351
|
Fucikova J, Coosemans A, Orsulic S, Cibula D, Vergote I, Galluzzi L, Spisek R. Immunological configuration of ovarian carcinoma: features and impact on disease outcome. J Immunother Cancer 2021; 9:jitc-2021-002873. [PMID: 34645669 PMCID: PMC8515436 DOI: 10.1136/jitc-2021-002873] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2021] [Indexed: 12/20/2022] Open
Abstract
Epithelial ovarian carcinoma (EOC) is a relatively rare malignancy but is the fifth-leading cause of cancer-related death in women, largely reflecting early, prediagnosis dissemination of malignant disease to the peritoneum. At odds with other neoplasms, EOC is virtually insensitive to immune checkpoint inhibitors, correlating with a tumor microenvironment that exhibits poor infiltration by immune cells and active immunosuppression. Here, we comparatively summarize the humoral and cellular features of primary and metastatic EOC, comparatively analyze their impact on disease outcome, and propose measures to alter them in support of treatment sensitivity and superior patient survival.
Collapse
Affiliation(s)
- Jitka Fucikova
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, Leuven Cancer Institute, KU Leuven, Leuven, Belgium
| | - Sandra Orsulic
- UCLA David Geffen School of Medicine and Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, USA
| | - David Cibula
- Gynecologic Oncology Center, Department of Obstetrics and Gynecology, 1st Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Ignace Vergote
- Division of Gynecological Oncology, Department of Obstetrics and Gynecology, University Hospital Leuven, Leuven, Belgium
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA
| | - Radek Spisek
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| |
Collapse
|
352
|
Bartoletti M, Musacchio L, Giannone G, Tuninetti V, Bergamini A, Scambia G, Lorusso D, Valabrega G, Mangili G, Puglisi F, Pignata S. Emerging molecular alterations leading to histology-specific targeted therapies in ovarian cancer beyond PARP inhibitors. Cancer Treat Rev 2021; 101:102298. [PMID: 34634660 DOI: 10.1016/j.ctrv.2021.102298] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 01/12/2023]
Abstract
After more than 30 years of a one-size-fits-all approach in the management of advanced ovarian cancer, in 2018 the SOLO1 trial results have introduced a new era of personalized medicine. A deeper knowledge of ovarian cancer biology and the development of new drugs targeting specific molecular pathways have led to biomarker-driven phase 3 trials with practice changing results. Thereafter, platinum-based combinations are no longer the only therapeutic options available in first line setting and poly-ADP ribose polymerase inhibitors maintenance therapy has become the mainstay in patients with tumor harboring a homologous recombination defect. However, most of the recent therapeutic breakthroughs regard high grade serous carcinoma, the most frequent ovarian cancer subtype, and only few improvements have occurred in the management of less common histotypes. Moving towards the next challenges, we aimed to investigate and review new potential molecular targets in ovarian cancer, according to histotype, starting from promising molecular drivers and matched drugs that have been investigated in early and late-stage clinical trials or conceptualized in preclinical studies.
Collapse
Affiliation(s)
- M Bartoletti
- Department of Medicine (DAME), University of Udine, Udine, Italy; Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano (PN), Italy
| | - L Musacchio
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - G Giannone
- Candiolo Cancer Institute, FPO- IRCCS, Candiolo (TO), Italy; Department of Oncology, University of Turin, Torino, Piemonte, Italy
| | - V Tuninetti
- Candiolo Cancer Institute, FPO- IRCCS, Candiolo (TO), Italy; Department of Oncology, University of Turin, Torino, Piemonte, Italy
| | - A Bergamini
- Department of Obstetrics and Gynecology, IRCCS, San Raffaele Hospital, Milan, Italy
| | - G Scambia
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Life Science and Public Health, Catholic University of Sacred Heart Largo Agostino Gemelli, Rome, Italy
| | - D Lorusso
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Department of Life Science and Public Health, Catholic University of Sacred Heart Largo Agostino Gemelli, Rome, Italy
| | - G Valabrega
- Candiolo Cancer Institute, FPO- IRCCS, Candiolo (TO), Italy; Department of Oncology, University of Turin, Torino, Piemonte, Italy
| | - G Mangili
- Department of Obstetrics and Gynecology, IRCCS, San Raffaele Hospital, Milan, Italy
| | - F Puglisi
- Department of Medicine (DAME), University of Udine, Udine, Italy; Unit of Medical Oncology and Cancer Prevention, Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, Aviano (PN), Italy
| | - S Pignata
- Department of Urology and Gynecology, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Naples, Italy.
