreatic Cancer Research, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
8
|
Congregado B, Rivero I, Osmán I, Sáez C, Medina López R. PARP Inhibitors: A New Horizon for Patients with Prostate Cancer. Biomedicines 2022; 10:1416. [PMID: 35740437 PMCID: PMC9220343 DOI: 10.3390/biomedicines10061416] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/16/2022] [Accepted: 05/23/2022] [Indexed: 11/23/2022] Open
Abstract
The introduction of PARP inhibitors (PARPi) in prostate cancer is a milestone and provides a pathway to hope in fighting this disease. It is the first time that drugs, based on the concept of synthetic lethality, have been approved for prostate cancer. In addition, it is also the first time that genetic mutation tests have been included in the therapeutic algorithm of this disease, representing a significant step forward for precision and personalized treatment of prostate cancer. The objectives of this review are: (1) understanding the mechanism of action of PARPi in monotherapy and combinations; (2) gaining insights on patient selection for PARPi; (3) exposing the pivotal studies that have allowed its approval, and; (4) offering an overview of the ongoing trials. Nevertheless, many unsolved questions remain, such as the number of patients who could potentially benefit from PARPi, whether to use PARPi in monotherapy or in combination, and when is the best time to use them in advanced or localized disease. To answer these and other questions, many clinical trials are underway. Some of them have recently demonstrated promising results that may favor the introduction of new combinations in metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Belén Congregado
- Urology and Nephrology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain; (I.R.); (I.O.); (R.M.L.)
| | - Inés Rivero
- Urology and Nephrology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain; (I.R.); (I.O.); (R.M.L.)
| | - Ignacio Osmán
- Urology and Nephrology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain; (I.R.); (I.O.); (R.M.L.)
| | - Carmen Sáez
- Department of Pathology, Biomedical Institute of Seville (IBIS), 41013 Seville, Spain;
| | - Rafael Medina López
- Urology and Nephrology Department, University Hospital Virgen del Rocío, 41013 Seville, Spain; (I.R.); (I.O.); (R.M.L.)
| |
Collapse
|
9
|
Sutton TL, Grossberg A, Ey F, O’Reilly EM, Sheppard BC. Multimodality therapy in metastatic pancreas cancer with a BRCA mutation and durable long-term outcome: biology, intervention, or both? Cancer Biol Ther 2021; 22:532-536. [PMID: 34696697 PMCID: PMC8726708 DOI: 10.1080/15384047.2021.1991739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/09/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022] Open
Abstract
Metastatic pancreatic adenocarcinoma (PDAC) is a rapidly lethal disease, with less than half of patients surviving 12 months, and 5-year survival approximately 3%. These outcomes are in large part due to a lack of effective medical and surgical therapies for metastatic PDAC. Herein, we present the case of a patient with oligometastatic liver recurrence of BRCA2-mutated PDAC following a curative-intent resection. Through a combination of systemic chemotherapy, metastasectomy, radiotherapy, and subsequent targeted therapy with olaparib, the patient is asymptomatic four years following metastatic diagnosis with stable low-volume disease. This patient's excellent outcome is attributable to the multi-disciplinary care received, all aspects of which were informed by new evidence surrounding metastasectomy for metastatic PDAC, the unique biology and medical treatment of BRCA-mutated PDAC, and the role of radiotherapy in controlling locoregional recurrence. We provide a review of this evidence, while highlighting the importance of evaluating disease biology through somatic and germline genetic testing as well as monitoring response to systemic chemotherapy.
