201
|
Simion L, Rotaru V, Cirimbei C, Stefan DC, Gherghe M, Ionescu S, Tanase BC, Luca DC, Gales LN, Chitoran E. Analysis of Efficacy-To-Safety Ratio of Angiogenesis-Inhibitors Based Therapies in Ovarian Cancer: A Systematic Review and Meta-Analysis. Diagnostics (Basel) 2023; 13:diagnostics13061040. [PMID: 36980348 PMCID: PMC10046967 DOI: 10.3390/diagnostics13061040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/28/2023] [Accepted: 03/02/2023] [Indexed: 03/30/2023] Open
Abstract
(1) Background: Among new anti-angiogenesis agents being developed and ever-changing guidelines indications, the question of the benefits/safety ratio remains unclear. (2) Methods: We performed a systematic review combined with a meta-analysis of 23 randomized controlled trials (12,081 patients), evaluating overall survival (OS), progression free survival (PFS) and toxicity (grade ≥ 3 toxic effects, type, and number of all adverse effects. (3) Results: The analysis showed improvement of pooled-PFS (HR, 0.71; 95% CI, 0.64-0.78; I2 = 77%; p < 0.00001) in first-line (HR, 0.85; 95% CI, 0.78-0.93; p = 0.0003) or recurrent cancer (HR, 0.62; 95% CI, 0.56-0.70; p < 0.00001) and regardless of the type of anti-angiogenesis drug used (Vascular endothelial growth factor (VEGF) inhibitors, VEGF-receptors (VEGF-R) inhibitors or angiopoietin inhibitors). Improved OS was also observed (HR, 0.95; 95% CI, 0.90-0.99; p = 0.03). OS benefits were only observed in recurrent neoplasms, both platinum-sensitive and platinum-resistant neoplasms. Grade ≥ 3 adverse effects were increased across all trials. Anti-angiogenetic therapy increased the risk of hypertension, infection, thromboembolic/hemorrhagic events, and gastro-intestinal perforations but not the risk of wound-related issues, anemia or posterior leukoencephalopathy syndrome. (4) Conclusions: Although angiogenesis inhibitors improve PFS, there are little-to-no OS benefits. Given the high risk of severe adverse reactions, a careful selection of patients is required for obtaining the best results possible.
Collapse
Affiliation(s)
- Laurentiu Simion
- Department of Surgery, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology "Prof. Dr. Al. Trestioreanu", 022328 Bucharest, Romania
| | - Vlad Rotaru
- Department of Surgery, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology "Prof. Dr. Al. Trestioreanu", 022328 Bucharest, Romania
| | - Ciprian Cirimbei
- Department of Surgery, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology "Prof. Dr. Al. Trestioreanu", 022328 Bucharest, Romania
| | - Daniela-Cristina Stefan
- Department of Surgery, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Mirela Gherghe
- Nuclear Medicine Department, Bucharest Institute of Oncology "Prof. Dr. Al. Trestioreanu", 022328 Bucharest, Romania
| | - Sinziana Ionescu
- Department of Surgery, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology "Prof. Dr. Al. Trestioreanu", 022328 Bucharest, Romania
| | - Bogdan Cosmin Tanase
- Thoracic Surgery Department, Bucharest Institute of Oncology "Prof. Dr. Al. Trestioreanu", 022328 Bucharest, Romania
| | - Dan Cristian Luca
- Department of Surgery, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology "Prof. Dr. Al. Trestioreanu", 022328 Bucharest, Romania
| | - Laurentia Nicoleta Gales
- Department of Surgery, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Medical Oncology Department, Bucharest Institute of Oncology "Prof. Dr. Al. Trestioreanu", 022328 Bucharest, Romania
| | - Elena Chitoran
- Department of Surgery, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- General Surgery and Surgical Oncology Department I, Bucharest Institute of Oncology "Prof. Dr. Al. Trestioreanu", 022328 Bucharest, Romania
| |
Collapse
|
202
|
Abstract
Ovarian clear cell carcinoma is a rare subtype of epithelial ovarian cancer with unique clinicopathological features. The most common genetic aberration observed is loss of function ARID1A mutations. Advanced and recurrent ovarian clear cell carcinoma is characterized by resistance to standard-of-care cytotoxic chemotherapy and a poor prognosis. Despite the distinct molecular features of ovarian clear cell carcinoma, current treatments for this subtype of epithelial ovarian cancer are based on clinical trials which predominantly recruited patients with high grade serous ovarian carcinoma. These factors have encouraged researchers to develop novel treatment strategies specifically for ovarian clear cell carcinoma which are currently being tested in the context of clinical trials. These new treatment strategies currently focus on three key areas: immune checkpoint blockade, targeting angiogenesis, and exploiting ARID1A synthetic lethal interactions. Rational combinations of these strategies are being assessed in clinical trials. Despite the progress made in identifying new treatments for ovarian clear cell carcinoma, predictive biomarkers to better define those patients likely to respond to new treatments remain to be elucidated. Additional future challenges which may be addressed through international collaboration include the need for randomized trials in a rare disease and establishing the relative sequencing of these novel treatments.
Collapse
Affiliation(s)
- James Stewart
- Royal Marsden Hospital NHS Trust, London, UK
- Gene Function Laboratory, Insitute of Cancer Research, London, UK
| | | | - Susana Banerjee
- Royal Marsden Hospital NHS Trust, London, UK
- Division of Clinical Studies, Institute of Cancer Research, London, UK
| |
Collapse
|
203
|
Segal BH, Giridharan T, Suzuki S, Khan ANH, Zsiros E, Emmons TR, Yaffe MB, Gankema AAF, Hoogeboom M, Goetschalckx I, Matlung HL, Kuijpers TW. Neutrophil interactions with T cells, platelets, endothelial cells, and of course tumor cells. Immunol Rev 2023; 314:13-35. [PMID: 36527200 PMCID: PMC10174640 DOI: 10.1111/imr.13178] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neutrophils sense microbes and host inflammatory mediators, and traffic to sites of infection where they direct a broad armamentarium of antimicrobial products against pathogens. Neutrophils are also activated by damage-associated molecular patterns (DAMPs), which are products of cellular injury that stimulate the innate immune system through pathways that are similar to those activated by microbes. Neutrophils and platelets become activated by injury, and cluster and cross-signal to each other with the cumulative effect of driving antimicrobial defense and hemostasis. In addition, neutrophil extracellular traps are extracellular chromatin and granular constituents that are generated in response to microbial and damage motifs and are pro-thrombotic and injurious. Although neutrophils can worsen tissue injury, neutrophils may also have a role in facilitating wound repair following injury. A central theme of this review relates to how critical functions of neutrophils that evolved to respond to infection and damage modulate the tumor microenvironment (TME) in ways that can promote or limit tumor progression. Neutrophils are reprogrammed by the TME, and, in turn, can cross-signal to tumor cells and reshape the immune landscape of tumors. Importantly, promising new therapeutic strategies have been developed to target neutrophil recruitment and function to make cancer immunotherapy more effective.
Collapse
Affiliation(s)
- Brahm H Segal
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
- Department of Medicine, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Thejaswini Giridharan
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Sora Suzuki
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Anm Nazmul H Khan
- Department of Internal Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Emese Zsiros
- Department of Gynecologic Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, New York, USA
| | - Tiffany R Emmons
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Michael B Yaffe
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Surgical Oncology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Angela A F Gankema
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark Hoogeboom
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Ines Goetschalckx
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Hanke L Matlung
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Department of Molecular Hematology, Sanquin Research, University of Amsterdam, Amsterdam, The Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, Emma Children's Hospital Amsterdam University Medical Center (Amsterdam UMC), University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
204
|
Manning-Geist BL, Gnjatic S, Aghajanian C, Konner J, Kim SH, Sarasohn D, Soldan K, Tew WP, Sarlis NJ, Zamarin D, Kravetz S, Laface I, Rasalan-Ho T, Qi J, Wong P, Sabbatini PJ, O’Cearbhaill RE. Phase I Study of a Multivalent WT1 Peptide Vaccine (Galinpepimut-S) in Combination with Nivolumab in Patients with WT1-Expressing Ovarian Cancer in Second or Third Remission. Cancers (Basel) 2023; 15:1458. [PMID: 36900251 PMCID: PMC10001251 DOI: 10.3390/cancers15051458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/03/2023] Open
Abstract
We examined the safety and immunogenicity of sequential administration of a tetravalent, non-HLA (human leukocyte antigen) restricted, heteroclitic Wilms' Tumor 1 (WT1) peptide vaccine (galinpepimut-S) with anti-PD-1 (programmed cell death protein 1) nivolumab. This open-label, non-randomized phase I study enrolled patients with WT1-expressing ovarian cancer in second or third remission from June 2016 to July 2017. Therapy included six (every two weeks) subcutaneous inoculations of galinpepimut-S vaccine adjuvanted with Montanide, low-dose subcutaneous sargramostim at the injection site, with intravenous nivolumab over 12 weeks, and up to six additional doses until disease progression or toxicity. One-year progression-free survival (PFS) was correlated to T-cell responses and WT1-specific immunoglobulin (Ig)G levels. Eleven patients were enrolled; seven experienced a grade 1 adverse event, and one experienced a grade ≥3 adverse event considered a dose-limiting toxicity. Ten (91%) of eleven patients had T-cell responses to WT1 peptides. Seven (88%) of eight evaluable patients had IgG against WT1 antigen and full-length protein. In evaluable patients who received >2 treatments of galinpepimut-S and nivolumab, the 1-year PFS rate was 70%. Coadministration of galinpepimut-S and nivolumab demonstrated a tolerable toxicity profile and induced immune responses, as indicated by immunophenotyping and WT1-specific IgG production. Exploratory analysis for efficacy yielded a promising 1-year PFS rate.
Collapse
Affiliation(s)
- Beryl L. Manning-Geist
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sacha Gnjatic
- Immune Monitoring Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Tisch Cancer Institute, Precision Immunology Institute, Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carol Aghajanian
- Department of Medicine, Weill Cornell Medical Center, New York, NY 10065, USA
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jason Konner
- Department of Medicine, Weill Cornell Medical Center, New York, NY 10065, USA
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sarah H. Kim
- Gynecology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Debra Sarasohn
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Krysten Soldan
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - William P. Tew
- Department of Medicine, Weill Cornell Medical Center, New York, NY 10065, USA
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | | | - Dmitriy Zamarin
- Department of Medicine, Weill Cornell Medical Center, New York, NY 10065, USA
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Sara Kravetz
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ilaria Laface
- Tisch Cancer Institute, Precision Immunology Institute, Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Teresa Rasalan-Ho
- Immune Monitoring Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Jingjing Qi
- Immune Monitoring Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Tisch Cancer Institute, Precision Immunology Institute, Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Phillip Wong
- Immune Monitoring Facility, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Paul J. Sabbatini
- Department of Medicine, Weill Cornell Medical Center, New York, NY 10065, USA
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Roisin E. O’Cearbhaill
- Department of Medicine, Weill Cornell Medical Center, New York, NY 10065, USA
- Gynecologic Medical Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Medicine, University of Galway, H91 YR71 Galway, Ireland
| |
Collapse
|
205
|
Truxova I, Cibula D, Spisek R, Fucikova J. Targeting tumor-associated macrophages for successful immunotherapy of ovarian carcinoma. J Immunother Cancer 2023; 11:jitc-2022-005968. [PMID: 36822672 PMCID: PMC9950980 DOI: 10.1136/jitc-2022-005968] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 02/25/2023] Open
Abstract
Epithelial ovarian cancer (EOC) is among the top five causes of cancer-related death in women, largely reflecting early, prediagnosis dissemination of malignant cells to the peritoneum. Despite improvements in medical therapies, particularly with the implementation of novel drugs targeting homologous recombination deficiency, the survival rates of patients with EOC remain low. Unlike other neoplasms, EOC remains relatively insensitive to immune checkpoint inhibitors, which is correlated with a tumor microenvironment (TME) characterized by poor infiltration by immune cells and active immunosuppression dominated by immune components with tumor-promoting properties, especially tumor-associated macrophages (TAMs). In recent years, TAMs have attracted interest as potential therapeutic targets by seeking to reverse the immunosuppression in the TME and enhance the clinical efficacy of immunotherapy. Here, we review the key biological features of TAMs that affect tumor progression and their relevance as potential targets for treating EOC. We especially focus on the therapies that might modulate the recruitment, polarization, survival, and functional properties of TAMs in the TME of EOC that can be harnessed to develop superior combinatorial regimens with immunotherapy for the clinical care of patients with EOC.
