1
|
Bassoy EY, Raja R, Rubino TE, Coscia F, Goergen K, Magtibay P, Butler K, Schmitt A, Oberg AL, Curtis M. Identification of TTLL8, POTEE, and PKMYT1 as immunogenic cancer-associated antigens and potential immunotherapy targets in ovarian cancer. Oncoimmunology 2025; 14:2460276. [PMID: 39891409 PMCID: PMC11792853 DOI: 10.1080/2162402x.2025.2460276] [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: 08/29/2024] [Revised: 12/27/2024] [Accepted: 01/24/2025] [Indexed: 02/03/2025] Open
Abstract
Most high-grade serous ovarian cancers (OC) do not respond to current immunotherapies. To identify potential new actionable tumor antigens in OC, we performed immunopeptidomics on a human OC cell line expressing the HLA-A02:01 haplotype, which is commonly expressed across many racial and ethnic groups. From this dataset, we identified TTLL8, POTEE, and PKMYT1 peptides as candidate tumor antigens with low expression in normal tissues and upregulated expression in OC. Using tissue microarrays, we assessed the protein expression of TTLL8 and POTEE and their association with patient outcomes in a large cohort of OC patients. TTLL8 was found to be expressed in 56.7% of OC and was associated with a worse overall prognosis. POTEE was expressed in 97.2% of OC patients and had no significant association with survival. In patient TILs, increases in cytokine production and tetramer-positive populations identified antigen-specific CD8 T cell responses, which were dependent on antigen presentation by HLA class I. Antigen-specific T cells triggered cancer cell killing of antigen-pulsed OC cells. These findings suggest that TTLL8, POTEE, and PKMYT1 are potential targets for the development of antigen-targeted immunotherapy in OC.
Collapse
Affiliation(s)
| | - Remya Raja
- Department of Immunology, Mayo Clinic, Phoenix, AZ, USA
| | | | - Fabian Coscia
- Max-Delbruck-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Krista Goergen
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Paul Magtibay
- Department of Obstetrics and Gynecology, Mayo Clinic, Phoenix, AZ, USA
| | - Kristina Butler
- Department of Obstetrics and Gynecology, Mayo Clinic, Phoenix, AZ, USA
- College of Medicine and Science, Mayo Clinic, Phoenix, AZ, USA
| | - Alessandra Schmitt
- Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Ann L. Oberg
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Marion Curtis
- Department of Immunology, Mayo Clinic, Phoenix, AZ, USA
- College of Medicine and Science, Mayo Clinic, Phoenix, AZ, USA
- Department of Cancer Biology, Mayo Clinic, Phoenix, AZ, USA
- Mayo Clinic Comprehensive Cancer Center, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
2
|
Cui M, Zhou M, Zhou L, Zhou G, Liu Y. Tertiary lymphoid structures achieve 'cold' to 'hot' transition by remodeling the cold tumor microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189312. [PMID: 40189114 DOI: 10.1016/j.bbcan.2025.189312] [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: 06/11/2024] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025]
Abstract
Immune checkpoint blockade (ICB) therapies have demonstrated significant clinical efficacy in immune-infiltrated tumors such as melanoma and non-small cell lung cancer. However, "cold tumors"-including ovarian cancer, pancreatic cancer, and gliomas-exhibit insufficient immune infiltration, leading to poor therapeutic responses to ICBs and limited improvement in patient prognosis. Recent studies have shown that tumor-associated tertiary lymphoid structures (TLSs) can induce strong local immune responses within the tumor microenvironment (TME), serving as important biological markers for predicting ICB therapy efficacy. Notably, preclinical and clinical studies on cold tumors have confirmed that TLSs can potently enhance ICB efficacy through TME remodeling-a breakthrough that has attracted considerable attention. Here, we systematically examine the immunological profile of cold tumors and decipher the mechanistic basis for their impaired immune cell infiltration. We further delineate the distinctive features of tumor-associated TLSs in generating antitumor immunity and establish criteria for their identification. Significantly, we emphasize the unique capability of TLSs to reprogram the immunosuppressive tumor microenvironment characteristic of cold tumors. Based on these insights, we evaluate clinical evidence supporting TLS-mediated enhancement of ICB efficacy and discuss emerging strategies for exogenous TLSs induction.
Collapse
Affiliation(s)
- Mengke Cui
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Mengfan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Lu Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Gan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China; National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, 110 Xiangya Road, Changsha, Hunan 410008, PR China.
| | - Yingzi Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China.
| |
Collapse
|
3
|
Porter JM, Jin C, Churchman M, Croy I, Gourley CM, Glyn-Wright O, Stewart M, Brownsell E, Monks B, Sanderson P, Nirsimloo R, Herrington CS, Gourley C, Hollis RL. Antibody drug conjugate targets are highly differentially expressed across the major types of ovarian cancer. Eur J Cancer 2025; 224:115522. [PMID: 40446758 DOI: 10.1016/j.ejca.2025.115522] [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: 03/24/2025] [Revised: 05/15/2025] [Accepted: 05/20/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND Antibody-drug conjugates (ADCs) are emerging anti-cancer agents. The folate receptor alpha (FOLRα)-directed ADC mirvetuximab soravtansine recently demonstrated clinical activity in platinum-resistant ovarian cancer, with other ADCs currently in development. The relative expression of FOLRα and other ADC targets is largely unknown across ovarian cancer histotypes. METHODS Expression levels of the ADC targets FOLRα, TROP2 and B7-H4 were assessed by immunohistochemistry in patient cohorts using tumour tissue microarrays of the major ovarian cancer histotypes: high grade serous (HGSOC, n = 331); endometrioid (EnOC, n = 101) and clear cell ovarian carcinoma (CCOC, n = 60). Degree of expression was quantified by membrane histoscore. RESULTS We observed differences in ADC target expression patterns across ovarian cancer histotypes. FOLRα expression was highest in HGSOC, with few EnOC or CCOC demonstrating positivity (HGSOC: 70.9 % FOLRα histoscore ≥50 vs 21.1 % and 29.3 % in EnOC and CCOC). B7-H4 was expressed in HGSOC, EnOC and CCOC (99.7 %, 89.8 % and 80.7 % with histoscore ≥50). CCOC were mostly TROP2 negative (89.3 % with histoscore <50); a subset of HGSOC and EnOC expressed TROP2 (54.8 % and 57.7 % with histoscore ≥50, respectively). There was no significant association between ADC target expression and molecular subtypes of HGSOC (BRCA1/2-mutant, CCNE1-gained, other) or EnOC (TP53-mutant, CTNNB1-mutant, POLE-mutant, MMR deficient, no specific molecular profile). In CCOC, ARID1A/B mutation was associated with lower B7-H4 expression (P-adj=0.024). CONCLUSION EnOC and CCOC are usually FOLRα negative, while HGSOC, EnOC and CCOC frequently express B7-H4. TROP2 positivity is limited to HGSOC and EnOC. Careful consideration of histotype and ADC target expression levels is warranted when designing and analysing clinical studies of ADCs.
Collapse
Affiliation(s)
- Joanna M Porter
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Cici Jin
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Michael Churchman
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Ian Croy
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Catriona M Gourley
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Oni Glyn-Wright
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Marianne Stewart
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Elizabeth Brownsell
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Basil Monks
- Royal Infirmary of Edinburgh, NHS Lothian, Edinburgh, UK
| | | | - Rachel Nirsimloo
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh, UK
| | - C Simon Herrington
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Charlie Gourley
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Robert L Hollis
- The Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
4
|
Li Y, Yuan X, Yin XF, Zheng D, Shi F, Liu D, Hu L, Shi X, Wen N, He QY, Yang H, Zhang CZ. Proteomics analysis and immune profiling reveal regulators of PD-L1 in oesophageal squamous cell carcinoma. Br J Cancer 2025:10.1038/s41416-025-03068-4. [PMID: 40490504 DOI: 10.1038/s41416-025-03068-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 05/05/2025] [Accepted: 05/16/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Proteomics studies have advanced our comprehension of cancer biology, accelerated targeted therapy, and improved patient outcomes. METHODS High-resolution mass spectrometry and immune profiling based on immunohistochemistry and multiple immunohistochemistry were employed to investigate proteomic and immune landscapes in oesophageal squamous cell carcinoma (ESCC) and explore the regulators of PD-L1 in ESCC. Molecular validation was performed using qRT-PCR, western blotting, and in vitro functional assays. RESULTS Proteomic profiling of 89 treatment-naive ESCC specimens identified over 9300 proteins, with 6900 proteins detected across most samples. Proteome-based stratification identified three subtypes related to diverse clinical and molecular features. Combined proteomics and immune analyses revealed core proteins associated with the immune landscape in ESCC. Further, integrated proteomics, transcriptomics, and immune profiling nominated COTL1 as a potential regulator of PD-L1 in ESCC. Overexpression of COTL1 upregulated both mRNA and protein levels of PD-L1 and promoted cell proliferation in ESCC. Patients with high COTL1 protein expression were likely to have a poor prognosis, along with increased infiltration of CD4+CD8+ and CD4+GrB+ cells. CONCLUSIONS Collectively, our integrative analysis enables a more comprehensive understanding of the proteomic and immune landscape of ESCC and implicates COTL1 as a potential modulator of PD-L1 and immune cell infiltration.
Collapse
Affiliation(s)
- Yuying Li
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xiaoyi Yuan
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xing-Feng Yin
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Dandan Zheng
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Fujin Shi
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Danya Liu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Liling Hu
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xinyu Shi
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Nengqiao Wen
- Department of Pathology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Qing-Yu He
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China.
| | - Hong Yang
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Guangdong Esophageal Cancer Institute, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou City, China.
| | - Chris Zhiyi Zhang
- MOE Key Laboratory of Tumor Molecular Biology and State Key Laboratory of Bioactive Molecules and Druggability Assessment, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China.
| |
Collapse
|
5
|
Alim L, Adityan S, Chen R, Neilson T, Coleborn E, Wilkinson AN, He Y, Irgam G, Bhavsar C, Lourie R, Rogers R, Cabraal N, Jagasia N, Chetty N, Perrin L, Hooper JD, Steptoe R, Wu SY. Antigen presentation potential is variable among human ovarian tumour and syngeneic murine models and dictates pre-clinical outcomes of immunotherapy. Biomed Pharmacother 2025; 187:118141. [PMID: 40347847 DOI: 10.1016/j.biopha.2025.118141] [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: 11/27/2024] [Revised: 04/24/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
High grade serous ovarian carcinoma (HGSC) is a fatal gynaecological malignancy with limited therapeutic options. Immunotherapies targeting MHC-I-dependent antigen presentation offer potential. Currently, the antigen presentation machinery (APM) of widely used syngeneic murine HGSC models remains poorly characterised, limiting translational relevance. Here, we systematically evaluate APM gene expression in syngeneic murine and patient samples. Tap1 and Psmb8 were identified as critical APM markers, deficient in murine models and strongly correlating with MHC-I expression. Hierarchical clustering correlation analysis using these markers revealed that ID8-p53⁻/⁻BRCA1⁻/⁻ was the most strongly correlated model and aligned with the largest patient subset. Moreover, ID8-ip1 correlated to the smaller second patient subset strongly. The low MHC-I expressing IG10 model was unique clustering alongside patient derived LP28 tumour and not fitting either patient subset. In vivo test of a novel combination immune therapy consisting of Flt3L, Poly(I:C), and paclitaxel therapy demonstrated significantly reduced tumour burden in high APM models (p53⁻/⁻BRCA1⁻/⁻, ID8-ip1; p < 0.01), but not IG10. Furthermore, high expressing MHC-I models were linked to enhanced DC expansion, CD8⁺ T-cell infiltration, and effector differentiation (131 % increase in ID8-ip1), alongside improved CD8⁺ T-cell activation and CD86⁺ B-cell co-stimulation. These findings establish MHC-I as a predictive biomarker for immunotherapy response and underscore the need for APM-enhancing strategies in antigen-poor tumours. By bridging murine models to human APM heterogeneity, this work provides a framework for optimising preclinical immunotherapy evaluation and patient stratification, advancing tailored therapeutic approaches for HGSC.
Collapse
Affiliation(s)
- Louisa Alim
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Siddharth Adityan
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Rui Chen
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Trent Neilson
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Elaina Coleborn
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Andrew N Wilkinson
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Yaowu He
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland 4102, Australia
| | - Gowri Irgam
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Chintan Bhavsar
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Rohan Lourie
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia; Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - Rebecca Rogers
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia; Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - Nimithri Cabraal
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia; Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - Nisha Jagasia
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia; Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - Naven Chetty
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia; Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - Lewis Perrin
- Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia; Mater Health Services, South Brisbane, Queensland 4101, Australia
| | - John D Hooper
- Mater Research Institute - The University of Queensland, Translational Research Institute, Woolloongabba, Queensland 4102, Australia; Mater Ovarian Cancer Research Collaborative, Mater Adult Hospital, South Brisbane, Queensland 4101, Australia
| | - Raymond Steptoe
- Frazer Institute, University of Queensland, Brisbane, Australia
| | - Sherry Y Wu
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia.
| |
Collapse
|
6
|
Wu CH, Lin H, Ou YC, Fu HC, Yang MY, Huang CC. Molecular Interplay Between PTEN, ARID1A, PD-L1, and MMR in Asian Ovarian Clear Cell Carcinoma: Implications for Immunotherapy Response and Patient Stratification. Int J Mol Sci 2025; 26:4915. [PMID: 40430056 PMCID: PMC12112434 DOI: 10.3390/ijms26104915] [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: 04/13/2025] [Revised: 05/10/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
Ovarian clear cell carcinoma (OCCC) represents a distinct histological subtype with a high prevalence in Asian populations and poor chemotherapy response. This study investigated molecular interactions between phosphatase and tensin homolog (PTEN), AT-rich interactive domain 1A (ARID1A), programmed death-ligand 1 (PD-L1), and mismatch repair (MMR) proteins in Asian patients with OCCC. Immunohistochemical analysis was performed on tissue microarrays from 69 OCCC cases. The expression of PTEN, ARID1A, PD-L1, and four MMR proteins was evaluated alongside clinical data. A high prevalence of PTEN loss (78.3%) and ARID1A deficiency (48.8%), with PD-L1 expression in 26.1% and MMR deficiency in 10.1% of cases, was observed. All PD-L1-positive tumors demonstrated concurrent PTEN loss (p = 0.007). MMR deficiency was significantly associated with ARID1A loss (p = 0.049). PTEN loss correlated with worse progression-free survival (PFS) in early-stage disease (p = 0.039). PTEN and ARID1A alterations represent early pathogenic events in Asian OCCC, with PTEN loss significantly impacting PFS in early-stage disease. The correlation between PTEN loss and PD-L1 expression, alongside ARID1A-MMR deficiency association, provides insights into OCCC's immunological landscape and therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Chen-Hsuan Wu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan; (C.-H.W.); (H.L.); (Y.-C.O.); (H.-C.F.)
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
| | - Hao Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan; (C.-H.W.); (H.L.); (Y.-C.O.); (H.-C.F.)
| | - Yu-Che Ou
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan; (C.-H.W.); (H.L.); (Y.-C.O.); (H.-C.F.)
| | - Hung-Chun Fu
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan; (C.-H.W.); (H.L.); (Y.-C.O.); (H.-C.F.)
| | - Ming-Yu Yang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Department of Otolaryngology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
| | - Chao-Cheng Huang
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan;
- Department of Anatomic Pathology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
| |
Collapse
|
7
|
Aoki D, Yamauchi M, Izawa M, Ito Y, Hamada M, Ozaki M, Maekawa S, Muro K. Effectiveness and safety of pembrolizumab for the treatment of Japanese patients with microsatellite instability-high tumors excluding colorectal cancer: a post-marketing surveillance. Jpn J Clin Oncol 2025:hyaf064. [PMID: 40382714 DOI: 10.1093/jjco/hyaf064] [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: 12/20/2024] [Accepted: 04/08/2025] [Indexed: 05/20/2025] Open
Abstract
BACKGROUND We aimed to assess the real-world effectiveness and safety of pembrolizumab monotherapy in Japanese patients with high-frequency microsatellite instability (MSI-H) solid tumors except colorectal cancer. METHODS This multicenter, observational, post-marketing surveillance had a 12-month observation period. We included all patients with locally advanced or metastatic MSI-H solid tumors, except colorectal cancer, in whom standard treatment was difficult or who had shown tumor progression after conventional chemotherapies and had started treatment with pembrolizumab by 31 December 2019. RESULTS In total, 403 patients were enrolled, and 396 and 376 patients were included in the safety and effectiveness analysis sets, respectively. The numbers of patients and frequencies of tumor types occurring in ≥20 cases were: endometrial, 162/403 (40.2%); gastric, 61/403 (15.1%); biliary tract, 42/403 (10.4%); pancreatic, 29/403 (7.2%); and ovarian, 20/403 (5.0%). The objective response rate was 50.3% (189/376) and the disease control rate was 71.5% (269/376). The 12-month progression-free survival (PFS) rate was 42.1% and the median PFS was 8.8 months (95% confidence interval, 6.4-11.5). The 12-month overall survival (OS) rate was 75.1%, and median OS was not reached. Treatment-related adverse events (AEs) of special interest of any grade occurred in 128/396 (32.3%) patients, and those of Grade ≥ 3, in 54/396 (13.6%) patients. One patient with esophageal cancer experienced a Grade 5 AE. No new safety signals were observed. CONCLUSIONS This study confirmed the real-world effectiveness and safety of pembrolizumab monotherapy in patients with MSI-H solid tumors except colorectal cancer in Japan.
