1
|
Newell A, Andac-Jones E, Gonzalo M, Wright AA, Palesh O, Fortune EE, Szamreta E. Patients with advanced ovarian cancer and their experiences and perceptions of sleep disturbance and fatigue across the treatment trajectory: A qualitative study. Gynecol Oncol 2025; 198:42-48. [PMID: 40411968 DOI: 10.1016/j.ygyno.2025.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/11/2025] [Accepted: 05/17/2025] [Indexed: 05/27/2025]
Abstract
OBJECTIVE Sleep disturbance and fatigue are prevalent among patients with advanced ovarian cancer (OC) and associated with poor health-related quality of life (HRQoL). This study aimed to illustrate the lived experiences of OC patients with sleep disturbance and fatigue and the range of factors that they perceive as contributing to these symptoms. METHODS Individual semi-structured interviews were conducted with twenty patients with Stage III and IV OC diagnosed and treated within three years (17/20 receiving treatment). Interview transcripts were analyzed based on emergent themes in the data and study goals. RESULTS Participants reported high levels of sleep disturbance and fatigue, which interfered with their cognitive, social, and physical function. The nature of their sleep and fatigue issues varied across the treatment trajectory. The majority (16/20) used chemotherapy as a benchmark to assess their current symptom burden. Fatigue and sleep disturbance were described as more intense and cyclic, with the highest symptom burden immediately following chemotherapy infusions followed by periodic improvement between treatments, whereas symptoms were less intense, but more persistent during maintenance therapy. Participants pursued a variety of strategies to manage symptoms, but fatigue persisted among thirteen of twenty participants. Participants were dissatisfied with providers' lack of communication and recommendations to manage symptoms. CONCLUSION Findings underscore that while sleep disturbance and fatigue are intense among participants with OC, the lived experiences of these symptoms are qualitatively distinct at different points during treatment. Participants' dissatisfaction with providers' communication suggests the need for improved screening and scaled interventions for advanced OC patients.
Collapse
Affiliation(s)
- Abigail Newell
- Research, Cancer Support Community, Washington, DC, USA.
| | | | - Maria Gonzalo
- Research, Cancer Support Community, Washington, DC, USA
| | - Alexi A Wright
- Dana Farber Cancer Institute, Harvard School of Medicine, Boston, MA, USA
| | - Oxana Palesh
- Department of Psychiatry, Massey Comprehensive Cancer Center, VCU School of Medicine, Richmond, VA, USA
| | | | | |
Collapse
|
2
|
Lindemann K, Kildal W, Kleppe A, Tobin KAR, Pradhan M, Mascialino B, Schneider D, Edvardsen H, Sørlie T, Kristensen GB, Askautrud HA. Real-world outcomes in molecular subgroups for patients with advanced or recurrent endometrial cancer treated with platinum-based chemotherapy. Int J Gynecol Cancer 2025; 35:101618. [PMID: 40189984 DOI: 10.1016/j.ijgc.2024.101618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 05/11/2025] Open
Abstract
OBJECTIVE There is scarce real-world evidence on patients with advanced/recurrent endometrial cancer treated with platinum-based chemotherapy. We assessed the oncological outcome in groups by molecular classification. METHODS This retrospective cohort study included patients with advanced/recurrent endometrial cancer treated with platinum-based chemotherapy after hysterectomy at The Norwegian Radium Hospital, Oslo University Hospital, Norway, between January 2006 and December 2017. Patients were molecularly classified as pathogenic POLE mutated, mismatch repair deficient, p53 abnormal, or no specific molecular profile. Time-to-recurrence and cancer-specific survival were calculated. RESULTS We identified 264 advanced-stage patients (stage III/IV) and 96 patients with recurrent disease. The molecular classification was prognostic for time-to-recurrence (p < .0001) and cancer-specific survival (p < .0001) in patients with advanced disease, but the outcome did not differ significantly by molecular groups in recurrent patients. In all molecular groups, patients with stage III disease had longer time-to-recurrence and cancer-specific survival compared to patients with stage IV disease. The worst outcome was observed in patients with p53 abnormal tumors with an HR of 1.57 (95% CI 1.07 to 2.30) for time-to-recurrence and HR of 1.78 (95% CI 1.19 to 2.65) for cancer-specific survival in stage III/IV disease and an HR of 1.45 (95% CI 0.83 to 2.52) for time-to-recurrence and HR of 1.60 (95% CI 0.99 to 2.68) for cancer-specific survival in patients with recurrent disease. The few patients with POLE mutated tumors had favorable outcomes despite the advanced/recurrent disease status. CONCLUSIONS Oncological outcomes differ by molecular groups, in particular among patients with advanced disease. Patients with p53 abnormal tumors have the worst outcome, while patients with POLE mutated tumors have favorable outcomes even with recurrent disease. Implementation of the addition of immunotherapy to chemotherapy is expected to lead to substantial improvement of outcome, particularly in patients with mismatch repair deficient advanced/recurrent disease. There is still a high unmet need in advanced/recurrent patients with p53 abnormal and no specific molecular profile tumors.
Collapse
Affiliation(s)
- Kristina Lindemann
- Oslo University Hospital, The Norwegian Radium Hospital, Section for Gynecological Oncology, Department of Surgical Oncology, Oslo, Norway; University of Oslo, Faculty of Medicine, Institute of Clinical Medicine, Oslo, Norway.
| | - Wanja Kildal
- Oslo University Hospital, Institute for Cancer Genetics and Informatics, Oslo, Norway
| | - Andreas Kleppe
- Oslo University Hospital, Institute for Cancer Genetics and Informatics, Oslo, Norway; University of Oslo, Department of Informatics, Oslo, Norway; UiT The Arctic University of Norway, Centre for Research-based Innovation Visual Intelligence, Tromsø, Norway
| | - Kari Anne R Tobin
- Oslo University Hospital, Institute for Cancer Genetics and Informatics, Oslo, Norway
| | - Manohar Pradhan
- Oslo University Hospital, Institute for Cancer Genetics and Informatics, Oslo, Norway
| | | | | | | | - Therese Sørlie
- University of Oslo, Faculty of Medicine, Institute of Clinical Medicine, Oslo, Norway; Oslo University Hospital, Institute for Cancer Research, Department of Cancer Genetics, Oslo, Norway
| | - Gunnar B Kristensen
- Oslo University Hospital, The Norwegian Radium Hospital, Section for Gynecological Oncology, Department of Surgical Oncology, Oslo, Norway; Oslo University Hospital, Institute for Cancer Genetics and Informatics, Oslo, Norway
| | - Hanne A Askautrud
- Oslo University Hospital, Institute for Cancer Genetics and Informatics, Oslo, Norway
| |
Collapse
|
3
|
Bryan S, Rockall A, Tookman L. ESR Bridges: imaging and treatment of ovarian cancer-a multidisciplinary view. Eur Radiol 2025; 35:2415-2417. [PMID: 39422723 PMCID: PMC12021942 DOI: 10.1007/s00330-024-11072-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 07/05/2024] [Accepted: 09/03/2024] [Indexed: 10/19/2024]
Affiliation(s)
- Stacey Bryan
- Department of Gynaecological Oncology, Imperial College NHS Trust, London, UK
| | - Andrea Rockall
- Department of Cancer and Surgery, Faculty of Medicine, Imperial College, London, UK.
- Department of Radiology, Imperial College Healthcare NHS Trust, London, UK.
| | - Laura Tookman
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
4
|
Arafa AT, Yadav S, Marshall CH, Mauer E, Huang M, Yilma B, van der Pol Y, Fragkogianni S, Teslow EA, Kellen S, Boytim E, Luo C, Ludwig M, Zhang W, Jayaraj A, Armstrong DK, Isaacs WB, Drake JM, Nguyen HD, Huang RS, Chao CY, Lou E, Dehm SM, Couch FJ, Hwang JH, Antonarakis ES. Germline-Somatic Interactions in BRCA-Associated Cancers: Unique Molecular Profiles and Clinical Outcomes Linking ATM to TP53 Synthetic Essentiality. Clin Cancer Res 2025; 31:1730-1745. [PMID: 40019487 PMCID: PMC12045718 DOI: 10.1158/1078-0432.ccr-24-2058] [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: 06/29/2024] [Revised: 09/27/2024] [Accepted: 02/27/2025] [Indexed: 03/01/2025]
Abstract
PURPOSE Germline alterations in homologous recombination repair (gHRR) genes affect the pathogenesis, treatment options, and survival of patients with cancer. However, distinct gHRR gene alterations may differentially affect treatment response and oncogenic signaling. In this study, we interrogated genomic and transcriptomic data and assessed clinical outcomes of patients with gHRR mutations across four BRCA-associated cancers (breast, ovarian, pancreatic, and prostate cancers) to identify therapeutic vulnerabilities. EXPERIMENTAL DESIGN We assessed 24,309 patients undergoing matched tumor/normal next-generation DNA and RNA sequencing. Annotated gHRR gene variants [germline BRCA1, germline BRCA2, germline PALB2, germline ATM (gATM), and germline CHEK2] were analyzed. HRs were used to assess survival outcomes comparing germline versus sporadic groups. Somatic alterations and their frequencies were compared across gHRR-altered groups. Differential gene expression and gene set enrichment analysis were used to compare transcriptomic profiles. RESULTS Somatic TP53 mutations were depleted in gATM carriers (P < 0.05) across all four BRCA-associated cancers by up to 2.5-fold. Tumors with germline BRCA1/2 mutations were associated with improved survival in patients with ovarian cancer and had consistent enrichment of TP53 mutations in all four cancers. gATM mutations displayed elevated p53 transcriptional activity in all four cancers, with significance reached in breast and prostate cancers (P < 0.01). In breast, ovarian, and prostate cancers, gATM tumors demonstrated significantly increased inflammatory pathways (P < 0.001). Finally, using gene dependency data, we found that cell lines that were highly dependent on ATM were co-dependent on canonical p53 function. CONCLUSIONS gATM-associated cancers seem to require intact p53 activity and this synthetic essentiality may be used to guide targeted therapies that perturb canonical TP53 function.
Collapse
Affiliation(s)
- Ali T. Arafa
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Catherine H. Marshall
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | | | - Samuel Kellen
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ella Boytim
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Christine Luo
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Megan Ludwig
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Weijie Zhang
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Deborah K. Armstrong
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - William B. Isaacs
- Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Justin M. Drake
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Hai Dang Nguyen
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - R. Stephanie Huang
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, MN 55455, USA
- Bioinformatics and Computational Biology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Emil Lou
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Scott M. Dehm
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | - Fergus J. Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Justin H. Hwang
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA
| | | |
Collapse
|
5
|
Habaka M, Daly GR, Shinyanbola D, Alabdulrahman M, McGrath J, Dowling GP, Hehir C, Huang HYR, Hill ADK, Varešlija D, Young LS. PARP Inhibitors in the Neoadjuvant Setting; A Comprehensive Overview of the Rationale for their Use, Past and Ongoing Clinical Trials. Curr Oncol Rep 2025; 27:533-551. [PMID: 40192976 DOI: 10.1007/s11912-025-01669-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2025] [Indexed: 05/16/2025]
Abstract
PURPOSEOF REVIEW Poly (ADP-ribose) polymerases (PARPs) are enzymes essential for detecting and repairing DNA damage through poly-ADP-ribosylation. In cancer, cells with deficiencies in homologous recombination repair mechanisms often become more dependent on PARP-mediated repair mechanisms to effectively repair dsDNA breaks. As such, PARP inhibitors (PARPis) were introduced into clinical practice, serving as a key targeted therapy option through synthetic lethality in the treatment of cancers with homologous recombination repair deficiency (HRD). Though PARPis are currently approved in the adjuvant setting for several cancer types such as ovarian, breast, prostate and pancreatic cancer, their potential role in the neoadjuvant setting remains under investigation. This review outlines the rationale for using PARPi in the neoadjuvant setting and evaluates findings from early and ongoing clinical trials. RECENT FINDINGS Our analysis indicates that numerous studies have explored PARPi as a neoadjuvant treatment for HRD-related cancers. The majority of neoadjuvant PARPi trials have been performed in breast and ovarian cancer, while phase II/III evidence supporting efficacy in prostate and pancreatic cancers remains limited. Studies are investigating PARPi in the neoadjuvant setting of HRD-related cancers. Future research should prioritize combination strategies with immune checkpoint inhibitors and expand outcome measures to include patient satisfaction and quality-of-life metrics.
Collapse
Affiliation(s)
- Minatoullah Habaka
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland.
| | - Gordon R Daly
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Deborah Shinyanbola
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | | | - Jason McGrath
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Gavin P Dowling
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Cian Hehir
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
| | - Helen Ye Rim Huang
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Arnold D K Hill
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Department of Surgery, Beaumont Hospital, Dublin, Ireland
- Beaumont RCSI Cancer Centre, Beaumont Hospital, Dublin, Ireland
| | - Damir Varešlija
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Beaumont RCSI Cancer Centre, Beaumont Hospital, Dublin, Ireland
| | - Leonie S Young
- Department of Surgery, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- Beaumont RCSI Cancer Centre, Beaumont Hospital, Dublin, Ireland
| |
Collapse
|
6
|
Duraki D, Jabeen M, Mao C, Wang L, Ghosh S, Dai X, Zhu J, Boudreau MW, Nelson ER, Hergenrother PJ, Cheng G, Shapiro DJ. A necrosis inducer promotes an immunogenic response and destroys ovarian cancers in mouse xenografts and patient ascites organoids. Cancer Lett 2025; 625:217738. [PMID: 40311911 DOI: 10.1016/j.canlet.2025.217738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/24/2025] [Accepted: 04/22/2025] [Indexed: 05/03/2025]
Abstract
Most ovarian cancer patients present with advanced disease and there are few targeted therapies; consequently, five-year survival for ovarian cancer remains below 50%. We described the anticipatory unfolded protein response (a-UPR) hyperactivator, ErSO, which induced profound and often complete regression of breast cancer in mouse models. Here we explore the effectiveness of ErSO against ovarian cancer. ErSO induced death of human PEO4 and Caov-3 ovarian cancer cells in vitro. In mouse xenografts, injected ErSO induced rapid complete, or near complete, regression of orthotopic metastatic PEO4 tumors and of Caov-3 ovarian tumors. Ovarian cancer patients often develop malignant ascites containing ovarian cancer organoids that drive metastasis. ErSO showed activity against 7/7 fresh patient derived ascites organoids (PDAOs). Low nanomolar ErSO destroyed 2/7 PDAOs. ErSO-mediated cell death in PDAOs occurred through the same a-UPR activation mechanism seen in cell culture. Moreover, ErSO family compound-induced a-UPR activation in ovarian cancer cells triggers necrotic cell death and release of damage associated molecular patterns (DAMPs), which strongly activated human macrophage and induced monocyte migration. These studies suggest ErSO has unusual potential for treatment of advanced ovarian cancer.