| |
Collapse
|
353
|
Xie H, Wang W, Qi W, Jin W, Xia B. Targeting DNA Repair Response Promotes Immunotherapy in Ovarian Cancer: Rationale and Clinical Application. Front Immunol 2021; 12:661115. [PMID: 34712221 PMCID: PMC8546337 DOI: 10.3389/fimmu.2021.661115] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 09/22/2021] [Indexed: 01/21/2023] Open
Abstract
Immune checkpoint inhibitors (ICI) have emerged as a powerful oncologic treatment modality for patients with different solid tumors. Unfortunately, the efficacy of ICI monotherapy in ovarian cancer is limited, and combination therapy provides a new opportunity for immunotherapy in ovarian cancer. DNA damage repair (DDR) pathways play central roles in the maintenance of genomic integrity and promote the progression of cancer. A deficiency in DDR genes can cause different degrees of DNA damage that enhance local antigen release, resulting in systemic antitumor immune responses. Thus, the combination of DDR inhibitors with ICI represents an attractive therapeutic strategy with the potential to improve the clinical outcomes of patients with ovarian cancer. In this review, we provide an overview of the interconnectivity between DDR pathway deficiency and immune response, summarize available clinical trials on the combination therapy in ovarian cancer, and discuss the potential predictive biomarkers that can be utilized to guide the use of combination therapy.
Collapse
Affiliation(s)
- Hongyu Xie
- Clinical Research Center, Women’s Hospital School of Medicine Zhejiang University, Hangzhou, China
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wenjie Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Harbin Medical University, Harbin, China
| | - Wencai Qi
- Department of Gynecology Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of University of Science and Technology, Hefei, China
| | - Weilin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Bairong Xia
- Department of Gynecology Oncology, Division of Life Sciences and Medicine, The First Affiliated Hospital of University of Science and Technology, Hefei, China
| |
Collapse
|
354
|
Bronger H. Immunology and Immune Checkpoint Inhibition in Ovarian Cancer - Current Aspects. Geburtshilfe Frauenheilkd 2021; 81:1128-1144. [PMID: 34629492 PMCID: PMC8494520 DOI: 10.1055/a-1475-4335] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 04/05/2021] [Indexed: 12/17/2022] Open
Abstract
In the last decade immunotherapies such as immune checkpoint blockade (ICB) against the PD-1/PD-L1 system have revolutionised the treatment of numerous entities. To date, ovarian cancer has benefited very little from this success story. Possible causes include a rather low mutational burden compared to other tumour types, inadequate presentation of (neo-)antigens, and increased infiltration with immunosuppressive immune cells such as regulatory T cells and tumour-associated macrophages. In the clinical trials completed to date, the response rates to PD-1/PD-L1 checkpoint inhibitors have therefore been disappointingly low as well, although isolated long-term remissions have also been observed in ovarian cancer. The task now is to find suitable predictive biomarkers as well as to identify combination partners for ICB therapy that can increase the immunogenicity of ovarian cancer or overcome immunosuppressive resistance mechanisms. This paper provides an overview of the immune milieu in ovarian cancer, its impact on the effect of ICB, and summarises the clinical trial data available to date on ICB in ovarian cancer.