Collapse
Affiliation(s)
- Daisuke Aoki
- International University of Health and Welfare Graduate School, W 4-1-26 Akasaka, Minato-ku, Tokyo 107-8402, Japan
- Akasaka Sanno Medical Center, W 4-1-26 Akasaka, Minato-ku, Tokyo 107-8402, Japan
| | - Mai Yamauchi
- Medical Affairs Oncology, MSD K.K., Kitanomaru Square, 1-13-12 Kudan-kita, Chiyoda-ku, Tokyo 102-8667, Japan
| | - Makiko Izawa
- Medical Affairs Oncology, MSD K.K., Kitanomaru Square, 1-13-12 Kudan-kita, Chiyoda-ku, Tokyo 102-8667, Japan
| | - Yuichiro Ito
- Medical Affairs Oncology, MSD K.K., Kitanomaru Square, 1-13-12 Kudan-kita, Chiyoda-ku, Tokyo 102-8667, Japan
| | - Masahiro Hamada
- Pharmacovigilance, MSD K.K., Kitanomaru Square, 1-13-12 Kudan-kita, Chiyoda-ku, Tokyo 102-8667, Japan
| | - Masahiko Ozaki
- Pharmacovigilance, MSD K.K., Kitanomaru Square, 1-13-12 Kudan-kita, Chiyoda-ku, Tokyo 102-8667, Japan
| | - Shinichiroh Maekawa
- Pharmacovigilance, MSD K.K., Kitanomaru Square, 1-13-12 Kudan-kita, Chiyoda-ku, Tokyo 102-8667, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center Hospital, 1-1 Kanokoden, Chikusa-ku, Nagoya 464-8681, Japan
| |
Collapse
|
8
|
Park J, Joung JG, Lim MC, Lee J, Kim BG, Kim JW, Shin SJ, Kim S, Park E, Choi CH, Kim HS, Park SY, Lee JY. Neoadjuvant Chemotherapy with Dual Immune Checkpoint Inhibitors for Advanced-Stage Ovarian Cancer: Final Analysis of TRU-D Phase II Nonrandomized Clinical Trial. Clin Cancer Res 2025; 31:1865-1876. [PMID: 40043003 DOI: 10.1158/1078-0432.ccr-24-3753] [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: 11/09/2024] [Revised: 01/12/2025] [Accepted: 03/03/2025] [Indexed: 05/16/2025]
Abstract
PURPOSE This open-label, investigator-initiated, phase II study was conducted to evaluate the safety, survival, and neoadjuvant outcomes of neoadjuvant chemotherapy (NAC) combined with dual immune checkpoint inhibitors in advanced-stage epithelial ovarian cancer (EOC). PATIENTS AND METHODS Between June 2019 and July 2021, 45 patients with unresectable stage III to IV EOC were enrolled. The patients received three cycles of NAC combined with durvalumab and tremelimumab. All patients underwent interval debulking surgery and received three cycles of durvalumab and adjuvant chemotherapy, followed by 12 cycles of durvalumab as maintenance therapy. The primary endpoint was the 12-month progression-free survival (PFS) rate; the secondary endpoints were the objective response rate after NAC, a chemotherapy response score, pathologic complete response, overall survival, and safety. The preplanned exploratory analyses assessed the lymphocyte infiltration, PD-L1 expression, and genomic profiles of pretreatment tumors. RESULTS The 12-month PFS rate was 65.9% [95% confidence interval (CI), 52.8-not estimated (NE)], whereas the 24- and 30-month PFS rates were 38.6% (95% CI, 26.7-NE) and 36.4% (95% CI, 24.7-NE), respectively. After NAC, the objective response rate was 86.7%, whereas 14 patients (31.1%) had a chemotherapy response score of three, and five (11.1%) achieved pathologic complete response. The 30-month overall survival rate was 87.7%. The most common grade ≥3 adverse event was neutropenia (26.7%). In an exploratory analysis, patients with pre-NAC tumors showing PD-L1 (combined positive score) ≥1, high Mutation Signature 3, and a high extracellular matrix signature demonstrated improved PFS outcomes. CONCLUSIONS NAC combined with dual immune checkpoint inhibitors is feasible for advanced-stage EOC and shows promising activity with a durable clinical response.
Collapse
Affiliation(s)
- Junsik Park
- Department of Obstetrics and Gynecology, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, South Korea
| | - Je-Gun Joung
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam, South Korea
| | - Myong Cheol Lim
- Center for Gynecologic Cancer, National Cancer Center, Goyang, South Korea
| | - Jungbok Lee
- Department of Clinical Epidemiology and Biostatistics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Byoung-Gie Kim
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| | - Jae-Weon Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - So Jin Shin
- Department of Obstetrics and Gynecology, Keimyung University Dongsan Medical Center, Daegu, South Korea
| | - Sunghoon Kim
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, South Korea
| | - Eunhyang Park
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Chel Hun Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University, Seoul, South Korea
| | - Hee Seung Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Yoon Park
- Center for Gynecologic Cancer, National Cancer Center, Goyang, South Korea
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
9
|
Kuang W, Zeng J, Tong L, Liu Q, Sun H, Feng M, Liang D, Wang W, Wang C. Frequency of microsatellite instability in gynecologic cancers and the efficacy of immune checkpoint inhibitors treated: real-world data from a single gynecologic center. Front Immunol 2025; 16:1567824. [PMID: 40416974 PMCID: PMC12098365 DOI: 10.3389/fimmu.2025.1567824] [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: 01/28/2025] [Accepted: 04/15/2025] [Indexed: 05/27/2025] Open
Abstract
Objective This study evaluated the incidence of Microsatellite Instability-High (MSI-H) in patients with gynecologic cancers in a single gynecologic center and investigated the effect of immune checkpoint inhibitors (ICIs) in treating MSI-H in advanced or recurrent gynecologic cancers. Methods We conducted a retrospective study of patients diagnosed with gynecological cancers between June 2021 and May 2024. We investigated their clinicopathological information, the results of microsatellite instability (MSI), the immunohistochemistry staining PD-L1 analyses, the molecular classification testing, and the tumor response to treatment with ICIs. Results Among 1333 patients included in the analysis, the frequency of MSI-H was 1.3% (3/223) in cervical cancer, 25.7% (280/1091) in endometrial cancer, and 10.5% (2/19) in ovarian or tubal and peritoneal cancer. When the patients were evaluated by histologic type, the frequency of MSI-H was 26.1% (241/921) in endometrioid adenocarcinoma and 35.1% (20/57) in mixed adenocarcinoma. Molecular classification results for the 1020 cases that successfully underwent the tests were 71 for the POLE mutation (POLEmut) subtype, 271 for MMR-deficiency (MMRd) subtype, 571 for the non-specific molecular profile (NSMP) subtype, and 107 for the p53 abnormality (p53abn) subtype. Thirty-five patients were treated with ICIs for at least one cycle. The objective response rate (ORR) was 34.3% (95% CI, 19.1% to 52.2%). Among the patients who achieved an objective response, the median time to respond was 2.65 months, and the median duration of response had not been reached. The median progression-free survival (PFS) was 9 months (95% CI, 4 to 10), and the median overall survival (OS) had not been reached. Additionally, in the patients with endometrial cancer, the median PFS in MSI-H patients was 5 months versus 3 months in microsatellite stable (MSS) patients (Δ = 2 months; p=0.92), and the median OS in both MSI-H and MSS patients had not been reached (p=0.89). Conclusion This study had shown the MSI-H frequencies for the three major types of gynecological tumors and demonstrated the clinical benefit of treatment with ICIs in patients with advanced or recurrent gynecologic cancer. Among endometrial cancer patients, the effects of immunotherapy may be consistent regardless of MSI status.
Collapse
Affiliation(s)
- Wei Kuang
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Jing Zeng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lingling Tong
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Qianqi Liu
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Huanxin Sun
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Min Feng
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Dongni Liang
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Wei Wang
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Wang
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Liu J, Jiao X, Mu W, Li H, Xia Y, Wu Y, Zhu L, Zhong Q, Pan W, Liu X, Xiang M, Cheng J, Lin H, Zhao X, Ding Z, Hu G, Mills GB, Ma D, Gao Q, Fang Y. Mitigating T cell DNA damage during PARP inhibitor treatment enhances antitumor efficacy. Sci Transl Med 2025; 17:eadr5861. [PMID: 40333991 DOI: 10.1126/scitranslmed.adr5861] [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: 10/01/2024] [Accepted: 03/12/2025] [Indexed: 05/09/2025]
Abstract
Poly(ADP-ribose) polymerase inhibitors (PARPis) are a class of agents targeting DNA damage repair that have become standard therapy for epithelial ovarian cancer (EOC) and multiple other solid tumors. In addition to targeting DNA damage repair, PARPis actively modulate antitumor immune responses, with efficacy being partially dependent on T cell activity. Here, we found that patient T cells sustain DNA damage during PARPi treatment, which reduces treatment efficacy. Leveraging paired pre- and posttreatment tumor samples from a clinical trial of patients with EOC treated with neoadjuvant niraparib as monotherapy, we showed that the PARPi caused DNA damage, slowed proliferation, and increased apoptosis in T cells, which we validated both in vitro and in mouse models. A genome-wide CRISPR (clustered regularly interspaced short palindromic repeats) knockout screen in primary human T cells identified PARP1 as the principal mediator of PARPi-induced T cell death. T cell-specific deletion of PARP1 or mutating Parp1 at its binding sites in transgenic mice led to reduced T cell DNA damage during PARPi treatment, resulting in improved efficacy of PARPis, alone or in combination with immune checkpoint inhibition. We then engineered PARPi-tolerant CAR T cells using cytosine base editing, which decreased PARPi-induced PARP1 trapping and led to reduced PARPi-induced DNA damage, resulting in superior antitumor efficacy in xenograft models compared with parental CAR T cells. This study highlights the relevance of PARPi-induced DNA damage to T cells and suggests opportunities to improve the efficacy of PARPis as monotherapy or in combination with immunotherapy.
Collapse
Affiliation(s)
- Jiahao Liu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiaofei Jiao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wei Mu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huayi Li
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yu Xia
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yijie Wu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Li Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qing Zhong
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Wen Pan
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xingzhe Liu
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Minghua Xiang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jiali Cheng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Haolong Lin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xuejiao Zhao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- Division of Oncological Sciences, Oregon Heath and Sciences University, Portland, OR 97201, USA
- Knight Cancer Institute, Portland, OR 97201, USA
| | - Zhiyong Ding
- Mills Institute for Personalized Cancer Care, Fynn Biotechnologies Ltd., Jinan, 250101, China
| | - Guang Hu
- Nanjing IASO Biotherapeutics Ltd., Nanjing, 210043, China
| | - Gordon B Mills
- Division of Oncological Sciences, Oregon Heath and Sciences University, Portland, OR 97201, USA
- Knight Cancer Institute, Portland, OR 97201, USA
| | - Ding Ma
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qinglei Gao
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yong Fang
- Department of Gynecological Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
- National Clinical Research Center for Obstetrics and Gynecology, Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
11
|
McGinty MT, Putelo AM, Kolli SH, Feng TY, Dietl MR, Hatzinger CN, Bajgai S, Poblete MK, Azar FN, Mohammad A, Kumar P, Rutkowski MR. TLR5 Signaling Causes Dendritic Cell Dysfunction and Orchestrates Failure of Immune Checkpoint Therapy against Ovarian Cancer. Cancer Immunol Res 2025; 13:696-711. [PMID: 39932226 PMCID: PMC12048257 DOI: 10.1158/2326-6066.cir-24-0513] [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: 05/30/2024] [Revised: 11/05/2024] [Accepted: 01/24/2025] [Indexed: 02/23/2025]
Abstract
Ovarian cancer accounts for more deaths than any other cancer of the female reproductive system. Patients who have ovarian tumors infiltrated with high frequencies of T cells are associated with a greater survival probability. However, therapies to revitalize tumor-associated T cells, such as PD-L1/PD-1 or CTLA4 blockade, are ineffective for the treatment of ovarian cancer. In this study, we demonstrate that for ovarian cancer, Toll-like receptor 5 (TLR5) signaling, for which the only known ligand is bacterial flagellin, governed failure of PD-L1 and CTLA4 blockade. Mechanistically, chronic TLR5 signaling on CD11c+ cells in vivo and in vitro impaired the differentiation of functional IL-12-producing XCR1+CD103+ conventional type 1 dendritic cells, biasing CD11c+ precursor cells toward myeloid subsets expressing high levels of PD-L1. This culminated in impaired activation of CD8+ T cells, reducing CD8+ T-cell function and ability to persist within the ovarian tumor microenvironment. Expansion of XCR1+CD103+ conventional type 1 dendritic cells in situ using Flt3L-Ig in combination with PD-L1 blockade achieved significant survival benefit in TLR5 knockout mice bearing ovarian tumors, whereas no benefit was observed in the presence of TLR5 signaling. Thus, we have identified a host-intrinsic mechanism leading to the failure of PD-L1 blockade for ovarian cancer, demonstrating that chronic TLR5 signaling on CD11c+ cells is a barrier limiting the efficacy of checkpoint therapy.
Collapse
Affiliation(s)
- Mitchell T. McGinty
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Audrey M. Putelo
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Sree H. Kolli
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Tzu-Yu Feng
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Madison R. Dietl
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Cara N. Hatzinger
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Simona Bajgai
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Mika K. Poblete
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Francesca N. Azar
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| | - Anwaruddin Mohammad
- Bioinformatics Core, University of Virginia School of Medicine, Charlottesville, VA
| | - Pankaj Kumar
- Bioinformatics Core, University of Virginia School of Medicine, Charlottesville, VA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA
| | - Melanie R. Rutkowski
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, VA, USA
| |
Collapse
|
12
|
Miller CH, Vemuri A, Lengyel E, Lastra RR. Adaptive immune response and PD-1/ PD-L1 status in chemotherapy treated high grade serous carcinoma is dependent on chemotherapy response score. Hum Pathol 2025; 159:105800. [PMID: 40389122 DOI: 10.1016/j.humpath.2025.105800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 05/16/2025] [Accepted: 05/16/2025] [Indexed: 05/21/2025]
Abstract
BACKGROUND Platinum-based chemotherapy and debulking surgery is the standard of care for patients with advanced tubo-ovarian high-grade serous carcinoma (HGSC). The chemotherapy response scoring (CRS) system is a histopathologic scoring system developed to measure response to neoadjuvant chemotherapy with prognostic implications. Omental samples with high CRS have greater inflammatory cell infiltrates, but the immunophenotype of infiltrating immune cells and PD-L1 expression of the residual tumor has not been well-defined. DESIGN Twenty cases of patients with FIGO stage IIIA to IIIC HGSC undergoing interval debulking after receiving 3-4 rounds of chemotherapy were selected. 6/20 cases of omental samples were graded as CRS 1, 7/20 were graded CRS 2, and 7/20 were graded CRS 3. The following immunohistochemical stains were performed: CD8, CD4, Foxp3, PD1, and PD-L1. The total number of tumor-infiltrating lymphocytes was recorded, and each case was given a PD-L1 combined positive score (CPS) and tumor proportion score (TPS). RESULTS There was a significantly greater number of CD8+ T cells, PD-1+ T cells, CD4+ T cells, and Foxp3+ T cells in CRS 3-scored cases compared to CRS 1 scored cases (p-values: 0.0018, 0.0224, 0.0071, and 0.0136, respectively). CRS 3-scored cases had a greater PD-L1 CPS (CRS 3 CPS 13 ± 8.2 versus CRS 1 CPS 0 ± 0; p-value: 0.0485). CONCLUSIONS Tubo-ovarian high-grade serous carcinoma with greater response to neoadjuvant treatment have significantly greater T cell infiltrate and greater PD-L1 combined positive score, highlighting a potential role of the CRS as a predictive biomarker for immune checkpoint blockade therapy.
Collapse
Affiliation(s)
| | - Anusha Vemuri
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology/ Section of Gynecologic Oncology, University of Chicago, IL, USA
| | - Ricardo R Lastra
- Department of Pathology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
13
|
Stur E, Peng F, Teng PN, Bayraktar E, Hu M, Corvigno S, Brown DJ, Lee S, Moore KN, Bateman NW, Darcy KM, Maxwell GL, P Conrads T, Sahni N, Vázquez-García I, Shah SP, Celestino J, D Fleming N, Navin NE, Wang L, Sood AK. The dynamic immune behavior of primary and metastatic ovarian carcinoma. NPJ Precis Oncol 2025; 9:120. [PMID: 40281242 PMCID: PMC12032089 DOI: 10.1038/s41698-025-00818-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 01/17/2025] [Indexed: 04/29/2025] Open
Abstract
Patients with high-grade serous ovarian carcinoma (HGSC) are usually diagnosed with advanced-stage disease, and the tumors often have immunosuppressive characteristics. Together, these factors are important for disease progression, drug resistance, and mortality. In this study, we used a combination of single-cell sequencing and spatial transcriptomics to identify the molecular mechanisms that lead to immunosuppression in HGSC. Primary tumors consistently showed a more active immune microenvironment than did omental tumors. In addition, we found that untreated primary tumors were mostly populated by dysfunctional CD4 and CD8 T cells in later stages of differentiation; this, in turn, was correlated with expression changes in the interferon α and γ pathways in epithelial cells, showing that cross-communication between the epithelial and immune compartments is important for immune suppression in HGSC. These findings could have implications for the design of clinical trials with immune-modulating drugs.