Collapse
Affiliation(s)
- Darjan Duraki
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Musarrat Jabeen
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Chengjian Mao
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Lawrence Wang
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Santanu Ghosh
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Xinyi Dai
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Junyao Zhu
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Matthew W Boudreau
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Erik R Nelson
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Paul J Hergenrother
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Carl R. Woese Institute for Genomic Biology University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Georgina Cheng
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Gynecologic Oncology, Carle Health, Urbana IL, 61801, USA; Department of Clinical Sciences, Carle Illinois College of Medicine, Urbana, IL, 61801, USA
| | - David J Shapiro
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA; Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
7
|
Poveda A, Lheureux S, Colombo N, Cibula D, Elstrand M, Weberpals J, Bjurberg M, Oaknin A, Sikorska M, González-Martín A, Madry R, Rubio Pérez MJ, Ledermann J, Romero I, Özgören O, Barnicle A, Marshall H, Bashir Z, Škof E. Maintenance olaparib monotherapy in patients with platinum-sensitive relapsed ovarian cancer without a germline BRCA1 and/or BRCA2 mutation: Final overall survival results from the OPINION trial. Gynecol Oncol 2025; 197:74-82. [PMID: 40300425 DOI: 10.1016/j.ygyno.2025.04.580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 05/01/2025]
Abstract
OBJECTIVE Maintenance olaparib demonstrated clinical activity for progression-free survival in patients without a germline BRCA1 and/or BRCA2 mutation (non-gBRCAm) who had platinum-sensitive relapsed ovarian cancer in the phase IIIb, open-label, single-arm, non-comparator, international OPINION trial (NCT03402841). We report final overall survival (OS; secondary endpoint), prespecified secondary endpoint updates and ad hoc OS analysis by homologous recombination deficiency (HRD) and somatic BRCAm (sBRCAm) status. METHODS Patients with non-gBRCAm platinum-sensitive relapsed ovarian cancer, ≥2 prior lines of platinum-based chemotherapy, and in response following their last platinum-based chemotherapy received 300 mg olaparib tablets twice daily until disease progression or unacceptable toxicity. RESULTS 279 patients were enrolled and treated. With a median follow-up in patients censored for OS of 33.1 months (data cut-off September 17, 2021), median OS was 32.7 months (95 % CI 29.5-35.3); the 24-month OS rate was 65.8 %. In ad hoc subgroup analyses, OS rates tended to be higher in patients with HRD-positive tumors; 24-month OS rates were 81.5 %, 74.2 %, 72.0 % and 55.8 % in the sBRCAm, HRD-positive including sBRCAm, HRD-positive excluding sBRCAm, and HRD-negative subgroups, respectively. Grade ≥ 3 treatment-emergent adverse events were reported in 82 patients (29.4 %), most commonly anemia (13.6 %). Overall, two cases of myelodysplastic syndrome were reported (no new cases since the primary analysis). CONCLUSION These data provide additional evidence of olaparib as maintenance therapy in patients with non-gBRCAm platinum-sensitive relapsed ovarian cancer, with longer OS observed in those with HRD-positive tumors. The safety profile was consistent with the primary analysis and known safety profile of olaparib, with no new safety findings.
Collapse
Affiliation(s)
| | | | - Nicoletta Colombo
- University of Milan-Bicocca and European Institute of Oncology IRCCS, Milan, Italy
| | - David Cibula
- General University Hospital in Prague, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Mari Elstrand
- Oslo University Hospital, Norwegian Radium Hospital, Oslo, Norway
| | | | - Maria Bjurberg
- Skåne University Hospital, Lund University, Lund, Sweden
| | - Ana Oaknin
- Medical Oncology Service, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | | | | | | | | | - Jonathan Ledermann
- UCL Cancer Institute, University College London and UCL Hospitals, London, UK
| | | | | | | | | | | | - Erik Škof
- Institute of Oncology Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
8
|
Haraga H, Nakayama K, Razia S, Ishikawa M, Yamashita H, Kanno K, Nagase M, Ishibashi T, Katagiri H, Shimomura R, Otsuki Y, Nakayama S, Kyo S. Exploring the Genetic and Clinical Landscape of Dedifferentiated Endometrioid Carcinoma. Int J Mol Sci 2025; 26:4137. [PMID: 40362376 PMCID: PMC12071752 DOI: 10.3390/ijms26094137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/21/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Dedifferentiated endometrioid carcinoma (DDEC) is rare, has a poor prognosis, and the genes responsible for dedifferentiation remain unclear. This study aimed to clarify the characteristics of DDEC in Japanese patients and develop treatment strategies. Eighteen DDEC cases were included; their clinicopathological features and prognoses were analyzed and compared to those of other histological subtypes. The samples were divided into well-differentiated and undifferentiated components; immunostaining and whole-exome sequencing (n = 3 cases) were conducted. The incidence of DDEC was 2.0% among endometrial cancers. The 5-year progression-free survival and the 5-year overall survival for DDEC was approximately 40% and 30%, respectively. Immunohistochemistry showed that 66.7% of patients were mismatch repair deficient. The rate of p53 mutations was higher than that reported in previous studies, and patients with p53 mutations in the undifferentiated components had a poor prognosis. Whole-exome sequencing revealed different gene mutations and mutation signatures between well-differentiated and undifferentiated components. New genetic mutations in undifferentiated regions were uncommon in all three cases. One case (case 1) exhibited homologous recombination deficiency, whereas the other two showed microsatellite instability-high and hypermutator phenotypes. Genetic analysis suggests that immune checkpoint and poly (ADP-ribose) polymerase inhibitors and drugs targeting the p53 pathway may be effective against DDEC.
Collapse
Affiliation(s)
- Hikaru Haraga
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University, 89-1, Enya-Cho, Izumo 693-8501, Shimane, Japan; (H.H.); (M.I.); (H.Y.); (K.K.)
| | - Kentaro Nakayama
- Department of Obstetrics and Gynecology, Nagoya City University East Medical Center, Nagoya 464-8547, Aichi, Japan;
| | - Sultana Razia
- Department of Legal Medicine, Faculty of Medicine, Shimane University, 89-1, Enya-Cho, Izumo 693-8501, Shimane, Japan;
| | - Masako Ishikawa
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University, 89-1, Enya-Cho, Izumo 693-8501, Shimane, Japan; (H.H.); (M.I.); (H.Y.); (K.K.)
| | - Hitomi Yamashita
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University, 89-1, Enya-Cho, Izumo 693-8501, Shimane, Japan; (H.H.); (M.I.); (H.Y.); (K.K.)
| | - Kosuke Kanno
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University, 89-1, Enya-Cho, Izumo 693-8501, Shimane, Japan; (H.H.); (M.I.); (H.Y.); (K.K.)
| | - Mamiko Nagase
- Department of Pathology, Faculty of Medicine, Shimane University, 89-1, Enya-Cho, Izumo 693-8501, Shimane, Japan;
| | - Tomoka Ishibashi
- Department of Obstetrics and Gynecology, Nagoya City University East Medical Center, Nagoya 464-8547, Aichi, Japan;
| | - Hiroshi Katagiri
- Department of Obstetrics and Gynecology, Masuda Red Cross Hospital, I103-1, Otoyoshi-Cho, Masuda 698-8501, Shimane, Japan;
| | - Ryoichi Shimomura
- Department of Pathology, Masuda Red Cross Hospital, I103-1, Otoyoshi-Cho, Masuda 698-8501, Shimane, Japan;
| | - Yoshiro Otsuki
- Department of Pathology, Seirei Hamamatsu General Hospital, 2-12-12, Sumiyoshi, Chuo-ku, Hamamatsu 430-8558, Shizuoka, Japan;
| | - Satoru Nakayama
- Department of Obstetrics and Gynecology, Seirei Hamamatsu General Hospital, 2-12-12, Sumiyoshi, Chuo-ku, Hamamatsu 430-8558, Shizuoka, Japan;
| | - Satoru Kyo
- Department of Obstetrics and Gynecology, Faculty of Medicine, Shimane University, 89-1, Enya-Cho, Izumo 693-8501, Shimane, Japan; (H.H.); (M.I.); (H.Y.); (K.K.)
| |
Collapse
|
9
|
Trewin-Nybråten CB, Leithe S, Paulsen T, Langseth H, Fortner RT. Ovarian cancer survival by residual disease following cytoreductive surgery: a nationwide study in Norway. Br J Cancer 2025:10.1038/s41416-025-03018-0. [PMID: 40287590 DOI: 10.1038/s41416-025-03018-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 03/25/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Residual disease (RD) following cytoreductive surgery is prognostic for epithelial ovarian cancer (EOC) patients. Few studies have evaluated RD and survival by tumor histotype and across continuous RD diameter. METHODS 2608 individuals with stages III-IV invasive EOC diagnosed between 2013 and 2022 were identified using the Cancer Registry of Norway. In flexible parametric models, we compared excess mortality comparing RD versus no macroscopic residual disease (NMRD); systemic anti-cancer therapy was evaluated in a sub-cohort from 2019. Excess mortality was assessed across continuous RD size using restricted cubic splines. RESULTS Among 1849 patients with cytoreductive surgery, survival was worse for individuals with RD (vs. NMRD), excess hazard ratio (EHR) = 2.62 (95% confidence interval = (2.27-3.01)); no heterogeneity was observed by histotype (p = 0.21). Patients with 0.1-0.4 cm RD had 2-fold higher risk of death (EHR = 2.09 (1.63-2.68)) relative to women with NMRD; ~3-fold higher risk was observed for all other categories (e.g., 0.5-0.9 cm, EHR = 2.97 (2.26-3.89); 3.0-20 cm, 2.75 (2.05-3.70)). No significant difference in three-year survival was observed across continuous RD diameter (p ≥ 0.17). NMRD was associated with better survival regardless of neoadjuvant chemotherapy. DISCUSSION Achieving NMRD resulted in the best survival outcomes. Among patients with RD, we observed no significant difference in survival by RD diameter.
Collapse
Affiliation(s)
- Cassia B Trewin-Nybråten
- Department of Registration, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | - Sigrid Leithe
- Department of Registration, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
| | - Torbjørn Paulsen
- Department of Gynecological Oncology, Division of Cancer Medicine, Oslo University Hospital, Oslo, Norway
| | - Hilde Langseth
- Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - Renée Turzanski Fortner
- Department of Research, Cancer Registry of Norway, Norwegian Institute of Public Health, Oslo, Norway.
- Division of Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany.
| |
Collapse
|
10
|
Micha JP, Rettenmaier MA, Bohart R, Gorman JP, Goldstein BH. Clinical differences among the PARP inhibitors in the first-line treatment of advanced-stage ovarian carcinoma. Expert Rev Anticancer Ther 2025:1-5. [PMID: 40249727 DOI: 10.1080/14737140.2025.2496519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/03/2025] [Accepted: 04/17/2025] [Indexed: 04/20/2025]
Abstract
INTRODUCTION Despite favorable patient response rates to first-line chemotherapy, 5-year overall survival rates in ovarian cancer are dismal. Fortunately, the inclusion of PARP inhibitors (e.g. olaparib, niraparib, and rucaparib) following the completion of primary induction chemotherapy, has conferred improved progression-free survival rates. AREAS COVERED There are treatment outcome differences among the various PARP inhibitors that coincide with a patient's specific homologous recombination deficit (HRD) status; moreover, only single-agent olaparib and olaparib with bevacizumab have conferred a 5-year overall survival benefit. In the current review, we recount the clinical differences associated with the available PARP inhibitors in the management of advanced-stage ovarian carcinoma. EXPERT OPINION The inclusion of PARP inhibitors has significantly improved survival benefits in advanced-stage ovarian cancer, especially among patients with an identifiable HRD. While there are tolerability differences inherent to the specific PARP inhibitors, not to mention approval distinctions, olaparib is the only PARP inhibitor that has demonstrated consistent overall survival benefits.
Collapse
Affiliation(s)
- John P Micha
- Department of Gynecologic and Breast Oncology, Women's Cancer Research Foundation, Laguna Beach, CA, USA
| | - Mark A Rettenmaier
- Department of Gynecologic and Breast Oncology, Women's Cancer Research Foundation, Laguna Beach, CA, USA
| | - Randy Bohart
- Department of Pharmacology, Oso Home Care, Inc, Irvine, CA, USA
| | - Joshua P Gorman
- Department of Health Sciences, University of Arizona College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Bram H Goldstein
- Department of Gynecologic and Breast Oncology, Women's Cancer Research Foundation, Laguna Beach, CA, USA
| |
Collapse
|
11
|
Wickramasinghe C, Kim S, Jiang Y, Bao X, Yue Y, Jiang J, Hong A, Sanai N, Li J. Population Pharmacokinetic Modeling of Total and Unbound Pamiparib in Glioblastoma Patients: Insights into Drug Disposition and Dosing Optimization. Pharmaceutics 2025; 17:524. [PMID: 40284519 PMCID: PMC12030534 DOI: 10.3390/pharmaceutics17040524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/10/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
Background: This study aimed to develop a population pharmacokinetic (PK) model that characterized the plasma concentration-time profiles of the total and unbound pamiparib, a PARP inhibitor, in glioblastoma patients and identified patient factors influencing the PK. Methods: The total and unbound pamiparib plasma concentration data were obtained from 41 glioblastoma patients receiving 60 mg of pamiparib twice daily. Nonlinear mixed-effects modeling was performed using Monolix (2024R1) to simultaneously fit the total and unbound drug plasma concentration data. The covariate model was developed by covariate screening using generalized additive modeling followed by stepwise covariate modeling. Model simulations were performed following oral doses of 10-60 mg BID. Results: The total and unbound pamiparib plasma concentration-time profiles were best described by a one-compartment model with first-order absorption and elimination. Creatinine clearance and age were the significant covariates on the apparent volume of distribution (V/F) and apparent clearance (CL/F), respectively, explaining ~22% and ~5% of IIV of V/F and CL/F. Population estimates of the absorption rate constant (Ka), V/F, CL/F, and unbound fraction for the total drug were 1.58 h-1, 44 L, 2.59 L/h, and 0.041. Model simulations suggested that doses as low as 20 mg BID may be adequate for therapeutic effects in a general patient population, assuming that a target engagement ratio (i.e., unbound Css,min/IC50) of 5 or above is sufficient for full target engagement. Conclusions: The total and unbound pamiparib plasma PK are well characterized by a linear one-compartment model, with creatinine clearance as the significant covariate on V/F. Model simulations support further clinical investigation into dose reduction to optimize the benefit-to-risk ratio of pamiparib, particularly in combination therapies.
Collapse
Affiliation(s)
- Charuka Wickramasinghe
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Seongho Kim
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yuanyuan Jiang
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Xun Bao
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yang Yue
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Jun Jiang
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Amy Hong
- Barrow Neurological Institute, St. Joseph’s Hospital & Medical Center, Phoenix, AZ 85013, USA
| | - Nader Sanai
- Barrow Neurological Institute, St. Joseph’s Hospital & Medical Center, Phoenix, AZ 85013, USA
| | - Jing Li
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
12
|
Martires LCM, Ahronian LG, Pratt CB, Das NM, Zhang X, Whittington DA, Zhang H, Shen B, Come J, McCarren P, Liu MS, Min C, Feng T, Jahic H, Ali JA, Aird DR, Li F, Andersen JN, Huang A, Mallender WD, Nicholson HE. LIG1 Is a Synthetic Lethal Target in BRCA1 Mutant Cancers. Mol Cancer Ther 2025; 24:618-627. [PMID: 39868490 PMCID: PMC11962389 DOI: 10.1158/1535-7163.mct-24-0598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/08/2024] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Synthetic lethality approaches in BRCA1/2-mutated cancers have focused on PARP inhibitors, which are subject to high rates of innate or acquired resistance in patients. In this study, we used CRISPR/Cas9-based screening to identify DNA ligase I (LIG1) as a novel target for synthetic lethality in BRCA1-mutated cancers. Publicly available data supported LIG1 hyperdependence of BRCA1 mutant cells across a variety of breast and ovarian cancer cell lines. We used CRISPRn, CRISPRi, RNAi, and protein degradation to confirm the lethal effect of LIG1 inactivation at the DNA, RNA, and protein level in BRCA1 mutant cells in vitro. LIG1 inactivation resulted in viability loss across multiple BRCA1-mutated cell lines, whereas no effect was observed in BRCA1/2 wild-type cell lines, demonstrating target selectivity for the BRCA1 mutant context. On-target nature of the phenotype was demonstrated through rescue of viability with exogenous wild-type LIG1 cDNA. Next, we demonstrated a concentration-dependent relationship of LIG1 protein expression and BRCA1 mutant cell viability using a titratable, degradable LIG1 fusion protein. BRCA1 mutant viability required LIG1 catalytic activity, as catalytically dead mutant LIG1K568A failed to rescue viability loss caused by endogenous LIG1 depletion. LIG1 perturbation produced proportional increases in PAR staining in BRCA1 mutant cells, indicating a mechanism consistent with the function of LIG1 in sealing ssDNA nicks. Finally, we confirmed LIG1 hyperdependence in vivo using a xenograft model in which LIG1 loss resulted in tumor stasis in all mice. Our cumulative findings demonstrate that LIG1 is a promising synthetic lethal target for development in patients with BRCA1-mutant cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jon Come
- Tango Therapeutics Inc., Boston, Massachusetts
| | | | - Mu-Sen Liu
- Tango Therapeutics Inc., Boston, Massachusetts
| | | | | | - Haris Jahic
- Tango Therapeutics Inc., Boston, Massachusetts
| | | | | | - Fang Li
- Tango Therapeutics Inc., Boston, Massachusetts
| | | | - Alan Huang
- Tango Therapeutics Inc., Boston, Massachusetts
| | | | | |
Collapse
|
13
|
Hu Y, Zhou J, Ling X, Zhao K, Huang P, Gao M, Li X, Sun M, Zou Y, Feng G. KPNA5 Suppresses Malignant Progression of Ovarian Cancer Through Importing the PTPN4 Into the Nucleus. Cancer Med 2025; 14:e70731. [PMID: 40145330 PMCID: PMC11947769 DOI: 10.1002/cam4.70731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/29/2025] [Accepted: 02/15/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Abnormal protein localization due to disrupted nucleoplasmic transport is common in tumor cells, but its mechanisms are not well understood. Nuclear pore complexes and nuclear transporter proteins are crucial for protein transport between the nucleus and cytoplasm. Evidence increasingly shows that abnormal expression of karyopherin family proteins disrupts protein translocation, affecting processes like cell differentiation, proliferation, apoptosis, and transcriptional regulation. However, their functions and roles in ovarian cancer remain unclear. METHODS The expression level of KPNA5 in ovarian cancer tissues and cells was detected by IHC, Western blot, and qPCR. CCK-8 and colony formation assays were used to assess cell proliferation ability. Transwell assay was conducted to determine cell migration and invasion capacity. A xenograft model was used to assess the effect of KPNA5 on tumor growth in vivo. RESULTS KPNA5 expression is downregulated in ovarian cancer (OC) tissues. Low KPNA5 levels were associated with poor survival in OC patients, validated by an OC tissue sample cohort. Overexpression of KPNA5 significantly suppressed OC cell proliferation, tumor growth, and invasion in both in vitro and in vivo studies. Mechanistically, KPNA5 recognizes nuclear localization signals (NLSs) in PTPN4, mediating its nuclear transport and inhibiting STAT3 phosphorylation and its downstream signaling pathway. Similarly, PTPN4 overexpression reduced OC cell viability and invasion, also suppressing STAT3 phosphorylation. CONCLUSIONS Our findings identify KPNA5 as a tumor suppressor in OC, presenting a potential therapeutic target for OC treatment.