Collapse
Affiliation(s)
| | - David Cibula
- Gynecologic Oncology Center, Department of Obstetrics and Gynecology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Radek Spisek
- Sotio Biotech, Prague, Czech Republic,Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Jitka Fucikova
- Sotio Biotech, Prague, Czech Republic .,Department of Immunology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| |
Collapse
|
206
|
Goto M, Tsubamoto H, Isono-Taniguchi R, Takimoto Y, Tashima L, Hori K, Ito K. Combination therapy of oral cyclophosphamide and bevacizumab for patients with recurrent ovarian and peritoneal cancer. Medicine (Baltimore) 2023; 102:e32880. [PMID: 36827071 PMCID: PMC11309712 DOI: 10.1097/md.0000000000032880] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/11/2023] [Accepted: 01/18/2023] [Indexed: 02/25/2023] Open
Abstract
Chemotherapy for patients with recurrent cancer aims to obtain survival benefits, relieve symptoms, and improve quality of life. We used oral cyclophosphamide and bevacizumab (BEV) combination therapy in recurrent ovarian and peritoneal cancer cases, where standard chemotherapy was infeasible. Subsequently, we evaluated the safety and efficacy of this treatment. Between August 2014 and June 2020, patients received the following regimen: oral cyclophosphamide 50 mg daily and intravenous cyclic BEV 15 mg/kg every 3 weeks. Data from 2 facilities were retrospectively analyzed. Twenty-two patients were enrolled (20 with ovarian cancer and two with peritoneal cancer). The median follow-up period and age were 18.9 months (range, 5.0-51.5) and 60 years (range 37-81), respectively. Sixteen patients had platinum resistance. The median number of previous chemotherapy regimens was 2.5 (range 0-5). The median implementation cycle was five (range 2-14). Eighteen patients discontinued treatment due to side effects (3 patient) and disease progression (15 patient). Grade 2 toxicities included neutropenia (1 patient), proteinuria (1 patient), hypertension (2 patient), and esophagitis (1 patient). Two patients had complete response and one had a partial response. Five patients had stable disease. The response rate in platinum-sensitive recurrence was 33.3%, and 7.1% in platinum-resistant recurrence, and a clinical benefit was found in 8 (36.3%) patients. The median PFS and overall survival from cyclophosphamide and BEV initiation was 5.3 months (range, 0.8-23.5) and 9.2 months (range, 4.8-51.5), respectively. The combination of oral cyclophosphamide and BEV does not have a high response rate, but is well-tolerated and can be used safely in patients who are difficult to treat after second-line chemotherapy. Data from 2 facilities were retrospectively analyzed.
Collapse
Affiliation(s)
- Mayako Goto
- Department of Obstetrics and Gynecology, Kansai Rousai Hospital, Amagasaki, Japan
| | - Hiroshi Tsubamoto
- Department of Obstetrics and Gynecology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Roze Isono-Taniguchi
- Department of Obstetrics and Gynecology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Yumi Takimoto
- Department of Obstetrics and Gynecology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Lena Tashima
- Department of Obstetrics and Gynecology, Kansai Rousai Hospital, Amagasaki, Japan
| | - Kensuke Hori
- Department of Obstetrics and Gynecology, Kansai Rousai Hospital, Amagasaki, Japan
| | - Kimihiko Ito
- Department of Obstetrics and Gynecology, Kansai Rousai Hospital, Amagasaki, Japan
| |
Collapse
|
207
|
Discordance of PD-L1 expression in primary and metastatic ovarian high-grade serous carcinoma and its correlation with CD8 + tumor-infiltrating lymphocytes and patient prognosis. Virchows Arch 2023; 482:755-766. [PMID: 36806916 DOI: 10.1007/s00428-023-03512-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/16/2023] [Accepted: 02/03/2023] [Indexed: 02/23/2023]
Abstract
Differential expression of programmed death-1 ligand (PD-L1) and its clinical significance in primary and metastatic ovarian high-grade serous carcinoma (HGSC) have not been defined. Thus, we investigated the PD-L1 expression of paired ovarian primary and omental metastatic HGSC and its correlation with CD8 + tumor-infiltrating lymphocyte (TILs) and patient survival. A total of 212 cases of ovarian HGSCs with matched primary ovarian and metastatic omental tumors accessioned between 2003 and 2018 were selected for further analysis. Using immunohistochemistry, we evaluated the density of CD8 + TILs and expression of PD-L1 on whole tissue sections. Applying tumor proportion score (TPS, cutoff 1%) and combined positive score (CPS, cutoff 1), the prevalence of PD-L1 expression was similar but with significant discordance in ovarian and omental tumor. Using TPS, patients with PD-L1-positive tumors demonstrated significantly worse recurrence free survival (RFS) and overall survival (OS) than patients with PD-L1-negative tumors. Using CPS, patients with PD-L1-positive ovarian tumors demonstrated significantly worse OS while no significant difference in RFS was found. Patients with PD-L1-positive omental tumors demonstrated significantly worse RFS and OS. Patients with omental PD-L1-positive tumors (TPS) were associated with poorer RFS and OS, while patients with ovarian PD-L1-positive tumors (TPS) were associated with OS not RFS, in COX multivariant analysis. Nonetheless, ovarian and omental high CD8 TILs density was not associated with worse OS in univariant and COX multivariant analysis. PD-L1 expression in ovarian and omental tumor associated with an increased CD8 + TILs density. PD-L1 expression by TPS was better correlated with survival than by CPS, and PD-L1 expression in omental tumors was a stronger prognostic indicator than that in ovarian tumors.
Collapse
|
208
|
Liu L, Wan H, Liu L, Wang J, Tang Y, Cui S, Li Y. Deep Learning Provides a New Magnetic Resonance Imaging-Based Prognostic Biomarker for Recurrence Prediction in High-Grade Serous Ovarian Cancer. Diagnostics (Basel) 2023; 13:diagnostics13040748. [PMID: 36832236 PMCID: PMC9954966 DOI: 10.3390/diagnostics13040748] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 01/13/2023] [Accepted: 01/28/2023] [Indexed: 02/18/2023] Open
Abstract
This study aims to use a deep learning method to develop a signature extract from preoperative magnetic resonance imaging (MRI) and to evaluate its ability as a non-invasive recurrence risk prognostic marker in patients with advanced high-grade serous ovarian cancer (HGSOC). Our study comprises a total of 185 patients with pathologically confirmed HGSOC. A total of 185 patients were randomly assigned in a 5:3:2 ratio to a training cohort (n = 92), validation cohort 1 (n = 56), and validation cohort 2 (n = 37). We built a new deep learning network from 3839 preoperative MRI images (T2-weighted images and diffusion-weighted images) to extract HGSOC prognostic indicators. Following that, a fusion model including clinical and deep learning features is developed to predict patients' individual recurrence risk and 3-year recurrence likelihood. In the two validation cohorts, the consistency index of the fusion model was higher than both the deep learning model and the clinical feature model (0.752, 0.813 vs. 0.625, 0.600 vs. 0.505, 0.501). Among the three models, the fusion model had a higher AUC than either the deep learning model or the clinical model in validation cohorts 1 or 2 (AUC = was 0.986, 0.961 vs. 0.706, 0.676/0.506, 0.506). Using the DeLong method, the difference between them was statistically significant (p < 0.05). The Kaplan-Meier analysis distinguished two patient groups with high and low recurrence risk (p = 0.0008 and 0.0035, respectively). Deep learning may be a low-cost, non-invasive method for predicting risk for advanced HGSOC recurrence. Deep learning based on multi-sequence MRI serves as a prognostic biomarker for advanced HGSOC, which provides a preoperative model for predicting recurrence in HGSOC. Additionally, using the fusion model as a new prognostic analysis means that can use MRI data can be used without the need to follow-up the prognostic biomarker.
Collapse
Affiliation(s)
- Lili Liu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
- Department of Radiology, Chongqing General Hospital, Chongqing 401120, China
| | - Haoming Wan
- College of Computer and Information Science, Chongqing Normal University, Chongqing 400016, China
| | - Li Liu
- Department of Radiology, The People’s Hospital of Yubei District of Chongqing, Chongqing 401120, China
| | - Jie Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yibo Tang
- College of Computer and Information Science, Chongqing Normal University, Chongqing 400016, China
| | - Shaoguo Cui
- College of Computer and Information Science, Chongqing Normal University, Chongqing 400016, China
- Correspondence: (S.C.); (Y.L.)
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing 400016, China
- Correspondence: (S.C.); (Y.L.)
| |
Collapse
|
209
|
Falcinelli M, Al-Hity G, Baron S, Mampay M, Allen MC, Samuels M, Jones W, Cilibrasi C, Flaherty RL, Giamas G, Thaker PH, Flint MS. Propranolol reduces IFN-γ driven PD-L1 immunosuppression and improves anti-tumour immunity in ovarian cancer. Brain Behav Immun 2023; 110:1-12. [PMID: 36796704 DOI: 10.1016/j.bbi.2023.02.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
The immune system plays an important role in controlling epithelial ovarian cancer (EOC). EOC is considered to be a "cold tumour," a tumour that has not triggered a strong response by the immune system. However, tumour infiltrating lymphocytes (TILs) and the expression of programmed cell death ligand (PD-L1) are used as prognostic indicators in EOC. Immunotherapy such as PD-(L)1 inhibitors have shown limited benefit in EOC. Since the immune system is affected by behavioural stress and the beta-adrenergic signalling pathway, this study aimed to explore the impact of propranolol (PRO), a beta-blocker, on anti-tumour immunity in both in vitro and in vivo EOC models. Noradrenaline (NA), an adrenergic agonist, did not directly regulate PD-L1 expression but PD-L1 was significantly upregulated by IFN-γ in EOC cell lines. IFN-γ also increased PD-L1 on extracellular vesicles (EVs) released by ID8 cells. PRO significantly decreased IFN-γ levels in primary immune cells activated ex vivo and showed increased viability of the CD8+ cell population in an EV-immune cell co-incubation. In addition, PRO reverted PD-L1 upregulation and significantly decreased IL-10 levels in an immune-cancer cell co-culture. Chronic behavioural stress increased metastasis in mice while PRO monotherapy and the combo of PRO and PD-(L)1 inhibitor significantly decreased stress-induced metastasis. The combined therapy also reduced tumour weight compared to the cancer control group and induced anti-tumour T-cell responses with significant CD8 expression in tumour tissues. In conclusion, PRO showed a modulation of the cancer immune response by decreasing IFN-γ production and, in turn, IFN-γ-mediated PD-L1 overexpression. The combined therapy of PRO and PD-(L)1 inhibitor decreased metastasis and improved anti-tumour immunity offering a promising new therapy.
Collapse
Affiliation(s)
- M Falcinelli
- University of Brighton, School of Pharmacy & Biosciences, Brighton BN2 4GJ, UK
| | - G Al-Hity
- University of Brighton, School of Pharmacy & Biosciences, Brighton BN2 4GJ, UK
| | - S Baron
- University of Brighton, School of Pharmacy & Biosciences, Brighton BN2 4GJ, UK
| | - M Mampay
- University of Brighton, School of Pharmacy & Biosciences, Brighton BN2 4GJ, UK
| | - M C Allen
- University of Brighton, School of Pharmacy & Biosciences, Brighton BN2 4GJ, UK
| | - M Samuels
- University of Sussex, Department for Biochemistry and Biomedicine, Falmer, Brighton BN1 9QG, UK
| | - W Jones
- University of Sussex, Department for Biochemistry and Biomedicine, Falmer, Brighton BN1 9QG, UK
| | - C Cilibrasi
- University of Sussex, Department for Biochemistry and Biomedicine, Falmer, Brighton BN1 9QG, UK
| | - Renee L Flaherty
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, SW3 6JG London, UK
| | - G Giamas
- University of Sussex, Department for Biochemistry and Biomedicine, Falmer, Brighton BN1 9QG, UK
| | - P H Thaker
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Washington University School of Medicine, St Louis, MO 63110, United States
| | - M S Flint
- University of Brighton, School of Pharmacy & Biosciences, Brighton BN2 4GJ, UK.
| |
Collapse
|
210
|
Immunotherapeutic Approaches in Ovarian Cancer. Curr Issues Mol Biol 2023; 45:1233-1249. [PMID: 36826026 PMCID: PMC9955550 DOI: 10.3390/cimb45020081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer (OC) is gynecological cancer, and diagnosis and treatment are continuously advancing. Next-generation sequencing (NGS)-based diagnoses have emerged as novel methods for identifying molecules and pathways in cancer research. The NGS-based applications have expanded in OC research for early detection and identification of aberrant genes and dysregulation pathways, demonstrating comprehensive views of the entire transcriptome, such as fusion genes, genetic mutations, and gene expression profiling. Coinciding with advances in NGS-based diagnosis, treatment strategies for OC, such as molecular targeted therapy and immunotherapy, have also advanced. Immunotherapy is effective against many other cancers, and its efficacy against OC has also been demonstrated at the clinical phase. In this review, we describe several NGS-based applications for therapeutic targets of OC, and introduce current immunotherapeutic strategies, including vaccines, checkpoint inhibitors, and chimeric antigen receptor (CAR)-T cell transplantation, for effective diagnosis and treatment of OC.
Collapse
|
211
|
Molecular portraits of clear cell ovarian and endometrial carcinoma with comparison to clear cell renal cell carcinoma. Gynecol Oncol 2023; 169:164-171. [PMID: 36333181 DOI: 10.1016/j.ygyno.2022.10.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 10/16/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Advanced clear cell gynecologic malignancies remain among the most challenging diseases to manage. We evaluated ovarian and endometrial clear cell carcinoma (OCCC and ECCC) specimens using comprehensive sequencing technology to identify mutational targets and compared their molecular profiles to histologically similar clear cell renal cell carcinoma (ccRCC). METHODS Using next-generation sequencing (NGS), fragment analysis (FA), and in situ hybridization (ISH), 164 OCCC, 75 ECCC and 234 ccRCC specimens from 2015 to 2018 were evaluated and compared. RESULTS The highest mutation rates in ECCC and OCCC were noted in: ARID1A (75.0%, 87.5%), TP53 (34.8%, 11.1%), PIK3CA (25.0%, 46.8%), PPP2R1A (8.7%, 16.7%), MSI-high (8.8%, 6.4%) and PTEN (8.3%, 7.1%). Among these mutations, there was no significant difference between OCCC and ECCC mutation prevalence except in TP53, with higher mutation rates in ECCC versus OCCC (34.8 vs. 11.1%, respectively, p < 0.05). ccRCC demonstrated different mutation profiles with higher mutation rates in VHL (80.3%), PBRM1 (43.9%), SETD2 (31.1%), and KDM5C (29.2%). By contrast, VHL, PBRM1, and SETD2 mutations were not found in ECCC and OCCC (0.0%). Compared to ccRCC and ECCC, OCCC was found to have a significantly higher tumor mutation burden (TMB) (19.1%). CONCLUSION Gynecologic and renal CCC demonstrate separate and disparate somatic profiles. However, OCCC and ECCC are diseases with similar profiles. TMB and MSI analyses indicate that a subset of OCCC may benefit from immunotherapy. Prospective clinical trials are needed and are underway to examine targeted therapies in these gynecologic disease subtypes.