Collapse
Affiliation(s)
- Elaine Stur
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fuduan Peng
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pang-Ning Teng
- Gynecologic Cancer Center of Excellence, The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Walter Reed National Military Medical Center, Bethesda, MD, 20889, USA
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Emine Bayraktar
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Min Hu
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sara Corvigno
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David J Brown
- Obstetrics and Gynecology, Stephenson Cancer Center, Stephenson Cancer Center at the University of Oklahoma Health Sciences Center/Sarah Cannon Research Institute, Oklahoma City, OK, USA
| | - Sanghoon Lee
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kathleen N Moore
- Obstetrics and Gynecology, Stephenson Cancer Center, Stephenson Cancer Center at the University of Oklahoma Health Sciences Center/Sarah Cannon Research Institute, Oklahoma City, OK, USA
| | - Nicholas W Bateman
- Gynecologic Cancer Center of Excellence, The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Walter Reed National Military Medical Center, Bethesda, MD, 20889, USA
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - Kathleen M Darcy
- Gynecologic Cancer Center of Excellence, The Henry M. Jackson Foundation for the Advancement of Military Medicine Inc., Walter Reed National Military Medical Center, Bethesda, MD, 20889, USA
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | - George L Maxwell
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA
- Women's Health Integrated Research Center, Women's Service Line, Inova Health System, Falls Church, VA, USA
| | - Thomas P Conrads
- Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD, USA
- Women's Health Integrated Research Center, Women's Service Line, Inova Health System, Falls Church, VA, USA
| | - Nidhi Sahni
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Quantitative and Computational Biosciences Program, Baylor College of Medicine, Houston, TX, USA
| | - Ignacio Vázquez-García
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- The Halvorsen Center for Computational Oncology, Memorial Sloan Kettering Cancer Center, Boston, MA, USA
- Irving Institute for Cancer Dynamics, Columbia University, Cambridge, MA, 10027, USA
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sohrab P Shah
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joseph Celestino
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicole D Fleming
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas E Navin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, MA, USA
| | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- The James P. Allison Institute, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
- The Institute for Data Science in Oncology (IDSO), The University of Texas MD, Anderson Cancer Center, Houston, TX, 77030, USA.
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences (GSBS), Houston, TX, 77030, USA.
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
14
|
Kordowitzki P, Lange B, Elias KM, Haigis MC, Mechsner S, Braicu IE, Sehouli J. Transforming treatment paradigms: Focus on personalized medicine for high-grade serous ovarian cancer. CA Cancer J Clin 2025. [PMID: 40252048 DOI: 10.3322/caac.70008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 04/21/2025] Open
Abstract
High-grade serous ovarian cancer (HGSOC) is the most common and aggressive subtype of ovarian cancer, accounting for approximately 70% of all ovarian cancer cases and contributing significantly to the high mortality rates associated with this disease. Because of the asymptomatic nature of early stage disease, most patients are diagnosed at advanced stages when the cancer has already spread into the abdominal cavity, requiring complex and intensive surgical and chemotherapeutic interventions followed by maintenance therapies. Although a minority of cases are associated with well defined genetic syndromes, specific risk factors and a clear etiology in many cases remain elusive. HGSOC tumors are characterized by a high frequency of somatic gene copy number alterations, often associated with defects in homologous recombination repair of DNA. All attempts to introduce an effective screening for HGSOC to date have been unsuccessful. This review elucidates the complexities surrounding HGSOC and encompasses its etiology, epidemiology, classification, pathogenesis, and the current array of treatment strategies. Understanding molecular underpinnings is crucial for the development of targeted therapies and personalized multimodal treatment approaches in centralized therapeutic structures. This review also examines the importance of the tumor microenvironment. In addition, the authors' objective is to underscore the critical importance of placing the patient's perspective and diversity at the forefront of therapeutic strategies, thereby fostering a genuinely participatory decision-making process and ultimately improving patient quality of life.
Collapse
Affiliation(s)
- Pawel Kordowitzki
- Department of Preclinical and Basic Sciences, Nicolaus Copernicus University, Torun, Poland
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
- Department of Gynecology, Center of Oncological Surgery, European Competence Center for Ovarian Cancer, Charité-University Medicine Berlin, Berlin, Germany
| | - Britta Lange
- Institute for Cultural Studies, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Kevin M Elias
- Section of Gynecologic Oncology, Obstetrics and Gynecology Institute, Taussig Cancer Institute, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Marcia C Haigis
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, USA
| | - Sylvia Mechsner
- Department of Gynecology, Center of Oncological Surgery, European Competence Center for Ovarian Cancer, Charité-University Medicine Berlin, Berlin, Germany
| | - Ioana Elena Braicu
- Department of Gynecology, Center of Oncological Surgery, European Competence Center for Ovarian Cancer, Charité-University Medicine Berlin, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynecology, Center of Oncological Surgery, European Competence Center for Ovarian Cancer, Charité-University Medicine Berlin, Berlin, Germany
| |
Collapse
|
15
|
Martin SD, Thornton S, Chow C, Milne K, de Barros JS, Morris KA, Leung S, Jamieson A, Nelson BH, Cochrane DR, Huntsman DG, Gilks CB, Hoang L, McAlpine JN, Zhang AW. Activated immune infiltrates expand opportunities for targeted therapy in p53-abnormal endometrial carcinoma. J Pathol 2025. [PMID: 40223796 DOI: 10.1002/path.6429] [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: 10/18/2024] [Revised: 02/12/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025]
Abstract
Tumor protein p53 mutated/abnormal (p53abn) endometrial carcinomas account for over 50% of deaths but comprise only 15% of all endometrial carcinomas. Most patients show limited response to standard-of-care chemotherapy with or without radiotherapy, and only a minority of cases are amenable to targeted therapies like poly-ADP ribose polymerase (PARP) inhibitors and HER2-directed therapies. Recent immunotherapy clinical trials have demonstrated remarkable efficacy, not only in mismatch repair deficient (MMRd) tumors but also in a subset of mismatch repair-proficient (MMRp) tumors. However, the immune microenvironment and its relationship to other therapeutic targets in MMRp endometrial carcinoma remains poorly understood. Here, we characterize the immune microenvironment of p53abn endometrial carcinoma, the most clinically aggressive subtype of MMRp endometrial carcinoma, and correlate antitumor immune signatures with other targetable alterations. We accrued 256 treatment-naïve p53abn endometrial carcinomas and systemically profiled T-cell, B-cell, myeloid, and tumor-cell populations with multiplex immunofluorescence to assess the tissue localization and functional status of immune cells. Shallow whole-genome sequencing was performed on a subset of 126 cases. Patterns of immune infiltration were compared to survival outcomes and mutational signatures. Mixture modeling divided p53abn endometrial carcinoma into tumor-infiltrating lymphocyte (TIL)-rich and TIL-poor subsets. Over 50% of tumors were TIL-rich. TIL-rich cases overexpressed targetable immune evasion molecules and were associated with longer overall and disease-specific survival in multivariate analysis. This effect was particularly pronounced in advanced stage disease and in patients who did not receive adjuvant chemotherapy. TIL did not associate with homologous recombination deficient mutational signatures or HER2 amplification. Our findings demonstrate a biological rationale for immunotherapy in a substantial subset of patients with p53abn endometrial cancer and may help inform combination therapies with immune checkpoint inhibition, PARP inhibitors, and anti-HER2 agents. © 2025 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Spencer D Martin
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Shelby Thornton
- Molecular and Advanced Pathology Core (MAPcore), The University of British Columbia, Vancouver, Canada
| | - Christine Chow
- Molecular and Advanced Pathology Core (MAPcore), The University of British Columbia, Vancouver, Canada
| | - Katy Milne
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - Juliana Sobral de Barros
- Department of Molecular Oncology, British Columbia Cancer Agency, The University of British Columbia, Vancouver, Canada
| | - Kayleigh A Morris
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
| | - Samuel Leung
- Department of Molecular Oncology, British Columbia Cancer Agency, The University of British Columbia, Vancouver, Canada
| | - Amy Jamieson
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, The University of British Columbia, Vancouver, Canada
| | - Brad H Nelson
- Trev and Joyce Deeley Research Centre, British Columbia Cancer Agency, Victoria, Canada
- Department of Medical Genetics, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Dawn R Cochrane
- Department of Molecular Oncology, British Columbia Cancer Agency, The University of British Columbia, Vancouver, Canada
| | - David G Huntsman
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - C Blake Gilks
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Lien Hoang
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| | - Jessica N McAlpine
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, The University of British Columbia, Vancouver, Canada
| | - Allen W Zhang
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, The University of British Columbia, Vancouver, Canada
| |
Collapse
|
16
|
Aronson SL, Thijssen B, Lopez-Yurda M, Koole SN, van der Leest P, León-Castillo A, Harkes R, Seignette IM, Sanders J, Alkemade M, Kemper I, Holtkamp MJ, Mandjes IAM, Broeks A, Lahaye MJ, Rijlaarsdam MA, van den Broek D, Wessels LFA, Horlings HM, van Driel WJ, Sonke GS. Neo-adjuvant pembrolizumab in stage IV high-grade serous ovarian cancer: the phase II Neo-Pembro trial. Nat Commun 2025; 16:3520. [PMID: 40229272 PMCID: PMC11997049 DOI: 10.1038/s41467-025-58440-y] [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: 10/22/2024] [Accepted: 03/24/2025] [Indexed: 04/16/2025] Open
Abstract
While immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, their efficacy in high-grade serous ovarian cancer (HGSOC) remains limited. Some patients, however, achieve lasting responses, emphasizing the need to understand how tumor microenvironment and molecular characteristics influence ICI response. The phase 2 Neo-Pembro study (NCT03126812) included 33 untreated stage IV HGSOC patients, who were scheduled for 6 cycles of carboplatin-paclitaxel and interval cytoreductive surgery. Pembrolizumab (pembro) was added from cycle two and continued for one year. The primary objective was to assess intratumoral immune activation using multiplexed immunofluorescence and immune-related gene expression. Our findings show immune activation, evidenced by an increase in CD3 + , CD8 + , CD8 + /FOXP3+ ratio, TNF-α and interferon-γ signaling. Treatment was well-tolerated. We observed major pathologic responses in 9/33 patients (27%, 95%CI 14-46), with pathologic response strongly associated with immune activation and OS. At a median follow-up of 52.8 months, 8/9 major responders were alive, with 6 patients recurrence-free. In contrast, 4/24 minor responders survived, including one recurrence-free. ctDNA clearance was observed in all major responders and was associated with prolonged PFS and OS. PD-L1 expression and homologous recombination deficiency were predictive of major response and may serve as biomarkers, warranting further exploration. These results suggest major responders may benefit from neo-adjuvant pembro.
Collapse
Affiliation(s)
- S L Aronson
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Center for Gynecologic Oncology Amsterdam, Department of Gynecologic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - B Thijssen
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - M Lopez-Yurda
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - S N Koole
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Center for Gynecologic Oncology Amsterdam, Department of Gynecologic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - P van der Leest
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - A León-Castillo
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - R Harkes
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - I M Seignette
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - J Sanders
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - M Alkemade
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - I Kemper
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M J Holtkamp
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - I A M Mandjes
- Department of Biometrics, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - A Broeks
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - M J Lahaye
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - M A Rijlaarsdam
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - D van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - L F A Wessels
- Division of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, Netherlands
| | - H M Horlings
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - W J van Driel
- Center for Gynecologic Oncology Amsterdam, Department of Gynecologic Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - G S Sonke
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
17
|
Liu X, Wang S, Lv H, Chen E, Yan L, Yu J. Advances in the relationship of immune checkpoint inhibitors and DNA damage repair. Curr Res Transl Med 2025; 73:103494. [PMID: 39824061 DOI: 10.1016/j.retram.2025.103494] [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: 01/22/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
Cancer immunotherapy, alongside surgery, radiation therapy, and chemotherapy, has emerged as a key treatment modality. Immune checkpoint inhibitors (ICIs) represent a promising immunotherapy that plays a critical role in the management of various solid tumors. However, the limited efficacy of ICI monotherapy and the development of primary or secondary resistance to combination therapy remain a challenge. Consequently, identifying molecular markers for predicting ICI efficacy has become an area of active clinical research. Notably, the correlation between DNA damage repair (DDR) mechanisms and the effectiveness of ICI treatment has been established. This review outlines the two primary pathways of DDR, namely, the homologous recombination repair pathway and the mismatch repair pathway. The relationship between these key genes and ICIs has been discussed and the potential of these genes as molecular markers for predicting ICI efficacy summarized.
Collapse
Affiliation(s)
- Xiaolin Liu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Shan Wang
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Hongwei Lv
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Enli Chen
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China
| | - Li Yan
- School of Humanities, Beijing University of Chinese Medicine, Beijing, PR China
| | - Jing Yu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, PR China.
| |
Collapse
|
18
|
Kristeleit R, Devlin MJ, Clamp A, Gourley C, Roux R, Hall M, Nirsimloo R, Kounnis V, Sage L, Narayanan P, Herrington CS, Arora R, Farrelly L, Hughes L, Counsell N, Miller RE. Pembrolizumab in Patients With Advanced Clear Cell Gynecological Cancer: A Phase 2 Nonrandomized Clinical Trial. JAMA Oncol 2025; 11:377-385. [PMID: 39913118 PMCID: PMC11803509 DOI: 10.1001/jamaoncol.2024.6797] [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: 08/15/2024] [Accepted: 12/10/2024] [Indexed: 02/07/2025]
Abstract
Importance Advanced clear cell gynecological cancers (CCGCs) have a poor prognosis, with response rates to second-line chemotherapy less than 8%. Preliminary clinical activity with programmed cell death 1 protein (PD-1) inhibitors reported in CCGC merits further investigation. Objective To assess the clinical benefit of pembrolizumab in patients with previously treated advanced CCGC. Design, Setting, and Participants The PEACOCC trial is a single-arm multicenter phase 2 trial conducted at 5 UK centers investigating the clinical benefit and safety of pembrolizumab. PD-1 inhibitor-naive patients with histologically confirmed advanced CCGC, radiological disease progression following 1 or more prior courses of chemotherapy, and an Eastern Cooperative Oncology Group (ECOG) performance status (PS) score of 0 to 1 were included. Patients were enrolled from March 2019 to October 2021, with data collected until July 2024. Interventions Pembrolizumab, 200 mg, intravenously every 21 days up to 2 years until progression, discontinuation due to toxic effects, or patient/clinician decision. Up to 1 year of retreatment on diseases progression, if stable disease, partial response, or complete response at 2 years. Main Outcomes and Measures The primary end point was progression-free survival (PFS) rate at 12 weeks using Response Evaluation Criteria in Solid Tumors version 1.1 to detect a 12-week PFS rate of 33% or greater and exclude a PFS rate of less than 15%, with 90% power and 1-sided 5% significance level. Secondary end points included objective response rate, duration of response, PFS, overall survival, safety, and quality of life. Results A total of 48 patients were eligible. The median (range) age was 58.5 (32-77) years, and 26 (54%) had an ECOG PS score of 0 and 22 (46%) had an ECOG PS score of 1; 41 (85%) had ovarian, 6 (13%) had endometrial, and 1 (2%) had cervical advanced CCGC. The median (range) courses prior therapy was 3 (1-6); 19 patients (40%) received prior anti-angiogenic therapy, and 19 (40%) had a platinum-free interval of more than 12 months. Grade 3 treatment-related adverse events were observed in 9 patients (19%), and no patients had grade 4 or 5 adverse events. A total of 45 of 46 patients (98%) had mismatch repair-proficient tumors. The 12-week PFS rate was 42% (95% CI, 28-57), and the best objective response rate was 25% (95% CI, 14-40), with 12 partial responses. After a median follow-up of 46.9 months (95% CI, 43.4-55.0), the median PFS was 2.7 months (95% CI, 1.3-5.4), and the median overall survival was 14.8 months (95% CI, 6.7-28.2). Conclusions and Relevance The PEACOCC trial showed clinical benefit with pembrolizumab in patients with previously treated advanced CCGC, of whom all except 1 had MMR-proficient disease. Clinical outcomes were durable with an overall tolerable safety profile, justifying further evaluation of pembrolizumab monotherapy for advanced CCGC in a randomized clinical trial. Trial Registration ClinicalTrials.gov Identifier: NCT03425565.
Collapse
MESH Headings
- Humans
- Female
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Middle Aged
- Aged
- Adult
- Genital Neoplasms, Female/drug therapy
- Genital Neoplasms, Female/pathology
- Genital Neoplasms, Female/mortality
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Adenocarcinoma, Clear Cell/drug therapy
- Adenocarcinoma, Clear Cell/pathology
- Immune Checkpoint Inhibitors/therapeutic use
- Immune Checkpoint Inhibitors/adverse effects
- Progression-Free Survival
- Treatment Outcome
Collapse
Affiliation(s)
- Rebecca Kristeleit
- Guy’s and St Thomas’ NHS Foundation Trust and Comprehensive Cancer Centre, King’s College London, London, United Kingdom
| | | | - Andrew Clamp
- The Christie NHS Foundation Trust and University of Manchester, Manchester, United Kingdom
| | - Charlie Gourley
- Edinburgh Cancer Centre, Western General Hospital, Edinburgh, United Kingdom
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research United Kingdom Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - René Roux
- Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, United Kingdom
| | - Marcia Hall
- Hillingdon Hospitals NHS Foundation Trust, Mount Vernon Hospital, Middlesex, United Kingdom
| | - Rachel Nirsimloo
- Edinburgh Cancer Centre, Western General Hospital, Edinburgh, United Kingdom
| | - Valentinos Kounnis
- Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, United Kingdom
| | - Lesley Sage
- Gynae-Oncology Trials Group United Kingdom and Ovacome, London, United Kingdom
| | | | - C. Simon Herrington
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research United Kingdom Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Rupali Arora
- University College London Hospitals, London, United Kingdom
| | - Laura Farrelly
- Cancer Research United Kingdom & UCL Cancer Trials Centre, University College London, London, United Kingdom
| | - Laura Hughes
- Cancer Research United Kingdom & UCL Cancer Trials Centre, University College London, London, United Kingdom
| | - Nicholas Counsell
- Cancer Research United Kingdom & UCL Cancer Trials Centre, University College London, London, United Kingdom
| | - Rowan E. Miller
- Queen Mary University of London, London, United Kingdom
- University College London Hospitals, London, United Kingdom
| |
Collapse
|
19
|
Santoro A, Angelico G, Travaglino A, Inzani F, Spadola S, Pettinato A, Mazzucchelli M, Bragantini E, Maccio L, Zannoni GF. The multiple facets of ovarian high grade serous carcinoma: A review on morphological, immunohistochemical and molecular features. Crit Rev Oncol Hematol 2025; 208:104603. [PMID: 39732305 DOI: 10.1016/j.critrevonc.2024.104603] [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: 09/15/2024] [Revised: 12/06/2024] [Accepted: 12/20/2024] [Indexed: 12/30/2024] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the most aggressive subtype of epithelial ovarian cancer and a leading cause of mortality among gynecologic malignancies. This review aims to comprehensively analyze the morphological, immunohistochemical, and molecular features of HGSOC, highlighting its pathogenesis and identifying biomarkers with diagnostic, prognostic, and therapeutic significance. Special emphasis is placed on the role of tumor microenvironment (TME) and genomic instability in shaping the tumor's behavior and therapeutic vulnerabilities. Key advancements, such as the identification of TP53 and BRCA mutations, the classification of homologous recombination repair (HRR) deficiencies, and the clinical implications of biomarkers like folate receptor alpha (FRα) and PD-L1 are discussed. These findings reveal actionable insights into targeted therapies, including immune checkpoint inhibitors and PARP inhibitors, which hold promise for improving outcomes in HGSOC. This synthesis of knowledge aims to bridge gaps in understanding HGSOC's multifaceted biology, enhance clinical decision-making, and foster the development of precision therapies.