Collapse
Affiliation(s)
- Yanming Hu
- Suzhou Cancer Center Core LaboratoryThe Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolSuzhouChina
| | - Jing Zhou
- Department of OncologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhouJiangsuChina
| | - Xinru Ling
- Department of Obstetrics and GynecologyThe Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolSuzhouChina
| | - Kun Zhao
- Department of OncologyHuaian Hospital of Huaian CityHuaianJiangsuChina
| | - Peng Huang
- Suzhou Cancer Center Core LaboratoryThe Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolSuzhouChina
| | - Max Gao
- Department of Computer Science and EngineeringUniversity of CaliforniaDavisCaliforniaUSA
| | - Xiaoqing Li
- Suzhou Cancer Center Core LaboratoryThe Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolSuzhouChina
| | - Ming Sun
- Suzhou Cancer Center Core LaboratoryThe Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolSuzhouChina
| | - Yanfen Zou
- Department of Obstetrics and GynecologyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Guannan Feng
- Department of Obstetrics and GynecologyThe Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu SchoolSuzhouChina
| |
Collapse
|
14
|
Turaga SM, Hembruff SL, Savelieff MG, Ghosh A, Puri RV, Pathak HB, Paradiso LJ, Myers TJ, Li A, Godwin AK. Dual targeting of Aurora Kinase A and poly (ADP-ribose) polymerase as a therapeutic option for patients with ovarian cancer: preclinical evaluations. J Cancer Res Clin Oncol 2025; 151:124. [PMID: 40138020 PMCID: PMC11946953 DOI: 10.1007/s00432-025-06152-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
PURPOSE Epithelial ovarian cancers (EOCs) are often diagnosed at an advanced stage, leading to poor survival outcomes despite chemotherapeutic and surgical advances. Precision oncology strategies have been developed to treat EOCs characterized by BRCA1 and BRCA2 inactivation with consequent homologous recombination (HR) repair defects. HR deficiency enhances tumor sensitivity to poly (ADP-ribose) polymerase (PARP) inhibitors (PARPis), approved for EOCs as maintenance therapy, although they have been discontinued as recurrent EOC monotherapy. However, combination treatment with PARPis may be a viable alternate strategy for EOCs. Moreover, EOC patients with wild-type BRCA are ineligible for PARPs, necessitating novel approaches. We previously discovered that inhibiting Aurora kinase A (AURKA) downregulates PARP and BRCA1/2 expression in EOCs and may constitute a viable approach for EOCs. METHODS Herein, we evaluated combined PARPi olaparib with the selective AURKA inhibitor (AURKAi) VIC-1911 in six different patient-derived xenograft (PDX) EOC models, including two with mutant BRCA1, two with mutant BRCA2, one with mutant BRCA1/2, and one with wild-type BRCA1/2. RESULTS We found that combined olaparib + VIC-1911 treatment reduced tumor volumes and weights by up 90% in some PDX models, with synergistic effect compared to olaparib and VIC-1911 monotherapy. Additionally, combined olaparib + VIC-1911 treatment improved survival of mice harboring both mutant BRCA1 and wild-type BRCA1/2 PDXs. Generally, mice tolerated the drug combinations well during treatment, though loss of body weight was observed at higher drug dosages and with intensive treatment regimens. CONCLUSION Our studies indicate a synergistic benefit from combined PARPi and AURKAi in mutant and wild-type BRCA EOC tumors.
Collapse
Affiliation(s)
- Soumya M Turaga
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Masha G Savelieff
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA
| | - Arnab Ghosh
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Zoology, Rajiv Gandhi University, Itanagar, Arunachal Pradesh, India
| | - Rajni V Puri
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Harsh B Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | | | | | - Ao Li
- JS Innopharm LTD, Shanghai, China
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.
- University of Kansas Cancer Center, Kansas City, KS, USA.
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
15
|
Huang X, Zhou J, Li Z, Ye M, Hou C, Zhang Q, Chen Y, Li Q, Li F, Zhu X, Jiang J. EM-12, a natural sesquiterpene lactone extracted from Elephantopus mollis, promotes cancer cell apoptosis by activating ER stress. Med Oncol 2025; 42:115. [PMID: 40100452 DOI: 10.1007/s12032-025-02654-z] [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: 11/17/2024] [Accepted: 02/25/2025] [Indexed: 03/20/2025]
Abstract
The Elephantopus mollis H.B.K. contains various sesquiterpene lactones that have shown anti-proliferative and proapoptotic effects in various cancers, although the underlying mechanisms are partially understood. Inducing of excessive ER stress is a potential cancer therapeutic strategy. However, ER stress activator remain limited in current clinical applications. In this study, we identified that EM-12, an uncovered sesquiterpene lactone isolated from Elephantopus mollis H.B.K., as a BiP ATPase activity inhibitor through BiP ATPase activity assay in vitro. This molecule also exhibits significantly greater cytotoxicity in numerous ovarian cancer cell lines, including paclitaxel-resistance ovarian cancer cell line, compared to transformed ovarian epithelial cell lines. In addition, EM-12 exerts broad-spectrum cytotoxicity against various human cancer cell lines, including liver, nasopharyngeal, and breast cancer cell lines. Mechanically, EM-12 promotes ER stress and ER-stress-related apoptosis to against cancer cells through inhibiting BiP ATPase activity.
Collapse
Affiliation(s)
- Xiang Huang
- Medical College of Jiaying University, Meizhou, 514031, China
- Department of Medical Bioinformatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Junzhen Zhou
- Department of Breast Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Zhenhua Li
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou, 510630, China
| | - Meijun Ye
- Department of Medical Bioinformatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Changyan Hou
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou, 510630, China
| | - Qing Zhang
- Department of Breast Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Yuanhong Chen
- Department of Gynecology, Dongguan Eastern Central Hospital, The Sixth Affiliated Hospital of Jinan University, Dongguan, 523560, China
| | - Qiang Li
- Department of General Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
| | - Fengying Li
- Department of Gynecology, The Affiliated Shunde Hospital of Jinan University, Foshan, 528000, China
| | - Xiaofeng Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China.
| | - Jianwei Jiang
- Department of Biochemistry, Basic Medical College, Jinan University, Guangzhou, 510630, China.
| |
Collapse
|
16
|
Matsumoto K, Matsumoto Y, Wada J. PARylation-mediated post-transcriptional modifications in cancer immunity and immunotherapy. Front Immunol 2025; 16:1537615. [PMID: 40134437 PMCID: PMC11933034 DOI: 10.3389/fimmu.2025.1537615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Poly-ADP-ribosylation (PARylation) is a post-translational modification in which ADP-ribose is added to substrate proteins. PARylation is mediated by a superfamily of ADP-ribosyl transferases known as PARPs and influences a wide range of cellular functions, including genome integrity maintenance, and the regulation of proliferation and differentiation. We and others have recently reported that PARylation of SH3 domain-binding protein 2 (3BP2) plays a role in bone metabolism, immune system regulation, and cytokine production. Additionally, PARylation has recently gained attention as a target for cancer treatment. In this review, we provide an overview of PARylation, its involvement in several signaling pathways related to cancer immunity, and the potential of combination therapies with PARP inhibitors and immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | - Yoshinori Matsumoto
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Faculty of
Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | | |
Collapse
|
17
|
Bacares R, Soslow R, Olvera N, Levine DA, Zhang L. A Rapid and Reliable Test for BRCA1 Promoter Hypermethylation in Paraffin Tissue Using Pyrosequencing. Diagnostics (Basel) 2025; 15:601. [PMID: 40075848 PMCID: PMC11898801 DOI: 10.3390/diagnostics15050601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/06/2025] [Accepted: 02/16/2025] [Indexed: 03/14/2025] Open
Abstract
Background: Ovarian cancers harboring inactivating mutations in BRCA1 or BRCA2 demonstrate increased sensitivity to poly (ADP-ribose) polymerase inhibitors (PARPis). BRCA1 promoter methylation could serve as a more precise biomarker for therapy response, as it reflects a dynamic mechanism, compared with genomic scarring, which remains persistent and lacks real-time prediction of sensitivity after prior lines of treatment. Additionally, the BRCA1 promoter methylation may provide a more precise biomarker for identifying homologous recombination deficiency compared to genomic scars. In this study, we describe the validation of a pyrosequencing method to assess BRCA1 promoter methylation status. Methods: Tumor DNA from high-grade serous ovarian carcinoma was tested targeting 11 CpG sites adjacent to the BRCA1 transcription start site. All cases had concordant results compared with TCGA methylation data or real-time PCR results. To determine the sensitivity of this assay, we performed a dilution series experiment using seven mixtures of methylated DNA and unmethylated genomic DNA (100%, 50%, 25%, 12.5%, 6.25%, 3.125%, and 1.56%). Results: We observed a high degree of correlation (R2 = 0.9945) between predicted and observed results. Intra- and inter-run reproducibility was established by performing six cases in triplicate in the same run and in three different runs. Conclusions: By applying 10% as the cutoff for detection of methylation, the PyroMark Q24 pyrosequencing assay demonstrated 100% concordance across all the ovarian cancer cases included in this validation. This assay has been approved by the New York State Department of Health as a laboratory-specific assay for clinical use.
Collapse
Affiliation(s)
- Ruben Bacares
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.B.); (R.S.)
| | - Robert Soslow
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.B.); (R.S.)
- Department of Pathology, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Narciso Olvera
- Departments of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (N.O.)
- Laura and Issac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, NY 10016, USA
| | - Douglas A. Levine
- Departments of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (N.O.)
- Global Clinical Development, Merck Research Laboratories, Rahway, NJ 07065, USA
| | - Liying Zhang
- Departments of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (R.B.); (R.S.)
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
18
|
Rodríguez Sagrado MA, Alvarez Criado J, Arenaza Peña AE, Escudero-Vilaplana V, Folguera Olias C, Herrero Fernandez M, Martinez Nieto C, Rubio Salvador AR, Sanmartin Fenollera P, Vazquez Castillo MJ. Niraparib Maintenance Therapy in Patients with Platinum-Sensitive Recurrent Ovarian Cancer: Real-World Experience at Hospitals in Spain. Target Oncol 2025; 20:319-327. [PMID: 39853564 DOI: 10.1007/s11523-024-01121-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2024] [Indexed: 01/26/2025]
Abstract
BACKGROUND The reported benefit of poly (ADP-ribose) polymerase inhibitor (PARPi) maintenance in patients with newly diagnosed and platinum (Pt)-sensitive recurrent ovarian cancer (OC) included in randomized clinical trials needs to be corroborated in a less selected population. OBJECTIVE The aim is to increase the evidence on niraparib in a real-world setting. METHODS This is a retrospective observational study including women with platinum-sensitive relapsed high-grade serous OC who started niraparib maintenance between August 2019 (marketing data, Spain) and May 2022. Patients received ≥ 2 previous lines of therapy with complete or partial response to prior chemotherapy. Patient characteristics, niraparib dose, adequacy of dose individualization, effectiveness (progression-free survival [PFS] and overall survival), safety, and economic savings with an individualized starting dose (ISD) strategy were assessed. RESULTS The study included 217 patients with a median of 8.9 months of niraparib duration: breast cancer gene (BRCA) wild-type OC, 70%; two prior treatment lines, 49%; Research on Adverse Drug Events and Reports (RADAR) criteria, 82% (receiving mainly 200 mg of niraparib, 79%). Median PFS was 10.8 months (95% confidence interval [CI], 8.4-14.8) without statistically significant differences based on starting dose strategy, contrary to what was observed on the basis of prior lines, response to prior chemotherapy, BRCA mutational status, and International Federation of Gynecology and Obstetrics (FIGO) stage at diagnosis. The last three variables also showed a statistically significant predictive prognostic value for effectiveness. Dose interruptions due to toxicity were required in 7% of patients, and dose adjustments in 56% were mainly due to hematologic toxicities. The actual dose of niraparib reveals economic savings versus the theoretical cost. CONCLUSION This large real-world analysis corroborates the tolerability and activity of niraparib maintenance for platinum-sensitive recurrent OC and economic savings.
Collapse
Affiliation(s)
| | | | | | - Vicente Escudero-Vilaplana
- Pharmacy Service, Hospital General Universitario Gregorio Marañón. Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
19
|
Alvarez Secord A, Lewin SN, Murphy CG, Cecere SC, Barquín A, Gálvez-Montosa F, Mathews CA, Konecny GE, Ray-Coquard I, Oaknin A, Rubio Pérez MJ, Bonaventura A, Diver EJ, Ayuk SM, Wang Y, Corr BR, Salutari V. The efficacy and safety of mirvetuximab soravtansine in FRα-positive, third-line and later, recurrent platinum-sensitive ovarian cancer: the single-arm phase II PICCOLO trial. Ann Oncol 2025; 36:321-330. [PMID: 39617145 DOI: 10.1016/j.annonc.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/13/2024] [Accepted: 11/23/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Mirvetuximab soravtansine-gynx (MIRV) is a first-in-class, folate receptor alpha (FRα)-targeting antibody-drug conjugate with United States Food and Drug Administration approval for FRα-positive platinum-resistant ovarian cancer. PICCOLO is a phase II, global, open-label, single-arm trial of MIRV as third-line or greater (≥3L) treatment in patients with FRα-positive (≥75% of cells with ≥2+ staining intensity) recurrent platinum-sensitive ovarian cancer (PSOC). PATIENTS AND METHODS Participants received MIRV (6 mg/kg adjusted ideal body weight every 3 weeks) until progressive disease (PD), unacceptable toxicity, withdrawal of consent, or death. Primary endpoint was investigator-assessed objective response rate (ORR). Key secondary endpoint was investigator-assessed duration of response (DOR). Additional endpoints included investigator-assessed progression-free survival (PFS), overall survival (OS), and safety. Analyses of subgroups by disease characteristics (e.g. platinum-free interval) and treatment history [e.g. prior bevacizumab and poly (adenosine diphosphate [ADP]-ribose) polymerase inhibitor (PARPi) treatment] were exploratory. RESULTS Seventy-nine participants were enrolled and efficacy assessable. The primary endpoint was met; ORR was 51.9% [95% confidence interval (CI) 40.4% to 63.3%]. Median DOR was 8.25 months (95% CI 5.55-10.78 months) and median PFS was 6.93 months (95% CI 5.85-9.59 months). OS was not mature at data cut-off. ORR was 45.8% (95% CI 32.7% to 59.2%) in participants with PD while on/within 30 days of prior PARPi (n = 59) and 60.0% (95% CI 14.7% to 94.7%) in those without PD with prior PARPi (n = 5). No new safety signals occurred; most common treatment-emergent adverse events (TEAEs) were gastrointestinal, neurosensory, and resolvable ocular events. TEAEs led to discontinuation in 13 participants (16%) and death in 2 participants (3%). CONCLUSIONS MIRV as ≥3L treatment in heavily pretreated recurrent FRα-positive PSOC demonstrated notable efficacy and tolerable safety, including among those with prior PD on or within 30 days of PARPi (NCT05041257).