Collapse
Affiliation(s)
- Thomas L. Sutton
- Department of Surgery, Oregon Heath & Science University (OHSU), Portland, Oregon, USA
| | - Aaron Grossberg
- Department of Radiation Medicine, OHSU, Portland, Oregon, USA
| | - Frederick Ey
- Department of Hematology/Oncology, OHSU, Portland, Oregon, USA
| | - Eileen M. O’Reilly
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Brett C. Sheppard
- Department of Surgery, Oregon Heath & Science University (OHSU), Portland, Oregon, USA
| |
Collapse
|
10
|
Pettitt SJ, Frankum JR, Punta M, Lise S, Alexander J, Chen Y, Yap TA, Haider S, Tutt ANJ, Lord CJ. Clinical BRCA1/2 Reversion Analysis Identifies Hotspot Mutations and Predicted Neoantigens Associated with Therapy Resistance. Cancer Discov 2020; 10:1475-1488. [PMID: 32699032 PMCID: PMC7611203 DOI: 10.1158/2159-8290.cd-19-1485] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/16/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
Reversion mutations in BRCA1 or BRCA2 are associated with resistance to PARP inhibitors and platinum. To better understand the nature of these mutations, we collated, codified, and analyzed more than 300 reversions. This identified reversion "hotspots" and "deserts" in regions encoding the N and C terminus, respectively, of BRCA2, suggesting that pathogenic mutations in these regions may be at higher or lower risk of reversion. Missense and splice-site pathogenic mutations in BRCA1/2 also appeared less likely to revert than truncating mutations. Most reversions were <100 bp deletions. Although many deletions exhibited microhomology, this was not universal, suggesting that multiple DNA-repair processes cause reversion. Finally, we found that many reversions were predicted to encode immunogenic neopeptides, suggesting a route to the treatment of reverted disease. As well as providing a freely available database for the collation of future reversion cases, these observations have implications for how drug resistance might be managed in BRCA-mutant cancers. SIGNIFICANCE: Reversion mutations in BRCA genes are a major cause of clinical platinum and PARP inhibitor resistance. This analysis of all reported clinical reversions suggests that the position of BRCA2 mutations affects the risk of reversion. Many reversions are also predicted to encode tumor neoantigens, providing a potential route to targeting resistance.This article is highlighted in the In This Issue feature, p. 1426.
Collapse
Affiliation(s)
- Stephen J Pettitt
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, United Kingdom.
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Jessica R Frankum
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, United Kingdom
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Marco Punta
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Stefano Lise
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - John Alexander
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Yi Chen
- Scientific Computing Team, The Institute of Cancer Research, London, United Kingdom
| | - Timothy A Yap
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Andrew N J Tutt
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| | - Christopher J Lord
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London, United Kingdom.
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
11
|
Swan K, Dougherty KC, Myers SW. Somatic Testing and Germline Genetic Status: Implications for Cancer Treatment Decisions and Genetic Counseling. CURRENT GENETIC MEDICINE REPORTS 2020. [DOI: 10.1007/s40142-020-00192-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
12
|
Vidula N, Rich TA, Sartor O, Yen J, Hardin A, Nance T, Lilly MB, Nezami MA, Patel SP, Carneiro BA, Fan AC, Brufsky AM, Parker BA, Bridges BB, Agarwal N, Maughan BL, Raymond VM, Fairclough SR, Lanman RB, Bardia A, Cristofanilli M. Routine Plasma-Based Genotyping to Comprehensively Detect Germline, Somatic, and Reversion BRCA Mutations among Patients with Advanced Solid Tumors. Clin Cancer Res 2020; 26:2546-2555. [DOI: 10.1158/1078-0432.ccr-19-2933] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/17/2019] [Accepted: 02/03/2020] [Indexed: 11/16/2022]
|
13
|
Kondrashova O, Ho GY, Au-Yeung G, Leas L, Boughtwood T, Alsop K, Zapparoli G, Dobrovic A, Ko YA, Hsu AL, Love CJ, Lunke S, Wakefield MJ, McNally O, Quinn M, Ananda S, Neesham D, Hamilton A, Grossi M, Freimund A, Kanjanapan Y, Rischin D, Traficante N, Bowtell D, Scott CL, Christie M, Taylor GR, Mileshkin L, Waring PM. Clinical Utility of Real-Time Targeted Molecular Profiling in the Clinical Management of Ovarian Cancer: The ALLOCATE Study. JCO Precis Oncol 2019; 3:1-18. [DOI: 10.1200/po.19.00019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE The ALLOCATE study was designed as a pilot to demonstrate the feasibility and clinical utility of real-time targeted molecular profiling of patients with recurrent or advanced ovarian cancer for identification of potential targeted therapies. PATIENTS AND METHODS A total of 113 patients with ovarian cancer of varying histologies were recruited from two tertiary hospitals, with 99 patient cases suitable for prospective analysis. Targeted molecular and methylation profiling of fresh biopsy and archived tumor samples were performed by screening for mutations or copy-number variations in 44 genes and for promoter methylation