Collapse
MESH Headings
- Humans
- Female
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/diagnosis
- Ovarian Neoplasms/therapy
- Cystadenocarcinoma, Serous/pathology
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/diagnosis
- Cystadenocarcinoma, Serous/therapy
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- Tumor Microenvironment
- Neoplasm Grading
- Immunohistochemistry
- Prognosis
- Carcinoma, Ovarian Epithelial/pathology
- Carcinoma, Ovarian Epithelial/genetics
- Carcinoma, Ovarian Epithelial/metabolism
- Mutation
Collapse
Affiliation(s)
- Angela Santoro
- Pathology Institute, Catholic University of Sacred Heart, Rome 00168, Italy; Pathology Unit, Department of Woman and Child's Health and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy
| | - Giuseppe Angelico
- Department of Medicine and Surgery, Kore University of Enna, Enna 94100, Italy
| | - Antonio Travaglino
- Pathology Unit, Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Frediano Inzani
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS San Matteo Hospital, Pavia 27100, Italy
| | - Saveria Spadola
- Department of Medicine and Surgery, Kore University of Enna, Enna 94100, Italy
| | - Angela Pettinato
- Department of Pathological Anatomy, A.O.E. Cannizzaro, Via Messina, 829, Catania 95126, Italy
| | - Manuel Mazzucchelli
- Department of Medical and Surgical Sciences and Advanced Technologies "G.F. Ingrassia", Anatomic Pathology, University of Catania, Catania, Italy
| | - Emma Bragantini
- Unit of Surgical Pathology, Santa Chiara Hospital, APSS, Trento, Italy
| | - Livia Maccio
- Unit of Surgical Pathology, Santa Chiara Hospital, APSS, Trento, Italy
| | - Gian Franco Zannoni
- Pathology Institute, Catholic University of Sacred Heart, Rome 00168, Italy; Pathology Unit, Department of Woman and Child's Health and Public Health Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome 00168, Italy.
| |
Collapse
|
20
|
Maurac Pašalić I, Sabol M, Prtenjača E, Puvačić Solomun L, Pavić M. Mature cystic teratoma and their malignant transformation. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109641. [PMID: 40348473 DOI: 10.1016/j.ejso.2025.109641] [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: 12/05/2024] [Revised: 01/17/2025] [Accepted: 01/24/2025] [Indexed: 05/14/2025]
Abstract
Mature cystic teratomas (MCT) are benign germ cells tumors which are fairly common and are often diagnosed incidentally. In most cases they don't present a significant risk to survival, but in some rare cases they can progress to a malignant phenotype. In such cases the treatment must be adjusted to the new conditions and tailored to suit the malignant phenotype. In this review, we aim to describe the main clinical and biological features of various types of recorded malignant transformations and/or complications, as well as stress their differences in clinical behavior and therapeutic options. The management of benign vs. malignant MCT differs greatly and a specific subtype of transformation should be taken into consideration when designing therapy, as it can greatly improve the survival and quality of life.
Collapse
Affiliation(s)
- Ivana Maurac Pašalić
- School of Medicine, University of Zagreb, Zagreb, Croatia; Department of Gynecology and Obstetrics, University Hospital Centre Zagreb, Zagreb, Croatia.
| | - Maja Sabol
- Laboratory for Hereditary Cancer, Division of Molecular Medicine, Ruđer Bošković Institute, Zagreb, Croatia.
| | | | | | - Mato Pavić
- School of Medicine, University of Zagreb, Zagreb, Croatia; Department of Gynecology and Obstetrics, University Hospital Centre Zagreb, Zagreb, Croatia.
| |
Collapse
|
21
|
Ying F, Zhou X, Chen M, Huang L, Gao L, Zhao Q, Zhang Y. Preclinical study of inetetamab combined with atezolizumab to synergistically inhibit HER2 and PD-L1 in the treatment of ovarian cancer. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200938. [PMID: 40034965 PMCID: PMC11874541 DOI: 10.1016/j.omton.2025.200938] [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: 07/18/2024] [Revised: 12/14/2024] [Accepted: 01/14/2025] [Indexed: 03/05/2025]
Abstract
Epithelial ovarian cancer (EOC) is the deadliest gynecological malignancy. Precision treatments are crucial for improving patient survival. This research explored the potential anti-tumor effects of combining inetetamab and atezolizumab for HER2+ EOC patients. The expressions of human epidermal growth factor receptor 2 (HER2) and programmed cell death ligand 1 (PD-L1) in EOC cells were evaluated. EOC cell-derived subcutaneous and peritoneal dissemination mouse models were used to evaluate the anti-tumor effects of inetetamab, with or without atezolizumab. The correlations between the expressions of HER2 and PD-L1 as well as the infiltration of T cells in tumors from patients and mice were analyzed by immunohistochemistry. Inetetamab suppressed the growth of HER2+ tumors in mouse models. HER2 overexpression increased PD-L1 levels in EOC cells. The expression level of HER2 is positively related to that of PD-L1 in the tumors of EOC patients as well as the infiltration of both CD4+ and CD8+ T cells. The combination of inetetamab and atezolizumab impeded the growth of HER2+ EOC tumors in vivo and induced a long-term anti-tumor effect with the elevated infiltration of CD103+CD8+ cells. These findings suggest that the combination of inetetamab and atezolizumab could be a promising precision treatment strategy for HER2+ EOC patients.
Collapse
Affiliation(s)
- Feiquan Ying
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xuyang Zhou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Department of Medical Ultrasound, Yueyang Central Hospital, Yueyang 414000, China
| | - Mengqing Chen
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lin Huang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lingling Gao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qing Zhao
- Department of Obstetrics and Gynecology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
| | - Yuan Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
22
|
Scattolin T, Cavarzerani E, Alessi D, Mauceri M, Botter E, Tonon G, Caligiuri I, Repetto O, Kamensek U, Brezar SK, Dalla Pozza M, Palazzolo S, Cemazar M, Canzonieri V, Demitri N, Nolan SP, Gasser G, Visentin F, Rizzolio F. Unlocking the potential of organopalladium complexes for high-grade serous ovarian cancer therapy. Dalton Trans 2025; 54:4685-4696. [PMID: 39967474 DOI: 10.1039/d5dt00194c] [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: 02/20/2025]
Abstract
High-Grade Serous Ovarian Cancer (HGSOC) is the most common and lethal subtype of ovarian cancer, known for its high aggressiveness and extensive genomic alterations. Typically diagnosed at an advanced stage, HGSOC presents formidable challenges in drug therapy. The limited efficacy of standard treatments, development of chemoresistance, scarcity of targeted therapies, and significant tumor heterogeneity render this disease incurable with current treatment options, highlighting the urgent need for novel therapeutic approaches to improve patient outcomes. In this study we report a straightforward and stereoselective synthetic route to novel Pd(II)-vinyl and -butadienyl complexes bearing a wide range of monodentate and bidentate ligands. Most of the synthesized complexes exhibited good to excellent in vitro anticancer activity against ovarian cancer cells. Particularly promising is the water-soluble complex bearing two PTA (1,3,5-triaza-7-phosphaadamantane) ligands and the Pd(II)-butadienyl fragment. This compound combines excellent cytotoxicity towards cancer cells with substantial inactivity towards non-cancerous ones. This derivative was selected for further studies on ex vivo tumor organoids and in vivo mouse models, which demonstrate its remarkable efficacy with surprisingly low collateral toxicity even at high dosages. Moreover, this class of compounds appears to operate through a ferroptotic mechanism, thus representing the first such example for an organopalladium compound.
Collapse
Affiliation(s)
- Thomas Scattolin
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Marzolo 1, 35131 Padova, Italy.
| | - Enrico Cavarzerani
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Dario Alessi
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Marzolo 1, 35131 Padova, Italy.
| | - Matteo Mauceri
- Dipartimento di Scienze Chimiche, Università degli Studi di Padova, via Marzolo 1, 35131 Padova, Italy.
| | - Eleonora Botter
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Giovanni Tonon
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Isabella Caligiuri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Ombretta Repetto
- Immunopathology and Cancer Biomarkers, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Urska Kamensek
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Simona Kranjc Brezar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Maria Dalla Pozza
- ChimieParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Stefano Palazzolo
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana, Ljubljana, 1000, Slovenia
| | - Vincenzo Canzonieri
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
- Department of Medical, Surgical and Health Sciences, Università degli Studi di Trieste, Strada di Fiume 447, Trieste, Italy
| | - Nicola Demitri
- Area Science Park, Elettra-Sincrotrone Trieste, S.S. 14 Km 163.5, Basovizza, 34149, Trieste, Italy
| | - Steven P Nolan
- Department of Chemistry and Centre for Sustainable Chemistry, Ghent University Krijgslaan 281, S-3, 9000 Ghent, Belgium
| | - Gilles Gasser
- ChimieParisTech, PSL University, CNRS, Institute of Chemistry for Life and Health Sciences, Laboratory for Inorganic Chemical Biology, 75005, Paris, France
| | - Fabiano Visentin
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
| | - Flavio Rizzolio
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174 Venezia-Mestre, Italy.
- Pathology Unit, Centro di Riferimento Oncologico di Aviano (C.R.O.) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| |
Collapse
|
23
|
Gaillard S, Verma N, Berg M, Harrison J, Huang P, Leatherman JM, Doucet M, Sen R, Suru A, Cai H, Durham J, Jelovac D, Cimino-Mathews A, Cherry C, Ganguly S, Emens LA. A clinical study of tremelimumab, alone or in combination with olaparib, for recurrent epithelial ovarian cancer. Gynecol Oncol 2025; 194:41-47. [PMID: 39951918 PMCID: PMC11993342 DOI: 10.1016/j.ygyno.2025.01.015] [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: 12/17/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVE PARP inhibitors may work synergistically to improve the efficacy of immunotherapy in patients with epithelial ovarian cancer (EOC). We performed a parallel-arm study of tremelimumab, alone or with olaparib, in patients with recurrent EOC. METHODS Eligibility criteria included measurable disease and progression <12 months from last platinum. Participants were randomized to Arm A (tremelimumab monotherapy, 10 mg/kg/dose intravenously [IV]) or Arm B (dose level 1 [DL1] olaparib orally 150 mg twice daily with tremelimumab IV 3 mg/kg/dose and DL2 olaparib orally 150 mg twice daily with tremelimumab IV 10 mg/kg/dose). Primary objectives were safety, change in peripheral ICOS+ T cells, and identification of optimal dose combination. RESULTS Among 24 total patients (12 on Arm A, 6 on Arm B-DL1, 6 on Arm B-DL2), the most common grade 3 toxicities were rash (13 %), immune-mediated hepatitis (8 %), and colitis (8 %). No grade ≥ 4 toxicities were identified. No dose-limiting toxicities were identified. One patient (Arm B-DL2) experienced a partial response; no complete responses were observed. Ten patients (7 on Arm A, 2 on Arm B-DL2, and 1 on Arm B-DL1) had a best response of stable disease. There was a significant increase in CD4+ICOS+ and CD8+ICOS+ T cells at both C1D15 and C1D22 in groups treated with tremelimumab IV 10 mg/kg/dose, but not in those treated with tremelimumab 3 mg/kg/dose. CONCLUSIONS Tremelimumab IV 10 mg/kg/dose with olaparib 150 mg orally twice daily was safe and feasible. Tremelimumab 10 mg/kg/dose (as opposed to 3 mg/kg/dose) was required for immune activation, although this did not translate into clinical responses.
Collapse
Affiliation(s)
- Stéphanie Gaillard
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America; Department of Gynecology and Obstetrics Division of Gynecologic Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America.
| | - Neha Verma
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Maureen Berg
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Jeanne Harrison
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Peng Huang
- Department of Oncology Division of Biostatistics and Bioinformatics, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - James M Leatherman
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Michele Doucet
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Rupashree Sen
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Aditya Suru
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Hongyan Cai
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Jennifer Durham
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Danijela Jelovac
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Ashley Cimino-Mathews
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America; Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Christopher Cherry
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Sudipto Ganguly
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| | - Leisha A Emens
- Department of Oncology Johns Hopkins School of Medicine, Baltimore, MD, United States of America
| |
Collapse
|
24
|
Lu Y, Zhang Y, Li W, Jiang H, Wang J, Guo X. Tumor Cell-Expressed Herpesvirus Entry Mediator Regulates Proliferation and Adaptive Immunity in Ovarian Cancer. Immun Inflamm Dis 2025; 13:e70175. [PMID: 40105652 PMCID: PMC11921469 DOI: 10.1002/iid3.70175] [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: 05/12/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Ovarian cancer (OvCa) is a prevalent gynecological malignancy with an increasing incidence and high mortality rate. Although the role of the herpesvirus entry mediator (HVEM), encoded by the TNFRSF14 gene, is currently considered pivotal in various types of cancer, the regulation of tumor cell-expressed HVEM in OvCa remains inadequately understood. METHODS Specimens were used to detect HVEM expression via quantitative RT-PCR and flow cytometry. The proliferation of the murine OvCa cell line ID8 was determined using the Cell Counting Kit-8, colony formation, and EdU staining assays. The immune constituents within the ascites fluid and spleen of tumor-bearing mice were analyzed by flow cytometry. Bioinformatics analysis was performed to explore cytokines, chemokines, and signaling pathways regulated by HVEM, and differential expression levels were confirmed via quantitative RT-PCR and western blot analysis. RESULTS Herein, we identified a significant upregulation of HVEM in OvCa tissues compared with that in benign tissues and observed dominant expression of HVEM in CD45⁻EpCAM⁺ subsets in OvCa specimens. Tumor cell-expressed HVEM was found to promote OvCa cell proliferation by partly activating spliced X-box-binding protein 1 (XBP1s)-c-Myc signaling. In mouse models, knockdown of Tnfrsf14 in ID8 cells alleviated OvCa progression and specifically affected the frequency and function of T cells in the ascites fluid and spleen. In addition, tumor cell-expressed HVEM altered chemokine expression (CXCL1/9/10/11 and CCL2/4/5) and STAT signal activation (STAT5 and STAT6) in ID8 cells. CONCLUSION This study investigated the effects of HVEM on OvCa and validated its potential as a therapeutic marker for treating OvCa.
Collapse
Affiliation(s)
- Yun Lu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Yijun Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Wenxuan Li
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Haonan Jiang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Jiapo Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal‐Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| | - Xiaoqing Guo
- Department of Gynecological Oncology, Shanghai First Maternity and Infant Hospital, School of MedicineTongji UniversityShanghaiChina
| |
Collapse
|
25
|
Zhong Y, Wang Y, Wang C, Cao K, Wang X, Xu X, Yang M, Zhang G, Liu H, Lu J. Targeting mesothelin-CD24 axis repolarizes tumor-associated macrophages to potentiate PD-1 blockade therapy in high-grade serous ovarian cancer. J Immunother Cancer 2025; 13:e011230. [PMID: 40010770 PMCID: PMC11873354 DOI: 10.1136/jitc-2024-011230] [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: 12/05/2024] [Accepted: 02/03/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND High-grade serous ovarian cancer (HGSOC) is a highly aggressive malignancy marked by an immunosuppressive tumor microenvironment that hinders effective immune responses. A key feature of this environment is the extensive infiltration of myeloid cells, which contributes to immune evasion. This study explored how mesothelin (MSLN), a tumor-associated antigen, modulates the expression of CD24, an emerging target for immune modulation, and its role in promoting immune evasion in HGSOC. Understanding these underlying mechanisms is crucial for enhancing the efficacy of immune checkpoint blockade (ICB) therapies and improving outcomes in patients with HGSOC. METHODS We analyzed the expression of MSLN in HGSOC samples and examined its correlation with clinical outcome. In vitro and in vivo models were used to explore how MSLN influences CD24 expression and the polarization of tumor-associated macrophages (TAMs). We also investigated the role of MSLN in the activation of Wnt/β-catenin signaling and its impact on T-cell function and antitumor immunity. The effects of Msln knockdown on CD24 expression and the response to anti-programmed cell death protein-1 (PD-1) therapy were evaluated in syngeneic mouse models. RESULTS MSLN expression was found to be significantly elevated in HGSOC, with high MSLN levels correlating with poor prognosis and resistance to ICB. MSLN upregulated CD24 and promoted the protumorigenic polarization of TAMs, contributing to T-cell dysfunction. Mechanistically, MSLN activated Wnt/β-catenin signaling, which in turn enhanced CD24 expression. This activation forms a positive feedback loop that further promotes MSLN transcription. In contrast, Msln knockdown reduced CD24 expression, relieved cytotoxic T-cell suppression, and significantly improved the efficacy of anti-PD-1 therapy in syngeneic models. CONCLUSIONS This study elucidates the critical role of MSLN in immune evasion in HGSOC and its underlying mechanisms. Targeting MSLN in combination with ICB is a promising strategy to enhance the efficacy of immunotherapy and improve patient outcomes in HGSOC.