Collapse
MESH Headings
- Humans
- Female
- Middle Aged
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/mortality
- Ovarian Neoplasms/metabolism
- Folate Receptor 1/metabolism
- Folate Receptor 1/antagonists & inhibitors
- Aged
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/pathology
- Immunoconjugates/adverse effects
- Immunoconjugates/therapeutic use
- Immunoconjugates/administration & dosage
- Maytansine/analogs & derivatives
- Maytansine/adverse effects
- Maytansine/therapeutic use
- Maytansine/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Adult
- Drug Resistance, Neoplasm/drug effects
- Progression-Free Survival
- Aged, 80 and over
Collapse
Affiliation(s)
- A Alvarez Secord
- Duke Cancer Institute, Duke University School of Medicine, Durham, USA.
| | - S N Lewin
- Holy Name Medical Center Regional Cancer Center, Teaneck, USA
| | - C G Murphy
- Bon Secours Hospital Cork, Cork, Ireland; Cancer Trials Ireland, Dublin, Ireland
| | - S C Cecere
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy; Multicenter Italian Trials in Ovarian Cancer and Gynecologic Malignancies (MITO), Naples, Italy
| | - A Barquín
- Gynecological, Genitourinary, and Skin Cancer Unit, Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - F Gálvez-Montosa
- Medical Oncology Department, Hospital Universitario de Jaén, Jaén, Spain
| | - C A Mathews
- Women & Infants Hospital, Legorreta Cancer Center, The Warren Alpert Medical School of Brown University, Providence, USA
| | - G E Konecny
- Department of Medical Oncology, University of California Los Angeles Medical Center, Santa Monica, USA
| | - I Ray-Coquard
- Leon Berard Center, Lyon, France; GINECO Group, Lyon, France
| | - A Oaknin
- Medical Oncology Service, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - M J Rubio Pérez
- Grupo Español de Investigación en Cáncer de Ovario (GEICO), Madrid, Spain; Department of Medical Oncology, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - A Bonaventura
- Newcastle Private Hospital, New Lambton Heights, Australia
| | | | | | - Y Wang
- ImmunoGen, Inc, Waltham, USA
| | - B R Corr
- Division of Gynecologic Oncology, University of Colorado Cancer Center, Aurora, USA
| | - V Salutari
- Policlinico Universitario Fondazione Agostino Gemelli IRCCS, Rome, Italy
| |
Collapse
|
20
|
Kołakowska K, Kiśluk J, Nikliński J. A Novel Insight into the Role of Obesity-Related Adipokines in Ovarian Cancer-State-of-the-Art Review and Future Perspectives. Int J Mol Sci 2025; 26:1857. [PMID: 40076482 PMCID: PMC11900017 DOI: 10.3390/ijms26051857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/17/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Ovarian cancer (OC) is one of the most fatal gynecological neoplasms. Meta-analyses have shown that the relationship between body mass index (BMI) and ovarian cancer incidence was detected in some types of ovarian cancer. Chronic inflammation and excessive accumulation of free fatty acids are key adipose tissue-derived factors initiating cancer development. Cancer cells transform adipose-derived stem cells into cancer-associated adipocytes, which produce adipokines and interleukins. It was revealed that adipokines exert a pleiotropic role in ovarian cancer pathogenesis. Chemerin presents both pro-cancer and anti-cancer action in ovarian cancer development. Chemerin induces angiogenesis and increases programmed death ligand-1 (PD-L1) expression, leading to enhanced proliferation and migration of OC cells. Apelin impacts cancer cell migration and acts as a mitogenic factor. Moreover, apelin exerts influence on lipid uptake into cancer cells and accelerates fatty acid oxidation, which provides energy for cancer cells. Visfatin induces matrix metallopeptidase 2 (MMP2) expression involved in extracellular matrix degradation and suppresses claudin 3 and 4 expression. Visfatin also induces a shift to anaerobic glucose metabolism and influences poly-ADP ribose polymerase (PARP). Resistin induces MMP2 and vascular endothelial growth factor (VEGF) expression and contributes to cisplatin-resistance development. A substantial body of evidence indicates that antagonists of adipokines mitigate OC progression, and adipokines are gaining gradual recognition as a potential therapeutic aim in ovarian cancer targeted therapy.
Collapse
Affiliation(s)
| | - Joanna Kiśluk
- Department of Clinical Molecular Biology, Medical University of Białystok, 15-269 Białystok, Poland; (K.K.)
| | | |
Collapse
|
21
|
Backes FJ, Calderón Boyle TA, Lim J, Hartman J, Schilder JM, Hurteau JA, Perhanidis J, Golembesky A, Salani R. Real-world duration of first-line maintenance niraparib monotherapy in patients with epithelial ovarian cancer in the United States: the CHAR1ZMA study. Int J Gynecol Cancer 2025; 35:100044. [PMID: 39971430 DOI: 10.1016/j.ijgc.2024.100044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 02/21/2025] Open
Abstract
OBJECTIVE The CHAR1ZMA study described real-world duration of first-line maintenance niraparib monotherapy among patients with epithelial ovarian cancer. METHODS This retrospective study used a US nationwide, electronic-health-record-derived, de-identified database. Eligible patients were aged ≥18 years with epithelial ovarian cancer who initiated first-line maintenance niraparib monotherapy (January 2017-December 2022 [inclusive]) following first-line platinum-based chemotherapy. Niraparib monotherapy duration was measured as the time to treatment discontinuation, estimated using Kaplan-Meier methodology, overall and stratified by treatment duration (early discontinuers [≤90 days]; non-early discontinuers [>90 days or did not discontinue]). Analyses were repeated in a sub-group of patients with homologous recombination-deficient tumors. RESULTS Toxicity was the most common reason for niraparib discontinuation among early discontinuers (67.7%) and was less frequent among non-early discontinuers (18.1%). Dose modifications were less frequent among early discontinuers (29.8%) than non-early discontinuers (53.6%). Disease progression was the most common reason for niraparib discontinuation among non-early discontinuers (74.8%) and was less frequent among early discontinuers (30.4%). The observed median treatment duration was 7.2 months (95% CI 6.0 to 8.1) overall (N = 560) and was 4.5 months longer among non-early discontinuers (11.7 months [95% CI 9.8 to 14.7]; n = 399). In the homologous recombination-deficient sub-group (n = 144), the observed median treatment duration was 11.6 months (95% CI 7.8. to 16.1) and was 5.1 months longer among non-early discontinuers (16.7 months [95% CI 12.0 to 22.8]; n = 114). CONCLUSION In this real-world study of patients with epithelial ovarian cancer receiving first-line maintenance niraparib monotherapy, drug toxicity was the most common reason for treatment discontinuation in early discontinuers. Effective and early clinical management of drug toxicity may help mitigate these adverse events, allowing patients to remain on niraparib maintenance treatment longer and experience the potential full therapeutic benefit.
Collapse
Affiliation(s)
- Floor J Backes
- The Ohio State University Wexner Medical Center and James Hospital Comprehensive Cancer Center, Division of Gynecologic Oncology, Columbus, OH, USA.
| | | | | | | | | | | | | | | | - Ritu Salani
- David Geffen School of Medicine at University of California, Los Angeles, Department of Obstetrics and Gynecology, Los Angeles, CA, USA
| |
Collapse
|
22
|
Tang Z, Pan Y, Li W, Ma R, Wang J. Unlocking the future: mitochondrial genes and neural networks in predicting ovarian cancer prognosis and immunotherapy response. Transl Cancer Res 2025; 14:512-521. [PMID: 39974375 PMCID: PMC11833377 DOI: 10.21037/tcr-24-1233] [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: 07/18/2024] [Accepted: 11/13/2024] [Indexed: 02/21/2025]
Abstract
Background Mitochondrial genes are involved in the tumor metabolism of ovarian cancer (OC), affecting immune cell infiltration and treatment response. We aimed to utilize mitochondrial genes to predict OC prognosis and immunotherapy response. Methods The prognosis data, immunotherapy efficacy and next generation sequencing data of OC patients were downloaded from The Cancer Genome Atlas Program (TCGA) and Gene Expression Omnibus (GEO). Mitochondrial genes were sourced from the MitoCarta3.0 database. Seventy percent of the patients were randomly selected as the discovery cohort for model construction, while the remaining 30% constituted the validation cohort for model assessment. Using the expression of mitochondrial genes as the predictor variable and based on the neural network algorithm, the overall survival (OS) time and immunotherapy efficacy (complete or partial response) of the included patients were predicted. Results There were 375 OC patients included to construct the prognostic model, and 26 patients were included to construct the immune efficacy model. The average area under the receiver operating characteristic curve (AUC) of the prognostic model was: 0.7268 [95% confidence interval (CI), 0.7258-0.7278] in the discovery cohort and 0.6475 (95% CI: 0.6466-0.6484) in the validation cohort. The average AUC of the immunotherapy efficacy model was: 0.9444 (95% CI: 0.8333-1.0000) in the discovery cohort and 0.9167 (95% CI: 0.6667-1.0000) in the validation cohort. Conclusions The application of mitochondrial genes and neural networks shows potential in predicting the prognosis and immunotherapy response in OC patients. And this approach could provide valuable insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Zhijian Tang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
| | - Yuanming Pan
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Wei Li
- Department of Thoracic Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ruiqiong Ma
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
| | - Jianliu Wang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
23
|
Ji S, Chen L, Yu Y, Chen X, Wei L, Gou L, Shi C, Zhuang S. A comprehensive comparison of PARP inhibitors as maintenance therapy in platinum-sensitive recurrent ovarian cancer: a systematic review and network meta-analysis. J Ovarian Res 2025; 18:18. [PMID: 39885555 PMCID: PMC11780803 DOI: 10.1186/s13048-025-01599-1] [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: 04/18/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND PARP inhibitors (PARPis) have shown promising effectiveness for ovarian cancer. This network meta-analysis (PROSPERO registration number CRD42024503390) comprehensively evaluated the effectiveness and safety of PARPis in platinum-sensitive recurrent ovarian cancer (PSROC). METHODS Articles published before January 6, 2024 were obtained from electronic databases. The study assessed and compared survival outcomes including overall survival (OS), progression-free survival (PFS), second progression-free survival (PFS2), time to first subsequent treatment (TFST), time to second subsequent treatment (TSST), and chemotherapy-free interval (CFI). Additionally, safety outcomes were investigated, specifically focusing on grade 3-4 treatment-emergent adverse effects (TEAEs). The evaluation of OS and PFS was also conducted based on the BRCA and HRD (homologous recombination deficiency) statuses. RESULTS Six randomized controlled trials were examined and the four PARPis (olaparib, niraparib, rucaparib and fuluzolparib) have been found to significantly increase the PFS in entire population as well as in subgroups of HRD and BRCAm (BRCA mutation). Only olaparib demonstrated a substantial improvement in OS compared to placebo in entire population (hazard ratio [HR] 0.73; 95% confidence interval [CI] 0.60-0.90), as well as in the subgroup of BRCAm. All analyzed PARPis had significant efficacy in prolonging PFS2, TFST, TSST and CFI. For safety concerns, PARPis could significantly increase incidence of TEAEs (grade3-4), while olaparib had least haematological TEAEs (grade3-4) events compared to other PARPis. CONCLUSION All included PARPis showed various degrees of benefit in survival outcomes and safety profile was acceptable for PSROC patients. Among them olaparib had the best performance in both efficacy and safety.
Collapse
Affiliation(s)
- Shiya Ji
- Department of Health Education, Nanjing Municipal Center for Disease Control and Prevention, No.3, Zizhulin Road, Nanjing, Jiangsu Province, 210003, China.
| | - Lu Chen
- Clinical Medicine College, Yangzhou University, Yangzhou, China
| | - Yebo Yu
- Department of Social Medicine and Health Education, School of Public Health, Peking University, Beijing, China
| | - Xupeng Chen
- Department of Health Education, Nanjing Municipal Center for Disease Control and Prevention, No.3, Zizhulin Road, Nanjing, Jiangsu Province, 210003, China
| | - Liwen Wei
- Department of Health Education, Nanjing Municipal Center for Disease Control and Prevention, No.3, Zizhulin Road, Nanjing, Jiangsu Province, 210003, China
| | - Lili Gou
- Department of Health Education, Nanjing Municipal Center for Disease Control and Prevention, No.3, Zizhulin Road, Nanjing, Jiangsu Province, 210003, China
| | - Cheng Shi
- Department of Health Education, Nanjing Municipal Center for Disease Control and Prevention, No.3, Zizhulin Road, Nanjing, Jiangsu Province, 210003, China
| | - Susu Zhuang
- Department of Health Education, Nanjing Municipal Center for Disease Control and Prevention, No.3, Zizhulin Road, Nanjing, Jiangsu Province, 210003, China
| |
Collapse
|
24
|
Tang ZJ, Pan YM, Li W, Ma RQ, Wang JL. Unlocking the future: Mitochondrial genes and neural networks in predicting ovarian cancer prognosis and immunotherapy response. World J Clin Oncol 2025; 16:94813. [PMID: 39867736 PMCID: PMC11528894 DOI: 10.5306/wjco.v16.i1.94813] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/17/2024] [Accepted: 06/05/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Mitochondrial genes are involved in tumor metabolism in ovarian cancer (OC) and affect immune cell infiltration and treatment responses. AIM To predict prognosis and immunotherapy response in patients diagnosed with OC using mitochondrial genes and neural networks. METHODS Prognosis, immunotherapy efficacy, and next-generation sequencing data of patients with OC were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus. Mitochondrial genes were sourced from the MitoCarta3.0 database. The discovery cohort for model construction was created from 70% of the patients, whereas the remaining 30% constituted the validation cohort. Using the expression of mitochondrial genes as the predictor variable and based on neural network algorithm, the overall survival time and immunotherapy efficacy (complete or partial response) of patients were predicted. RESULTS In total, 375 patients with OC were included to construct the prognostic model, and 26 patients were included to construct the immune efficacy model. The average area under the receiver operating characteristic curve of the prognostic model was 0.7268 [95% confidence interval (CI): 0.7258-0.7278] in the discovery cohort and 0.6475 (95%CI: 0.6466-0.6484) in the validation cohort. The average area under the receiver operating characteristic curve of the immunotherapy efficacy model was 0.9444 (95%CI: 0.8333-1.0000) in the discovery cohort and 0.9167 (95%CI: 0.6667-1.0000) in the validation cohort. CONCLUSION The application of mitochondrial genes and neural networks has the potential to predict prognosis and immunotherapy response in patients with OC, providing valuable insights into personalized treatment strategies.