Collapse
|
212
|
Abiko K, Hamanishi J, Matsumura N, Mandai M. Dynamic host immunity and PD-L1/PD-1 blockade efficacy: developments after "IFN-γ from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer". Br J Cancer 2023; 128:461-467. [PMID: 36068276 PMCID: PMC9938281 DOI: 10.1038/s41416-022-01960-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/01/2022] [Accepted: 08/11/2022] [Indexed: 11/09/2022] Open
Abstract
In the article titled "IFN-γ from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer" in 2015, we showed that PD-L1 expression is induced by IFN-γ from lymphocytes in the tumour microenvironment. This article proposed that PD-L1 expression in cancer cells is not stable but varies among cases, or even within a case, which is influenced by the stromal infiltration of cytotoxic lymphocytes. Immune-checkpoint inhibitors, especially anti-PD-1/PD-L1 therapies, are now widely used to treat various types of cancer. Predictive biomarkers for the efficacy of immune-checkpoint inhibitors include PD-L1 expression, MSI/mismatch repair deficiency and high tumour mutation burden. However, clinical trials have proven that their use in ovarian cancer is still challenging. Reliable biomarkers and new treatment strategies may be sought by elucidating the complex immune microenvironment of ovarian cancer. Although the interaction between cytotoxic lymphocytes and PD-1/PD-L1 on tumour cells is at the centre of therapeutic targets, other immune checkpoints and various immunosuppressive cells also play important roles in ovarian cancer. Targeting these role players in combination with PD-1/PD-L1 blockade may be a promising therapeutic strategy.
Collapse
Affiliation(s)
- Kaoru Abiko
- Department of Obstetrics and Gynecology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan.
| | - Junzo Hamanishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Obstetrics and Gynecology, Kindai University Faculty of Medicine, Osaka-sayama, Osaka prefecture, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
213
|
Li Q, Yang C, Tian H, Jiang J, Li P, Zhu X, Lei T, Yin R, Ding P, Bai P, Li Q. Development of a personalized dendritic cell vaccine and single-cell RNA sequencing-guided assessment of its cell type composition. Cytotherapy 2023; 25:210-219. [PMID: 36443171 DOI: 10.1016/j.jcyt.2022.10.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/11/2022] [Accepted: 10/28/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND AIMS Dendritic cell (DC)-based immunotherapy is a promising approach to treat cancer; however, there is no consensus on the manufacturing processes. Cell type heterogeneity in products manufactured by various methods is understudied and may elicit safety concerns from the regulatory perspective. METHODS We characterized the cell type composition of a recently developed DC vaccine, CUD-002, consisting of DCs loaded with mRNA encoding personalized tumor neoantigens (NCT05270720). RESULTS Using single-cell transcriptomic analysis as an unbiased approach, we found that >80% cells in the final product were DCs and the rest primarily comprised myelocytes and lymphocytes. Subsequent fluorescence-activated cell sorting analyses confirmed these cellular identities. These results indicate that unintended cells originate from leukapheresis, the first step of the manufacturing process, and thus likely safe. Consistently, no overt toxicity or tumorigenicity was observed in mice inoculated with CUD-002. CONCLUSIONS Considering that leukapheresis is a widely used procedure for collecting diverse peripheral blood cell types to manufacture various cytotherapies, this study establishes a workflow to analyze and address regulatory considerations on cell type heterogeneity.
Collapse
Affiliation(s)
- Qingli Li
- Departments of Obstetrics & Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, College of Life Sciences, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Chao Yang
- Departments of Obstetrics & Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, College of Life Sciences, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Huan Tian
- Department of Forensic Genetics, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Jinfeng Jiang
- Non-coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu 610500, Sichuan, China
| | - Ping Li
- Departments of Obstetrics & Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, College of Life Sciences, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiaofeng Zhu
- Departments of Obstetrics & Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, College of Life Sciences, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Tingjun Lei
- Division of Immunology, Sichuan Cunde Therapeutics, Chengdu 610093, Sichuan, China
| | - Rutie Yin
- Departments of Obstetrics & Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, College of Life Sciences, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China
| | - Ping Ding
- Non-coding RNA and Drug Discovery Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu 610500, Sichuan, China
| | - Peng Bai
- Department of Forensic Genetics, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Qintong Li
- Departments of Obstetrics & Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Center of Growth, Metabolism and Aging, College of Life Sciences, State Key Laboratory of Biotherapy and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
214
|
Efficacy of immune checkpoint inhibitor monotherapy or combined with other small molecule-targeted agents in ovarian cancer. Expert Rev Mol Med 2023; 25:e6. [PMID: 36691778 DOI: 10.1017/erm.2023.3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Ovarian cancer is the most lethal female reproductive system tumour. Despite the great advances in surgery and systemic chemotherapy over the past two decades, almost all patients in stages III and IV relapse and develop resistance to chemotherapy after first-line treatment. Ovarian cancer has an extraordinarily complex immunosuppressive tumour microenvironment in which immune checkpoints negatively regulate T cells activation and weaken antitumour immune responses by delivering immunosuppressive signals. Therefore, inhibition of immune checkpoints can break down the state of immunosuppression. Indeed, Immune checkpoint inhibitors (ICIs) have revolutionised the therapeutic landscape of many solid tumours. However, ICIs have yielded modest benefits in ovarian cancer. Therefore, a more comprehensive understanding of the mechanistic basis of the immune checkpoints is needed to improve the efficacy of ICIs in ovarian cancer. In this review, we systematically introduce the mechanisms and expression of immune checkpoints in ovarian cancer. Moreover, this review summarises recent updates regarding ICI monotherapy or combined with other small-molecule-targeted agents in ovarian cancer.
Collapse
|
215
|
Li Y, Fang T, Shan W, Gao Q. Identification of a Novel Model for Predicting the Prognosis and Immune Response Based on Genes Related to Cuproptosis and Ferroptosis in Ovarian Cancer. Cancers (Basel) 2023; 15:cancers15030579. [PMID: 36765541 PMCID: PMC9913847 DOI: 10.3390/cancers15030579] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/20/2023] Open
Abstract
(1) Background: Ovarian cancer (OV) presents a high degree of malignancy and a poor prognosis. Cell death is necessary to maintain tissue function and morphology. Cuproptosis and ferroptosis are two novel forms of death, and we look forward to finding their relationship with OV and providing guidance for treatment. (2) Methods: We derived information about OV from public databases. Based on cuproptosis-related and ferroptosis-related genes, a risk model was successfully constructed, and exceptional subtypes were identified. Next, various methods are applied to assess prognostic value and treatment sensitivity. Besides, the comprehensive analysis of the tumor environment, together with immune cell infiltration, immune function status, immune checkpoint, and human HLA genes, is expected to grant assistance for the prognosis and treatment of OV. (3) Results: Specific molecular subtypes and models possessed excellent potential to predict prognosis. Immune infiltration abundance varied between groups. The susceptibility of individuals to different chemotherapy drugs and immunotherapies could be predicted based on specific groups. (4) Conclusions: Our molecular subtypes and risk model, with strong immune prediction and prognostic prediction capabilities, are committed to guiding ovarian cancer treatment.
Collapse
Affiliation(s)
- Ying Li
- Key Laboratory of the Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tian Fang
- Key Laboratory of the Ministry of Education, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wanying Shan
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Correspondence: (W.S.); (Q.G.)
| | - Qinglei Gao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Correspondence: (W.S.); (Q.G.)
| |
Collapse
|
216
|
Schweer D, Anand N, Anderson A, McCorkle J, Neupane K, Nail AN, Harvey B, Hill KS, Ueland F, Richards C, Kolesar J. Human macrophage-engineered vesicles for utilization in ovarian cancer treatment. Front Oncol 2023; 12:1042730. [PMID: 36713536 PMCID: PMC9875020 DOI: 10.3389/fonc.2022.1042730] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023] Open
Abstract
Background Ovarian cancer is a deadly female malignancy with a high rate of recurrent and chemotherapy-resistant disease. Tumor-associated macrophages (TAMs) are a significant component of the tumor microenvironment and include high levels of M2-protumor macrophages that promote chemoresistance and metastatic spread. M2 macrophages can be converted to M1 anti-tumor macrophages, representing a novel therapeutic approach. Vesicles engineered from M1 macrophages (MEVs) are a novel method for converting M2 macrophages to M1 phenotype-like macrophages. Methods Macrophages were isolated and cultured from human peripheral blood mononuclear cells. Macrophages were stimulated to M1 or M2 phenotypes utilizing LPS/IFN-γ and IL-4/IL-13, respectively. M1 MEVs were generated with nitrogen cavitation and ultracentrifugation. Co-culture of ovarian cancer cells with macrophages and M1 MEVs was followed by cytokine, PCR, and cell viability analysis. Murine macrophage cell line, RAW264.7 cells were cultured and used to generate M1 MEVs for use in ovarian cancer xenograft models. Results M1 MEVs can effectively convert M2 macrophages to an M1-like state both in isolation and when co-cultured with ovarian cancer cells in vitro, resulting in a reduced ovarian cancer cell viability. Additionally, RAW264.7 M1 MEVs can localize to ovarian cancer tumor xenografts in mice. Conclusion Human M1 MEVs can repolarize M2 macrophages to a M1 state and have anti-cancer activity against ovarian cancer cell lines. RAW264.7 M1 MEVs localize to tumor xenografts in vivo murine models.
Collapse
Affiliation(s)
- David Schweer
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Namrata Anand
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States,Department of Pharmacy and Practice, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Abigail Anderson
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - J. Robert McCorkle
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Khaga Neupane
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, United States
| | - Alexandra N. Nail
- Department of Pharmacy and Practice, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Brock Harvey
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, United States
| | - Kristen S. Hill
- Markey Cancer Center, University of Kentucky, Lexington, KY, United States
| | - Frederick Ueland
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Christopher Richards
- Department of Chemistry, College of Arts and Sciences, University of Kentucky, Lexington, KY, United States
| | - Jill Kolesar
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, College of Medicine, University of Kentucky, Lexington, KY, United States,Department of Pharmacy and Practice, College of Pharmacy, University of Kentucky, Lexington, KY, United States,*Correspondence: Jill Kolesar,
| |
Collapse
|
217
|
Devlin MJ, Miller RE. Disparity in the era of personalized medicine for epithelial ovarian cancer. Ther Adv Med Oncol 2023; 15:17588359221148024. [PMID: 36643655 PMCID: PMC9837277 DOI: 10.1177/17588359221148024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023] Open
Abstract
The treatment of high-grade serous ovarian cancer and high-grade endometrioid ovarian cancer has seen significant improvements in recent years, with BRCA1/2 and homologous recombination status guiding a personalized approach which has resulted in improved patient outcomes. However, for other epithelial ovarian cancer subtypes, first-line treatment remains unchanged from the platinum-paclitaxel trials of the early 2000s. In this review, we explore novel therapeutic approaches being adopted in the treatment of clear cell, mucinous, carcinosarcoma and low-grade serous ovarian cancer and the biological rational behind them. We discuss why such disparities exist, the challenges faced in conducting dedicated trials in these rarer histologies and look towards new approaches being adopted to overcome them.
Collapse
Affiliation(s)
| | - Rowan E. Miller
- Department of Medical Oncology, St Bartholomew’s Hospital, London, UK,Department of Medical Oncology, University College London Hospital, London, UK
| |
Collapse
|
218
|
Hwang C, Lee HJ, Na JY, Kim KH, Song YJ, Kim JY, Kim K, Shin DH, Park JY, Kim SY, Lee JH, Choi KU. The stromal tumor-infiltrating lymphocytes, cancer stemness, epithelial-mesenchymal transition, and B7-H4 expression in ovarian serous carcinoma. J Ovarian Res 2023; 16:3. [PMID: 36609273 PMCID: PMC9825048 DOI: 10.1186/s13048-022-01076-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 12/12/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND B7-H4 is expressed in various types of cancers and its expression inversely correlates with the degree of tumor-infiltrating lymphocytes (TILs). Studies have shown the relationship between B7-H4, cancer stem cell (CSC) properties, and epithelial-mesenchymal transition (EMT) in various cancers. However, very few studies have investigated the relationship between B7-H4, TILs, cancer stemness, and EMT in epithelial ovarian cancer (EOC). The present study aimed to elucidate whether B7-H4 is involved in immune evasion and examine whether B7-H4 is associated with cancer stemness or EMT in ovarian serous carcinoma, the most common type of EOC. The clinical significance of B7-H4 was also investigated to evaluate its potential as a therapeutic target. METHODS A total of 145 patients included in this study. The degree of stromal TILs was evaluated using hematoxylin and eosin (H&E)-stained slides. Immunohistochemical analysis of B7-H4, CSC-related biomarkers (CD24, CD44s, CD133, and ALDH1), and EMT-related biomarkers (E-cadherin, N-cadherin, and vimentin) was performed using tissue microarray. qRT-PCR for VTCN1, CD24, CD44, PROM1, ALDH1, CDH1, CDH2, and VIM genes was performed on 38 frozen tissue samples. The mRNA expression levels were analyzed using Gene Expression Profiling Interactive Analysis (GEPIA) online analysis tool. RESULTS B7-H4 protein expression positively correlated with the degree of stromal TILs. CD24, CD44s, and CD133 expression showed a positive correlation with B7-H4 expression at both the protein and mRNA levels, but ALDH1 correlated only at the protein level. E-cadherin expression was positively correlated with B7-H4 expression at both the protein and mRNA levels. N-cadherin and vimentin expression was inversely related to B7-H4 expression only at the mRNA level. B7-H4 positive patients were associated with higher tumor grade and lower overall survival rate than B7-H4 negative patients, especially in ovarian serous carcinoma with low stromal TILs. CONCLUSIONS The present study demonstrates that B7-H4 may not be involved in the immune evasion mechanism, but is involved in cancer stemness and mesenchymal-epithelial transition. In addition, B7-H4 may be a therapeutic target for the treatment of ovarian serous carcinoma, especially with low stromal TILs.