Collapse
Affiliation(s)
- Yujing Zhong
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yiying Wang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Obstetrics and Gynecology Hospital, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Chenyang Wang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Kankan Cao
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xueling Wang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xuyao Xu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Moran Yang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Obstetrics and Gynecology Hospital, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Guodong Zhang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Obstetrics and Gynecology Hospital, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Haiou Liu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Obstetrics and Gynecology Hospital, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jiaqi Lu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Obstetrics and Gynecology Hospital, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
26
|
Huang R, Nakamura B, Senguttuvan R, Li YJ, Martincuks A, Bakkar R, Song M, Ann DK, Rodriguez-Rodriguez L, Yu H. A Critical Role of Intracellular PD-L1 in Promoting Ovarian Cancer Progression. Cells 2025; 14:314. [PMID: 39996786 PMCID: PMC11853747 DOI: 10.3390/cells14040314] [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: 01/14/2025] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
Disrupting the interaction between tumor-cell surface PD-L1 and T cell membrane PD-1 can elicit durable clinical responses. However, only about 10% of ovarian cancer patients respond to PD-1/PD-L1 blockade. Here, we show that PD-L1 expression in ovarian cancer-patient tumors is predominantly intracellular. Notably, PARP inhibitor treatment highly increased intracellular PD-L1 accumulation in both ovarian cancer-patient tumor samples and cell lines. We investigated whether intracellular PD-L1 might play a critical role in ovarian cancer progression. Mutating the PD-L1 acetylation site in PEO1 and ID8Brca1-/- ovarian cancer cells significantly decreased PD-L1 levels and impaired colony formation, which was accompanied by cell cycle G2/M arrest and apoptosis induction. PEO1 and ID8Brca1-/- tumors with PD-L1 acetylation site mutation also exhibited significantly reduced growth in mice. Furthermore, targeting intracellular PD-L1 with a cell-penetrating antibody effectively decreased ovarian tumor-cell intracellular PD-L1 level and induced tumor-cell growth arrest and apoptosis, as well as enhanced DNA damage and STING activation, both in vitro and in vivo. In conclusion, we have shown the critical role of intracellular PD-L1 in ovarian cancer progression.
Collapse
Affiliation(s)
- Rui Huang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.H.); (Y.-J.L.); (A.M.)
| | - Brad Nakamura
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (B.N.); (R.S.); (M.S.)
| | - Rosemary Senguttuvan
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (B.N.); (R.S.); (M.S.)
| | - Yi-Jia Li
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.H.); (Y.-J.L.); (A.M.)
| | - Antons Martincuks
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.H.); (Y.-J.L.); (A.M.)
| | - Rania Bakkar
- Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Mihae Song
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (B.N.); (R.S.); (M.S.)
| | - David K. Ann
- Department of Diabetes Complication and Metabolism, Arthur Riggs Diabetes & Metabolism Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA;
| | - Lorna Rodriguez-Rodriguez
- Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA; (B.N.); (R.S.); (M.S.)
| | - Hua Yu
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA; (R.H.); (Y.-J.L.); (A.M.)
| |
Collapse
|
27
|
Li J, Zhou X, Wu L, Ma J, Tan Y, Wu S, Zhu J, Wang Q, Shi Q. Optimal early endpoint for second-line or subsequent immune checkpoint inhibitors in previously treated advanced solid cancers: a systematic review. BMC Cancer 2025; 25:293. [PMID: 39966752 PMCID: PMC11837729 DOI: 10.1186/s12885-025-13712-0] [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: 06/01/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND The administration of second-line or subsequent immune checkpoint inhibitors (ICIs) in previously treated patients with advanced solid cancers has been clinically investigated. However, previous clinical trials lacked an appropriate primary endpoint for efficacy assessment. This systematic review aimed to explore the most optimal early efficacy endpoint for such trials. METHODS Phase 2 or 3 clinical trials involving patients with advanced solid cancers with disease progression following standard first-line therapy receiving second-line or subsequent ICI administration, with adequate survival outcome data, were included from PubMed, Embase, Web of Science, and Cochrane Library databases before February 2023. Quality assessment was conducted using the Cochrane tool and Newcastle-Ottawa Quality Assessment Scale for Cohort Studies for randomized controlled trials (RCTs) and non-randomized trials, respectively. Objective response rate (ORR) and progression-free survival (PFS) at 3, 6, and 9 months were investigated as potential early efficacy endpoint candidates for 12-month overall survival (OS), with a strong correlation defined as Pearson's correlation coefficient r ≥ 0.8. RESULTS A total of 64 RCTs comprising 22,725 patients and 106 non-randomized prospective trials involving 10,608 participants were eligible for modeling and external validation, respectively. RCTs examined 15 different cancer types, predominantly non-small-cell lung cancer (NSCLC) (17, 28%), melanoma (9, 14%), and esophageal squamous cell carcinoma (5, 8%). The median sample size of RCTs was 124 patients, and the median follow-up time was 3.2-57.7 months. The ORR (r = 0.38; 95% confidence interval [CI], 0.18-0.54) and PFS (r = 0.42; 95% CI, 0.14-0.64) exhibited weak trial-level correlations with OS. Within ICI treatment arms, the r values of ORR and 3-, 6-, and 9-month PFS with 12-month OS were 0.61 (95% CI, 0.37-0.79), 0.78 (95% CI, 0.62-0.88), 0.84 (95% CI, 0.77-0.90), and 0.86 (95% CI, 0.79-0.90), respectively. External validation of 6-month PFS indicated an acceptable discrepancy between actual and predicted 12-month OS. CONCLUSIONS In non-randomized phase 2 trials on second-line or subsequent ICI therapy in patients with advanced solid cancers, 6-month PFS could serve as an early efficacy endpoint. However, early efficacy endpoints are not recommended in RCTs to replace OS.
Collapse
Affiliation(s)
- Jingqiu Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoding Zhou
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Wu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiabao Ma
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Tan
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Songke Wu
- Department of Oncology, People'S Hospital of Cangxi County, Guangyuan, China.
| | - Jie Zhu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| | - Qifeng Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| | - Qiuling Shi
- Center for Cancer Prevention Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| |
Collapse
|
28
|
Bao Y, Cruz G, Zhang Y, Qiao Y, Mannan R, Hu J, Yang F, Gondal M, Shahine M, Kang S, Mahapatra S, Chu A, Choi JE, Yu J, Lin H, Miner SJ, Robinson DR, Wu YM, Zheng Y, Cao X, Su F, Wang R, Hosseini N, Cieslik M, Kryczek I, Vaishampayan U, Zou W, Chinnaiyan AM. The UBA1-STUB1 Axis Mediates Cancer Immune Escape and Resistance to Checkpoint Blockade. Cancer Discov 2025; 15:363-381. [PMID: 39540840 PMCID: PMC11803397 DOI: 10.1158/2159-8290.cd-24-0435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/12/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024]
Abstract
SIGNIFICANCE Our study reveals UBA1 as a predictive biomarker for clinical outcomes in ICB cohorts, mediating cancer immune evasion and ICB resistance. We further highlight JAK1 stabilization as a key mechanism of UBA1 inhibition and nominate the UBA1-STUB1 axis as an immuno-oncology therapeutic target to improve the efficacy of ICB.
Collapse
Affiliation(s)
- Yi Bao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Gabriel Cruz
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yuping Zhang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yuanyuan Qiao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
| | - Rahul Mannan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Jing Hu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fan Yang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Mahnoor Gondal
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Miriam Shahine
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Sarah Kang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
| | - Somnath Mahapatra
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Alec Chu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Jae Eun Choi
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Jiali Yu
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, Michigan
| | - Heng Lin
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, Michigan
| | - Stephanie J. Miner
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Dan R. Robinson
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yi-Mi Wu
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Yang Zheng
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Xuhong Cao
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Fengyun Su
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Rui Wang
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Noshad Hosseini
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Marcin Cieslik
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan
| | - Ilona Kryczek
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, Michigan
| | - Ulka Vaishampayan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Weiping Zou
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Department of Surgery, University of Michigan, Ann Arbor, Michigan
- Center of Excellence for Cancer Immunology and Immunotherapy, University of Michigan, Ann Arbor, Michigan
| | - Arul M. Chinnaiyan
- Michigan Center for Translational Pathology, University of Michigan, Ann Arbor, Michigan
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan
- Howard Hughes Medical Institute, University of Michigan, Ann Arbor, Michigan
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
29
|
Block MS, Clubb JHA, Mäenpää J, Pakola S, Quixabeira DCA, Kudling T, Jirovec E, Haybout L, van der Heijden M, Zahraoui S, Grönberg-Vähä-Koskela S, Raatikainen S, Arias V, Basnet S, Ojala N, Pellinen T, Hemmes A, Välimäki K, Pasanen A, Alanko T, Adamo D, Ramadan S, Sormunen J, Kononen J, Cohen JW, Chisamore MJ, Goldfinch J, Sorsa S, Havunen R, Kistler C, Kalervo A, Cervera-Carrascon V, Dos Santos JM, Hemminki A. The oncolytic adenovirus TILT-123 with pembrolizumab in platinum resistant or refractory ovarian cancer: the phase 1a PROTA trial. Nat Commun 2025; 16:1381. [PMID: 39910037 PMCID: PMC11799410 DOI: 10.1038/s41467-025-56482-w] [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: 05/21/2024] [Accepted: 01/17/2025] [Indexed: 02/07/2025] Open
Abstract
Immune checkpoint inhibitors have demonstrated modest efficacy as a monotherapy in ovarian cancer. Originally developed to improve efficacy of T-cell therapies such as immune checkpoint inhibitors and adoptive cell transfer, TILT-123 (Ad5/3-E2F-D24-hTNFα-IRES-hIL-2) is a serotype chimeric oncolytic adenovirus encoding tumor necrosis factor alpha and interleukin-2. Here we report results from phase 1a of PROTA, a single-arm, multicentre dose escalation trial with TILT-123 and pembrolizumab in female patients with platinum resistant or refractory ovarian cancer (NCT05271318). The primary endpoint was safety. Secondary endpoints included efficacy, tolerability, virus persistence and anti-viral immunity. Patients (n = 15) received intravenous and intraperitoneal and/or intratumoral injections of TILT-123 as well as intravenous pembrolizumab. Treatment was well tolerated, and no dose-limiting toxicities were observed. The most frequent adverse events were fever (40%), fatigue (40%) and nausea (40%). Disease control was achieved in 64% of evaluable patients (9/14). Median progression-free survival and overall survival were 98 and 190 days respectively. Clinical responses were associated with higher serum anti-adenovirus neutralizing antibody titer at baseline and post-treatment. The phase 1b investigating TILT-123, pembrolizumab and PEGylated liposomal doxorubicin in a similar patient population is underway.
Collapse
Affiliation(s)
| | - James Hugo Armstrong Clubb
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Johanna Mäenpää
- Docrates Cancer Center, Helsinki, Finland
- Tampere University, Faculty of Medicine and Medical Technology, Tampere, Finland
| | - Santeri Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Dafne Carolina Alves Quixabeira
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Tatiana Kudling
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Elise Jirovec
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Lyna Haybout
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Mirte van der Heijden
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | | | | | | | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Saru Basnet
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Nea Ojala
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Teijo Pellinen
- Digital Microscopy and Molecular Pathology Unit, Institute for Molecular Medicine Finland, Helsinki, Finland
| | - Annabrita Hemmes
- Digital Microscopy and Molecular Pathology Unit, Institute for Molecular Medicine Finland, Helsinki, Finland
| | - Katja Välimäki
- Digital Microscopy and Molecular Pathology Unit, Institute for Molecular Medicine Finland, Helsinki, Finland
| | - Annukka Pasanen
- Department of Pathology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | | | | | | | | | | | | | | | - Suvi Sorsa
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Riikka Havunen
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | | | | | - Víctor Cervera-Carrascon
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - João Manuel Dos Santos
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Akseli Hemminki
- TILT Biotherapeutics Ltd, Helsinki, Finland.
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.
- Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
30
|
Dong S, Ye C, Li B, Lv F, Zhang L, Yang S, Wang F, Zhu M, Zhou M, Guo F, Li Z, Peng L, Ji C, Lu X, Cheng Y, Ren X, Chen Y, Zhou J, Yang J, Zhang Y. Targeting of Tumoral NAC1 Mitigates Myeloid-Derived Suppressor Cell-Mediated Immunosuppression and Potentiates Anti-PD-1 Therapy in Ovarian Cancer. Cancer Immunol Res 2025; 13:286-302. [PMID: 39531476 DOI: 10.1158/2326-6066.cir-24-0084] [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: 01/24/2024] [Revised: 06/01/2024] [Accepted: 11/12/2024] [Indexed: 11/16/2024]
Abstract
Epithelial ovarian cancer is the most common type of ovarian cancer with a low rate of response to immunotherapy such as immune checkpoint blockade therapy. In this study, we report that nucleus accumbens-associated protein 1 (NAC1), a putative driver of epithelial ovarian cancer, has a critical role in immune evasion. We showed in murine ovarian cancer models that depleting or inhibiting tumoral NAC1 reduced the recruitment and immunosuppressive function of myeloid-derived suppressor cells (MDSC) in the tumor microenvironment, led to significant increases of cytotoxic tumor-infiltrating CD8+ T cells, and promoted antitumor immunity and suppressed tumor progression. We further showed that tumoral NAC1 directly enhanced the transcription of CXCL16 by binding to CXCR6, thereby promoting MDSC recruitment to the tumor. Moreover, lipid C20:1T produced by NAC1-expressing tumor cells fueled oxidative metabolism of MDSCs and promoted their immune-suppressive function. We also showed that NIC3, a small-molecule inhibitor of NAC1, was able to sensitize mice bearing NAC1-expressing ovarian tumors to anti-PD-1 therapy. Our study reveals a critical role for NAC1 in controlling tumor infiltration of MDSCs and in modulating the efficacy of immune checkpoint blockade therapy. Thus, targeting of NAC1 may be exploited to sensitize ovarian cancer to immunotherapy.
Collapse
Affiliation(s)
- Shunli Dong
- Center of Translational Medicine, First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Cong Ye
- Center of Translational Medicine, First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Bin Li
- Department of Central Laboratory, Suzhou Ninth Hospital Affiliated to Soochow University, Soochow University, Suzhou, China
| | - Fanglin Lv
- Center of Translational Medicine, First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Lu Zhang
- Center of Translational Medicine, First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Shumin Yang
- Center of Translational Medicine, First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Fang Wang
- Department of Gynecology and Obstetrics, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Mingxian Zhu
- Center of Translational Medicine, First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Mingxuan Zhou
- Center of Translational Medicine, First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Fanfan Guo
- Center of Translational Medicine, First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zhenyun Li
- Center of Translational Medicine, First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Lei Peng
- Center of Translational Medicine, First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Cheng Ji
- Department of Respiratory Medicine, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Xialiang Lu
- Department of Pathology, Suzhou Ninth Hospital Affiliated to Soochow University, Soochow University, Suzhou, China
| | - Yan Cheng
- Center of Translational Medicine, First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xingcong Ren
- Department of Cancer Biology and Toxicology, Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Youguo Chen
- Department of Gynecology and Obstetrics, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Jinhua Zhou
- Department of Gynecology and Obstetrics, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Jinming Yang
- Department of Cancer Biology and Toxicology, Markey Cancer Center, College of Medicine, University of Kentucky, Lexington, Kentucky
| | - Yi Zhang
- Center of Translational Medicine, First People's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang, China
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| |
Collapse
|
31
|
Doro Pereira L, Wielgos-Bonvallet M, Misirlioglu S, Khodadai-Jamayran A, Jelinic P, Levine DA. PARP Inhibitors Differentially Regulate Immune Responses in Distinct Genetic Backgrounds of High-Grade Serous Tubo-Ovarian Carcinoma. CANCER RESEARCH COMMUNICATIONS 2025; 5:339-348. [PMID: 39851178 PMCID: PMC11836641 DOI: 10.1158/2767-9764.crc-24-0515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 12/02/2024] [Accepted: 01/21/2025] [Indexed: 01/26/2025]
Abstract
SIGNIFICANCE This work highlights how different PARPis, especially talazoparib, modulate immune-related gene expression in ovarian cancer cells, independent of the cGAS-STING pathway. These findings may improve our understanding of how different PARPis affect the immune system in various genetic backgrounds.