Collapse
Affiliation(s)
- Zhi-Jian Tang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing 100044, China
| | - Yuan-Ming Pan
- Cancer Research Center, Beijing Chest Hospital, Beijing 101149, China
| | - Wei Li
- Cancer Research Center, Beijing Chest Hospital, Beijing 101149, China
- Department of Thoracic Surgery, Sichuan Provincial People's Hospital, Chengdu 610072, Sichuan Province, China
| | - Rui-Qiong Ma
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing 100044, China
| | - Jian-Liu Wang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing 100044, China
| |
Collapse
|
25
|
Hillmann J, Maass N, Bauerschlag DO, Flörkemeier I. Promising new drugs and therapeutic approaches for treatment of ovarian cancer-targeting the hallmarks of cancer. BMC Med 2025; 23:10. [PMID: 39762846 PMCID: PMC11706140 DOI: 10.1186/s12916-024-03826-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Ovarian cancer remains the most lethal gynecological malignancy. Despite the approval of promising targeted therapy such as bevacizumab and PARP inhibitors, 5-year survival has not improved significantly. Thus, there is an urgent need for new therapeutics. New advancements in therapeutic strategies target the pivotal hallmarks of cancer. This review is giving an updated overview of innovative and upcoming therapies for the treatment of ovarian cancer that focuses specific on the hallmarks of cancer. The hallmarks of cancer constitute a broad concept to reenact complexity of malignancies and furthermore identify possible targets for new treatment strategies. For this purpose, we analyzed approvals and current clinical phase III studies (registered at ClinicalTrials.gov (National Library of Medicine, National Institutes of Health; U.S. Department of Health and Human Services, 2024)) for new drugs on the basis of their mechanisms of action and identified new target approaches. A broad spectrum of new promising drugs is currently under investigation in clinical phase III studies targeting mainly the hallmarks "self-sufficiency in growth signals," "genomic instability," and "angiogenesis." The benefit of immune checkpoint inhibitors in ovarian cancer has been demonstrated for the first time. Besides, targeting the tumor microenvironment is of growing interest. Replicative immortality, energy metabolism, tumor promoting inflammation, and the microbiome of ovarian cancer are still barely targeted by drugs. Nevertheless, precision medicine, which focuses on specific disease characteristics, is becoming increasingly important in cancer treatment.
Collapse
Affiliation(s)
- Julia Hillmann
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Nicolai Maass
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany
| | - Dirk O Bauerschlag
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany.
- Department of Gynaecology, Jena University Hospital, Jena, Germany.
| | - Inken Flörkemeier
- Department of Gynaecology and Obstetrics, University and University Medical Center Schleswig-Holstein Campus Kiel, Kiel, Germany.
| |
Collapse
|
26
|
Nitecki Wilke R, Liu J, Westin SN, Fellman BM, Sims TT, Pham M, Rangel K, Sey E, Rauh-Hain JA, Lu KH, Sood AK, Fleming ND. The association of the chemotherapy response score and homologous recombination deficiency in patients undergoing interval tumor reductive surgery following neoadjuvant chemotherapy. Int J Gynecol Cancer 2025:ijgc-2024-005893. [PMID: 39414313 DOI: 10.1136/ijgc-2024-005893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2024] Open
Abstract
OBJECTIVES In patients undergoing interval tumor reductive surgery, a good response to neoadjuvant chemotherapy may limit available tumor for homologous recombination deficiency testing. The objective of this study was to assess whether the chemotherapy response score predicts homologous recombination status. METHODS We identified patients with advanced epithelial ovarian cancer (diagnosed January 2019 to 20 June 2023) who received neoadjuvant chemotherapy, underwent interval surgery, and for whom a chemotherapy response score was reported (1=no or minimal tumor response, 2=appreciable tumor response, 3=complete or near complete response with no residual tumor). Comparisons were made using ANOVAs or Kruskal-Wallis test for continuous variables and χ2 or Fisher's exact test for categorical variables. RESULTS The cohort consisted of 234 patients with advanced ovarian cancer who underwent interval surgery following neoadjuvant chemotherapy. Of those who underwent germline genetic testing, 22% (51/232) had a pathogenic BRCA1 or BRCA2 mutation and of those with tumors sent for testing, 65% were found to have homologous recombination deficiency (66/146). With increasing chemotherapy response scores, a higher likelihood of a complete gross resection was observed (50% (chemotherapy response score, CRS 1) vs 77% (CRS 2) vs 88% (CRS 3), p<0.001). On multivariable analysis, CRS 2 (adjusted odds ratio=3.28, 95% CI 1.12 to 9.60, p=0.03) and CRS 3 (5.83, 1.79 to 18.93, p=0.003) were independently associated with homologous recombination deficiency compared with CRS 1. CONCLUSION A positive response to chemotherapy at the time of interval tumor reductive surgery defined by the chemotherapy response score was associated with homologous recombination status and the likelihood of achieving a complete gross resection.
Collapse
Affiliation(s)
- Roni Nitecki Wilke
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jinsong Liu
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, Teaxs, USA
| | - Shannon Neville Westin
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Bryan M Fellman
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Travis T Sims
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Melissa Pham
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kelly Rangel
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Esther Sey
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jose Alejandro Rauh-Hain
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Karen H Lu
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nicole D Fleming
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
27
|
Tsilingiri K, Chalari A, Christopoulou G, Voutsina A, Constantoulakis P, Potaris Κ, Vamvakaris I, Hatzidaki D, Zachou G, Vatsellas G, Georgoulias V, Kotsakis A, Klinakis A. Genomic scarring score predicts the response to PARP inhibitors in non-small cell lung cancer. NPJ Precis Oncol 2024; 8:291. [PMID: 39725687 DOI: 10.1038/s41698-024-00777-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024] Open
Abstract
PARP inhibitors (PARPi) have shown efficacy in tumours harbouring mutations in homologous recombination repair (HRR) genes. Somatic HRR mutations have been described in patients with Non-Small Cell Lung Cancer (NSCLC), but PARP inhibitors (PARPi) are not yet a therapeutic option. Here we assessed the homologous recombination status of early-stage NSCLC and explored the therapeutic benefit of PARPi in preclinical models. The Genomic Scarring Score GSS (GSS) and HRR mutation profile of 136 patients were assessed. High GSS (h-GSS) was observed in 39 (28.7%) patients half of which carried pathogenic/likely pathogenic somatic HRR mutations. TP53 mutations were significantly enriched in h-GSS tumours (p < 0.001). Olaparib significantly delayed tumour growth in h-GSS but not l-GSS Patient-derived Xenografts (PDXs), while patients with h-GSS/TP53mut tumours respond favourably to adjuvant platinum-based chemotherapy. Our functional data clearly support the idea that the use of GSS rather than the mutational status of HRR genes could select patients for administration of PARPi.
Collapse
Affiliation(s)
| | - Anna Chalari
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Georgia Christopoulou
- Genotypos MSA, Private Molecular Biology and Cytogenetics Diagnostic Center, Athens, Greece
| | - Alexandra Voutsina
- Institute of Chemical Biology, National Hellenic Research Foundation, Athens, Greece
| | | | | | | | | | | | - Giannis Vatsellas
- Greek Genome Centre, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | | - Athanasios Kotsakis
- Department of Medical Oncology, University General Hospital of Larisa, Larisa, Greece
| | - Apostolos Klinakis
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
28
|
Li Z, Li N, Ndzie Noah ML, Shao Q, Zhan X. Pharmacoproteomics reveals energy metabolism pathways as therapeutic targets of ivermectin in ovarian cancer toward 3P medical approaches. EPMA J 2024; 15:711-737. [PMID: 39635022 PMCID: PMC11612093 DOI: 10.1007/s13167-024-00385-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024]
Abstract
Objective Ovarian cancer is the malignant tumor with the highest mortality rate in the female reproductive system, enormous socio-economic burden, and limited effective drug therapy. There is an urgent need to find novel effective drugs for ovarian cancer therapy. Our previous in vitro studies demonstrate that ivermectin effectively inhibits ovarian cancer cells and affects energy metabolism pathways. This study aims to clarify in vivo mechanisms and therapeutic targets of ivermectin in the treatment of ovarian cancer to establish predictive biomarkers, guide personalized treatments, and improve preventive strategies in the framework of 3P medicine. Methods A TOV-21G tumor-bearing mouse model was constructed based on histopathological data and biochemical parameters. TMT-based proteomic analysis was performed on tumor tissues from the different treatment groups. All significantly differentially abundant proteins were characterized by hierarchical clustering, Gene Ontology (GO) enrichment analyses, and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. In addition, the data were integrated and analyzed with the proteomic data of clinical ovarian cancer tissues from our previous study and the proteomic data of ivermectin intervention in ovarian cancer cells to identify key regulators of ivermectin. Results Ivermectin (10 mg/kg) had a significant anti-ovarian cancer effect in mice, with a tumor inhibitory rate of 61.5%. Molecular changes in tumor tissue of ivermectin-treated mice were established, and protein-protein interaction (PPI) analysis showed that the main differential pathway networks included the TCA cycle, propanoate metabolism, 2-0xocarboxyacid metabolism, and other pathways. Integrating our previous clinical ovarian cancer tissue and cell experimental data, this study found that ivermectin significantly interfered with the energy metabolic pathways of ovarian cancer, including glycolysis, TCA cycle, oxidative phosphorylation, and other related pathways. Conclusions This study evaluated the anti-ovarian cancer effect in vitro and in vivo, and its specific regulatory effect on energy metabolism. The expressions of drug target molecules in the energy metabolism pathway of ovarian cancer will be used to guide the diagnosis and prevention of ovarian cancer. The significant efficacy of ivermectin will be applied to the treatment of ovarian cancer and personalized medication. This has guiding significance for the clinical diagnosis, treatment, personalized medication, and prognosis evaluation of ovarian cancer. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-024-00385-1.
Collapse
Affiliation(s)
- Zhijun Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Na Li
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Marie Louise Ndzie Noah
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Qianwen Shao
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
| | - Xianquan Zhan
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong 250117 People’s Republic of China
- Shandong Provincial Key Medical and Health Laboratory of Ovarian Cancer Multiomics, & Jinan Key Laboratory of Cancer Multiomics, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 6699 Qingao Road, Jinan, Shandong 250117 People’s Republic of China
| |
Collapse
|
29
|
Kanbergs A, Rauh-Hain JA, Wilke RN. Differential Receipt of Genetic Services Among Patients With Gynecologic Cancer and Their Relatives: A Review of Challenges to Health Equity. Clin Obstet Gynecol 2024; 67:666-671. [PMID: 39331025 DOI: 10.1097/grf.0000000000000893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
Abstract
Up to 14% of endometrial cancers and 23% of epithelial ovarian cancers are associated with genetic predispositions. Referral for genetic testing and counseling can significantly impact a patient's oncologic outcomes. However, significant disparities in genetic referral and testing exist within medically underserved and minority populations in the United States. These disparities in care and access to care are multifactorial, often involving patient-level, health care-level, and system-level factors. In this review, we focus on disparities in genetic testing among patients with ovarian and uterine cancer, and the missed opportunities for primary cancer prevention among their relatives.
Collapse
Affiliation(s)
- Alexa Kanbergs
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | | |
Collapse
|
30
|
Ravindranathan R, Somuncu O, da Costa AABA, Mukkavalli S, Lamarre BP, Nguyen H, Grochala C, Jiao Y, Liu J, Kochupurakkal B, Parmar K, Shapiro GI, D’Andrea AD. PARG inhibitor sensitivity correlates with accumulation of single-stranded DNA gaps in preclinical models of ovarian cancer. Proc Natl Acad Sci U S A 2024; 121:e2413954121. [PMID: 39546575 PMCID: PMC11588084 DOI: 10.1073/pnas.2413954121] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/11/2024] [Indexed: 11/17/2024] Open
Abstract
Poly (ADP-ribose) glycohydrolase (PARG) is a dePARylating enzyme which promotes DNA repair by removal of poly (ADP-ribose) (PAR) from PARylated proteins. Loss or inhibition of PARG results in replication stress and sensitizes cancer cells to DNA-damaging agents. PARG inhibitors are now undergoing clinical development for patients having tumors with homologous recombination deficiency (HRD), such as cancer patients with germline or somatic BRCA1/2-mutations. PARP inhibitors kill BRCA-deficient cancer cells by increasing single-stranded DNA gaps (ssGAPs) during replication. Here, we report that, like PARP inhibitor (PARPi), PARG inhibitor (PARGi) treatment also causes an accumulation of ssGAPs in sensitive cells. PARGi exposure increased accumulation of S-phase-specific PAR, a marker for Okazaki fragment processing (OFP) defects on lagging strands and induced ssGAPs, in sensitive cells but not in resistant cells. PARGi also caused accumulation of PAR at the replication forks and at the ssDNA sites in sensitive cells. Additionally, PARGi exhibited monotherapy activity in specific HR-deficient, as well as HR-proficient, patient-derived, or patient-derived xenograft (PDX)-derived organoids of ovarian cancer, and drug sensitivity directly correlated with the accumulation of ssGAPs. Taken together, PARGi treatment results in toxic accumulation of PAR at replication forks resulting in ssGAPs due to OFP defects during replication. Regardless of the BRCA/HRD-status, the induction of ssGAPs in preclinical models of ovarian cancer cells correlates with PARGi sensitivity. Patient-derived organoids (PDOs) may be a useful model system for testing PARGi sensitivity and functional biomarkers.
Collapse
Affiliation(s)
- Ramya Ravindranathan
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Ozge Somuncu
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Alexandre André B. A. da Costa
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Sirisha Mukkavalli
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Benjamin P. Lamarre
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Huy Nguyen
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Carter Grochala
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Yuqing Jiao
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Joyce Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Bose Kochupurakkal
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Kalindi Parmar
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Geoffrey I. Shapiro
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| | - Alan D. D’Andrea
- Center for DNA Damage and Repair, Dana-Farber Cancer Institute, Boston, MA02215
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA02215
| |
Collapse
|
31
|
Huang R, Ji F, Huang L, Qin Y, Liang Z, Huang M, Li C, Ban J. Efficacy and safety of angiogenesis inhibitors combined with poly ADP ribose polymerase inhibitors in the maintenance treatment of advanced ovarian cancer: a meta-analysis. Front Oncol 2024; 14:1477105. [PMID: 39624625 PMCID: PMC11609078 DOI: 10.3389/fonc.2024.1477105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/31/2024] [Indexed: 01/03/2025] Open
Abstract
INTRODUCTION This meta-analysis was performed to evaluate the efficacy and safety of angiogenesis inhibitors (Ais) combined with poly ADP ribose polymerase inhibitors (PARPi) in the maintenance treatment of advanced ovarian cancer (OC). MATERIALS AND METHODS A systematic search was conducted in four databases (Pubmed, Embase, Web of Science, and Cochrane) for articles published from the inception of the databases until January 15, 2024. The focus of the search was on articles investigating the combination of Ais with PARPi in the maintenance treatment of ovarian cancer. Meta-analyses were conducted to assess the objective response rate (ORR), progression-free survival (PFS), overall survival (OS), and the risk of Grade ≥ 3 adverse events (Grade≥ 3 AEs). RESULTS Totally nine studies were included for meta-analysis. The overall pooled ORR of Ais combined with PARPi was 57% (95% CI, 35% to 77%). Subgroup analyses showed that the ORR for patients with platinum-resistant recurrent ovarian cancer, platinum-sensitive recurrent ovarian cancer and newly diagnosed advanced ovarian cancer were 30% (95% CI, 12% to 52%), 70% (95% CI, 61% to 78%) and 59% (95% CI, 55% to 63%), respectively. The median PFS was 5.8 months (95% CI, 5.3 to 7.1), 12.4 months (95% CI, 10.6 to 13.2) and 22.4 months (95% CI, 21.5 to 24.2), respectively. The median OS was 15.5 months (95% CI, 12.3 to 24.8), 40.8 months (95% CI, 33.4 to 45.2) and 56.3 months (95% CI, 49.0 to 62.0), respectively. The rate Grade≥ 3 TRAEs rate was found to be 0.22 (95% CI, 0.13 to 0.33). CONCLUSIONS Our results confirmed that PARPi plus Ais was a feasible and safe option for the maintenance treatment of advanced ovarian cancer. The combination therapy should be recommended as the first-line maintenance treatment for patients with advanced ovarian cancer. PARPi plus Ais yielded more favorable oncological prognosis for patients with platinum-sensitive recurrent ovarian cancer, compared to patients with platinum-resistant recurrent ovarian cancer. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42024543590, identifier CRD42024543590.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jian Ban
- The First Affiliated Hospital of Guangxi University of Science and Technology, Guangxi University of Science and Technology, Liuzhou, China
| |
Collapse
|
32
|
O'Neill K, Pleasance E, Fan J, Akbari V, Chang G, Dixon K, Csizmok V, MacLennan S, Porter V, Galbraith A, Grisdale CJ, Culibrk L, Dupuis JH, Corbett R, Hopkins J, Bowlby R, Pandoh P, Smailus DE, Cheng D, Wong T, Frey C, Shen Y, Lewis E, Paulin LF, Sedlazeck FJ, Nelson JMT, Chuah E, Mungall KL, Moore RA, Coope R, Mungall AJ, McConechy MK, Williamson LM, Schrader KA, Yip S, Marra MA, Laskin J, Jones SJM. Long-read sequencing of an advanced cancer cohort resolves rearrangements, unravels haplotypes, and reveals methylation landscapes. CELL GENOMICS 2024; 4:100674. [PMID: 39406235 PMCID: PMC11605692 DOI: 10.1016/j.xgen.2024.100674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/26/2024] [Accepted: 09/18/2024] [Indexed: 11/16/2024]
Abstract
The Long-Read Personalized OncoGenomics (POG) dataset comprises a cohort of 189 patient tumors and 41 matched normal samples sequenced using the Oxford Nanopore Technologies PromethION platform. This dataset from the POG program and the Marathon of Hope Cancer Centres Network includes DNA and RNA short-read sequence data, analytics, and clinical information. We show the potential of long-read sequencing for resolving complex cancer-related structural variants, viral integrations, and extrachromosomal circular DNA. Long-range phasing facilitates the discovery of allelically differentially methylated regions (aDMRs) and allele-specific expression, including recurrent aDMRs in the cancer genes RET and CDKN2A. Germline promoter methylation in MLH1 can be directly observed in Lynch syndrome. Promoter methylation in BRCA1 and RAD51C is a likely driver behind homologous recombination deficiency where no coding driver mutation was found. This dataset demonstrates applications for long-read sequencing in precision medicine and is available as a resource for developing analytical approaches using this technology.