Collapse
Affiliation(s)
- Chungsu Hwang
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Hyun Jung Lee
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Ju-Young Na
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Ki Hyung Kim
- grid.412588.20000 0000 8611 7824Department of Obstetrics and Gynecology, Pusan National University Hospital, 179 Gudeok-Ro, Seo-Gu, Busan, 49241 South Korea
| | - Yong Jung Song
- grid.412591.a0000 0004 0442 9883Department of Obstetrics and Gynecology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Jee Yeon Kim
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Kyungbin Kim
- grid.262229.f0000 0001 0719 8572Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, 179 Gudeok-Ro, Seo-Gu, Busan, 49241 South Korea
| | - Dong Hoon Shin
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Joon Young Park
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - So Young Kim
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Jung Hee Lee
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Kyung Un Choi
- grid.262229.f0000 0001 0719 8572Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, 179 Gudeok-Ro, Seo-Gu, Busan, 49241 South Korea
| |
Collapse
|
219
|
Lee SM, Lee S, Cho HW, Min KJ, Hong JH, Song JY, Lee JK, Lee NW. Application of Immune Checkpoint Inhibitors in Gynecological Cancers: What Do Gynecologists Need to Know before Using Immune Checkpoint Inhibitors? Int J Mol Sci 2023; 24:974. [PMID: 36674491 PMCID: PMC9865129 DOI: 10.3390/ijms24020974] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/30/2022] [Accepted: 12/30/2022] [Indexed: 01/06/2023] Open
Abstract
Standard treatments for gynecological cancers include surgery, chemotherapy, and radiation therapy. However, there are limitations associated with the chemotherapeutic drugs used to treat advanced and recurrent gynecological cancers, and it is difficult to identify additional treatments. Therefore, immune checkpoint inhibitor (ICI) therapy products, including PD-1/PD-L1 inhibitors and CTLA-4 inhibitors, are in the spotlight as alternatives for the treatment of advanced gynecological cancers. Although the ICI monotherapy response rate in gynecological cancers is lower than that in melanoma or non-small cell lung cancer, the response rates are approximately 13-52%, 7-22%, and 4-17% for endometrial, ovarian, and cervical cancers, respectively. Several studies are being conducted to compare the outcomes of combining ICI therapy with chemotherapy, radiation therapy, and antiangiogenesis agents. Therefore, it is critical to determine the mechanism underlying ICI therapy-mediated anti-tumor activity and its application in gynecological cancers. Additionally, understanding the possible immune-related adverse events induced post-immunotherapy, as well as the appropriate management of diagnosis and treatment, are necessary to create a quality environment for immunotherapy in patients with gynecological cancers. Therefore, in this review, we summarize the ICI mechanisms, ICIs applied to gynecological cancers, and appropriate diagnosis and treatment of immune-related side effects to help gynecologists treat gynecological cancers using immunotherapy.
Collapse
Affiliation(s)
- Seon-Mi Lee
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Koreadae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Sanghoon Lee
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Koreadae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Hyun-Woong Cho
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 148, Gurodong-ro, Guro-gu, Seoul 08308, Republic of Korea
| | - Kyung-Jin Min
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si 15355, Gyeonggi-do, Republic of Korea
| | - Jin-Hwa Hong
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 148, Gurodong-ro, Guro-gu, Seoul 08308, Republic of Korea
| | - Jae-Yun Song
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 73 Koreadae-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Jae-Kwan Lee
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 148, Gurodong-ro, Guro-gu, Seoul 08308, Republic of Korea
| | - Nak-Woo Lee
- Department of Obstetrics and Gynecology, Korea University College of Medicine, 123, Jeokgeum-ro, Danwon-gu, Ansan-si 15355, Gyeonggi-do, Republic of Korea
| |
Collapse
|
220
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
221
|
Loss of Major Histocompatibility Complex Class I, CD8 + Tumor-infiltrating Lymphocytes, and PD-L1 Expression in Ovarian Clear Cell Carcinoma. Am J Surg Pathol 2023; 47:124-130. [PMID: 36221308 DOI: 10.1097/pas.0000000000001975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Ovarian clear cell carcinoma (OCCC), a chemoresistant ovarian cancer, shows a modest response to anti-programmed death-1/programmed death ligand-1 (PD-1/PD-L1) therapies. The effects of anti-PD-1/PD-L1 therapies rely on cytotoxic T-cell response, which is triggered by antigen presentation mediated by major histocompatibility complex (MHC) class I. The loss of MHC class I with simultaneous PD-L1 expression has been noted in several cancer types; however, these findings and their prognostic value have rarely been evaluated in OCCC. We collected data from 76 patients with OCCC for clinicopathologic analysis. Loss of MHC class I expression was seen in 44.7% of the cases including 39.3% to 47.4% of the PD-L1 + cases and was associated with fewer CD8 + tumor-infiltrating lymphocytes (TILs). PD-L1 positivity was associated with a higher number of CD8 + TILs. Cox proportional hazard models showed that high (≥50/mm 2 ) CD8 + TILs was associated with shorter disease-specific survival (hazard ratio [HR]=3.447, 95% confidence interval [CI]: 1.222-9.720, P =0.019) and overall survival (HR=3.053, 95% CI: 1.105-8.43, P =0.031). PD-L1 positivity using Combined Positive Score was associated with shorter progression-free survival (HR=3.246, 95% CI: 1.435-7.339, P =0.005), disease-specific survival (HR=4.124, 95% CI: 1.403-12.116, P =0.010), and overall survival (HR=4.489, 95% CI: 1.553-12.972, P =0.006). Loss of MHC class I may contribute to immune evasion and resistance to anti-PD-1/PD-L1 therapies in OCCC, and CD8 + TILs and PD-L1 positivity using Combined Positive Score may have a negative prognostic value.
Collapse
|
222
|
Singh N, Jayraj AS, Sarkar A, Mohan T, Shukla A, Ghatage P. Pharmacotherapeutic treatment options for recurrent epithelial ovarian cancer. Expert Opin Pharmacother 2023; 24:49-64. [PMID: 35968667 DOI: 10.1080/14656566.2022.2112030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Almost 80% of epithelial ovarian cancer present in advanced stage at diagnosis and despite excellent response to surgery and chemotherapy, more than 70% cancers recur. Subsequent therapies become decreasingly effective in controlling the disease, with each successful therapy being effective for a shorter duration. As a result, there is a need for novel therapeutic strategies to effectively treat recurrence. AREAS COVERED In this extensive literature review of high-quality articles, we have focused on surveillance strategy to detect recurrence early, classification of recurrence based on timeline, role of surgery, chemotherapy, and targeted agents such as anti-angiogenetic drugs, PARP inhibitors, and immune checkpoint inhibitors in platinum-sensitive and platinum-resistant disease, respectively. EXPERT OPINION Recurrent ovarian cancers (ROC) are represented by a heterogenous group of patient population in terms of platinum-free interval (PFI), histology, molecular characteristics and immune recognition. In today's era of precision medicine, chemotherapy should be combined with appropriate targeted agent in a multipronged approach to prolong survival and provide better quality of life outcomes by minimizing side effects.
Collapse
Affiliation(s)
- Nilanchali Singh
- Department of Obstetrics and Gynaecology, AIIMS, New Delhi, India
| | | | - Avir Sarkar
- Department of Obstetrics and Gynaecology, AIIMS, New Delhi, India
| | - Trishala Mohan
- Department of Obstetrics and Gynaecology, AIIMS, New Delhi, India
| | - Amlin Shukla
- Division of Reproductive Biology, Indian Council of Medical Research, New Delhi, India
| | - Prafull Ghatage
- Gynaecologic Oncology, Tom Baker Cancer Centre, Calgary, Alberta, Canada
| |
Collapse
|
223
|
Cioffi R, Galli F, Rabaiotti E, Candiani M, Pella F, Candotti G, Bocciolone L, De Marzi P, Mangili G, Bergamini A. Experimental drugs for fallopian cancer: promising agents in the clinical trials and key stumbling blocks for researchers. Expert Opin Investig Drugs 2022; 31:1339-1357. [PMID: 36537209 DOI: 10.1080/13543784.2022.2160313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Fallopian tube carcinoma (FC) as a single entity is a rare disease. Although its diagnosis is increasing thanks to the widespread use of prophylactic salpingectomy, there are no clinical trials exclusively designed for FC. AREAS COVERED This review aims at identifying the most promising trials and future therapeutic pathways in the setting of FC. EXPERT OPINION Hot topics in FC treatment include the consequences of using PARP inhibitors (PARPi) as first-line therapy, ways to overcome platinum resistance, and the role of immunotherapy. Patient selection is a key point for future development of target therapies. Next-generation sequencing (NGS) is one of the most investigated technologies both for drug discovery and identification of reverse mutations, involved in resistance to PARPi and platinum. New, promising molecular targets are emerging. Notwithstanding the disappointing outcomes when used by itself, immunotherapy in FC treatment could still have a role in combination with other agents, exploiting synergistic effects at the molecular level. The development of cancer vaccines is currently hampered by the high variability of tumor neoantigens in FC. Genomic profiling could be a solution, allowing the synthesis of individualized vaccines.
Collapse
Affiliation(s)
- Raffaella Cioffi
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Federica Galli
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Emanuela Rabaiotti
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Massimo Candiani
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Francesca Pella
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giorgio Candotti
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Luca Bocciolone
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Patrizia De Marzi
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Giorgia Mangili
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| | - Alice Bergamini
- Obstetrics and Gynecology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132, Milan, Italy
| |
Collapse
|
224
|
Xu T, Liu Z, Huang L, Jing J, Liu X. Modulating the tumor immune microenvironment with nanoparticles: A sword for improving the efficiency of ovarian cancer immunotherapy. Front Immunol 2022; 13:1057850. [PMID: 36532066 PMCID: PMC9751906 DOI: 10.3389/fimmu.2022.1057850] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
With encouraging antitumor effects, immunotherapy represented by immune checkpoint blockade has developed into a mainstream cancer therapeutic modality. However, only a minority of ovarian cancer (OC) patients could benefit from immunotherapy. The main reason is that most OC harbor a suppressive tumor immune microenvironment (TIME). Emerging studies suggest that M2 tumor-associated macrophages (TAMs), T regulatory cells (Tregs), myeloid-derived suppressor cells (MDSCs), and cancer-associated fibroblasts (CAFs) are enriched in OC. Thus, reversing the suppressive TIME is considered an ideal candidate for improving the efficiency of immunotherapy. Nanoparticles encapsulating immunoregulatory agents can regulate immunocytes and improve the TIME to boost the antitumor immune response. In addition, some nanoparticle-mediated photodynamic and photothermal therapy can directly kill tumor cells and induce tumor immunogenic cell death to activate antigen-presenting cells and promote T cell infiltration. These advantages make nanoparticles promising candidates for modulating the TIME and improving OC immunotherapy. In this review, we analyzed the composition and function of the TIME in OC and summarized the current clinical progress of OC immunotherapy. Then, we expounded on the promising advances in nanomaterial-mediated immunotherapy for modulating the TIME in OC. Finally, we discussed the obstacles and challenges in the clinical translation of this novel combination treatment regimen. We believe this resourceful strategy will open the door to effective immunotherapy of OC and benefit numerous patients.