Collapse
Affiliation(s)
- Luiza Doro Pereira
- Division of Gynecologic Oncology, Department of OB/GYN, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Monica Wielgos-Bonvallet
- Division of Gynecologic Oncology, Department of OB/GYN, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Selim Misirlioglu
- Division of Gynecologic Oncology, Department of OB/GYN, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Alireza Khodadai-Jamayran
- Department of Pathology, Applied Bioinformatics Laboratories, NYU School of Medicine, New York, New York
| | - Petar Jelinic
- Division of Gynecologic Oncology, Department of OB/GYN, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Douglas A. Levine
- Division of Gynecologic Oncology, Department of OB/GYN, Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| |
Collapse
|
32
|
Ricciuti J, Liu Q, Khan ANMNH, Joseph JM, Veuskens BR, Giridharan T, Suzuki S, Emmons TR, Yaffe MB, Kuijpers TW, Jongerius I, Brouwer MC, Pouw RB, Odunsi K, Frederick P, Mager KL, Lele S, Gaulin N, Hakim C, Edwards RP, Olawaiye AB, Sukumanovich P, Taylor S, Elishaev E, Zsiros E, Modugno F, Moysich KB, Segal BH. Prognostic significance of serum complement activation, neutrophil extracellular traps and extracellular DNA in newly diagnosed epithelial ovarian cancer. Gynecol Oncol 2025; 193:49-57. [PMID: 39764857 PMCID: PMC11871990 DOI: 10.1016/j.ygyno.2024.12.006] [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: 08/19/2024] [Revised: 11/25/2024] [Accepted: 12/05/2024] [Indexed: 02/02/2025]
Abstract
PURPOSE We observed that the tumor microenvironment (TME) in metastatic epithelial ovarian cancer (EOC) and in other solid tumors can reprogram normal neutrophils to acquire a complement-dependent suppressor phenotype characterized by inhibition of stimulated T cell activation. This study aims to evaluate whether serum markers of neutrophil activation and complement at diagnosis of EOC would be associated with clinical outcomes. EXPERIMENTAL DESIGN We conducted a two-center prospective study of patients with newly diagnosed EOC (N = 188). Blood and ascites fluid were collected at diagnosis for biomarker analysis. Patients were evaluated for progression-free survival (PFS) and overall survival (OS). RESULTS The median OS was 47 months (95 % CI: 34-58) and the median PFS was 12 months (95 % CI: 11-15). Pre-treatment serum levels of genomic DNA (gDNA), markers of neutrophil degranulation (myeloperoxidase [MPO]) and neutrophil extracellular traps (NETs) (citrullinated histone H3 [CitH3]), and complement activation (C3b/c) were each associated with worse OS in univariate analysis. In multivariate analyses controlling for age, stage, and optimal debulking, serum gDNA, MPO, and CitH3 remained associated with worse OS, while C3b/c levels were not. In an exploratory analysis, the largest magnitude of difference in 2-year OS occurred in patients with low C3b/c and low CitH3 compared to all other patients (87 % vs 46 % survival, respectively). In ascites fluid, increased factor H, a negative regulator of complement activation, was associated with improved OS in univariate analysis. CONCLUSIONS These results point to serum gDNA, NETs, and complement activation as potential prognostic biomarkers in patients with newly diagnosed EOC.
Collapse
Affiliation(s)
- Jason Ricciuti
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Dr. Ricciuti is currently in the Division of Gynecologic Oncology, Saint Louis University School of Medicine, Saint Louis, MO
| | - Qian Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - ANM Nazmul H. Khan
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Janine M. Joseph
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Bert R.J. Veuskens
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Institute for Immunology and Infection Diseases, Amsterdam, the Netherlands
| | | | - Sora Suzuki
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Tiffany R. Emmons
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Michael B. Yaffe
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts
- Division of Acute Care Surgery, Trauma and Surgical Critical Care, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Taco W. Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, the Netherlands
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Institute for Immunology and Infection Diseases, Amsterdam, the Netherlands
| | - Ilse Jongerius
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, the Netherlands
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Institute for Immunology and Infection Diseases, Amsterdam, the Netherlands
- Current affiliation Genmab, Utrecht, the Netherland
| | - Mieke C. Brouwer
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Institute for Immunology and Infection Diseases, Amsterdam, the Netherlands
| | - Richard B. Pouw
- Sanquin Research and Landsteiner Laboratory, Amsterdam University Medical Centre, Amsterdam Institute for Immunology and Infection Diseases, Amsterdam, the Netherlands
- Sanquin Diagnostic Services, Amsterdam, the Netherlands
| | - Kunle Odunsi
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, IL
| | - Peter Frederick
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Katherine LaVigne Mager
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Dr. Ricciuti is currently in the Division of Gynecologic Oncology, Saint Louis University School of Medicine, Saint Louis, MO
| | - Shashikant Lele
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Dr. Ricciuti is currently in the Division of Gynecologic Oncology, Saint Louis University School of Medicine, Saint Louis, MO
| | - Nicole Gaulin
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Dr. Ricciuti is currently in the Division of Gynecologic Oncology, Saint Louis University School of Medicine, Saint Louis, MO
| | - Christiane Hakim
- Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Robert P. Edwards
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Women’s Cancer Research Center, Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA
| | - Alexander B. Olawaiye
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Paniti Sukumanovich
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Sarah Taylor
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Women’s Cancer Research Center, Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA
| | - Esther Elishaev
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Women’s Cancer Research Center, Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA
| | - Emese Zsiros
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Dr. Ricciuti is currently in the Division of Gynecologic Oncology, Saint Louis University School of Medicine, Saint Louis, MO
| | - Francesmary Modugno
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health
- Women’s Cancer Research Center, Hillman Cancer Center and Magee-Womens Research Institute, Pittsburgh, PA
| | - Kirsten B. Moysich
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Brahm H. Segal
- Departments of Internal Medicine and Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| |
Collapse
|
33
|
Bogani G, Moore KN, Ray-Coquard I, Lorusso D, Matulonis UA, Ledermann JA, González-Martín A, Kurtz JE, Pujade-Lauraine E, Scambia G, Caruso G, Raspagliesi F, Colombo N, Monk BJ. Incorporating immune checkpoint inhibitors in epithelial ovarian cancer. Gynecol Oncol 2025; 193:30-40. [PMID: 39764856 DOI: 10.1016/j.ygyno.2024.12.011] [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: 10/09/2024] [Revised: 11/08/2024] [Accepted: 12/18/2024] [Indexed: 03/03/2025]
Abstract
OBJECTIVE Therapeutic interventions for epithelial ovarian cancer (EOC) have increased greatly over the last decade but improvements outside of biomarker selected therapies have been limited. There remains a pressing need for more effective treatment options that can prolong survival and enhance the quality of life of patients with EOC. In contrast to the significant benefits of immunotherapy with immune checkpoint inhibitors (CPI) seen in many solid tumors, initial experience in EOC suggests limited efficacy of CPIs monotherapy. METHODS A systematic review of phase III studies testing the role of CPIs in ovarian cancer was performed. RESULTS Seven randomized trials testing CPIs in newly diagnosed (n = 3) and recurrent (n = 4) EOC are evaluated. Overall, those trials included data of 5671 patients. Single-agent PD-L1 inhibitor trials have not shown significant efficacy in newly diagnosed ovarian cancer. Triplet maintenance with bevacizumab plus olaparib and durvalumab is associated with longer progression-free survival than maintenance with bevacizumab alone in patients without tumor BRCA mutations. CPIs were not effective in platinum-sensitive (n = 1031) and platinum-resistant (n = 1420) EOC. CONCLUSIONS The value of adding CPI to standard treatment including poly (ADP-ribose) polymerase (PARP) inhibitors with or without bevacizumab remains unclear and is being addressed in ongoing clinical trials. The combination of cytotoxic T-lymphocyte associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) inhibitors may enhance the efficacy of immunotherapy in EOC and studies are underway to investigate the combination of CPI with other emerging treatment modalities. PROSPERO registration ID: CRD42024536017.
Collapse
Affiliation(s)
- Giorgio Bogani
- Deaprtment of Gynecologic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy.
| | - Kathleen N Moore
- Stephenson Cancer Center at the University of Oklahoma Health Sciences Center/Sarah Cannon Research Institute, Oklahoma City, OK, United States.
| | - Isabelle Ray-Coquard
- Centre Anticancereux Léon Bérard, University Claude Bernard Lyon, GINECO Group, Lyon, France.
| | - Domenica Lorusso
- Department of Gynecologic Oncology, Humanitas San Pio X, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.
| | - Ursula A Matulonis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America.
| | | | - Antonio González-Martín
- Medical Oncology Department, Translational Oncology Group, CIMA, Universidad de Navarra, Cancer Center Clinica Universidad de Navarra, Madrid, and Grupo Español de Investigación en Cáncer ginecológicO (GEICO), Madrid, Spain.
| | - Jean-Emmanuel Kurtz
- Department of Medical and Surgical Oncology & Hematology, ICANS, Strasbourg, France.
| | - Eric Pujade-Lauraine
- Association de Recherche Cancers Gynécologiques (ARCAGY)-Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Paris, France; GINECO, Paris, France.
| | - Giovanni Scambia
- Department of Woman's and Child Health and Public Health Sciences, Gynecologic Oncology Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Catholic University of the Sacred Heart, Rome, Italy.
| | - Giuseppe Caruso
- Department of Gynecology, European Institute of Oncology, IEO, IRCCS, Milan, Italy; Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy.
| | - Francesco Raspagliesi
- Deaprtment of Gynecologic Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy.
| | - Nicoletta Colombo
- Gynecology Program, European Institute of Oncology, IEO, IRCCS, Milan, Italy; Department of Medicine and Surgery, University of Milan-Bicocca, Milan, Italy.
| | - Bradley J Monk
- GOG Foundation, Florida Cancer Specialists and Research Institute, West Palm Beach, FL 33401, United States of America.
| |
Collapse
|
34
|
Chen K, Wang J, Yang M, Deng S, Sun L. Immunotherapy in Recurrent Ovarian Cancer. Biomedicines 2025; 13:168. [PMID: 39857752 PMCID: PMC11762523 DOI: 10.3390/biomedicines13010168] [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: 11/11/2024] [Revised: 12/25/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES It remains challenging to treat recurrent ovarian cancer effectively as traditional interventions like chemotherapy and surgery have limited long-term efficacy, highlighting an urgent need for innovative approaches. Immunotherapy offers potential advantages in modulating the immune response against tumor cells and has emerged as a promising strategy in ovarian cancer management. This review discusses various immunotherapy modalities, including active and passive immune strategies, for recurrent ovarian cancer. METHODS We systematically reviewed recent immunotherapy advances for recurrent ovarian cancer, including the efficacy and mechanisms of single and dual immune checkpoint inhibitors, checkpoint inhibitor combinations with chemotherapy or radiotherapy, anti-angiogenic agents, PARP inhibitors, antibody-drug conjugates (ADC), tumor vaccines, and adoptive cell therapies (ACT). Additionally, we assessed emerging research on biomarkers predictive of immunotherapy responsiveness in ovarian cancer. RESULTS The findings indicate that immunotherapy, particularly combinations involving immune checkpoint inhibitors and other agents, demonstrates promising efficacy in recurrent ovarian cancer, with some therapies showing enhanced benefits in specific subtypes. The immune microenvironment in platinum-sensitive and -resistant cases exhibits distinct immunological profiles, influencing therapy outcomes. Several potential biomarkers have been identified, potentially aiding in patient stratification and treatment optimization. CONCLUSIONS Immunotherapy significantly advances recurrent ovarian cancer treatment, with various combinations potentially improving outcomes. Further research on predictive biomarkers and immune microenvironment characteristics is crucial for personalizing immunotherapy approaches and enhancing their efficacy in managing recurrent ovarian cancer.
Collapse
Affiliation(s)
| | | | | | | | - Li Sun
- Gynecology Department, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China; (K.C.); (J.W.); (M.Y.); (S.D.)
| |
Collapse
|
35
|
Lakhani NJ, Stewart D, Richardson DL, Dockery LE, Van Le L, Call J, Rangwala F, Wang G, Ma B, Metenou S, Huguet J, Offman E, Pandite L, Hamilton E. First-in-human phase I trial of the bispecific CD47 inhibitor and CD40 agonist Fc-fusion protein, SL-172154 in patients with platinum-resistant ovarian cancer. J Immunother Cancer 2025; 13:e010565. [PMID: 39800375 PMCID: PMC11749819 DOI: 10.1136/jitc-2024-010565] [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: 09/11/2024] [Accepted: 12/18/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND SL-172154 is a hexameric fusion protein adjoining the extracellular domain of SIRPα to the extracellular domain of CD40L via an inert IgG4-derived Fc domain. In preclinical studies, a murine equivalent SIRPα-Fc-CD40L fusion protein provided superior antitumor immunity in comparison to CD47- and CD40-targeted antibodies. A first-in-human phase I trial of SL-172154 was conducted in patients with platinum-resistant ovarian cancer. METHODS SL-172154 was administered intravenously at 0.1, 0.3, 1.0, 3.0, and 10.0 mg/kg. Dose escalation followed a modified toxicity probability interval-2 design. Objectives included evaluation of safety, dose-limiting toxicity, recommended phase II dose, pharmacokinetic (PK) and pharmacodynamic (PD) parameters, and antitumor activity. RESULTS 27 patients (median age 66 years (range, 33-85); median of 4 prior systemic therapies (range, 2-9)) with ovarian (70%), fallopian tube (15%), or primary peritoneal (15%) cancer received SL-172154. Treatment-emergent adverse events (TEAEs) were reported for 27 patients (100%), with 24 (88.9%) having a drug-related TEAE and infusion-related reactions being the most common. 12 patients (44.4%) had grade 3/4 TEAEs, and half of these patients (22.2%) had a drug-related grade 3/4 TEAE. There were no fatal adverse events, and no TEAEs led to drug discontinuation. SL-172154 Cmax and area under the curve increased with dose with greater than proportional exposure noted at 3.0 and 10.0 mg/kg. CD47 and CD40 target engagement on CD4+ T cells and B cells, respectively, approached 100% by 3.0 mg/kg. Dose-dependent responses in multiple cytokines (eg, interleukin 12 (IL-12), IP-10) approached a plateau at ≥3.0 mg/kg. Paired tumor biopsies demonstrated a shift in macrophages from an M2- to an M1-dominant phenotype and increased infiltration of CD8 T cells. PK/PD modeling showed near maximal margination of B cells and a dose-dependent production of IL-12 nearing a plateau at >3.0 mg/kg. The best response was stable disease in 6/27 (22%) patients. CONCLUSIONS SL-172154 was tolerable as monotherapy and induced, dose-dependent, and cyclical immune cell activation, increases in multiple serum cytokines, and trafficking of CD40-positive B cells and monocytes following each infusion. The safety, PK, and PD activity support 3.0 mg/kg as a safe and pharmacologically active dose. TRIAL REGISTRATION NUMBER NCT04406623.
Collapse
Affiliation(s)
- Nehal J Lakhani
- Clinical Research, START Midwest, Grand Rapids, Michigan, USA
| | - Daphne Stewart
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Debra L Richardson
- Division of Gynecologic Oncology, Stephenson Cancer Center and Sarah Cannon Research Institute/University of Oklahoma, Oklahoma City, Oklahoma, USA
| | - Lauren E Dockery
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Linda Van Le
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, The University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Justin Call
- Medical Oncology, START Mountain Region, West Valley City, Utah, USA
| | - Fatima Rangwala
- Clinical Research, Shattuck Labs R&D Office, Durham, North Carolina, USA
| | - Guanfang Wang
- Biometrics, Shattuck Labs R&D Office, Durham, North Carolina, USA
| | - Bo Ma
- Biometrics, Shattuck Labs R&D Office, Durham, North Carolina, USA
| | - Simon Metenou
- Translational Medicine, Shattuck Labs R&D Office, Durham, North Carolina, USA
| | - Jade Huguet
- Translational and Clinical Pharmacology, Certara, Toronto, Ontario, Canada
| | - Elliot Offman
- Translational and Clinical Pharmacology, Certara, Toronto, Ontario, Canada
| | - Lini Pandite
- Clinical Research, Shattuck Labs R&D Office, Durham, North Carolina, USA
| | - Erika Hamilton
- Medical Oncology, Sarah Cannon Research Institute, Nashville, Tennessee, USA
| |
Collapse
|
36
|
How JA, Dang M, Lee S, Fellman B, Westin SN, Sood AK, Fleming ND, Shafer A, Yuan Y, Liu J, Zhao L, Celestino J, Hajek R, Morgan MB, Parra ER, Laberiano Fernandez CD, Arrechedera CA, Solis Soto LM, Schmeler KM, Nick A, Lu KH, Coleman R, Wang L, Jazaeri AA. Pembrolizumab plus chemotherapy in frontline treatment of advanced ovarian cancer: Clinical and translational results from a phase 2 trial. MED 2025; 6:100494. [PMID: 39151421 PMCID: PMC11725453 DOI: 10.1016/j.medj.2024.07.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/05/2024] [Accepted: 07/23/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND The efficacy and feasibility of pembrolizumab combined with chemotherapy in frontline management of advanced high-grade epithelial ovarian cancer (EOC) is unknown. Additionally, modification of the tumor microenvironment following neoadjuvant therapy is not well understood. METHODS In this single-arm phase 2 trial (this study was registered at ClinicalTrials.gov: NCT02520154), eligible patients received up to 4 cycles of neoadjuvant chemotherapy followed by interval cytoreduction, 3 cycles of adjuvant intravenous carboplatin/weekly paclitaxel/pembrolizumab, and finally maintenance pembrolizumab until progression or toxicity (maximum 20 cycles). The primary endpoint was progression-free survival (PFS). Secondary endpoints included feasibility, toxicity, and overall survival (OS). PD-L1 staining, multiplex immunofluorescence staining, RNA sequencing, reverse-phase protein array analyses were performed on pre- and post-chemotherapy samples. FINDINGS Thirty-one eligible patients were enrolled. Median PFS and OS was 14.88 (95% CI 12.39-23.00) and 57.43 months (95% CI 30.88-not reached), respectively. Among those with PD-L1 combined positive score (CPS) ≥10, the median PFS and OS were not reached compared to those with CPS <10 (10.50 and 30.90 months, respectively). Feasibility was met, with all patients completing their planned adjuvant cycles. Treatment discontinuation due to immune-related toxicity occurred in 6 patients (20%). Chemotherapy resulted in an infiltration of anti-tumor immune cells in the tumor microenvironment. Samples of patients with the best PFS demonstrated increased expression of NF-κB, TGF-β, and β-catenin signaling. CONCLUSIONS Pembrolizumab with chemotherapy was feasible and resulted in PFS within the historical range for this EOC population. Patients with CPS ≥10 may benefit more from this regimen, and future studies should investigate this potential biomarker. FUNDING This investigator-initiated trial was funded by Merck.