Collapse
Affiliation(s)
- Kieran O'Neill
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Erin Pleasance
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Jeremy Fan
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Vahid Akbari
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Glenn Chang
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Katherine Dixon
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Veronika Csizmok
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Signe MacLennan
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Vanessa Porter
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Andrew Galbraith
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Cameron J Grisdale
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Luka Culibrk
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - John H Dupuis
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Richard Corbett
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - James Hopkins
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Reanne Bowlby
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Pawan Pandoh
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Duane E Smailus
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Dean Cheng
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Tina Wong
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Connor Frey
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Yaoqing Shen
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Eleanor Lewis
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Luis F Paulin
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Fritz J Sedlazeck
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX, USA
| | - Jessica M T Nelson
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Eric Chuah
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Karen L Mungall
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Richard A Moore
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Robin Coope
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Andrew J Mungall
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Melissa K McConechy
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Laura M Williamson
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada
| | - Kasmintan A Schrader
- Hereditary Cancer Program, BC Cancer, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Stephen Yip
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Marco A Marra
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada
| | - Janessa Laskin
- Department of Medical Oncology, BC Cancer, Vancouver, BC, Canada
| | - Steven J M Jones
- Canada's Michael Smith Genome Sciences Centre at BC Cancer, Vancouver, BC, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
33
|
Crymes A, Evans MG, Adeyelu T, Reid J, Ibe IO, Oberley MJ, Tseng JH. Case report: High grade serous fallopian tube carcinoma with rare NRG1 gene fusion presenting as widespread peritoneal carcinomatosis. Front Oncol 2024; 14:1472725. [PMID: 39575425 PMCID: PMC11580015 DOI: 10.3389/fonc.2024.1472725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/14/2024] [Indexed: 11/24/2024] Open
Abstract
The discovery of gene fusions involving Neuregulin-1 (NRG1) within solid tumors has important therapeutic implications, as they are being actively explored as targets for emerging ERBB/ERBB2/ERBB3-directed anti-cancer agents. NRG1 fusions are very uncommon across all tumor types and are infrequently documented in the medical literature. We report a female patient presenting with widespread peritoneal carcinomatosis diagnosed as high grade serous fallopian tube carcinoma, which harbored a previously undescribed MYH10::NRG1 fusion. Moreover, we queried the whole transcriptome sequencing results of neoplasms analyzed by a commercial laboratory (Caris Life Sciences) to determine the overall incidence of NRG1 fusions in carcinomas of the ovary, fallopian tube, and peritoneum (0.18%). Twenty-five additional tumors were found to demonstrate NRG1 fusions, including 20 new genes partners that had not been previously identified in gynecologic carcinomas. Overall, NRG1 fusion events are rare in ovarian, fallopian tube, and primary peritoneal carcinomas, but they may carry diagnostic significance in the context of borderline/low grade serous tumors, which demonstrated exclusively CLU::NRG1 fusions, and could have important predictive implications for response to ERBB/ERBB2/ERBB3-directed therapies.
Collapse
Affiliation(s)
- Anthony Crymes
- Department of Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Mark G. Evans
- Department of Pathology, Caris Life Sciences, Phoenix, AZ, United States
| | - Tolulope Adeyelu
- Department of Clinical and Translational Research, Caris Life Sciences, Phoenix, AZ, United States
| | - Jack Reid
- Department of Pathology, City of Hope National Cancer Center, Duarte, CA, United States
| | - Ifegwu O. Ibe
- Department of Pathology, Affiliated Pathologists Medical Group, Orange, CA, United States
| | - Matthew J. Oberley
- Department of Pathology, Caris Life Sciences, Phoenix, AZ, United States
| | - Jill H. Tseng
- Department of Obstetrics and Gynecology, University of California Irvine School of Medicine, Irvine, CA, United States
| |
Collapse
|
34
|
Bhatia T, Doshi G, Godad A. PARP inhibitors in ovarian cancer: Mechanisms, resistance, and the promise of combination therapy. Pathol Res Pract 2024; 263:155617. [PMID: 39357181 DOI: 10.1016/j.prp.2024.155617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/06/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
Current approaches to treating ovarian cancer focus mainly on surgical cytoreduction and chemotherapy using platinum-based drugs, while newer methods such as immunotherapy are being investigated to enhance treatment outcomes. Treating ovarian cancer is complicated by challenges such as late-stage detection, tumor diversity, and limited treatment choices. Therefore, innovative strategies such as precision medicine and targeted therapies like PARPi (Poly ADP-Ribose Polymerase inhibitors) are increasingly necessary. The article highlights the significance of an innovative therapeutic approach focusing on PARPi in revolutionizing ovarian cancer treatment and improving patient outcomes. It covers the basic knowledge of PARP, its structure, and its function in DNA repair. It further emphasizes how inhibiting PARP can help in treating ovarian cancer. It elaborates on the mechanism of action of PARPi. It covers the clinical trials governing PARPi and the combination of drugs used with PARPi. It mentions how the resistance is developed to PARPi and the strategies to overcome the resistance developed.
Collapse
Affiliation(s)
- Tejas Bhatia
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai 400056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai 400056, India
| | - Angel Godad
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, V L M Road, Vile Parle (w), Mumbai 400056, India.
| |
Collapse
|
35
|
Uchikura E, Fukuda T, Sengiku T, Noda T, Awazu Y, Wada T, Tasaka R, Yamauchi M, Yasui T, Sumi T. Role of Fyn expression in predicting the sensitivity to platinum‑based chemotherapy in patients with ovarian serous carcinoma. Oncol Lett 2024; 28:525. [PMID: 39268168 PMCID: PMC11391251 DOI: 10.3892/ol.2024.14658] [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: 07/23/2024] [Accepted: 08/15/2024] [Indexed: 09/15/2024] Open
Abstract
Ovarian serous carcinoma is a gynecological malignancy associated with a high mortality rate, which is commonly diagnosed in the first instance at a late stage and has a propensity to develop resistance to platinum-based chemotherapy. Identifying reliable biomarkers for platinum sensitivity is critical for improving patient outcomes. The present retrospective study included 64 patients with high-grade serous ovarian carcinoma (Federation of Gynecology and Obstetrics stages III or IV). Patients were classified as platinum-sensitive (no relapse within 6 months of the last platinum administration) or platinum-resistant (relapse within 6 months). Immunohistochemical analysis was performed to evaluate Fyn expression in tumor tissues, and Fyn knockdown experiments were performed using the OVSAHO ovarian cancer cell line to assess carboplatin sensitivity. Fyn expression was significantly higher in platinum-resistant patients compared with in platinum-sensitive patients (P<0.01). A weighted Fyn expression score was developed and a cutoff score of 6 was determined to predict platinum sensitivity with a specificity of 65.5% and a sensitivity of 62.9%. Patients with low Fyn expression (score ≤6) exhibited higher platinum sensitivity and longer overall survival (P<0.05). Multivariate analysis identified Fyn expression and postoperative residual tumor size as independent predictors of platinum sensitivity (P=0.033 and P=0.023, respectively). In vitro, Fyn knockdown significantly increased carboplatin sensitivity in ovarian cancer cells (P<0.05). Fyn, a member of the Src family of kinases, serves a crucial role in various cellular functions and has been implicated in chemotherapy resistance. The results demonstrated a notable association between Fyn expression and platinum sensitivity in ovarian serous carcinoma. The findings suggested that Fyn may serve as a predictive biomarker for response to platinum-based chemotherapy, offering the potential for more personalized treatment strategies. To the best of our knowledge, the present study is the first to establish an association between Fyn expression and platinum sensitivity in advanced ovarian serous carcinoma. Prospective studies with larger, multi-center cohorts and comprehensive biomarker analyses are recommended to validate and extend these results, ultimately improving therapeutic strategies and patient prognosis.
Collapse
Affiliation(s)
- Eijiro Uchikura
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, Osaka 5454-8585, Japan
| | - Takeshi Fukuda
- Department of Obstetrics and Gynecology, Osaka Metropolitan University Graduate School of Medicine, Osaka 5454-8585, Japan
| | - Tomoki Sengiku
- Department of Obstetrics and Gynecology, Osaka Metropolitan University Graduate School of Medicine, Osaka 5454-8585, Japan
| | - Takuya Noda
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, Osaka 5454-8585, Japan
| | - Yuichiro Awazu
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, Osaka 5454-8585, Japan
| | - Takuma Wada
- Department of Obstetrics and Gynecology, Osaka Metropolitan University Graduate School of Medicine, Osaka 5454-8585, Japan
| | - Reiko Tasaka
- Department of Obstetrics and Gynecology, Osaka Metropolitan University Graduate School of Medicine, Osaka 5454-8585, Japan
| | - Makoto Yamauchi
- Department of Obstetrics and Gynecology, Osaka Metropolitan University Graduate School of Medicine, Osaka 5454-8585, Japan
| | - Tomoyo Yasui
- Department of Obstetrics and Gynecology, Osaka Metropolitan University Graduate School of Medicine, Osaka 5454-8585, Japan
| | - Toshiyuki Sumi
- Department of Obstetrics and Gynecology, Osaka Metropolitan University Graduate School of Medicine, Osaka 5454-8585, Japan
| |
Collapse
|
36
|
Ren X, Sun P, Wang Y. PARP inhibitor-related acute renal failure: a real-world study based on the FDA adverse event reporting system database. Expert Opin Drug Saf 2024; 23:1463-1471. [PMID: 38967020 DOI: 10.1080/14740338.2024.2376690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/22/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Current clinical trial data on PARP inhibitors (PARPis)-related acute renal failure (ARF) are not entirely representative of real-world situations. Therefore, in this study, the US Food and Drug Administration Adverse Event Reporting System (FAERS) was used to evaluate PARPis-related ARF. RESEARCH DESIGN AND METHODS Data were obtained from 1 January 2015, to 30 September 2023. ARF event reports were analyzed based on four algorithms. The time-to-onset (TTO) and clinical outcomes of PARPis-associated ARF were assessed. RESULTS The total included cases were 2726. Significant signals were observed for olaparib, niraparib, and rucaparib (reporting odds ratio (ROR): 1.62, 95% confidence interval (CI): 1.49-1.78, 1.25, 95% CI: 1.19-1.32 and 1.59, 95% CI: 1.47-1.72 respectively). The median TTO of ARF onset was 57, 36, and 85 days for olaparib, niraparib, and rucaparib, respectively. The proportion of deaths with olaparib (9.88%) was significantly higher than for niraparib (2.52%) and rucaparib (2.94%) (p < 0.005). The proportion of life-threatening adverse events associated with niraparib (4.89%) was significantly higher than for rucaparib (0.98%) (p < 0.005). CONCLUSIONS ARF and PARPi were related, with the exception of talazoparib. More emphasis should be given to PARPis-related ARF due to the high proportion of serious AEs and delayed adverse reactions.
Collapse
Affiliation(s)
- Xiayang Ren
- Department of Pharmacy, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ping Sun
- Department of Cancer Prevention, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanfeng Wang
- Department of Comprehensive Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
37
|
Zhou Y, Xu J. Impact of PARP inhibitors on progression-free survival in platinum-sensitive recurrent epithelial ovarian cancer: a retrospective analysis. World J Surg Oncol 2024; 22:276. [PMID: 39434111 PMCID: PMC11492665 DOI: 10.1186/s12957-024-03562-8] [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: 06/10/2024] [Accepted: 10/16/2024] [Indexed: 10/23/2024] Open
Abstract
OBJECTIVE Poly (ADP-ribose) polymerase (PARP) inhibitors such as olaparib and niraparib have shown promise in extending progression-free survival (PFS) in patients with platinum-sensitive recurrent (PSR) epithelial ovarian cancer. In this retrospective study, we aimed to present our own data on the effect of PARP inhibitors on PFS in recurrent epithelial ovarian cancer. METHODS 82 patients diagnosed with PSR epithelial ovarian, tubal, or primary peritoneal cancer between May 2017 and September 2023 were initially enrolled from our hospital. However, 16 patients had prior exposure to PARP inhibitors during primary treatment, and 11 were lost to follow-up. Consequently, the study focused on 55 eligible patients. PFS was compared between patients receiving PARP inhibitor maintenance therapy and those who did not. RESULTS Among the 55 patients with PSR epithelial ovarian cancer, 18 received olaparib as maintenance therapy, 19 received niraparib, and 18 opted for observation. PARP inhibitor therapy significantly extended PFS (mean 24.0 months) compared to observation (mean 9.0 months, p = 0.0005), regardless of BRCA mutation status (HR = 0.20, 95% CI: 0.08-0.50). Subgroup analysis showed no statistical difference between olaparib and niraparib. Additionally, there was no PFS difference based on BRCA mutation status within both PARP inhibitor groups. CONCLUSION Our retrospective study demonstrates that PARP inhibitor maintenance therapy, including olaparib and niraparib, significantly prolongs PFS in patients with PSR epithelial ovarian, tubal, or primary peritoneal cancer, These findings support the broad utilization of PARP inhibitors as a standard maintenance therapy for PSR epithelial ovarian cancer irrespective of BRCA mutation status.
Collapse
Affiliation(s)
- Yumei Zhou
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China
| | - Junfen Xu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
38
|
Stiff PJ, Kertowidjojo E, Potkul RK, Banerjee S, Mehrotra S, Small W, Stack MS, Drakes ML. Cabozantinib inhibits tumor growth in mice with ovarian cancer. Am J Cancer Res 2024; 14:4788-4802. [PMID: 39553221 PMCID: PMC11560812 DOI: 10.62347/zswv1767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 08/07/2024] [Indexed: 11/19/2024] Open
Abstract
Ovarian cancer is usually detected in the advanced stages. Existing treatments for high grade serous ovarian cancer (HGSOC) are not adequate and approximately fifty percent of patients succumb to this disease and die within five years after diagnosis. We conducted pre-clinical studies in a mouse model of ovarian cancer to evaluate disease outcome in response to treatment with the multi-kinase inhibitor cabozantinib. Cabozantinib is a receptor tyrosine kinase inhibitor with multiple targets including vascular endothelial growth factor receptor-2 (VEGFR-2), associated with immune suppression in ovarian cancer. Mice (C57BL/6) were injected with ID8-RFP ovarian tumor cells and treated with cabozantinib. Studies investigated ascites development, tumor burden and regulation of anti-tumor immunity with treatment. Mice treated with cabozantinib had significantly decreased solid tumor burden and decreased malignant ascites as compared to untreated controls. Improved outcome in cabozantinib treated mice was associated with a significantly higher percentage of CD69 early activated T cells, a higher percentage of granzyme B secreting CD8 T cells, the enhanced release of cytokines and chemokines known to recruit CD8 T cells and amplify T cell function, as well as reduced VEGFR-2. Findings suggest that cabozantinib is an important clinical agent capable of improving ovarian cancer in mice potentially in part by priming the autologous immune system to promote anti-tumor immunity.