Collapse
Affiliation(s)
| | | | | | - Jing Jing
- *Correspondence: Xiaowei Liu, ; Jing Jing,
| | | |
Collapse
|
225
|
Ramnaraign BH, Lee JH, Ali A, Rogers SC, Fabregas JC, Thomas RM, Allegra CJ, Sahin I, DeRemer DL, George TJ, Chatzkel JA. Atezolizumab plus tivozanib for immunologically cold tumor types: the IMMCO-1 trial. Future Oncol 2022; 18:3815-3822. [PMID: 36399037 DOI: 10.2217/fon-2022-0392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Immune checkpoint inhibitor therapy represents a significant advance in cancer care. The interaction between PD-1 and PD-L1 induces immune tolerance and the inhibition of this interaction is an effective treatment strategy for numerous malignancies. Despite its demonstrated potential, immunotherapy is not clinically effective in immunogenically 'cold' tumors such as pancreatic cancer, prostate cancer and neuroendocrine tumors. Through the inhibition of VEGF, it may be possible to potentiate the effect of immune checkpoint blockade in tumors that have traditionally shown a lack of clinical response to immunotherapy. This signal-seeking, single-arm, prospective clinical trial aims to determine the objective response of tivozanib and atezolizumab in advanced immunogenically cold solid tumors. Clinical Trial Registration: NCT05000294 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Brian H Ramnaraign
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Ji-Hyun Lee
- Department of Biostatistics, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Azka Ali
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Sherise C Rogers
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Jesus C Fabregas
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Ryan M Thomas
- Department of Surgery, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Carmen J Allegra
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Ilyas Sahin
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - David L DeRemer
- Department of Pharmacy, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Thomas J George
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| | - Jonathan A Chatzkel
- Department of Medicine, University of Florida, 1600 SW Archer Rd, Gainesville, FL 32608, USA
| |
Collapse
|
226
|
Meng L, Zhang Z, Zhang X, Zhang X, Wei Y, Wu B, Xue X, Zhang X, He X, Xiao Y. Case report: Local cryoablation combined with pembrolizumab to eliminate lung metastases from ovarian clear cell carcinoma. Front Immunol 2022; 13:1006500. [DOI: 10.3389/fimmu.2022.1006500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 10/25/2022] [Indexed: 11/12/2022] Open
Abstract
Ovarian clear cell carcinoma has a high recurrence rate with poor prognosis and is generally not sensitive to conventional platinum-based chemotherapy. Its less frequent occurrence of mutations such as BRCA limited the targeted therapies. Immunotherapy is not currently recommended as a first-line agent for ovarian cancer, and most patients are not yet able to benefit from it. Cryoablation can be used to treat solid systemic tumors, including ovarian cancer metastases, and can produce a limited anti-tumor immune response. The anti-tumor effects of cryoablation combined with immunotherapy have not been adequately confirmed. This study reports a case of a patient with ovarian clear cell carcinoma who underwent conventional adjuvant chemotherapy after initially surgical resection of the tumor. Unfortunately, cancer recurred and metastasized to the abdominal wall. After a series of painful chemotherapy and a second surgery, the cancer was still not effectively controlled, and the patient developed extensive metastases in the lung. The patient’s PD-L1 expression level also did not support solo immunotherapy. We pioneered the use of cryoablation to first eradicate the most significant lesion in the upper lobe of the left lung and then combined it with the PD-L1 inhibitor pembrolizumab to treat the patient with immunotherapy, which resulted in the complete eradication of the other multiple metastases in the lung and saved the patient’s life. Although the precise mechanism of action has not yet been explored, we have reason to believe that the combination of cryoablation and immune checkpoint inhibitor has a powerful synergistic anti-tumor effect, which is yet to be confirmed by more basic research and clinical applications in the next step.
Collapse
|
227
|
Shah JB, Pueschl D, Wubbenhorst B, Fan M, Pluta J, D'Andrea K, Hubert AP, Shilan JS, Zhou W, Kraya AA, Llop Guevara A, Ruan C, Serra V, Balmaña J, Feldman M, Morin PJ, Nayak A, Maxwell KN, Domchek SM, Nathanson KL. Analysis of matched primary and recurrent BRCA1/2 mutation-associated tumors identifies recurrence-specific drivers. Nat Commun 2022; 13:6728. [PMID: 36344544 PMCID: PMC9640723 DOI: 10.1038/s41467-022-34523-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 10/27/2022] [Indexed: 11/09/2022] Open
Abstract
Recurrence is a major cause of death among BRCA1/2 mutation carriers with breast (BrCa) and ovarian cancers (OvCa). Herein we perform multi-omic sequencing on 67 paired primary and recurrent BrCa and OvCa from 27 BRCA1/2 mutation carriers to identify potential recurrence-specific drivers. PARP1 amplifications are identified in recurrences (False Discovery Rate q = 0.05), and PARP1 is significantly overexpressed across primary BrCa and recurrent BrCa and OvCa, independent of amplification status. RNA sequencing analysis finds two BRCA2 isoforms, BRCA2-201/Long and BRCA2-001/Short, respectively predicted to be sensitive and insensitive to nonsense-mediated decay. BRCA2-001/Short is expressed more frequently in recurrences and associated with reduced overall survival in breast cancer (87 vs. 121 months; Hazard Ratio = 2.5 [1.18-5.5]). Loss of heterozygosity (LOH) status is discordant in 25% of patient's primary and recurrent tumors, with switching between both LOH and lack of LOH found. Our study reveals multiple potential drivers of recurrent disease in BRCA1/2 mutation-associated cancer, improving our understanding of tumor evolution and suggesting potential biomarkers.
Collapse
Affiliation(s)
- Jennifer B Shah
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Dana Pueschl
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Bradley Wubbenhorst
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mengyao Fan
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - John Pluta
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kurt D'Andrea
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anna P Hubert
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jake S Shilan
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Wenting Zhou
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Adam A Kraya
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Alba Llop Guevara
- Experimental Therapeutics Group, Vall d'Hebron Institut d'Oncologia, Barcelona, Spain
| | - Catherine Ruan
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Violeta Serra
- Experimental Therapeutics Group, Vall d'Hebron Institut d'Oncologia, Barcelona, Spain
| | - Judith Balmaña
- Hereditary Cancer Genetics Group, Vall d'Hebron Institut d'Oncologia, Barcelona, Spain
- Department of Medical Oncology, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Michael Feldman
- Division of Surgical Pathology, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Pat J Morin
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Anupma Nayak
- Division of Surgical Pathology, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Kara N Maxwell
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
- Basser Center for BRCA, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Susan M Domchek
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
- Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
- Basser Center for BRCA, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Katherine L Nathanson
- Division of Translational Medicine and Human Genetics, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
- Basser Center for BRCA, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
228
|
Devlin MJ, Miller R, Laforets F, Kotantaki P, Garsed DW, Kristeleit R, Bowtell DD, McDermott J, Maniati E, Balkwill FR. The Tumor Microenvironment of Clear-Cell Ovarian Cancer. Cancer Immunol Res 2022; 10:1326-1339. [PMID: 36095166 PMCID: PMC9627265 DOI: 10.1158/2326-6066.cir-22-0407] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/01/2022] [Accepted: 09/02/2022] [Indexed: 01/07/2023]
Abstract
Some patients with advanced clear-cell ovarian cancer (CCOC) respond to immunotherapy; however, little is known about the tumor microenvironment (TME) of this relatively rare disease. Here, we describe a comprehensive quantitative and topographical analysis of biopsies from 45 patients, 9 with Federation Internationale des Gynaecologistes et Obstetristes (FIGO) stage I/II (early CCOC) and 36 with FIGO stage III/IV (advanced CCOC). We investigated 14 immune cell phenotype markers, PD-1 and ligands, and collagen structure and texture. We interrogated a microarray data set from a second cohort of 29 patients and compared the TMEs of ARID1A-wildtype (ARID1Awt) versus ARID1A-mutant (ARID1Amut) disease. We found significant variations in immune cell frequency and phenotype, checkpoint expression, and collagen matrix between the malignant cell area (MCA), leading edge (LE), and stroma. The MCA had the largest population of CD138+ plasma cells, the LE had more CD20+ B cells and T cells, whereas the stroma had more mast cells and αSMA+ fibroblasts. PD-L2 was expressed predominantly on malignant cells and was the dominant PD-1 ligand. Compared with early CCOC, advanced-stage disease had significantly more fibroblasts and a more complex collagen matrix, with microarray analysis indicating "TGFβ remodeling of the extracellular matrix" as the most significantly enriched pathway. Data showed significant differences in immune cell populations, collagen matrix, and cytokine expression between ARID1Awt and ARID1Amut CCOC, which may reflect different paths of tumorigenesis and the relationship to endometriosis. Increased infiltration of CD8+ T cells within the MCA and CD4+ T cells at the LE and stroma significantly associated with decreased overall survival.
Collapse
Affiliation(s)
- Michael-John Devlin
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
- Department of Medical Oncology, St Bartholomew's Hospital, London, United Kingdom
| | - Rowan Miller
- Department of Medical Oncology, St Bartholomew's Hospital, London, United Kingdom
- Department of Medical Oncology, University College London Hospital, London, United Kingdom
| | - Florian Laforets
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Panoraia Kotantaki
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Dale W. Garsed
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Rebecca Kristeleit
- Medical Oncology Department, Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom
| | | | - Jacqueline McDermott
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Eleni Maniati
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Frances R. Balkwill
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| |
Collapse
|
229
|
Kandalaft LE, Dangaj Laniti D, Coukos G. Immunobiology of high-grade serous ovarian cancer: lessons for clinical translation. Nat Rev Cancer 2022; 22:640-656. [PMID: 36109621 DOI: 10.1038/s41568-022-00503-z] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/28/2022] [Indexed: 11/09/2022]
Abstract
Treatment of high-grade serous ovarian cancer (HGSOC) remains challenging. Although HGSOC can potentially be responsive to immunotherapy owing to endogenous immunity at the molecular or T cell level, immunotherapy for this disease has fallen short of expectations to date. This Review proposes a working classification for HGSOC based on the presence or absence of intraepithelial T cells, and elaborates the putative mechanisms that give rise to such immunophenotypes. These differences might explain the failures of existing immunotherapies, and suggest that rational therapeutic approaches tailored to each immunophenotype might meet with improved success. In T cell-inflamed tumours, treatment could focus on mobilizing pre-existing immunity and strengthening the activation of T cells embedded in intraepithelial tumour myeloid niches. Conversely, in immune-excluded and immune-desert tumours, treatment could focus on restoring inflammation by reprogramming myeloid cells, stromal cells and vascular epithelial cells. Poly(ADP-ribose) polymerase (PARP) inhibitors, low-dose radiotherapy, epigenetic drugs and anti-angiogenesis therapy are among the tools available to restore T cell infiltration in HGSOC tumours and could be implemented in combination with vaccines and redirected T cells.
Collapse
Affiliation(s)
- Lana E Kandalaft
- Ludwig Institute for Cancer Research, Lausanne Branch, and Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Denarda Dangaj Laniti
- Ludwig Institute for Cancer Research, Lausanne Branch, and Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - George Coukos
- Ludwig Institute for Cancer Research, Lausanne Branch, and Department of Oncology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
230
|
Zhang J, Lu M, Xu H, Ren F, Zhu L. Molecular subtypes based on cuproptosis-related genes and tumor microenvironment infiltration characterization in ovarian cancer. Cancer Cell Int 2022; 22:328. [PMID: 36307842 PMCID: PMC9617300 DOI: 10.1186/s12935-022-02756-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/15/2022] [Indexed: 12/03/2022] Open
Abstract
Background Cuproptosis (copper death) is a recently found cell death type produced by copper iron; nonetheless, the properties of cuproptosis molecular subtypes and possible involvement of cuproptosis-related genes (CRGs) in the tumor microenvironment (TME) in ovarian cancer (OC) remain unknown. Methods CRG changes were characterized at the genomic and transcriptional levels in 656 OC samples, and their expression patterns were investigated using three different datasets. Results We identified three distinct molecular subtypes, and discovered that variations in molecular subtypes were linked to patient prognosis, TME cell infiltration characteristics, malignancy, and immune-related pathways. Then, in order to predict overall survival (OS), we created a risk score and tested its predictive potential in OC patients. As a result, we created a very accurate nomogram to increase risk score clinical applicability. Better OS, younger age, early stage, and immune activity were all associated with a low risk score. The hallmarks of a high-risk score are older age, advanced stage, immunosuppression, and a bad prognosis. Furthermore, risk score was linked to immune checkpoint expression (including PD-L1, CTLA4), targeted therapy gene expression (PARP, PDGFRA), cancer stem cell (CSC), chemotherapy and targeted medication sensitivity. Conclusions Our comprehensive analysis of CRGs in OC showed their potential role in TME, clinicopathological characteristics, chemotherapy and targeted drug screening and prognosis. These discoveries could help us better understand CRGs in OC, as well as pave the path for novel ways to assess prognosis and design more effective immunotherapy strategies. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02756-y.
Collapse
|
231
|
Xu Y, Zuo F, Wang H, Jing J, He X. The current landscape of predictive and prognostic biomarkers for immune checkpoint blockade in ovarian cancer. Front Immunol 2022; 13:1045957. [PMID: 36389711 PMCID: PMC9647049 DOI: 10.3389/fimmu.2022.1045957] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/14/2022] [Indexed: 11/29/2022] Open
Abstract
Immune checkpoint blockade (ICB) therapy has evoked a prominent shift in anticancer therapy. Durable clinical antitumor activity to ICB has been observed in patients with ovarian cancer (OC). However, only a subset of patients derive clinical benefit, and immune-related adverse events (irAEs) caused by ICB therapy can lead to permanent tissue damage and even fatal consequences. It is thus urgent to develop predictive biomarkers to optimize patient outcomes and minimize toxicity risk. Herein, we review current predictive and prognostic biomarkers for checkpoint immunotherapy in OC and highlight emerging biomarkers to guide treatment with ICB. The prevalent biomarkers, such as PD-L1 expression status, tumor-infiltrating lymphocytes, mutational burden, and immune gene signatures, are further discussed. We provide a state-of-the-art survey on prognostic and predictive biomarkers for checkpoint immunotherapy and offer valuable information for guiding precision immunotherapy
Collapse
Affiliation(s)
| | | | | | - Jing Jing
- *Correspondence: Jing Jing, ; Xiujing He,
| | - Xiujing He
- *Correspondence: Jing Jing, ; Xiujing He,
| |
Collapse
|
232
|
Liu Y, Du S, Yuan M, He X, Zhu C, Han K, Zhu Y, Yang Q, Tong R. Identification of a novel ferroptosis-related gene signature associated with prognosis, the immune landscape, and biomarkers for immunotherapy in ovarian cancer. Front Pharmacol 2022; 13:949126. [PMID: 36386203 PMCID: PMC9641277 DOI: 10.3389/fphar.2022.949126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 10/11/2022] [Indexed: 12/09/2022] Open
Abstract
Ferroptosis has been implicated in tumor progression and immunoregulation. Identification of ferroptosis-related prognostic gene is important for immunotherapy and prognosis in ovarian cancer (OV). We assessed the potential predictive power of a novel ferroptosis-related gene (FRG) signature for prognosis and immunotherapy in Asian and Caucasian OV populations. We collected gene expression profiles and clinicopathological data from public databases. The least absolute shrinkage and selection operator Cox regression algorithm was used to construct the FRG signature. Receiver operating characteristic (ROC) curve, Kaplan-Meier method, Cox regression model were used to evaluate the clinical benefits of FRG signature. Gene functional and gene set enrichment analyses were used for functional annotation and immune landscape analysis. A 15-FRG signature was constructed and used to stratify patients into two risk groups. Patients in the high-risk group had significantly worse survival. The risk score was a significant independent risk factor for OS. The area under the ROC curve indicated the good prediction performance of the FRG signature. Notably, the low-risk group showed a significant enrichment in immune-related pathways and a “hot” immune status. The risk score was found to be an efficient and robust predictor of response to immunotherapy. In conclusion, our study identified a novel 15-FRG prognostic signature that can be used for prognostic prediction and precision immunotherapy in Asian and Caucasian OV populations.