Collapse
MESH Headings
- Humans
- Female
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Middle Aged
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/mortality
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Aged
- Paclitaxel/administration & dosage
- Carboplatin/administration & dosage
- Adult
- Carcinoma, Ovarian Epithelial/drug therapy
- Carcinoma, Ovarian Epithelial/pathology
- Carcinoma, Ovarian Epithelial/mortality
- Tumor Microenvironment/drug effects
- Neoadjuvant Therapy/methods
- Progression-Free Survival
- Cytoreduction Surgical Procedures
- B7-H1 Antigen
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/therapeutic use
Collapse
Affiliation(s)
- Jeffrey A How
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Minghao Dang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sanghoon Lee
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bryan Fellman
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicole D Fleming
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aaron Shafer
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ying Yuan
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jinsong Liu
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Zhao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph Celestino
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard Hajek
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Margaret B Morgan
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin R Parra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caddie D Laberiano Fernandez
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Claudio A Arrechedera
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luisa Maren Solis Soto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kathleen M Schmeler
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Karen H Lu
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Linghua Wang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amir A Jazaeri
- Department of Gynecologic Oncology and Reproductive Medicine, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
37
|
Huffman BM, Rahma OE, Tyan K, Li YY, Giobbie-Hurder A, Schlechter BL, Bockorny B, Manos MP, Cherniack AD, Baginska J, Mariño-Enríquez A, Kao KZ, Maloney AK, Ferro A, Kelland S, Ng K, Singh H, Welsh EL, Pfaff KL, Giannakis M, Rodig SJ, Hodi FS, Cleary JM. A Phase I Trial of Trebananib, an Angiopoietin 1 and 2 Neutralizing Peptibody, Combined with Pembrolizumab in Patients with Advanced Ovarian and Colorectal Cancer. Cancer Immunol Res 2025; 13:9-22. [PMID: 39348472 DOI: 10.1158/2326-6066.cir-23-1027] [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: 12/12/2023] [Revised: 04/08/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024]
Abstract
Ovarian cancers and microsatellite stable (MSS) colorectal cancers are insensitive to anti-programmed cell death 1 (PD-1) immunotherapy, and new immunotherapeutic approaches are needed. Preclinical data suggest a relationship between immunotherapy resistance and elevated angiopoietin 2 levels. We performed a phase I dose escalation study of pembrolizumab and the angiopoietin 1/2 inhibitor trebananib (NCT03239145). This multicenter trial enrolled patients with metastatic ovarian cancer or MSS colorectal cancer. Trebananib was administered intravenously weekly for 12 weeks with 200 mg intravenous pembrolizumab every 3 weeks. The toxicity profile of this combination was manageable, and the protocol-defined highest dose level (trebananib 30 mg/kg weekly plus pembrolizumab 200 mg every 3 weeks) was declared the maximum tolerated dose. The objective response rate for all patients was 7.3% (90% confidence interval, 2.5%-15.9%). Three patients with MSS colorectal cancer had durable responses for ≥3 years. One responding patient's colorectal cancer harbored a POLE mutation. The other two responding patients had left-sided colorectal cancers, with no baseline liver metastases, and genomic analysis revealed that they both had KRAS wild-type, ERBB2-amplified tumors. After development of acquired resistance, biopsy of one patient's KRAS wild-type ERBB2-amplified tumor showed a substantial decline in tumor-associated T cells and an increase in immunosuppressive intratumoral macrophages. Future studies are needed to carefully assess whether clinicogenomic features, such as lack of liver metastases, ERBB2 amplification, and left-sided tumors, can predict increased sensitivity to PD-1 immunotherapy combinations.
Collapse
Affiliation(s)
- Brandon M Huffman
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Osama E Rahma
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kevin Tyan
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Yvonne Y Li
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anita Giobbie-Hurder
- Division of Biostatistics, Department of Data Sciences, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Benjamin L Schlechter
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Bruno Bockorny
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Michael P Manos
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Andrew D Cherniack
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- The Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Joanna Baginska
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Adrián Mariño-Enríquez
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Katrina Z Kao
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Anna K Maloney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Allison Ferro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Sarah Kelland
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kimmie Ng
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Harshabad Singh
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Emma L Welsh
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kathleen L Pfaff
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marios Giannakis
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- The Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Scott J Rodig
- Harvard Medical School, Boston, Massachusetts
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
- Center for Immuno-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- ImmunoProfile, Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, Massachusetts
| | - F Stephen Hodi
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - James M Cleary
- Harvard Medical School, Boston, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
38
|
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
|
39
|
Westbom-Fremer S, Tran L, Ebbesson A, Martin de la Fuente L, Jönsson JM, Kannisto P, Veerla S, Hedenfalk I. Tertiary lymphoid structures in high-grade serous tubo-ovarian carcinoma: anatomical site matters. Cancer Immunol Immunother 2025; 74:56. [PMID: 39751944 PMCID: PMC11699021 DOI: 10.1007/s00262-024-03911-2] [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: 06/05/2024] [Accepted: 11/29/2024] [Indexed: 01/04/2025]
Abstract
Tertiary lymphoid structures (TLS) in the tumor microenvironment are prognostically beneficial in many solid cancer types. Reports on TLS in high-grade serous tubo-ovarian carcinoma (HGSC) are few, and the prognostic impact is unclear. We investigated mature TLS (mTLS), immature TLS (iTLS) and lymphoid aggregates (LA) in primary adnexal tumors (PTs) and synchronous omental/peritoneal metastases (pMets) of HGSC. Whole H&E slides were scrutinized for mTLS and LA in a population-based cohort of 130 cases with stage III-IV HGSC. The immune cell tumor infiltration was evaluated with single chromogenic immunohistochemistry (IHC) on a tissue microarray (TMA) from the same cases. Selected whole slides (PT n = 11, pMet n = 10) of the cases most abundant in mTLS and LA were further investigated with multiplex IHC and immunofluorescence using digital image analysis (QuPath), to confirm TLS status and map the T and B lymphocyte subtypes. The results showed that mTLS were more common in pMets than in PTs but did not have an independent prognostic impact on overall or progression-free survival. The presence of mTLS correlated with intratumoral infiltration of CD8+ cytotoxic T cells, FOXP3+ regulatory T cells and PD-1+ lymphocytes in pMets only. Although overall mTLS cell composition was similar between PTs and pMets, the outer zones of mTLS in PTs were more immune cell-rich. In conclusion, our results indicate differences in TLS presence and cellular elements between primary adnexal tumors and synchronous peritoneal metastases, which are important to consider when conducting studies of the immune environment in HGSC.
Collapse
Affiliation(s)
- Sofia Westbom-Fremer
- Division of Oncology, Department of Clinical Sciences Lund, and Lund University Cancer Center, Lund University, Lund, Sweden.
| | - Lena Tran
- Division of Oncology, Department of Clinical Sciences Lund, and Lund University Cancer Center, Lund University, Lund, Sweden
| | - Anna Ebbesson
- Division of Oncology, Department of Clinical Sciences Lund, and Lund University Cancer Center, Lund University, Lund, Sweden
| | - Laura Martin de la Fuente
- Division of Oncology, Department of Clinical Sciences Lund, and Lund University Cancer Center, Lund University, Lund, Sweden
| | - Jenny-Maria Jönsson
- Division of Oncology, Department of Clinical Sciences Lund, and Lund University Cancer Center, Lund University, Lund, Sweden
| | - Päivi Kannisto
- Division of Obstetrics and Gynaecology, Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| | - Srinivas Veerla
- Division of Oncology, Department of Clinical Sciences Lund, and Lund University Cancer Center, Lund University, Lund, Sweden
| | - Ingrid Hedenfalk
- Division of Oncology, Department of Clinical Sciences Lund, and Lund University Cancer Center, Lund University, Lund, Sweden
| |
Collapse
|
40
|
Lee JY, Tan D, Ray-Coquard I, Lee JB, Kim BG, Van Nieuwenhuysen E, Huang RYJ, Tse KY, González-Martin A, Scott C, Hasegawa K, Wilkinson K, Yang EY, Lheureux S, Kristeleit R. Phase II randomized study of dostarlimab alone or with bevacizumab versus non-platinum chemotherapy in recurrent gynecological clear cell carcinoma (DOVE/APGOT-OV7/ENGOT-ov80). J Gynecol Oncol 2025; 36:e51. [PMID: 39710508 PMCID: PMC11790990 DOI: 10.3802/jgo.2025.36.e51] [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: 08/24/2024] [Revised: 10/27/2024] [Accepted: 11/03/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Recurrent gynecological clear cell carcinoma (rGCCC) has a low objective response rate (ORR) to chemotherapy. Previous preclinical and clinical data suggest a potential synergy between immune checkpoint inhibitors and bevacizumab in rGCCC. Dostarlimab, a humanized monoclonal antibody targeting programmed cell death protein 1 (PD-1), combined with the anti-angiogenic bevacizumab, presents a novel therapeutic approach. This study will investigate the efficacy of dostarlimab +/- bevacizumab in rGCCC. METHODS DOVE is a global, multicenter, international, open-label, randomized phase 2 study of dostarlimab +/- bevacizumab with standard chemotherapy in rGCCC. We will enroll 198 patients with rGCCC and assign them to one of three groups in a 1:1:1 ratio: arm A (dostarlimab monotherapy), B (dostarlimab + bevacizumab), and C (investigator's choice of chemotherapy [weekly paclitaxel, pegylated liposomal doxorubicin, doxorubicin, or gemcitabine]). Patients with disease progression in arm A or C will be allowed to cross over to arm B. Stratification factors include prior bevacizumab use, prior lines of therapy (1 vs. >1), and primary site (ovarian vs. non-ovarian). Key inclusion criteria are histologically proven recurrent or persistent clear cell carcinoma of the ovary, endometrium, cervix, vagina, or vulva; up to five prior lines of therapy; disease progression within 12 months after platinum-based chemotherapy; and measurable disease. Key exclusion criteria are prior treatment with an anti-PD-1, anti-programmed death-ligand 1, or anti-programmed death-ligand 2 agent. The primary endpoint is progression-free survival determined by investigators. Secondary endpoints are ORR, disease control rate, clinical benefit rate, progression-free survival 2, overall survival, and toxicity. Exploratory objectives include immune biomarkers. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT06023862.
Collapse
Affiliation(s)
- Jung-Yun Lee
- Yonsei Cancer Center and Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| | - David Tan
- National University Hospital, Singapore
- Cancer Science Institute Singapore and Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Isabelle Ray-Coquard
- Leon Berard Centre, University Claude Bernard Lyon 1, Lyon, and GINECO Group, France
| | - Jung Bok Lee
- Asan Medical Center and University of Ulsan College of Medicine, Seoul, Korea
| | | | | | - Ruby Yun-Ju Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ka Yu Tse
- School of Clinical Medicine, The University of Hong Kong, Hong Kong
| | | | - Clare Scott
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Kosei Hasegawa
- Saitama Medical University International Medical Center, Hidaka, Japan
| | - Katie Wilkinson
- Cancer Research UK & University College London Cancer Trials Centre, UCL - University College London, London, UK
| | - Eun Yeong Yang
- Yonsei Cancer Center and Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | | | - Rebecca Kristeleit
- Guy's and St Thomas's NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
41
|
Nasso C, Puglisi S, Rebuzzi SE, Errigo V, Rosa F, Chiola I, Lazzari C, Musizzano Y, Venturino E, Gastaldo A, Siccardi C, Volpi EO, Mammoliti S, Benasso M. Immune checkpoint inhibitors in gynecological cancers: a narrative review on the practice-changing trials. Immunotherapy 2025; 17:57-66. [PMID: 39893504 PMCID: PMC11834418 DOI: 10.1080/1750743x.2025.2460964] [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: 07/21/2024] [Accepted: 01/28/2025] [Indexed: 02/04/2025] Open
Abstract
During the last decades, the introduction of immune checkpoint inhibitors has radically changed the treatment landscape of several cancer types, improving the prognosis and the quality of life of cancer patients. Even for gynecological cancers, where the prognosis has historically been poor despite advancements in surgery, radiotherapy and oncological treatment, immunotherapy has represented a significant leap forward. In cervical and endometrial cancer, the introduction of immunotherapy has radically changed the treatment algorithm, especially for advanced disease. However, the scenario remains less promising for ovarian cancer, where, despite extensive research efforts, no consistent positive results have been achieved with immune checkpoint inhibitors, except for a few cases in rarer histological subtypes Here, we present a narrative review summarizing the most important practice-changing studies involving immune checkpoint inhibitors in gynecological cancers, particularly in cervical, endometrial, ovarian and vulvar cancer.
Collapse
Affiliation(s)
- Cecilia Nasso
- Medical Oncology Unit, Ospedale Santa Corona, Pietra Ligure, Italy
| | - Silvia Puglisi
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico, San Martino, Italy
| | | | - Veronica Errigo
- Anatomical Pathology Unit, Ospedale San Paolo, Savona, Italy
| | - Francesca Rosa
- Diagnostic Imaging Department, Ospedale San Paolo, Savona, Italy
| | - Ilaria Chiola
- Radiotherapy Unit, Ospedale San Paolo, Savona, Italy
| | - Caterina Lazzari
- Obstetrics and Gynecology Unit, Ospedale San Paolo, Savona, Italy
| | - Yuri Musizzano
- Anatomical Pathology Unit, Ospedale San Paolo, Savona, Italy
| | - Ezio Venturino
- Anatomical Pathology Unit, Ospedale San Paolo, Savona, Italy
| | | | | | | | | | - Marco Benasso
- Medical Oncology Unit, Ospedale Santa Corona, Pietra Ligure, Italy
- Medical Oncology Unit, Ospedale San Paolo, Savona, Italy
| |
Collapse
|
42
|
Tan R, Wen M, Yang W, Zhan D, Zheng N, Liu M, Zhu F, Chen X, Wang M, Yang S, Xie B, He Q, Yuan K, Sun L, Wang Y, Qin J, Zhang Y. Integrated proteomics and scRNA-seq analyses of ovarian cancer reveal molecular subtype-associated cell landscapes and immunotherapy targets. Br J Cancer 2025; 132:111-125. [PMID: 39548315 PMCID: PMC11723995 DOI: 10.1038/s41416-024-02894-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) represents the most lethal gynaecological malignancy, yet understanding the connections between its molecular subtypes and their therapeutic implications remains incomplete. METHODS We conducted mass spectrometry-based proteomics analyses of 154 EOC tumour samples and 29 normal fallopian tubes, and single-cell RNA sequencing (scRNA-seq) analyses of an additional eight EOC tumours to classify proteomic subtypes and assess their cellular ecosystems and clinical significance. The efficacy of identified therapeutic targets was evaluated in patient-derived xenograft (PDX) and orthotopic mouse models. RESULTS We identified four proteomic subtypes with distinct clinical relevance: malignant proliferative (C1), immune infiltrating (C2), Fallopian-like (C3) and differentiated (C4) subtypes. C2 subtype was characterized by lymphocyte infiltration, notably an increased presence of GZMK CD8+ T cells and phagocytosis-like MRC+ macrophages. Additionally, we identified CD40 as a specific prognostic factor for C2 subtype. The interaction between CD40+ phagocytosis-like macrophages and CD40RL+ IL17R CD4+ T cells was correlated with a favourable prognosis. Finally, we established a druggable landscape for non-immune EOC patients and verified a TYMP inhibitor as a promising therapeutic strategy. CONCLUSIONS Our study refines the current immune subtype for EOC, highlighting CD40 agonists as promising therapies for C2 subtype patients and targeting TYMP for non-immune patients.
Collapse
Affiliation(s)
- Rong Tan
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China.
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China.
| | - Ming Wen
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan key laboratory of aging biology, Xiangya Hospital, Central South University, Changsha, China
| | - Wenqing Yang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, Hunan, China
| | - Dongdong Zhan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
- Beijing Pineal Diagnostics Co., Ltd., Beijing, China
| | - Nairen Zheng
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Mingwei Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Fang Zhu
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, Hunan, China
| | - Xiaodan Chen
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, Hunan, China
| | - Meng Wang
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China
| | - Siyu Yang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, Hunan, China
| | - Bin Xie
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Qiongqiong He
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, Hunan, China
| | - Kai Yuan
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, China
| | - Lunquan Sun
- Xiangya Cancer Center, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Molecular Radiation Oncology Hunan Province, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, China
- Center for Molecular Imaging of Central South University, Xiangya Hospital, Changsha, China
| | - Yi Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
| | - Jun Qin
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institute of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Yu Zhang
- Department of Gynecology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
- Gynecological Oncology Research and Engineering Center of Hunan Province, Changsha, Hunan, China.
| |
Collapse
|
43
|
Mollaoglu G, Tepper A, Falcomatà C, Potak HT, Pia L, Amabile A, Mateus-Tique J, Rabinovich N, Park MD, LaMarche NM, Brody R, Browning L, Lin JR, Zamarin D, Sorger PK, Santagata S, Merad M, Baccarini A, Brown BD. Ovarian cancer-derived IL-4 promotes immunotherapy resistance. Cell 2024; 187:7492-7510.e22. [PMID: 39481380 PMCID: PMC11682930 DOI: 10.1016/j.cell.2024.10.006] [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: 09/17/2023] [Revised: 04/20/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024]
Abstract
Ovarian cancer is resistant to immunotherapy, and this is influenced by the immunosuppressed tumor microenvironment (TME) dominated by macrophages. Resistance is also affected by intratumoral heterogeneity, whose development is poorly understood. To identify regulators of ovarian cancer immunity, we employed a spatial functional genomics screen (Perturb-map), focused on receptor/ligands hypothesized to be involved in tumor-macrophage communication. Perturb-map recapitulated tumor heterogeneity and revealed that interleukin-4 (IL-4) promotes resistance to anti-PD-1. We find ovarian cancer cells are the key source of IL-4, which directs the formation of an immunosuppressive TME via macrophage control. IL-4 loss was not compensated by nearby IL-4-expressing clones, revealing short-range regulation of TME composition dictating tumor evolution. Our studies show heterogeneous TMEs can emerge from localized altered expression of cancer-derived cytokines/chemokines that establish immune-rich and immune-excluded neighborhoods, which drive clone selection and immunotherapy resistance. They also demonstrate the potential of targeting IL-4 signaling to enhance ovarian cancer response to immunotherapy.