Collapse
Affiliation(s)
- Patrick J Stiff
- Department of Medicine, Cardinal Bernardin Cancer Center, Loyola University ChicagoMaywood, IL 60153, USA
| | | | - Ronald K Potkul
- Department of Obstetrics and Gynecology, Cardinal Bernardin Cancer Center, Loyola University ChicagoMaywood, IL 60153, USA
| | - Swarnali Banerjee
- Center for Data Science and Consulting, and Department of Mathematics and Statistics, Loyola University ChicagoChicago, IL 60660, USA
| | - Swati Mehrotra
- Department of Pathology, Edward Hines Jr. VA HospitalHines, IL 60141, USA
| | - William Small
- Department of Radiation Oncology, Cardinal Bernardin Cancer Center, Loyola University ChicagoMaywood, IL 60153, USA
| | - M Sharon Stack
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre DameSouth Bend, IN 46617, USA
| | - Maureen L Drakes
- Department of Medicine, Cardinal Bernardin Cancer Center, Loyola University ChicagoMaywood, IL 60153, USA
| |
Collapse
|
39
|
Chaudhary U, Kumar P, Sharma P, Chikara A, Barua A, Mahiya K, Adhikari Subin J, Nath Yadav P, Raj Pokharel Y. Synthesis of 5-hydroxyisatin thiosemicarbazones, spectroscopic investigation, protein-ligand docking, and in vitro anticancer activity. Bioorg Chem 2024; 153:107872. [PMID: 39442462 DOI: 10.1016/j.bioorg.2024.107872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024]
Abstract
A series of novel modifications were performed at the N(4) position of 5-hydroxyisatin thiosemicarbazone (TSC). The structure-activity approach is applied to design and synthesize derivatives by condensing thiosemicarbazides with 5-hydroxy isatin. The TSCs were characterized by various spectroscopic techniques viz. FTIR, 1H NMR, 13C NMR, UV-Vis, HRMS data, CHN elemental analysis, and single crystal X-ray diffraction. Biological evaluation of the synthesized compounds revealed the anticancer potency of the TSC analogues against breast cancer (MD-AMD-231, MCF-7), lung cancer (A549, NCI-H460), prostate cancer (PC3), and skin cancer (A431). The molecules, L2, L3, and L6 showed activity in the micromolar range (IC50; 0.19-2.19 μM). L6 exhibited the highest potency against skin cancer A431 cell line, with an IC50 of 0.19 μM compared to 1.8 μM with triapine and showed low toxicity against PNT-2 cells with an SI index of >100 μM. The mechanistic study revealed that L6 inhibited cancer cell proliferation, colony formation, and 3-dimensional spheroid formation by targeting the Ras/MAPK axis. It induced DNA damage and impaired DNA damage repair machinery, which led to the accumulation of DSB. Also, it lowered the ERK1/2 expression, which affected the caspase 3 activity and showed higher binding affinity compared to the FDA-approved drug Lenalidomide in molecular docking studies. Our findings demonstrated the possible future anticancer drug potency of L6 in the skin cancer A431 cells.
Collapse
Affiliation(s)
- Upendra Chaudhary
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Piyush Kumar
- Faculty of Life Science and Biotechnology, South Asian University, Rajpur Rd, Maidan Garhi, New Delhi 110068, India
| | - Pratibha Sharma
- Faculty of Life Science and Biotechnology, South Asian University, Rajpur Rd, Maidan Garhi, New Delhi 110068, India
| | - Anshul Chikara
- Faculty of Life Science and Biotechnology, South Asian University, Rajpur Rd, Maidan Garhi, New Delhi 110068, India
| | - Ayanti Barua
- Faculty of Life Science and Biotechnology, South Asian University, Rajpur Rd, Maidan Garhi, New Delhi 110068, India
| | - Kuldeep Mahiya
- Department of Chemistry, F G M Government College, Adampur, Mandi Adampur, Hisar 125052, Haryana, India
| | - Jhashanath Adhikari Subin
- Scientific Research and Training Nepal P. Ltd., Bioinformatics and Cheminformatics Division, Kaushaltar, Bhaktapur, Nepal
| | - Paras Nath Yadav
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal.
| | - Yuba Raj Pokharel
- Faculty of Life Science and Biotechnology, South Asian University, Rajpur Rd, Maidan Garhi, New Delhi 110068, India.
| |
Collapse
|
40
|
Bondar D, Karpichev Y. Poly(ADP-Ribose) Polymerase (PARP) Inhibitors for Cancer Therapy: Advances, Challenges, and Future Directions. Biomolecules 2024; 14:1269. [PMID: 39456202 PMCID: PMC11506039 DOI: 10.3390/biom14101269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Poly(ADP-ribose) polymerases (PARPs) are crucial nuclear proteins that play important roles in various cellular processes, including DNA repair, gene transcription, and cell death. Among the 17 identified PARP family members, PARP1 is the most abundant enzyme, with approximately 1-2 million molecules per cell, acting primarily as a DNA damage sensor. It has become a promising biological target for anticancer drug studies. Enhanced PARP expression is present in several types of tumors, such as melanomas, lung cancers, and breast tumors, correlating with low survival outcomes and resistance to treatment. PARP inhibitors, especially newly developed third-generation inhibitors currently undergoing Phase II clinical trials, have shown efficacy as anticancer agents both as single drugs and as sensitizers for chemo- and radiotherapy. This review explores the properties, characteristics, and challenges of PARP inhibitors, discussing their development from first-generation to third-generation compounds, more sustainable synthesis methods for discovery of new anti-cancer agents, their mechanisms of therapeutic action, and their potential for targeting additional biological targets beyond the catalytic active site of PARP proteins. Perspectives on green chemistry methods in the synthesis of new anticancer agents are also discussed.
Collapse
Affiliation(s)
| | - Yevgen Karpichev
- Department of Chemistry and Biotechnology, Tallinn University of Technology (TalTech), Akadeemia tee 15, 12618 Tallinn, Estonia;
| |
Collapse
|
41
|
Wu J, Wang X, Yao Y, Du N, Duan L, Gong P. Design, synthesis and antitumor activities of phthalazinone derivatives as PARP-1 inhibitors and PARP-1/HDAC-1 inhibitors. Bioorg Chem 2024; 151:107556. [PMID: 39068717 DOI: 10.1016/j.bioorg.2024.107556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 07/30/2024]
Abstract
In recent years, poly(ADP-ribose)polymerase-1 (PARP-1) and histone deacetylase (HDAC) have emerged as significant targets in tumor therapy, garnering widespread attention. In this study, we designed and synthesized two novel phthalazinone PARP-1 inhibitors and dual PARP-1/HDAC-1 inhibitors, named DLC-1-46 containing dithiocarboxylate fragments and DLC-47-63 containing hydroxamic acid fragments, and evaluated their inhibitory activities on enzymes and cells. Among the PARP-1 inhibitors, most compounds exhibited high inhibitory activity against the PARP-1 enzyme, with DLC-1-6 being particularly notable, showing IC50 values <0.2 nM. Notably, DLC-1 demonstrated significant anti-proliferative activity, with IC50 values for inhibiting the proliferation of MDA-MB-436, MDA-MB-231, and MCF-7 cells reaching 0.08, 26.39, and 1.01 μM, respectively. Further investigation revealed that DLC-1 arrested MDA-MB-231 cells in the G1 phase and induced apoptosis in a dose-dependent manner. Among the designed dual PARP-1/HDAC-1 inhibitors, several compounds exhibited potent dual-target inhibitory activity, with DLC-49 displaying IC50 values of 0.53 nM and 17 nM for PARP-1 and HDAC-1, respectively. DLC-50 demonstrated the most potent anti-proliferative activity, with IC50 values for inhibiting the proliferation of MDA-MB-436, MDA-MB-231, and MCF-7 cells at 0.30, 2.70, and 2.41 μM, respectively. Cell cycle arrest and apoptosis assays indicated that DLC-50 arrested the cell cycle in the G2 phase and induced apoptosis in HCT-116 cells. Our findings present a novel avenue for further exploration of PARP-1 inhibitors and dual PARP-1/HDAC-1 inhibitors.
Collapse
Affiliation(s)
- Jie Wu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Xiaoqian Wang
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Yaning Yao
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Nan Du
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China
| | - Liancheng Duan
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| | - Ping Gong
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang 110016, China.
| |
Collapse
|
42
|
Şeker Karatoprak G, Dumlupınar B, Celep E, Kurt Celep I, Küpeli Akkol E, Sobarzo-Sánchez E. A comprehensive review on the potential of coumarin and related derivatives as multi-target therapeutic agents in the management of gynecological cancers. Front Pharmacol 2024; 15:1423480. [PMID: 39364049 PMCID: PMC11447453 DOI: 10.3389/fphar.2024.1423480] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/16/2024] [Indexed: 10/05/2024] Open
Abstract
Current treatments for gynecological cancers include surgery, radiotherapy, and chemotherapy. However, these treatments often have significant side effects. Phytochemicals, natural compounds derived from plants, offer promising anticancer properties. Coumarins, a class of benzopyrone compounds found in various plants like tonka beans, exhibit notable antitumor effects. These compounds induce cell apoptosis, target PI3K/Akt/mTOR signaling pathways, inhibit carbonic anhydrase, and disrupt microtubules. Additionally, they inhibit tumor multidrug resistance and angiogenesis and regulate reactive oxygen species. Specific coumarin derivatives, such as auraptene, praeruptorin, osthole, and scopoletin, show anti-invasive, anti-migratory, and antiproliferative activities by arresting the cell cycle and inducing apoptosis. They also inhibit metalloproteinases-2 and -9, reducing tumor cell migration, invasion, and metastasis. These compounds can sensitize tumor cells to radiotherapy and chemotherapy. Synthetic coumarin derivatives also demonstrate potent antitumor and anticancer activities with minimal side effects. Given their diverse mechanisms of action and minimal side effects, coumarin-class phytochemicals hold significant potential as therapeutic agents in gynecological cancers, potentially improving treatment outcomes and reducing side effects. This review will aid in the synthesis and development of novel coumarin-based drugs for these cancers.
Collapse
Affiliation(s)
| | - Berrak Dumlupınar
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Istanbul Okan University, İstanbul, Türkiye
| | - Engin Celep
- Department of Pharmacognosy, Faculty of Pharmacy, Acıbadem Mehmet Ali Aydinlar University, Atasehir, Istanbul, Türkiye
| | - Inci Kurt Celep
- Department of Biotechnology, Faculty of Pharmacy, Istanbul Okan University, Istanbul, Türkiye
| | - Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara, Türkiye
| | - Eduardo Sobarzo-Sánchez
- Instituto de Investigación y Postgrado Facultad de Ciencias de la Salud Universidad Central de Chile, Santiago, Chile
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
43
|
Chen M, Lei N, Guo R, Han L, Zhao Q, Zhao Y, Qiu L, Wu F, Jiang S, Tong N, Wang K, Li S, Chang L. Genetic landscape of homologous recombination repair and practical outcomes of PARPi therapy in ovarian cancer management. Ther Adv Med Oncol 2024; 16:17588359241271845. [PMID: 39246808 PMCID: PMC11378221 DOI: 10.1177/17588359241271845] [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: 03/28/2024] [Accepted: 07/02/2024] [Indexed: 09/10/2024] Open
Abstract
Background Genetic studies of ovarian cancer (OC) have historically focused on BRCA1/2 mutations, lacking other studies of homologous recombination repair (HRR). Poly (ADP-ribose) polymerase inhibitors (PARPi) exploit synthetic lethality to significantly improve OC treatment outcomes, especially in BRCA1/2 deficiency patients. Objectives Our study aims to construct a mutation map of HRR genes in OC and identify factors influencing the efficacy of PARPi. Design A retrospective observational analysis of HRR gene variation data from 695 OC patients from March 2019 to February 2022 was performed. Methods The HRR gene variation data of 695 OC patients who underwent next-generation sequencing (NGS) in the First Affiliated Hospital of Zhengzhou University were retrospectively collected. Clinical data on the use of PARPi in these patients were also gathered to identify factors that may interfere with the efficacy of PARPi. Results Out of 127 pathogenic variants in the BRCA1/2 genes, 104 (81.9%) were BRCA1 mutations, and 23 (18.1%) were BRCA2 mutations. Among the 59 variants of uncertain significance (VUS), 20 (33.9%) were BRCA1, while 39 (66.1%) were BRCA2 mutations. In addition to BRCA1/2, HRR gene results showed that 9 (69%) of 13 were HRR pathway pathogenic variants; and 16 (1.7%) of 116 VUS were Food and Drug Administration (FDA)-approved mutated HRR genes. Notably, the treatment regimen significantly influenced the effectiveness of PARPi, especially when using first-line maintenance therapy, leading to enhanced progression-free survival (PFS) compared to alternative protocols. Conclusion Focusing on HRR gene mutations and supporting clinical research about PARPi in OC patients is crucial for developing precision treatment strategies and enhancing prognosis.
Collapse
Affiliation(s)
- Mengyu Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ningjing Lei
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruixia Guo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liping Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qinghe Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Zhao
- The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Luojie Qiu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fengling Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shan Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ningyao Tong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kunmei Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Siyu Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lei Chang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, No. 1 East Jianshe Road, Erqi District, Zhengzhou, Henan 450000, China
| |
Collapse
|
44
|
DE Morais GP, Machado CB, Dias Nogueira BM, DE Pinho Pessoa FMC, DE Sousa Oliveira D, Ribeiro RM, DA Silva JBS, Seabra AD, Mello Júnior FAR, Burbano RR, Khayat AS, DE Moraes Filho MO, DE Moraes MEA, Moreira-Nunes CA. Association of PARP1 Expression Levels and Clinical Parameters in Different Leukemic Subtypes With BCR::ABL1 p190+ Translocation. CANCER DIAGNOSIS & PROGNOSIS 2024; 4:592-598. [PMID: 39238631 PMCID: PMC11372699 DOI: 10.21873/cdp.10368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 06/21/2024] [Accepted: 07/12/2024] [Indexed: 09/07/2024]
Abstract
Background/Aim Although the reciprocal translocation t(9;22)(q34;q11) is a hallmark of chronic myeloid leukemia (CML), it is also present in acute lymphoblastic leukemia (ALL) and acute myeloid leukemia (AML). Depending on the gene's breakpoint, it is possible to obtain three isoforms, among which p190 stands out for the poor prognosis it induces whenever it appears. Due to the genomic instability induced by BCR::ABL1, it is proposed to expand the applicability of poly-ADP-ribose polymerase-1 (PARP1) and its inhibitors in hematological neoplasms. Materials and Methods We measured the expression levels of PARP1 by quantitative real-time PCR (qPCR) using TaqMan®, correlating its expression with BCR::ABL1 p190+, to evaluate its influence in the clinic of adult patients. Results We found that PARP1 is expressed differently in ALL, AML and CML and that p190 transcripts do not follow a linear pattern in these populations. We also found that PARP1 expression is not correlated with age, white blood cell and the amount of p190 transcripts. Conclusion Despite the lack of statistical correlation between the variables analyzed, the role of PARP1 in BCR::ABL1 leukemia cannot be ruled out, given the instability profile promoted by this translocation. Finally, further studies involving a larger sample of patients are needed, as well as investigations into other molecular pathways that may impact on the pathogenesis of different BCR::ABL1 leukemic subtypes.