of BRCA1 and RAD51C. RESULTS Somatic genomic or methylation events were identified in 85% of all patient cases, with potentially actionable events with defined targeted therapies (including four resistance events) detected in 60% of all patient cases. On the basis of these findings, six patients received molecularly guided therapy, three patients had unsuspected germline cancer–associated BRCA1/ 2 mutations and were referred for genetic counseling, and two intermediate differentiated (grade 2) serous ovarian carcinomas were reclassified as low grade, leading to changes in clinical management. Additionally, secondary reversion mutations in BRCA1/ 2 were identified in fresh biopsy samples of two patients, consistent with clinical platinum/poly (ADP-ribose) polymerase inhibitor resistance. Timely reporting of results if molecular testing is done at disease recurrence, as well as early referral for patients with platinum-resistant cancers, were identified as factors that could improve the clinical utility of molecular profiling. CONCLUSION ALLOCATE molecular profiling identified known genomic and methylation alterations of the different ovarian cancer subtypes and was deemed feasible and useful in routine clinical practice. Better patient selection and access to a wider range of targeted therapies or clinical trials will further enhance the clinical utility of molecular profiling.
Collapse
Affiliation(s)
- Olga Kondrashova
- University of Melbourne, Melbourne, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- QIMR Berghofer Medical Research Institute, Queensland, Australia
| | - Gwo-Yaw Ho
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Royal Women’s Hospital, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - George Au-Yeung
- University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Leakhena Leas
- University of Melbourne, Melbourne, Victoria, Australia
| | | | - Kathryn Alsop
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Giada Zapparoli
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- La Trobe University, Bundoora, Victoria, Australia
| | - Alexander Dobrovic
- University of Melbourne, Melbourne, Victoria, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, Victoria, Australia
- La Trobe University, Bundoora, Victoria, Australia
| | - Yi-An Ko
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Arthur L. Hsu
- University of Melbourne, Melbourne, Victoria, Australia
| | - Clare J. Love
- University of Melbourne, Melbourne, Victoria, Australia
| | | | - Matthew J. Wakefield
- University of Melbourne, Melbourne, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Orla McNally
- University of Melbourne, Melbourne, Victoria, Australia
- Royal Women’s Hospital, Parkville, Victoria, Australia
| | - Michael Quinn
- Royal Women’s Hospital, Parkville, Victoria, Australia
| | - Sumitra Ananda
- University of Melbourne, Melbourne, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Royal Women’s Hospital, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - Anne Hamilton
- University of Melbourne, Melbourne, Victoria, Australia
- Royal Women’s Hospital, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Marisa Grossi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Alison Freimund
- University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Yada Kanjanapan
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Danny Rischin
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | - David Bowtell
- University of Melbourne, Melbourne, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Clare L. Scott
- University of Melbourne, Melbourne, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Royal Women’s Hospital, Parkville, Victoria, Australia
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael Christie
- University of Melbourne, Melbourne, Victoria, Australia
- Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Graham R. Taylor
- University of Melbourne, Melbourne, Victoria, Australia
- King’s College London, London, United Kingdom
| | - Linda Mileshkin
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
14
|
Griffiths JI, Cohen AL, Jones V, Salgia R, Chang JT, Bild AH. Opportunities for improving cancer treatment using systems biology. ACTA ACUST UNITED AC 2019; 17:41-50. [PMID: 32518857 DOI: 10.1016/j.coisb.2019.10.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Current cancer therapies target a limited set of tumor features, rather than considering the tumor as a whole. Systems biology aims to reveal therapeutic targets associated with a variety of facets in an individual's tumor, such as genetic heterogeneity and its evolution, cancer cell-autonomous phenotypes, and microenvironmental signaling. These disparate characteristics can be reconciled using mathematical modeling that incorporates concepts from ecology and evolution. This provides an opportunity to predict tumor growth and response to therapy, to tailor patient-specific approaches in real time or even prospectively. Importantly, as data regarding patient tumors is often available from only limited time points during treatment, systems-based approaches can address this limitation by interpolating longitudinal events within a principled framework. This review outlines areas in medicine that could benefit from systems biology approaches to deconvolve the complexity of cancer.