Collapse
Affiliation(s)
- Gurkan Mollaoglu
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Tepper
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Chiara Falcomatà
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hunter T Potak
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luisanna Pia
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Angelo Amabile
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jaime Mateus-Tique
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Matthew D Park
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nelson M LaMarche
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rachel Brody
- Department of Pathology, Molecular and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lindsay Browning
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jia-Ren Lin
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA; Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Dmitriy Zamarin
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA; Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Sandro Santagata
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Systems Biology, Harvard Medical School, Boston, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Miriam Merad
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alessia Baccarini
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Brian D Brown
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
44
|
Li J, Mei B, Feng L, Wang X, Wang D, Huang J, Zhang G. Amitriptyline revitalizes ICB response via dually inhibiting Kyn/Indole and 5-HT pathways of tryptophan metabolism in ovarian cancer. iScience 2024; 27:111488. [PMID: 39759009 PMCID: PMC11697709 DOI: 10.1016/j.isci.2024.111488] [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] [Revised: 07/26/2024] [Accepted: 11/25/2024] [Indexed: 01/07/2025] Open
Abstract
Reprogramming tryptophan metabolism (TRP) may be able to overcome immunosuppression and restore the immune checkpoint blockade (ICB) response in patients with epithelial ovarian cancer (EOC) resistant to ICB therapy because TRP metabolism is involved in the kynurenine/indole and serotonin pathways of tryptophan metabolism. Herein, employing amitriptyline (AMI), an antagonist of TLR4 and serotonin transporter (SERT), we revealed that AMI remodels the immunological landscape of EOC. In particular, AMI lowered the expression of IDO1, IL-4I1, and PD-L1, the quantity of KYN and indoles, and the level of immunosuppressive immune cells MDSC, Tregs, and CD8+CD39+/PD-1+ T cell. AMI boosted the killing potential of anti-PD-1-directed CD8+T cells and worked in concert with PD-1 inhibitors to suppress tumor growth and to prolong the survival of EOC-bearing mice. This work highlights AMI as an effective regulator of ICB response by manipulating EOC cell TRP metabolism, indicating it could be a potential strategy for improving EOC ICB therapy.
Collapse
Affiliation(s)
- Junyang Li
- Department Gynecological Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Bingjie Mei
- Department Gynecological Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Lu Feng
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Xiaoxin Wang
- Department Gynecological Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Dengfeng Wang
- Department Gynecological Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Jianming Huang
- Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| | - Guonan Zhang
- Department Gynecological Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu 610041, China
| |
Collapse
|
45
|
Moffitt LR, Karimnia N, Wilson AL, Stephens AN, Ho GY, Bilandzic M. Challenges in Implementing Comprehensive Precision Medicine Screening for Ovarian Cancer. Curr Oncol 2024; 31:8023-8038. [PMID: 39727715 PMCID: PMC11674382 DOI: 10.3390/curroncol31120592] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024] Open
Abstract
Precision medicine has revolutionised targeted cancer treatments; however, its implementation in ovarian cancer remains challenging. Diverse tumour biology and extensive heterogeneity in ovarian cancer can limit the translatability of genetic profiling and contribute to a lack of biomarkers of treatment response. This review addresses the barriers in precision medicine for ovarian cancer, including obtaining adequate and representative tissue samples for analysis, developing functional and standardised screening methods, and navigating data infrastructure and management. Ethical concerns related to patient consent, data privacy and health equity are also explored. We highlight the socio-economic complexities for precision medicine and propose strategies to overcome these challenges with an emphasis on accessibility and education amongst patients and health professionals and the development of regulatory frameworks to support clinical integration. Interdisciplinary collaboration is essential to drive progress in precision medicine to improve disease management and ovarian cancer patient outcomes.
Collapse
Affiliation(s)
- Laura R. Moffitt
- Hudson Institute of Medical Research, Clayton 3168, Australia; (L.R.M.); (N.K.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Nazanin Karimnia
- Hudson Institute of Medical Research, Clayton 3168, Australia; (L.R.M.); (N.K.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Amy L. Wilson
- Hudson Institute of Medical Research, Clayton 3168, Australia; (L.R.M.); (N.K.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Andrew N. Stephens
- Hudson Institute of Medical Research, Clayton 3168, Australia; (L.R.M.); (N.K.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| | - Gwo-Yaw Ho
- School of Clinical Sciences, Monash University, Clayton 3168, Australia;
- Department of Oncology, Monash Health, Bentleigh 3165, Australia
| | - Maree Bilandzic
- Hudson Institute of Medical Research, Clayton 3168, Australia; (L.R.M.); (N.K.); (A.L.W.); (A.N.S.)
- Department of Molecular and Translational Sciences, Monash University, Clayton 3168, Australia
| |
Collapse
|
46
|
Launonen IM, Niemiec I, Hincapié-Otero M, Erkan EP, Junquera A, Afenteva D, Falco MM, Liang Z, Salko M, Chamchougia F, Szabo A, Perez-Villatoro F, Li Y, Micoli G, Nagaraj A, Haltia UM, Kahelin E, Oikkonen J, Hynninen J, Virtanen A, Nirmal AJ, Vallius T, Hautaniemi S, Sorger PK, Vähärautio A, Färkkilä A. Chemotherapy induces myeloid-driven spatially confined T cell exhaustion in ovarian cancer. Cancer Cell 2024; 42:2045-2063.e10. [PMID: 39658541 DOI: 10.1016/j.ccell.2024.11.005] [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: 03/18/2024] [Revised: 08/30/2024] [Accepted: 11/07/2024] [Indexed: 12/12/2024]
Abstract
Anti-tumor immunity is crucial for high-grade serous ovarian cancer (HGSC) prognosis, yet its adaptation upon standard chemotherapy remains poorly understood. Here, we conduct spatial and molecular characterization of 117 HGSC samples collected before and after chemotherapy. Our single-cell and spatial analyses reveal increasingly versatile immune cell states forming spatiotemporally dynamic microcommunities. We describe Myelonets, networks of interconnected myeloid cells that contribute to CD8+ T cell exhaustion post-chemotherapy and show that M1/M2 polarization at the tumor-stroma interface is associated with CD8+ T cell exhaustion and exclusion, correlating with poor chemoresponse. Single-cell and spatial transcriptomics reveal prominent myeloid-T cell interactions via NECTIN2-TIGIT induced by chemotherapy. Targeting these interactions using a functional patient-derived immuno-oncology platform demonstrates that high NECTIN2-TIGIT signaling in matched tumors predicts responses to immune checkpoint blockade. Our discovery of clinically relevant myeloid-driven spatial T cell exhaustion unlocks immunotherapeutic strategies to unleash CD8+ T cell-mediated anti-tumor immunity in HGSC.
Collapse
Affiliation(s)
- Inga-Maria Launonen
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Iga Niemiec
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | | | | | - Ada Junquera
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Daria Afenteva
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Matias M Falco
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Zhihan Liang
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Matilda Salko
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Foteini Chamchougia
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Angela Szabo
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | | | - Yilin Li
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Giulia Micoli
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Ashwini Nagaraj
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Ulla-Maija Haltia
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Obstetrics and Gynecology, Department of Oncology, Clinical Trials Unit, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Essi Kahelin
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Pathology, University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Jaana Oikkonen
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Johanna Hynninen
- Department of Obstetrics and Gynecology, University of Turku and Turku University Hospital, Turku, Finland
| | - Anni Virtanen
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Pathology, University of Helsinki and HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Ajit J Nirmal
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Tuulia Vallius
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Sampsa Hautaniemi
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland
| | - Peter K Sorger
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA; Ludwig Center at Harvard, Boston, MA, USA
| | - Anna Vähärautio
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Foundation for the Finnish Cancer Institute, Helsinki, Finland.
| | - Anniina Färkkilä
- Research Program in Systems Oncology, University of Helsinki, Helsinki, Finland; Department of Obstetrics and Gynecology, Department of Oncology, Clinical Trials Unit, Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland; iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland; Institute for Molecular Medicine Finland, Helsinki Institute for Life Sciences, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
47
|
Brodeur MN, Dopeso H, Zhu Y, Longhini ALF, Gazzo A, Sun S, Koche RP, Qu R, Rosenberg L, Hamard PJ, Bykov Y, Green H, Gusain L, Chiappinelli KB, Ozsoy MA, Chui MH, Basili T, Gardner R, Walderich S, DeStanchina E, Greenbaum B, Gönen M, Vabret N, Weigelt B, Zamarin D. Interferon response and epigenetic modulation by SMARCA4 mutations drive ovarian tumor immunogenicity. SCIENCE ADVANCES 2024; 10:eadk4851. [PMID: 39630912 PMCID: PMC11616711 DOI: 10.1126/sciadv.adk4851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 09/24/2024] [Indexed: 12/07/2024]
Abstract
Cell-intrinsic mechanisms of immunogenicity in ovarian cancer (OC) are not well understood. Damaging mutations in the SWI/SNF chromatin remodeling complex, such as SMARCA4 (BRG1), are associated with improved response to immune checkpoint blockade; however, the mechanism by which this occurs is unclear. We found that SMARCA4 loss in OC models resulted in increased cancer cell-intrinsic immunogenicity, characterized by up-regulation of long-terminal RNA repeats, increased expression of interferon-stimulated genes, and up-regulation of antigen presentation machinery. Notably, this response was dependent on STING, MAVS, and IRF3 signaling but was independent of the type I interferon receptor. Mouse ovarian and melanoma tumors with SMARCA4 loss demonstrated increased infiltration and activation of cytotoxic T cells, NK cells, and myeloid cells in the tumor microenvironment. These results were recapitulated in BRG1 inhibitor-treated SMARCA4-proficient tumor models, suggesting that modulation of chromatin remodeling through targeting SMARCA4 may serve as a strategy to overcome cancer immune evasion.
Collapse
Affiliation(s)
| | - Higinio Dopeso
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yingjie Zhu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ana Leda F. Longhini
- Flow Cytometry Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrea Gazzo
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Siyu Sun
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Richard P. Koche
- Center for Epigenetic Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rui Qu
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laura Rosenberg
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Pierre-Jacques Hamard
- Center for Epigenetic Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yonina Bykov
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hunter Green
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laxmi Gusain
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Katherine B. Chiappinelli
- Department of Microbiology, Immunology and Tropical Medicine, The GW Cancer Center, The George Washington University, Washington, DC, USA
| | - Melih Arda Ozsoy
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
| | - M. Herman Chui
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thais Basili
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rui Gardner
- Flow Cytometry Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sven Walderich
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Elisa DeStanchina
- Antitumor Assessment Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Benjamin Greenbaum
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nicolas Vabret
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Britta Weigelt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Dmitriy Zamarin
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
48
|
Rosario SR, Long MD, Chilakapati S, Gomez EC, Battaglia S, Singh PK, Wang J, Wang K, Attwood K, Hess SM, McGray AJR, Odunsi K, Segal BH, Paragh G, Liu S, Wargo JA, Zsiros E. Integrative multi-omics analysis uncovers tumor-immune-gut axis influencing immunotherapy outcomes in ovarian cancer. Nat Commun 2024; 15:10609. [PMID: 39638782 PMCID: PMC11621351 DOI: 10.1038/s41467-024-54565-8] [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: 05/30/2024] [Accepted: 11/08/2024] [Indexed: 12/07/2024] Open
Abstract
Recurrent ovarian cancer patients, especially those resistant to platinum, lack effective curative treatments. To address this, we conducted a phase 2 clinical trial (NCT02853318) combining pembrolizumab with bevacizumab, to increase T cell infiltration into the tumor, and oral cyclophosphamide, to reduce the number of regulatory T cells. The trial accrued 40 heavily pretreated recurrent ovarian cancer patients. The primary endpoint, progression free survival, was extended to a median of 10.2 months. The secondary endpoints demonstrated an objective response rate of 47.5%, and disease control in 30% of patients for over a year while maintaining a good quality of life. We performed comprehensive molecular, immune, microbiome, and metabolic profiling on samples of trial patients. Here, we show increased T and B cell clusters and distinct microbial patterns with amino acid and lipid metabolism are linked to exceptional clinical responses. This study suggests the immune milieu and host-microbiome can be leveraged to improve antitumor response in future immunotherapy trials.
Collapse
Affiliation(s)
- Spencer R Rosario
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Mark D Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Shanmuga Chilakapati
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA, 02111, USA
| | - Eduardo Cortes Gomez
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Sebastiano Battaglia
- Computational Biology Office of Translational Research, Janssen Pharmaceuticals, Buffalo, NY, 14263, USA
| | - Prashant K Singh
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jianmin Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Katy Wang
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Kristopher Attwood
- Department of Clinical Research, American College of Radiology, Reston, VA, 20191, USA
| | - Suzanne M Hess
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - A J Robert McGray
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Kunle Odunsi
- Department of Obstetrics and Gynecology, University of Chicago Comprehensive Cancer Center, Chicago, IL, 60637, USA
| | - Brahm H Segal
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Gyorgy Paragh
- Department of Dermatology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Emese Zsiros
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
49
|
Kurnit KC, Odunsi K. Harnessing Antitumor Immunity in Ovarian Cancer. Cold Spring Harb Perspect Med 2024; 14:a041336. [PMID: 38621830 PMCID: PMC11610759 DOI: 10.1101/cshperspect.a041336] [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: 04/17/2024]
Abstract
Despite progress in other tumor types, immunotherapy is not yet part of the standard of care treatment for high-grade serous ovarian cancer patients. Although tumor infiltration by T cells is frequently observed in patients with ovarian cancer, clinical responses to immunotherapy remain low. Mechanisms for immune resistance in ovarian cancer have been explored and may provide insight into future approaches to improve response to immunotherapy agents. In this review, we discuss what is known about the immune landscape in ovarian cancer, review the available data for immunotherapy-based strategies in these patients, and provide possible future directions.
Collapse
Affiliation(s)
- Katherine C Kurnit
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, Illinois 60637, USA
| | - Kunle Odunsi
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, Illinois 60637, USA
- University of Chicago Medicine Comprehensive Cancer Center, Chicago, Illinois 60637, USA
| |
Collapse
|
50
|
Liao JB, Jejurikar NS, Hitchcock-Bernhardt KM, Gwin WR, Reichow JL, Dang Y, Childs JS, Coveler AL, Swensen RE, Goff BA, Disis ML, Salazar LG. Intraperitoneal immunotherapy with denileukin diftitox (ONTAK) in recurrent refractory ovarian cancer. Gynecol Oncol 2024; 191:74-79. [PMID: 39362046 PMCID: PMC11637896 DOI: 10.1016/j.ygyno.2024.09.019] [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: 08/09/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Denileukin diftitox (ONTAK) is a diphtheria/IL-2R fusion protein able to deplete regulatory T cells in peripheral blood. Regulatory T cells in the local immune microenvironment have been shown to be associated with poor prognosis in ovarian cancer. This study examined whether denileukin diftitox (ONTAK) could be safely administered intraperitoneal in patients with advanced refractory ovarian cancer and assessed its effects on regulatory T cells and tumor associated cytokines in ascites and peripheral blood. PATIENTS AND METHODS A phase I dose escalation study of intraperitoneal denileukin diftitox (ONTAK) enrolled 10 patients with advanced, refractory ovarian carcinoma at 3 doses (5 μg/kg, 15 μg/kg, and 25 μg/kg). Serial CA-125 measurements assessed clinical response. Regulatory T cells were quantified using RT-PCR and cytokine levels measured by Luminex. RESULTS The maximum tolerated dose was 15 μg/kg with a dose limiting toxicity observed in 1 out of 6 patients in the expansion group. The majority of adverse events were transient grades 1-2. One patient treated at the 25 μg/kg dose experienced cytokine storm with prolonged hospitalization. 3 patients had decreases in CA-125 after treatment but none met criteria for partial response. Treatment with denileukin diftitox (ONTAK) decreased regulatory T cells in peripheral blood and ascites. Treated patients did not show any significant changes in IL-8, TGF-β, sIL2Ra in ascites or peripheral blood. CONCLUSIONS Denileukin diftitox (ONTAK) can be safely administered intraperitoneally to recurrent refractory ovarian cancer patients. Regulatory T cells were reduced in ascites and peripheral blood, but there were no significant changes in cytokine levels. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov # NCT00357448.
Collapse
Affiliation(s)
- John B Liao
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Washington, 1959 NE Pacific St, Seattle, WA 98195, United States of America; UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America.
| | - Nikita S Jejurikar
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Washington, 1959 NE Pacific St, Seattle, WA 98195, United States of America; UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Katie M Hitchcock-Bernhardt
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Washington, 1959 NE Pacific St, Seattle, WA 98195, United States of America; UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - William R Gwin
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Jessica L Reichow
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Yushe Dang
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Jennifer S Childs
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Andrew L Coveler
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Ron E Swensen
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Washington, 1959 NE Pacific St, Seattle, WA 98195, United States of America; UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America; Valley Medical Center, 400 South 43(rd) Street, Renton, WA 98055, United States of America
| | - Barbara A Goff
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Washington, 1959 NE Pacific St, Seattle, WA 98195, United States of America
| | - Mary L Disis
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| | - Lupe G Salazar
- UW Medicine Cancer Vaccine Institute, University of Washington, 850 Republican St, Seattle, WA 98195, United States of America
| |
Collapse
|