Collapse
Affiliation(s)
| | - Caio Bezerra Machado
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Beatriz Maria Dias Nogueira
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Flávia Melo Cunha DE Pinho Pessoa
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | | | | | | | | | | | - Rommel Rodriguez Burbano
- Molecular Biology Laboratory, Ophir Loyola Hospital, Belem, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belem, Brazil
| | - André Salim Khayat
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belem, Brazil
| | - Manoel Odorico DE Moraes Filho
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | | | - Caroline Aquino Moreira-Nunes
- Unichristus University Center, Faculty of Biomedicine, Fortaleza, Brazil
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belem, Brazil
- Clementino Fraga Group, Central Unity, Molecular Biology Laboratory, Fortaleza, Brazil
| |
Collapse
|
45
|
Ribeiro R, Carvalho FM, Baiocchi G, Guindalini RSC, da Cunha JR, Anjos CHD, de Nadai Costa C, Gifoni ACLVC, Neto RC, Cagnacci AQC, Carneiro VCG, Calabrich A, Moretti-Marques R, Pinheiro RN, de Castro Ribeiro HS. Guidelines of the Brazilian Society of Surgical Oncology for anatomopathological, immunohistochemical, and molecular testing in female tumors. J Surg Oncol 2024; 130:882-895. [PMID: 39038206 DOI: 10.1002/jso.27717] [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: 05/01/2024] [Accepted: 05/04/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Precision medicine has revolutionized oncology, providing more personalized diagnosis, treatment, and monitoring for patients with cancer. In the context of female-specific tumors, such as breast, ovarian, endometrial, and cervical cancer, proper tissue collection and handling are essential for obtaining tissue, immunohistochemical (IHC), and molecular data to guide therapeutic decisions. OBJECTIVES To establish guidelines for the collection and handling of tumor tissue, to enhance the quality of samples for histopathological, IHC, genomic, and molecular analyses. These guidelines are fundamental in informing therapeutic decisions in cancer treatment. METHOD The guidelines were developed by a multidisciplinary panel of renowned specialists between June 12, 2013 and February 12, 2024. Initially, the panel deliberated on critical and controversial topics related to conducting precision medicine studies focusing on female tumors. Subsequently, 22 pivotal topics were identified within the framework and assigned to groups. These groups reviewed relevant literature and drafted preliminary recommendations. Following this, the recommendations were reviewed by the coordinators and received unanimous approval. Finally, the groups made the final adjustments, classified the level of evidence, and ranked the recommendations. CONCLUSION The collection of surgical samples requires minimum quality standards to enable histopathological, IHC, genomic, and molecular analyses. These analyses provide crucial data for informing therapeutic decisions, significantly impacting potential survival gains for patients with female tumors.
Collapse
Affiliation(s)
- Reitan Ribeiro
- Department of Gynecology Oncology, Erasto Gaertner Hospital, Curitiba, Paraná, Brazil
| | - Filomena Marino Carvalho
- Department of Pathology, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Glauco Baiocchi
- Department of Gynecologic Oncology, AC Camargo Cancer Center , São Paulo, São Paulo, Brazil
| | | | | | | | | | | | - Renato Cagnacci Neto
- Department of Mastology, Breast Cancer Reference Center, AC Camargo Cancer, CenterSão Paulo, São Paulo, Brazil
| | - Allyne Queiroz Carneiro Cagnacci
- Department of Oncology, Oncology Center, Hospital Alemão Oswaldo Cruz, São Paulo, São Paulo, Brazil
- Hereditary Cancer Department, Instituto do Câncer do Estado de São Paulo (ICESPSP), São Paulo, São Paulo, Brazil
| | - Vandré Cabral Gomes Carneiro
- Department of Gynecology Oncology, Instituto de Medicina Integral Professor Fernando Figueira (IMIP), Recife, Pernambuco, Brazil
- Research Department, Hospital de Câncer de Pernambuco, Recife, Brazil
- Department of Oncogenetic, Oncologia D'OR, Recife, Pernambuco, Brazil
| | - Aknar Calabrich
- Department of Oncology, Clínica AMO/DASA, Salvador, Bahia, Brazil
| | - Renato Moretti-Marques
- Department of Oncology, Albert Einstein Israelite Hospital, São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
46
|
Del Bufalo D, Damia G. Overview of BH3 mimetics in ovarian cancer. Cancer Treat Rev 2024; 129:102771. [PMID: 38875743 DOI: 10.1016/j.ctrv.2024.102771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/09/2024] [Accepted: 05/21/2024] [Indexed: 06/16/2024]
Abstract
Ovarian carcinoma is the leading cause of gynecological cancer-related death, still with a dismal five-year prognosis, mainly due to late diagnosis and the emergence of resistance to cytotoxic and targeted agents. Bcl-2 family proteins have a key role in apoptosis and are associated with tumor development/progression and response to therapy in different cancer types, including ovarian carcinoma. In tumors, evasion of apoptosis is a possible mechanism of resistance to therapy. BH3 mimetics are small molecules that occupy the hydrophobic pocket on pro-survival proteins, allowing the induction of apoptosis, and are currently under study as single agents and/or in combination with cytotoxic and targeted agents in solid tumors. Here, we discuss recent advances in targeting anti-apoptotic proteins of the Bcl-2 family for the treatment of ovarian cancer, focusing on BH3 mimetics, and how these approaches could potentially offer an alternative/complementary way to treat patients and overcome or delay resistance to current treatments.
Collapse
Affiliation(s)
- Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144 Rome, Italy.
| | - Giovanna Damia
- Laboratory of Gynecological Preclinical Oncology, Experimental Oncology Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via M. Negri 2, 20156 Milan, Italy.
| |
Collapse
|
47
|
Frederick MI, Abdesselam D, Clouvel A, Croteau L, Hassan S. Leveraging PARP-1/2 to Target Distant Metastasis. Int J Mol Sci 2024; 25:9032. [PMID: 39201718 PMCID: PMC11354653 DOI: 10.3390/ijms25169032] [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: 07/22/2024] [Revised: 08/14/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Poly (ADP-Ribose) Polymerase (PARP) inhibitors have changed the outcomes and therapeutic strategy for several cancer types. As a targeted therapeutic mainly for patients with BRCA1/2 mutations, PARP inhibitors have commonly been exploited for their capacity to prevent DNA repair. In this review, we discuss the multifaceted roles of PARP-1 and PARP-2 beyond DNA repair, including the impact of PARP-1 on chemokine signalling, immune modulation, and transcriptional regulation of gene expression, particularly in the contexts of angiogenesis and epithelial-to-mesenchymal transition (EMT). We evaluate the pre-clinical role of PARP inhibitors, either as single-agent or combination therapies, to block the metastatic process. Efficacy of PARP inhibitors was demonstrated via DNA repair-dependent and independent mechanisms, including DNA damage, cell migration, invasion, initial colonization at the metastatic site, osteoclastogenesis, and micrometastasis formation. Finally, we summarize the recent clinical advancements of PARP inhibitors in the prevention and progression of distant metastases, with a particular focus on specific metastatic sites and PARP-1 selective inhibitors. Overall, PARP inhibitors have demonstrated great potential in inhibiting the metastatic process, pointing the way for greater use in early cancer settings.
Collapse
Affiliation(s)
- Mallory I. Frederick
- Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3T5, Canada; (M.I.F.); (D.A.); (L.C.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), l’Institut de Cancer de Montreal, Montreal, QC H2X 0A9, Canada;
| | - Djihane Abdesselam
- Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3T5, Canada; (M.I.F.); (D.A.); (L.C.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), l’Institut de Cancer de Montreal, Montreal, QC H2X 0A9, Canada;
| | - Anna Clouvel
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), l’Institut de Cancer de Montreal, Montreal, QC H2X 0A9, Canada;
| | - Laurent Croteau
- Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3T5, Canada; (M.I.F.); (D.A.); (L.C.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), l’Institut de Cancer de Montreal, Montreal, QC H2X 0A9, Canada;
| | - Saima Hassan
- Faculty of Medicine, Université de Montréal, Montreal, QC H3C 3T5, Canada; (M.I.F.); (D.A.); (L.C.)
- Centre de Recherche du Centre Hospitalier de l’Université de Montréal (CRCHUM), l’Institut de Cancer de Montreal, Montreal, QC H2X 0A9, Canada;
- Division of Surgical Oncology, Department of Surgery, Centre Hospitalier de l’Université de Montréal (CHUM), Montreal, QC H2X 0C1, Canada
| |
Collapse
|
48
|
Tai Y, Chow A, Han S, Coker C, Ma W, Gu Y, Estrada Navarro V, Kandpal M, Hibshoosh H, Kalinsky K, Manova-Todorova K, Safonov A, Walsh EM, Robson M, Norton L, Baer R, Merghoub T, Biswas AK, Acharyya S. FLT1 activation in cancer cells promotes PARP-inhibitor resistance in breast cancer. EMBO Mol Med 2024; 16:1957-1980. [PMID: 38956205 PMCID: PMC11319505 DOI: 10.1038/s44321-024-00094-2] [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: 09/06/2023] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
Acquired resistance to PARP inhibitors (PARPi) remains a treatment challenge for BRCA1/2-mutant breast cancer that drastically shortens patient survival. Although several resistance mechanisms have been identified, none have been successfully targeted in the clinic. Using new PARPi-resistance models of Brca1- and Bard1-mutant breast cancer generated in-vivo, we identified FLT1 (VEGFR1) as a driver of resistance. Unlike the known role of VEGF signaling in angiogenesis, we demonstrate a novel, non-canonical role for FLT1 signaling that protects cancer cells from PARPi in-vivo through a combination of cell-intrinsic and cell-extrinsic pathways. We demonstrate that FLT1 blockade suppresses AKT activation, increases tumor infiltration of CD8+ T cells, and causes dramatic regression of PARPi-resistant breast tumors in a T-cell-dependent manner. Moreover, PARPi-resistant tumor cells can be readily re-sensitized to PARPi by targeting Flt1 either genetically (Flt1-suppression) or pharmacologically (axitinib). Importantly, a retrospective series of breast cancer patients treated with PARPi demonstrated shorter progression-free survival in cases with FLT1 activation at pre-treatment. Our study therefore identifies FLT1 as a potential therapeutic target in PARPi-resistant, BRCA1/2-mutant breast cancer.
Collapse
Affiliation(s)
- Yifan Tai
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Biology, McGill University, Montreal, Quebec, QC, H3G0B1, Canada
| | - Angela Chow
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Seoyoung Han
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Jacobs School of Medicine, University of Buffalo, New York, NY, USA
| | - Courtney Coker
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Wanchao Ma
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Yifan Gu
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Valeria Estrada Navarro
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Manoj Kandpal
- Centre for Clinical and Translational Science, Rockefeller University Hospital, 1230 York Ave, New York, NY, 10065, USA
| | - Hanina Hibshoosh
- Department of Pathology and Cell Biology, 630 W 168th St, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Kevin Kalinsky
- Winship Cancer Institute of Emory University, Emory University School of Medicine, 1365 Clifton Road NE, Atlanta, GA, 30322, USA
| | - Katia Manova-Todorova
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Drukier Institute for Children's Health, Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - Anton Safonov
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Elaine M Walsh
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
- Department of Medicine, Georgetown Lombardi Comprehensive Cancer Center, 3800 Reservoir Rd, NW, Washington DC, 20007, USA
| | - Mark Robson
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Larry Norton
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Richard Baer
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA
- Department of Pathology and Cell Biology, 630 W 168th St, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Taha Merghoub
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, 1300 York Avenue, New York, NY, 10065, USA
| | - Anup K Biswas
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, 630 W 168th St, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| | - Swarnali Acharyya
- Institute for Cancer Genetics, 1130 St Nicholas Avenue, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Department of Pathology and Cell Biology, 630 W 168th St, Columbia University Irving Medical Center, New York, NY, 10032, USA.
- Herbert Irving Comprehensive Cancer Center, 1130 St. Nicholas Ave, Columbia University Irving Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
49
|
Chase DM, Iadeluca L, Lim J, Tseng WY, Bulsara P, Patton G. Prescribers and patients drive maintenance therapy patterns in a community oncology practice: National guidelines versus the real-world experience. Gynecol Oncol Rep 2024; 54:101440. [PMID: 39040942 PMCID: PMC11261288 DOI: 10.1016/j.gore.2024.101440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/18/2024] [Accepted: 06/23/2024] [Indexed: 07/24/2024] Open
Abstract
Objective Previous studies have shown that first-line (1L) maintenance therapy (MT) with poly(ADP-ribose) polymerase inhibitors and/or bevacizumab improves outcomes among patients with advanced ovarian cancer (OC); however, these treatments are underutilized. This study aimed to provide a real-world understanding of MTs among patients with advanced OC who received 1L platinum-based chemotherapy (PBC). Methods A retrospective chart review using iKnowMed electronic health records to identify patients aged ≥18 years with advanced OC who initiated 1L PBC between January 1, 2018-December 31, 2020. Following 1L PBC, patients could have received MT or active surveillance (AS). Kaplan-Meier methods were used to estimate time to treatment discontinuation (TTD), real-world progression-free survival (rwPFS), and overall survival (OS). Results Of the 600 chart-reviewed patients included, 239 (39.8 %) received MT and 315 (52.5 %) received AS. Patients who were <65 years of age, or those with higher-stage disease or those who had received neoadjuvant treatment, were more likely to initiate MT than AS. Genetic testing rates were low across both cohorts. Median (95 % confidence interval [CI]) TTD for the MT cohort was 13.6 months (11.0, 21.2). Median (95 % CI) rwPFS was 26.9 months (21.3, not reached) and 11.3 months (9.5, 13.0) for the 1L MT and AS cohorts, respectively (p < 0.0001). OS at 36 months was 82.4 % in the 1L MT cohort and 58.0 % in the 1L AS cohort. Conclusions This study reinforces clinical trial findings that 1L MT improves outcomes in patients with advanced OC; however, genetic testing rates and 1L MT remained low.
Collapse
Affiliation(s)
- Dana M. Chase
- University of California, Los Angeles, 100 UCLA Medical Plaza, Suite 383, Los Angeles, CA 90024, USA
| | - Laura Iadeluca
- GSK, 2929 Walnut Street, Ste. 1700, Philadelphia, PA 19104, USA
| | - Jonathan Lim
- GSK, 2929 Walnut Street, Ste. 1700, Philadelphia, PA 19104, USA
| | - Wan-Yu Tseng
- Ontada, 33 Arch Street, 20th Floor, Boston, MA 02110, USA
| | - Purva Bulsara
- Ontada, 33 Arch Street, 20th Floor, Boston, MA 02110, USA
| | - Gregory Patton
- Ontada, 33 Arch Street, 20th Floor, Boston, MA 02110, USA
| |
Collapse
|
50
|
Carballo EV, Kim KH, Penn CA. Trends in estimated PARP inhibitor eligibility and benefit among US epithelial ovarian cancer patients. Gynecol Oncol 2024; 187:204-211. [PMID: 38795509 DOI: 10.1016/j.ygyno.2024.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/28/2024]
Abstract
OBJECTIVE To estimate the annual percentage of patients with epithelial ovarian cancer (EOC) who could be eligible for and benefit from PARP inhibitor therapy amidst changing US Food and Drug Administration (FDA)-approved indications. METHODS This is a simulated retrospective observational study using publicly available data on patients with advanced-stage EOC. PARPi eligibility is based on FDA approvals and withdrawals from 2014 through 2023, along with published demographic and genomic data. Clinical trial data is used to estimate treatment benefit. PARPi including olaparib, niraparib, and rucaparib are analyzed in aggregate with sub-analyses by molecular classification and treatment timing. Results are reported as the percentage of EOC patients appropriate for any cancer-directed therapy. RESULTS PARPi were approved for 9 different indications in EOC between 2014 and 2021; reduced to 6 indications by 2023. Eligibility increased from 2.0% (95% CI,1.3%-1.6%) in 2014 to a maximum of 93.4% (95% CI,90.1%-94.6%) in 2021. The maximum percentage of patients with 2-year PFS benefit was 22.0% (95% CI, 17.2%-26.8%) in 2021, projected to decrease to 13.0% (95% CI, 9.9%-15.9%) in 2024. Most of this decrease was seen in the homologous recombination deficient, BRCA wild-type population (8.4% to 4.0%). CONCLUSIONS PARPi eligibility increased at a greater rate than benefit resulting in a low population-level benefit-to-eligibility ratio until 2021. Recent FDA withdrawals improved this ratio with an accompanied decrease in the absolute number of patients benefiting. To further optimize population-level benefit-to-eligibility ratio of targeted therapies in ovarian cancer, we need to identify better biomarkers, treatment combinations, and novel therapeutic targets.
Collapse
Affiliation(s)
- Erica V Carballo
- Division of Gynecologic Oncology, Vanderbilt University Medical Center, United States of America.
| | - Kenneth H Kim
- Division of Gynecologic Oncology, Cedars-Sinai Medical Center, United States of America
| | - Courtney A Penn
- Division of Gynecologic Oncology, Vanderbilt University Medical Center, United States of America
| |
Collapse
|