Collapse
Affiliation(s)
- Yilong Liu
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Suya Du
- Department of Clinical Pharmacy, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Mengying Yuan
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xia He
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Changyu Zhu
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Ke Han
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yuyan Zhu
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qianwen Yang
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Personalized Drug Therapy Key Laboratory of Sichuan Province, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- *Correspondence: Rongsheng Tong,
| |
Collapse
|
233
|
Dumitru A, Dobrica EC, Croitoru A, Cretoiu SM, Gaspar BS. Focus on PD-1/PD-L1 as a Therapeutic Target in Ovarian Cancer. Int J Mol Sci 2022; 23:ijms232012067. [PMID: 36292922 PMCID: PMC9603705 DOI: 10.3390/ijms232012067] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/27/2022] [Accepted: 10/06/2022] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is considered one of the most aggressive and deadliest gynecological malignancies worldwide. Unfortunately, the therapeutic methods that are considered the gold standard at this moment are associated with frequent recurrences. Survival in ovarian cancer is associated with the presence of a high number of intra tumor infiltrating lymphocytes (TILs). Therefore, immunomodulation is considered to have an important role in cancer treatment, and immune checkpoint inhibitors may be useful for restoring T cell-mediated antitumor immunity. However, the data presented in the literature until now are not sufficient to allow for the identification and selection of patients who really respond to immunotherapy among those with ovarian cancer. Although there are some studies with favorable results, more prospective trials are needed in this sense. This review focuses on the current and future perspectives of PD-1/L1 blockade in ovarian cancer and analyzes the most important immune checkpoint inhibitors used, with the aim of achieving optimal clinical outcomes. Future studies and trials are needed to maximize the efficacy of immune checkpoint blockade therapy in ovarian cancer, as well as in all cancers, in general.
Collapse
Affiliation(s)
- Adrian Dumitru
- Department of Pathology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Pathology, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania
| | - Elena-Codruta Dobrica
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Dermatology, Elias University Hospital, 011461 Bucharest, Romania
| | - Adina Croitoru
- Department of Medical Oncology, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Department of Oncology, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Sanda Maria Cretoiu
- Department of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Correspondence:
| | - Bogdan Severus Gaspar
- Surgery Department, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
234
|
Luo X, Sun Y, Li J, Jiang Q, Yuan L, Li T, Chen M, Yao L. A durable response to programmed cell death 1 blockade in a multidrug-resistant recurrent ovarian cancer patient with HLA-B44 supertype: A case report. Front Immunol 2022; 13:951422. [PMID: 36275748 PMCID: PMC9582938 DOI: 10.3389/fimmu.2022.951422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/26/2022] [Indexed: 11/30/2022] Open
Abstract
Relapsed/refractory ovarian cancer, especially platinum resistance recurrence, remains a major therapeutic challenge. Here, we present the case of a patient with recurrent ovarian clear cell carcinoma (OCCC) who failed to respond to multiline chemotherapy and target therapy but achieved an immune complete response (iCR) with programmed cell death 1 (PD-1) inhibitor treatment. The overall survival (OS) was 59 months, and the recurrence-free survival (RFS) was 34 months after immunotherapy, which was counting. Meantime, molecular testing results revealed that traditional biomarkers for immunotherapy, including PD-L1 expression, microsatellite instability (MSI), and tumor mutational burden (TMB), were negative. HLA-B44 (B*18:01) supertype was confirmed by sequence-based HLA typing. This case raises the possibility that ovarian cancer patients with multidrug resistance may still benefit from PD-1 inhibitor therapy, even if PD-L1 pathology is negative.
Collapse
Affiliation(s)
- Xukai Luo
- Department of Gynecology Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yating Sun
- Department of Gynecology Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jiajia Li
- Department of Gynecology Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Qidi Jiang
- Department of Gynecology Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Lei Yuan
- Department of Gynecology Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Ting Li
- Department of Bioinformatics, Precision Scientific (Beijing) CO., Ltd., Beijing, China
| | - Mo Chen
- Department of Gynecology Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- *Correspondence: Mo Chen, ; Liangqing Yao,
| | - Liangqing Yao
- Department of Gynecology Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
- *Correspondence: Mo Chen, ; Liangqing Yao,
| |
Collapse
|
235
|
Ethier JL, Fuh KC, Arend R, Konecny GE, Konstantinopoulos PA, Odunsi K, Swisher EM, Kohn EC, Zamarin D. State of the Biomarker Science in Ovarian Cancer: A National Cancer Institute Clinical Trials Planning Meeting Report. JCO Precis Oncol 2022; 6:e2200355. [PMID: 36240472 PMCID: PMC9848534 DOI: 10.1200/po.22.00355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/22/2022] [Accepted: 08/26/2022] [Indexed: 01/21/2023] Open
Abstract
PURPOSE Despite therapeutic advances in the treatment of ovarian cancer (OC), 5-year survival remains low, and patients eventually die from recurrent, chemotherapy-resistant disease. The National Cancer Gynecologic Cancer Steering Committee identified the integration of scientifically defined subgroups as a top strategic priority in clinical trial planning. METHODS A group of experts was convened to review the scientific literature in OC to identify validated predictive biomarkers that could inform patient selection and treatment stratification. Here, we report on these findings and their potential for use in future clinical trial design on the basis of hierarchal evidence grading. RESULTS The biomarkers were classified on the basis of mechanistic targeting, including DNA repair and replication stress, immunotherapy and tumor microenvironment, oncogenic signaling, and angiogenesis. Currently, BRCA mutations and homologous recombination deficiency to predict poly (ADP-ribose) polymerase inhibitor response are supported in OC by the highest level of evidence. Additional biomarkers of response to agents targeting the pathways above have been identified but require prospective validation. CONCLUSION Although a number of biomarkers of response to various agents in OC have been described in the literature, high-level evidence for the majority is lacking. This report highlights the unmet need for identification and validation of predictive biomarkers to guide therapy and future trial design in OC.
Collapse
Affiliation(s)
- Josee-Lyne Ethier
- Department of Oncology, Cancer Centre of Southeastern Ontario, Queen's University, Kingston, ON, Canada
| | - Katherine C. Fuh
- Division of Gynecologic Oncology, Washington University St Louis, St Louis, MO
| | - Rebecca Arend
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingam, AL
| | - Gottfried E. Konecny
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
| | | | - Kunle Odunsi
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL
| | | | - Elise C. Kohn
- Clinical Investigations Branch of The Cancer Therapy Evaluation Program, National Cancer Institute, Rockville, ML
| | - Dmitriy Zamarin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
236
|
Wu Y, Zhang X, Li L, Yang W, Yan Z, Gu C, Zhang Z, Zhou J, Liu L, Ye M, Meng Y. Case Report and Review of Literature: Camrelizumab Combined with Fuzuloparib and Apatinib for Platinum-Resistant Recurrent Ovarian Cancer. Onco Targets Ther 2022; 15:973-979. [PMID: 36118677 PMCID: PMC9480580 DOI: 10.2147/ott.s375643] [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: 05/28/2022] [Accepted: 08/18/2022] [Indexed: 11/23/2022] Open
Abstract
Background The mortality rate of ovarian cancer (OC) ranks first among female genital tract malignant tumors, which seriously threatens women's life and health. Because of its insidious onset and poor prognosis, it has become a thorny problem in the clinic, especially for patients with platinum-resistant recurrent ovarian cancer (PROC). In recent years, the medical treatment of OC has made gratifying results, bringing hope to the patients. Case Description A 54-year-old OC patient who has failed previous neoadjuvant chemotherapy, cytoreductive surgery, and postoperative chemotherapy was diagnosed with PROC. Then she received combination treatment of fuzuloparib (100mg PO BID), apatinib (250mg PO QD), and camrelizumab (200mg IV Q3W) for every 3-week cycle in a Phase II study for PROC patients. In the phase II study, her condition stabilized, responded well to treatment with a sharp decrease by 91.14% of target lesions and disappearances of non-target lesions, and continued to receive regular treatment with progression-free survival exceeding 15 months and no serious adverse events. Conclusion The present case proves PROC patients might have a sustained response to triplet combination with camrelizumab, combined with fuzuloparib and apatinib.
Collapse
Affiliation(s)
- Yawen Wu
- School of Medicine, Nankai University, Tianjin, People's Republic of China.,Department of Gynecology and Obstetrics, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Xiaoyan Zhang
- School of Medicine, Nankai University, Tianjin, People's Republic of China.,Department of Gynecology and Obstetrics, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lian Li
- Department of Gynecology and Obstetrics, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Wen Yang
- Department of Gynecology and Obstetrics, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhifeng Yan
- Department of Gynecology and Obstetrics, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Chenglei Gu
- Department of Gynecology and Obstetrics, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhe Zhang
- Department of Gynecology and Obstetrics, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Jiahuan Zhou
- Department of Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, People's Republic of China
| | - Lulu Liu
- Department of Clinical Research & Development, Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, People's Republic of China
| | - Mingxia Ye
- Department of Gynecology and Obstetrics, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Yuanguang Meng
- Department of Gynecology and Obstetrics, The First Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| |
Collapse
|
237
|
Identification of Novel Immunologic Checkpoint Gene Prognostic Markers for Ovarian Cancer. JOURNAL OF ONCOLOGY 2022; 2022:8570882. [PMID: 36157232 PMCID: PMC9499758 DOI: 10.1155/2022/8570882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 12/24/2022]
Abstract
Ovarian cancer has a higher resistance to chemotherapy, displaying the highest mortality rate among gynecological cancers. Recently, immune checkpoint inhibitor therapy is an effective treatment for selected patients. However, a low response rate for immune checkpoint treatment was observed for ovarian cancer patients. Therefore, it is necessary to identify ovarian cancer patients who might gain benefits from immune checkpoint treatment. Datasets containing ovarian cancer samples with mRNA-seq and clinical follow-up data were downloaded from different databases like The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). The researchers applied the univariate analysis for selecting the immune checkpoint genes (ICGs) at a significance level of P < 0.05 as the candidate ICGs. The Spearman correlation coefficients were calculated to compare the correlation between tumor mutation burden and candidate ICGs, and the Kaplan-Meier plots were generated. They also assessed the external validation datasets and the results of immunohistochemical staining. 46 and 35 ICGs were extracted from the TCGA and GEO datasets, respectively, and we categorized the ICGs into 3 expression patterns. Nine (TCGA) and three (GEO) ICGs were significantly related to the prognosis. Univariate survival analysis indicated a significant prognostic relationship between the expression levels of ICOS, TIGIT, and TNFRSF8 and overall survival (OS). Moreover, the expression of ICOS and TIGIT also presented a significantly positive relationship with the CD8A expression. Importantly, patients with a higher CD8A and ICOS expression level (ICOS-H/CD8A-H) showed a better survival rate compared to other patients. Stratified analysis using TIGIT, TNFRSF8, and CD8A expression also showed an improved prognosis for the high TIGIT/high CD8A expression subgroup and the low TNFRSF8/low CD8A expression subgroup compared to the other subgroups. This study identified different immune subtypes that can predict the OS of ovarian cancer patients. This data could prove to be beneficial for making important clinical decisions and designing individual immunotherapeutic strategies.