Collapse
Affiliation(s)
- Jason I Griffiths
- Department of Mathematics, University of Utah, Salt Lake City, UT 84112, USA
| | - Adam L Cohen
- Huntsman Cancer Institute, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84112, USA
| | - Veronica Jones
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Andrea H Bild
- Department of Medical Oncology, Division of Molecular Pharmacology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
15
|
Na B, Yu X, Withers T, Gilleran J, Yao M, Foo TK, Chen C, Moore D, Lin Y, Kimball SD, Xia B, Ganesan S, Carpizo DR. Therapeutic targeting of BRCA1 and TP53 mutant breast cancer through mutant p53 reactivation. NPJ Breast Cancer 2019; 5:14. [PMID: 30993195 PMCID: PMC6465291 DOI: 10.1038/s41523-019-0110-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 03/15/2019] [Indexed: 12/28/2022] Open
Abstract
Triple negative breast cancer (TNBC) is an aggressive subset for which effective therapeutic approaches are needed. A significant proportion of TNBC patients harbor either germline or somatic mutations in BRCA1, or epigenetic silencing of BRCA1, which renders them deficient in DNA repair. Virtually all BRCA1 deficient breast cancers harbor mutations in TP53 suggesting that inactivation of p53 is a requirement for tumor progression in the setting of BRCA1 deficiency. Due to this dependency, we hypothesized that restoring wild type p53 function in BRCA1 deficient breast cancer would be therapeutic. The majority of TP53 mutations are missense, which generate a defective protein that potentially can be targeted with small molecules. Zinc metallochaperones (ZMCs) are a new class of anti-cancer drugs that specifically reactivate zinc-deficient mutant p53 by restoring zinc binding. Using ZMC1 in human breast cancer cell lines expressing the zinc deficient p53R175H, we demonstrate that loss of BRCA1 sensitizes cells to mutant p53 reactivation. Using murine breast cancer models with Brca1 deficiency, we demonstrate that ZMC1 significantly improves survival of mice bearing tumors harboring the zinc-deficient Trp53 R172H allele but not the Trp53 -/- allele. We synthesized a new formulation of ZMC1 (Zn-1), in which the drug is made in complex with zinc to improve zinc delivery, and demonstrate that Zn-1 has increased efficacy. Furthermore, we show that ZMC1 plus olaparib is a highly effective combination for p53R172H tumor growth inhibition. In conclusion, we have validated preclinically a new therapeutic approach for BRCA1 deficient breast cancer through reactivation of mutant p53.
Collapse
Affiliation(s)
- Bing Na
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
| | - Xin Yu
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
| | - Tracy Withers
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
| | - John Gilleran
- Rutgers University Biomedical Research Cores, Rutgers University, Piscataway, NJ USA
- Department of Medicinal Chemistry, Rutgers Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ USA
| | - Ming Yao
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
| | - Tzeh Keong Foo
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ USA
| | - Chunxia Chen
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
- Department of Biostatistics, Rutgers School of Public Health, Rutgers University, New Brunswick, NJ USA
| | - Dirk Moore
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
- Department of Biostatistics, Rutgers School of Public Health, Rutgers University, New Brunswick, NJ USA
| | - Yong Lin
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
- Department of Biostatistics, Rutgers School of Public Health, Rutgers University, New Brunswick, NJ USA
| | - S. David Kimball
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
- Rutgers University Biomedical Research Cores, Rutgers University, Piscataway, NJ USA
- Department of Medicinal Chemistry, Rutgers Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ USA
- Office of Innovation & Research Commercialization, Rutgers University, Piscataway, NJ USA
| | - Bing Xia
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
- Department of Radiation Oncology, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ USA
| | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
- Department of Pharmacology, Rutgers University, Piscataway, NJ USA
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ USA
| | - Darren R. Carpizo
- Department of Surgery, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ USA
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
- Department of Pharmacology, Rutgers University, Piscataway, NJ USA
- Z53 Therapeutics, Inc., Holmdel, NJ USA
| |
Collapse
|