Collapse
|
238
|
Bound NT, Vandenberg CJ, Kartikasari AER, Plebanski M, Scott CL. Improving PARP inhibitor efficacy in high-grade serous ovarian carcinoma: A focus on the immune system. Front Genet 2022; 13:886170. [PMID: 36159999 PMCID: PMC9505691 DOI: 10.3389/fgene.2022.886170] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 08/05/2022] [Indexed: 12/03/2022] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is a genomically unstable malignancy responsible for over 70% of all deaths due to ovarian cancer. With roughly 50% of all HGSOC harboring defects in the homologous recombination (HR) DNA repair pathway (e.g., BRCA1/2 mutations), the introduction of poly ADP-ribose polymerase inhibitors (PARPi) has dramatically improved outcomes for women with HR defective HGSOC. By blocking the repair of single-stranded DNA damage in cancer cells already lacking high-fidelity HR pathways, PARPi causes the accumulation of double-stranded DNA breaks, leading to cell death. Thus, this synthetic lethality results in PARPi selectively targeting cancer cells, resulting in impressive efficacy. Despite this, resistance to PARPi commonly develops through diverse mechanisms, such as the acquisition of secondary BRCA1/2 mutations. Perhaps less well documented is that PARPi can impact both the tumour microenvironment and the immune response, through upregulation of the stimulator of interferon genes (STING) pathway, upregulation of immune checkpoints such as PD-L1, and by stimulating the production of pro-inflammatory cytokines. Whilst targeted immunotherapies have not yet found their place in the clinic for HGSOC, the evidence above, as well as ongoing studies exploring the synergistic effects of PARPi with immune agents, including immune checkpoint inhibitors, suggests potential for targeting the immune response in HGSOC. Additionally, combining PARPi with epigenetic-modulating drugs may improve PARPi efficacy, by inducing a BRCA-defective phenotype to sensitise resistant cancer cells to PARPi. Finally, invigorating an immune response during PARPi therapy may engage anti-cancer immune responses that potentiate efficacy and mitigate the development of PARPi resistance. Here, we will review the emerging PARPi literature with a focus on PARPi effects on the immune response in HGSOC, as well as the potential of epigenetic combination therapies. We highlight the potential of transforming HGSOC from a lethal to a chronic disease and increasing the likelihood of cure.
Collapse
Affiliation(s)
- Nirashaa T. Bound
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Cancer Ageing and Vaccines (CAVA), Translational Immunology & Nanotechnology Research Program, School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Cassandra J. Vandenberg
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Apriliana E. R. Kartikasari
- Cancer Ageing and Vaccines (CAVA), Translational Immunology & Nanotechnology Research Program, School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Magdalena Plebanski
- Cancer Ageing and Vaccines (CAVA), Translational Immunology & Nanotechnology Research Program, School of Health & Biomedical Sciences, RMIT University, Bundoora, VIC, Australia
| | - Clare L. Scott
- Cancer Biology and Stem Cells, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
- Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Royal Women’s Hospital, Parkville, VIC, Australia
| |
Collapse
|
239
|
Li Y, Tian R, Liu J, Li J, Tan H, Wu Q, Fu X. Deciphering the immune landscape dominated by cancer-associated fibroblasts to investigate their potential in indicating prognosis and guiding therapeutic regimens in high grade serous ovarian carcinoma. Front Immunol 2022; 13:940801. [PMID: 36119108 PMCID: PMC9478207 DOI: 10.3389/fimmu.2022.940801] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022] Open
Abstract
Limited immunotherapeutic effect in high-grade serous ovarian carcinoma (HGSOC) propels exploration of the mechanics behind this resistance, which may be partly elucidated by investigating characters of cancer-associated fibroblasts (CAFs), a significant population in HGSOC involved in shaping tumor immune microenvironment. Herein, leveraging gene expression data of HGSOC samples from The Cancer Genome Atlas and Gene Expression Omnibus datasets, we suggested that CAFs detrimentally affected the outcomes of HGSOC patients. Subsequently, we performed weighted gene co-expression network analysis (WGCNA) to identify a CAFs-related module and screened out seven hub genes from this module, all of which were positively correlated with the infiltration of immunosuppressive macrophages. As one of the hub genes, the expression of fibrillin 1 (FBN1) and its relevance to CD206 were further verified by immunohistochemistry staining in HGSOC samples. Meanwhile, we extracted genes that correlated well with CAF signatures to construct a CAFscore. The capacity of the CAFscore as an independent prognostic factor was validated by Cox regression analyses, and its relevance to components as well as signals in the tumor immune microenvironment was also investigated. Under the evaluation by the CAFscore, HGSOC patients with relatively high CAFscore had worse outcomes, activated mesenchymal signaling pathways, and immune checkpoint blockade (ICB) resistance signatures, which was consistent with the fact that non-responders in anti-PD-1 treatment cohorts tended to have higher CAFscore. Besides, the possibility of CAFscore to guide the selection of sensitive chemotherapeutic agents was explored. In conclusion, individualized assessment of the CAFscore could uncover the extent of stroma activation and immunosuppression and inform therapeutic strategies to improve the benefit of therapies.
Collapse
Affiliation(s)
- Yimin Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruotong Tian
- Department of Pharmacology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jiaxin Liu
- Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Juanni Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
| | - Hong Tan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Xiaodan Fu, ; ; Qihui Wu, ; Hong Tan,
| | - Qihui Wu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- Department of Obstetrics and Gynecology, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Xiaodan Fu, ; ; Qihui Wu, ; Hong Tan,
| | - Xiaodan Fu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, China
- *Correspondence: Xiaodan Fu, ; ; Qihui Wu, ; Hong Tan,
| |
Collapse
|
240
|
McAlarnen LA, Gupta P, Singh R, Pradeep S, Chaluvally-Raghavan P. Extracellular vesicle contents as non-invasive biomarkers in ovarian malignancies. Mol Ther Oncolytics 2022; 26:347-359. [PMID: 36090475 PMCID: PMC9420349 DOI: 10.1016/j.omto.2022.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Ovarian cancer most commonly presents at an advanced stage where survival is approximately 30% compared with >80% if diagnosed and treated before disease spreads. Diagnostic capabilities have progressed from surgical staging via laparotomy to image-guided biopsies and immunohistochemistry staining, along with advances in technology and medicine. Despite improvements in diagnostic capabilities, population-level screening for ovarian cancer is not recommended. Extracellular vesicles (EVs) are 40–150 nm structures formed when the cellular lipid bilayer invaginates. These structures function in cell signaling, immune responses, cancer progression, and establishing the tumor microenvironment. EVs are found in nearly every bodily fluid, including serum, plasma, ascites, urine, and effusion fluid, and contain molecular cargo from their cell of origin. This cargo can be analyzed to yield information about a possible malignancy. In this review we describe how the cargo of EVs has been studied as biomarkers in ovarian cancer. We bring together studies analyzing evidence for various cargos as ovarian cancer biomarkers. Then, we describe the role of EVs in modulation of the tumor microenvironment. This review also summarizes the therapeutic and translational potential of EVs for their optimal utilization as non-invasive biomarkers for novel treatments against cancer.
Collapse
|
241
|
Liu J, Chen C, Geng R, Shao F, Yang S, Zhong Z, Ni S, Bai J. Pyroptosis-related gene expression patterns and corresponding tumor microenvironment infiltration characterization in ovarian cancer. Comput Struct Biotechnol J 2022; 20:5440-5452. [PMID: 36249562 PMCID: PMC9535418 DOI: 10.1016/j.csbj.2022.09.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 09/06/2022] [Accepted: 09/26/2022] [Indexed: 11/22/2022] Open
Abstract
Pyroptosis, a form of inflammatory programmed cell death, is accompanied by inflammation and participate in the body's immune response. The expression of pyroptosis-related genes (PRGs) is associated with tumor prognosis in ovarian cancer (OC), but it is still unknown whether pyroptosis can affect tumor immune microenvironment (TME) of OC. Based on 30 PRGs, we comprehensively assessed the pyroptosis patterns by using PRGscore and correlated them with TME features in 474 OC patients. Finally, we identified three pyroptosis modification patterns and TME immune characteristics of these patterns were in response to three immune phenotypes (immune-desert, immune-inflamed, and immune-excluded phenotypes). PRGscore can predict patient survival, staging, grading, and immunotherapy efficacy. Low PRGscore was associated with better survival advantage and increased mutation burden. Low PRGscore patients showed significantly better therapeutic effects and clinical results in chemotherapy and immunotherapy. Besides, the capability of PRGscore in predicting prognosis and immunotherapy sensitivity was further verified in other three tumor cohorts. In conclusion, the comprehensive assessment of OC pyroptosis modifications can help enhancing our understanding of TME immune infiltration and provide better personalized treatment tactics for OC patients.
Collapse
|
242
|
Ma B, Zhao L, Zhang Y, Zhang F, Gao Q. Complete remission of ovarian clear cell carcinoma achieved after pseudoprogression during PD-1 inhibitor therapy. Immunotherapy 2022; 14:1205-1209. [PMID: 36043370 DOI: 10.2217/imt-2021-0328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: Ovarian clear cell carcinoma (OCCC), which is resistant to traditional treatment, has a poor prognosis. Immune checkpoint inhibitors (ICIs) have been emerged in the past decade and are now widely used in clinics. However, OCCC reportedly responds poorly to ICIs, and ICI monotherapy is rarely used for patients with OCCC. Methodology & Results: We report the case of a patient with refractory OCCC who received an ICI (nivolumab) monotherapy treatment and achieved a complete response despite the occurrence of pseudoprogression. Nivolumab was discontinued after 2 years, and the patient remained in complete remission more than a year after treatment withdrawal. Conclusion: This is the first report of complete remission being achieved in a case of refractory OCCC after pseudoprogression during nivolumab monotherapy.
Collapse
Affiliation(s)
- Baozhen Ma
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Lingdi Zhao
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Yong Zhang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Fang Zhang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Quanli Gao
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| |
Collapse
|
243
|
Russell S, Lim F, Peters PN, Wardell SE, Whitaker R, Chang CY, Previs RA, McDonnell DP. Development and Characterization of a Luciferase Labeled, Syngeneic Murine Model of Ovarian Cancer. Cancers (Basel) 2022; 14:cancers14174219. [PMID: 36077756 PMCID: PMC9454869 DOI: 10.3390/cancers14174219] [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: 07/16/2022] [Revised: 08/20/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022] Open
Abstract
Despite advances in surgery and targeted therapies, the prognosis for women with high-grade serous ovarian cancer remains poor. Moreover, unlike other cancers, immunotherapy has minimally impacted outcomes in patients with ovarian cancer. Progress in this regard has been hindered by the lack of relevant syngeneic ovarian cancer models to study tumor immunity and evaluate immunotherapies. To address this problem, we developed a luciferase labeled murine model of high-grade serous ovarian cancer, STOSE.M1 luc. We defined its growth characteristics, immune cell repertoire, and response to anti PD-L1 immunotherapy. As with human ovarian cancer, we demonstrated that this model is poorly sensitive to immune checkpoint modulators. By developing the STOSE.M1 luc model, it will be possible to probe the mechanisms underlying resistance to immunotherapies and evaluate new therapeutic approaches to treat ovarian cancer.
Collapse
Affiliation(s)
- Shonagh Russell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
- Correspondence: (S.R.); (D.P.M.); Tel.: +1-919-684-6035 (D.P.M.)
| | - Felicia Lim
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Pamela N. Peters
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Suzanne E. Wardell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Regina Whitaker
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Rebecca A. Previs
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Donald P. McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
- Correspondence: (S.R.); (D.P.M.); Tel.: +1-919-684-6035 (D.P.M.)
| |
Collapse
|
244
|
Rocconi RP, Stanbery L, Tang M, Madeira da Silva L, Walter A, Monk BJ, Herzog TJ, Coleman RL, Manning L, Wallraven G, Horvath S, Bognar E, Senzer N, Brun S, Nemunaitis J. ENTPD1/CD39 as a predictive marker of treatment response to gemogenovatucel-T as maintenance therapy in newly diagnosed ovarian cancer. COMMUNICATIONS MEDICINE 2022; 2:106. [PMID: 36051466 PMCID: PMC9424215 DOI: 10.1038/s43856-022-00163-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 07/26/2022] [Indexed: 11/09/2022] Open
Abstract
Background Broadened use of predictive molecular and phenotypic profiling amongst oncologists has facilitated optimal integration of targeted- and immuno-therapeutics into clinical care. However, the use of predictive immunomarkers in ovarian cancer (OC) has not consistently translated into clinical benefit. Vigil (gemogenovatucel-T) is a novel plasmid engineered autologous tumor cell immunotherapy designed to knock down the tumor suppressor cytokines, TGFβ1 and TGFβ2, augment local immune function via increased GMCSF expression and enhance presentation of clonal neoantigen epitopes. Methods: All patients enrolled in the VITAL trial (NCT02346747) of maintenance Vigil vs. placebo as front-line therapy with homologous recombination proficient (HRP) stage IIIB-IV newly diagnosed ovarian cancer underwent NanoString gene expression analysis. Tissue was obtained from surgically resected ovarian tumor tissue following surgical debulking. A statistical algorithm was used to analyze the NanoString gene expression data. Results Using the NanoString Statistical Algorithm (NSA), we identify high expression of ENTPD1/CD39 (which functions as the rate-limiting step in the production of the immune suppressor adenosine from ATP to ADP) as a presumptive predictor of response to Vigil versus placebo regardless of HRP status on the basis of relapse free survival (median not achieved vs 8.1 months, p = 0.00007) and overall survival (median not achieved vs 41.4 months, p = 0.013) extension. Conclusion NSA should be considered for application to investigational targeted therapies in order to identify populations most likely to benefit from treatment, in preparation for efficacy conclusive trials.
Collapse
Affiliation(s)
- Rodney P. Rocconi
- University of Alabama at Birmingham, Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Mobile, USA
| | | | - Min Tang
- Stat-Beyond Consulting, Irvine, USA
| | - Luciana Madeira da Silva
- Mitchell Cancer Institute, Division of Gynecologic Oncology, University of South Alabama, Mobile, USA
| | - Adam Walter
- Gradalis, Inc, Carrollton, USA
- Promedica, Toledo, USA
| | - Bradley J. Monk
- Arizona Oncology, (US Oncology Network), University of Arizona, Creighton University, Phoenix, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
245
|
Awada A, Ahmad S, McKenzie ND, Holloway RW. Immunotherapy in the Treatment of Platinum-Resistant Ovarian Cancer: Current Perspectives. Onco Targets Ther 2022; 15:853-866. [PMID: 35982728 PMCID: PMC9379118 DOI: 10.2147/ott.s335936] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/02/2022] [Indexed: 11/23/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic cancer. The gold standard therapeutic approach is a combination of surgery plus chemotherapy. Unfortunately, 80% of patients with EOC suffer recurrence within 2-years and the overall response rate for platinum-resistant epithelial ovarian cancer to cytotoxic chemotherapy or poly-(adenosine diphosphate)-ribose polymerase (PARP) inhibitor is modest. New therapies are needed to improve overall survival. The role of immunotherapy has been established in endometrial and cervical cancers, however its effective use in EOC has been limited due to the intrinsic genomics and micro-immune environment associated with EOC. Studies evaluating immunotherapy, largely immune checkpoint inhibitors (ICI), have shown limited activity, yet some patients benefit greatly. Thus, significant efforts must be devoted to finding new strategies for the use of immunotherapy/immunomodulatory drugs (IMiDs). Immunotherapy has a well-tolerated safety profile; however, cost-effectiveness can be an obstacle. The aim of this article is to review the most recent research into the use of IMiDs in patients with platinum-resistant epithelial ovarian cancer.
Collapse
Affiliation(s)
- Ahmad Awada
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL, 32804, USA
| | - Sarfraz Ahmad
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL, 32804, USA
| | - Nathalie D McKenzie
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL, 32804, USA
| | - Robert W Holloway
- AdventHealth Cancer Institute, Gynecologic Oncology Program, Orlando, FL, 32804, USA
| |
Collapse
|
246
|
Bonadio RC, Tarantino P, Testa L, Punie K, Pernas S, Barrios C, Curigliano G, Tolaney SM, Barroso-Sousa R. Management of patients with early-stage triple-negative breast cancer following pembrolizumab-based neoadjuvant therapy: What are the evidences? Cancer Treat Rev 2022; 110:102459. [PMID: 35998514 DOI: 10.1016/j.ctrv.2022.102459] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 08/11/2022] [Accepted: 08/14/2022] [Indexed: 11/28/2022]
Abstract
New therapy options have changed the treatment landscape of early-stage triple-negative breast cancer (TNBC) in recent years. Most patients are candidates for neoadjuvant chemotherapy, which helps to downstage the tumor and tailor adjuvant systemic therapy based on pathologic response. Capecitabine, pembrolizumab, and olaparib have been incorporated into the armamentarium of adjuvant treatment for selected patients. The KEYNOTE-522 trial, that demonstrated the benefit of pembrolizumab, given in addition to neoadjuvant chemotherapy and adjuvantly after surgery, represented a paradigm shift for early-stage TNBC treatment. Pembrolizumab was continued in the adjuvant setting irrespective of response to neoadjuvant therapy, and other adjuvant therapies were not administered in the trial. Many questions were then raised on the selection of adjuvant therapy regimens for patients with residual disease (RD). Prior to the routine use of immune-checkpoint inhibitors (ICI), the value of adjuvant capecitabine for patients with RD after neoadjuvant polychemotherapy was demonstrated. Given the poor prognosis of some patients with RD after neoadjuvant chemo-immunotherapy, while the survival advantage of adding capecitabine during the adjuvant phase of pembrolizumab is unknown, it does appear safe and can be considered. Regarding patients harboring germline BRCA mutations with RD after neoadjuvant ICI-containing regimens, the combination of olaparib with pembrolizumab can be an option based on existing safety data.
Collapse
Affiliation(s)
- Renata Colombo Bonadio
- Instituto D'Or de Pesquisa e Ensino (IDOR), São Paulo, Brazil; Instituto do Câncer do Estado de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | - Paolo Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; European Institute of Oncology, IRCCS
| | - Laura Testa
- Instituto D'Or de Pesquisa e Ensino (IDOR), São Paulo, Brazil; Instituto do Câncer do Estado de São Paulo, Universidade de São Paulo, São Paulo, Brazil
| | - Kevin Punie
- Department of General Medical Oncology and Multidisciplinary Breast Centre, Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | - Sonia Pernas
- Catalan Institute of Oncology; IDIBELL, L'Hospitalet de Llobregat (Barcelona), Spain
| | - Carlos Barrios
- Latin American Cooperative Oncology Group (LACOG), Grupo Oncoclínicas, Porto Alegre, Brazil
| | | | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Romualdo Barroso-Sousa
- Oncology Center, Hospital Sirio-Libanes, Brasília, Brazil; DASA Oncology, Brasília, Brazil.
| |
Collapse
|
247
|
Hollis RL, Meynert AM, Michie CO, Rye T, Churchman M, Hallas-Potts A, Croy I, McCluggage WG, Williams AR, Bartos C, Iida Y, Okamoto A, Dougherty B, Barrett JC, March R, Matakidou A, Roxburgh P, Semple CA, Harkin DP, Kennedy R, Herrington CS, Gourley C. Multiomic Characterization of High-Grade Serous Ovarian Carcinoma Enables High-Resolution Patient Stratification. Clin Cancer Res 2022; 28:3546-3556. [PMID: 35696721 PMCID: PMC9662902 DOI: 10.1158/1078-0432.ccr-22-0368] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/13/2022] [Accepted: 06/09/2022] [Indexed: 01/07/2023]
Abstract
PURPOSE High-grade serous ovarian carcinoma (HGSOC) is the most common ovarian cancer type; most patients experience disease recurrence that accumulates chemoresistance, leading to treatment failure. Genomic and transcriptomic features have been associated with differential outcome and treatment response. However, the relationship between events at the gene sequence, copy number, and gene-expression levels remains poorly defined. EXPERIMENTAL DESIGN We perform multiomic characterization of a large HGSOC cohort (n = 362) with detailed clinical annotation to interrogate the relationship between patient subgroups defined by specific molecular events. RESULTS BRCA2-mutant (BRCA2m) and EMSY-overexpressing cases demonstrated prolonged survival [multivariable hazard ratios (HR) 0.40 and 0.51] and significantly higher first- and second-line chemotherapy response rate. CCNE1-gained (CCNE1g) cases demonstrated underrepresentation of FIGO stage IV cases, with shorter survival but no significant difference in treatment response. We demonstrate marked overlap between the TCGA- and Tothill-derived subtypes. IMR/C2 cases displayed higher BRCA1/2m frequency (25.5%, 32.5%) and significantly greater immune cell infiltration, whereas PRO/C5 cases had the highest CCNE1g rate (23.9%, 22.2%) and were uniformly low in immune cell infiltration. The survival benefit for cases with aberrations in homologous recombination repair (HRR) genes was apparent across all transcriptomic subtypes (HR range, 0.48-0.68). There was significant co-occurrence of RB loss and HRR gene aberrations; RB loss was further associated with favorable survival within HRR-aberrant cases (multivariable HR, 0.50). CONCLUSIONS These data paint a high-resolution picture of the molecular landscape in HGSOC, better defining patients who may benefit most from specific molecular therapeutics and highlighting those for whom novel treatment strategies are needed to improve outcomes.
Collapse
Affiliation(s)
- Robert L. Hollis
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- Corresponding Author: Robb L. Hollis, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Crewe Road South, Edinburgh EH4 2XU, Scotland, UK. E-mail:
| | - Alison M. Meynert
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Caroline O. Michie
- Edinburgh Cancer Centre, Western General Hospital, NHS Lothian, Edinburgh, UK
| | - Tzyvia Rye
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Michael Churchman
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Amelia Hallas-Potts
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Ian Croy
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | | | | | - Clare Bartos
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Yasushi Iida
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
- The Jikei University School of Medicine, Tokyo, Japan
| | - Aikou Okamoto
- The Jikei University School of Medicine, Tokyo, Japan
| | - Brian Dougherty
- Translational Medicine, Oncology R&D, AstraZeneca, Waltham, Massachusetts
| | - J. Carl Barrett
- Translational Medicine, Oncology R&D, AstraZeneca, Waltham, Massachusetts
| | - Ruth March
- Precision Medicine and Biosamples, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Athena Matakidou
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Patricia Roxburgh
- Institute of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Belfast, UK
- Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Colin A. Semple
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - D. Paul Harkin
- Almac Diagnostics, Craigavon, UK
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - Richard Kennedy
- Almac Diagnostics, Craigavon, UK
- Centre for Cancer Research and Cell Biology, Queen's University Belfast, Belfast, UK
| | - C. Simon Herrington
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Scotland Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
248
|
Porter RL, Sun S, Flores MN, Berzolla E, You E, Phillips IE, KC N, Desai N, Tai EC, Szabolcs A, Lang ER, Pankaj A, Raabe MJ, Thapar V, Xu KH, Nieman LT, Rabe DC, Kolin DL, Stover EH, Pepin D, Stott SL, Deshpande V, Liu JF, Solovyov A, Matulonis UA, Greenbaum BD, Ting DT. Satellite repeat RNA expression in epithelial ovarian cancer associates with a tumor-immunosuppressive phenotype. J Clin Invest 2022; 132:e155931. [PMID: 35708912 PMCID: PMC9374379 DOI: 10.1172/jci155931] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Aberrant expression of viral-like repeat elements is a common feature of epithelial cancers, and the substantial diversity of repeat species provides a distinct view of the cancer transcriptome. Repeatome profiling across ovarian, pancreatic, and colorectal cell lines identifies distinct clustering independent of tissue origin that is seen with coding gene analysis. Deeper analysis of ovarian cancer cell lines demonstrated that human satellite II (HSATII) satellite repeat expression was highly associated with epithelial-mesenchymal transition (EMT) and anticorrelated with IFN-response genes indicative of a more aggressive phenotype. SATII expression - and its correlation with EMT and anticorrelation with IFN-response genes - was also found in ovarian cancer RNA-Seq data and was associated with significantly shorter survival in a second independent cohort of patients with ovarian cancer. Repeat RNAs were enriched in tumor-derived extracellular vesicles capable of stimulating monocyte-derived macrophages, demonstrating a mechanism that alters the tumor microenvironment with these viral-like sequences. Targeting of HSATII with antisense locked nucleic acids stimulated IFN response and induced MHC I expression in ovarian cancer cell lines, highlighting a potential strategy of modulating the repeatome to reestablish antitumor cell immune surveillance.
Collapse
Affiliation(s)
- Rebecca L. Porter
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Division of Gynecologic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Siyu Sun
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Micayla N. Flores
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Emily Berzolla
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Eunae You
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Ildiko E. Phillips
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Neelima KC
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Niyati Desai
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Eric C. Tai
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Annamaria Szabolcs
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Evan R. Lang
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Amaya Pankaj
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
- Department of Surgery, Massachusetts General Hospital
| | - Michael J. Raabe
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Vishal Thapar
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Katherine H. Xu
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Linda T. Nieman
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Daniel C. Rabe
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - David L. Kolin
- Department of Pathology, Brigham and Women’s Hospital, and
| | - Elizabeth H. Stover
- Division of Gynecologic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - David Pepin
- Department of Surgery, Massachusetts General Hospital
| | - Shannon L. Stott
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joyce F. Liu
- Division of Gynecologic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Alexander Solovyov
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ursula A. Matulonis
- Division of Gynecologic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Benjamin D. Greenbaum
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - David T. Ting
- Mass General Cancer Center, Harvard Medical School, Charlestown, Massachusetts, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
249
|
Ren Y, Song J, Li X, Luo N. Rationale and Clinical Research Progress on PD-1/PD-L1-Based Immunotherapy for Metastatic Triple-Negative Breast Cancer. Int J Mol Sci 2022; 23:ijms23168878. [PMID: 36012144 PMCID: PMC9408844 DOI: 10.3390/ijms23168878] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/26/2022] [Accepted: 08/04/2022] [Indexed: 01/23/2023] Open
Abstract
Metastatic triple-negative breast cancer (mTNBC), a highly aggressive and malignant tumor, currently lacks an effective treatment. There has been some progress in the treatment of mTNBC with programmed death receptor-1/programmed death ligand-1 (PD-1/PD-L1) immunotherapy in recent years. The combination of PD-1/PD-L1 inhibitors with other therapies is a noteworthy treatment strategy. Immunotherapy in combination with chemotherapy or small-molecule inhibitors still faces many challenges. Additionally, there are some new immunotherapy targets in development. We aimed to further evaluate the effectiveness and usefulness of immunotherapy for treating mTNBC and to propose new immunotherapy strategies. This review explains the rationale and results of existing clinical trials evaluating PD-1/PD-L1 inhibitors alone or in combination for the treatment of mTNBC. For patients with aggressive tumors and poor health, PD-1/PD-L1 inhibitors, either alone or in combination with other modalities, have proven to be effective. However, more research is needed to explore more effective immunotherapy regimens that will lead to new breakthroughs in the treatment of mTNBC.
Collapse
|
250
|
Abstract
Despite advances in surgery and chemotherapy, ovarian cancer remains one of the most lethal malignancies. Hence, the implementation of novel treatment approaches is required to improve the outcomes of the disease. Immunotherapy has been proven to be effective in many tumors and has already been incorporated into clinical practice. In this review, we describe key strategies in immunotherapy of ovarian cancer and summarize data from clinical studies assessing immunological prospects which could improve ovarian cancer treatment approaches in the future. The most notable current strategies include checkpoint blockade agents, the use of vaccines, adoptive cell transfer, as well as various combinations of these methods. While several of these options are promising, large controlled randomized studies are still needed to implement new immunotherapeutic options into clinical practice.
Collapse
|