Collapse
Affiliation(s)
- Holger Bronger
- Klinik und Poliklinik für Frauenheilkunde, Klinikum rechts der Isar, Technische Universität München, München, Germany.,Deutsches Konsortium für Translationale Krebsforschung (DKTK), Partnerstandort München und Deutsches Krebsforschungszentrum (DKFZ), Heidelberg, Germany
| |
Collapse
|
355
|
Meng J, Peng J, Feng J, Maurer J, Li X, Li Y, Yao S, Chu R, Pan X, Li J, Zhang T, Liu L, Zhang Q, Yuan Z, Bu H, Song K, Kong B. Niraparib exhibits a synergistic anti-tumor effect with PD-L1 blockade by inducing an immune response in ovarian cancer. J Transl Med 2021; 19:415. [PMID: 34620163 PMCID: PMC8499579 DOI: 10.1186/s12967-021-03073-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/07/2021] [Indexed: 01/22/2023] Open
Abstract
Background Immune checkpoint blockades (ICBs) therapy showed limited efficacy in ovarian cancer management. Increasing evidence indicated that conventional and targeted therapies could affect tumor-associated immune responses and increase the effectiveness of immunotherapy. However, the effects of Niraparib, one of the poly (ADP) ribose polymerase (PARP) inhibitors, on the immune response remains unclear. Delineating the crosstalk between cytotoxic anticancer agents and cancer-associated immunity may lead to more efficient combinatorial strategies. Methods Programmed death ligand 1 (PD-L1) expression in human ovarian cancer cells after PARP inhibitors treatment was examined by western blotting (WB) and flow cytometry. The expression of poly ADP-ribose polymerase (PARP1), PD-L1, and CD8 in human ovarian cancer tissues was detected by immunohistochemistry(IHC). The effect of Niraparib and PD-L1 blockade in ovarian cancer progression was investigated in vivo. The changes of immune cells and cytokines in vitro and in vivo were detected by flow cytometry and enzyme-linked immunosorbent assay (ELISA). Changes of cGAS/STING signal pathway after Niraparib treatment were determined by WB, ELISA. Results Niraparib upregulated membrane PD-L1 and total PD-L1 expression in ovarian cancer cells and had a synergistic effect with PD-L1 blockade in vivo. In clinical patient samples, Niraparib augmented cytotoxic CD8+T cell proportion and function. In vivo and vitro, Niraparib can also increase the proportion of T cells and combined with PD-L1 blockade could further enhance the effect. Besides, Niraparib activated the cGAS-STING pathway, increasing the levels of cytokines such as CCL5 and CXCL10, which played a vital role in augmenting the infiltration and activation of cytotoxic T cells. Conclusions Niraparib could modulate the immune response via the activation of the cGAS/STING pathway, and combination with PD-L1 blockade could further enhance the effect. These results provide a sound theoretical basis for clinical treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-03073-0.
Collapse
Affiliation(s)
- Jinyu Meng
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Jin Peng
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Jie Feng
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China
| | - Jochen Maurer
- Department of Obstetrics and Gynecology, University Hospital Aachen (UKA), 52074, Aachen, Germany
| | - Xiao Li
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China
| | - Yan Li
- School of Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Shu Yao
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Ran Chu
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Xiyu Pan
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China
| | - Junting Li
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China
| | - Ting Zhang
- Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China.,Department of Obstetrics and Gynecology, The Sixth Hospital of Beijing, Beijing, China
| | - Lu Liu
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Qing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China
| | - Zeng Yuan
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China
| | - Hualei Bu
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China
| | - Kun Song
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China. .,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China.
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, Shandong, China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Jinan, Shandong, China
| |
Collapse
|
356
|
Gjorgoska M, Rižner TL. Estrogens and the Schrödinger's Cat in the Ovarian Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13195011. [PMID: 34638494 PMCID: PMC8508344 DOI: 10.3390/cancers13195011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/29/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Ovarian cancer is a complex pathology for which we require effective screening and therapeutical strategies. Apart from the cancer cell portion, there exist plastic immune and non-immune cell populations, jointly constituting the context-adaptive tumor microenvironment, which is pivotal in tumorigenesis. Estrogens might be synthesized in the ovarian tumor tissue and actively contribute to the shaping of an immunosuppressive microenvironment. Current immune therapies have limited effectiveness as a multitude of factors influence the outcome. A thorough understanding of the ovarian cancer biology is crucial in the efforts to reestablish homeostasis. Abstract Ovarian cancer is a heterogeneous disease affecting the aging ovary, in concert with a complex network of cells and signals, together representing the ovarian tumor microenvironment. As in the “Schrödinger’s cat” thought experiment, the context-dependent constituents of the—by the time of diagnosis—well-established tumor microenvironment may display a tumor-protective and -destructive role. Systemic and locally synthesized estrogens contribute to the formation of a pro-tumoral microenvironment that enables the sustained tumor growth, invasion and metastasis. Here we focus on the estrogen biosynthetic and metabolic pathways in ovarian cancer and elaborate their actions on phenotypically plastic, estrogen-responsive, aging immune cells of the tumor microenvironment, altogether highlighting the multicomponent-connectedness and complexity of cancer, and contributing to a broader understanding of the ovarian cancer biology.
Collapse
|
357
|
Pisano S, Lenna S, Healey GD, Izardi F, Meeks L, Jimenez YS, Velazquez OS, Gonzalez D, Conlan RS, Corradetti B. Assessment of the immune landscapes of advanced ovarian cancer in an optimized in vivo model. Clin Transl Med 2021; 11:e551. [PMID: 34709744 PMCID: PMC8506632 DOI: 10.1002/ctm2.551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Ovarian cancer (OC) is typically diagnosed late, associated with high rates of metastasis and the onset of ascites during late stage disease. Understanding the tumor microenvironment and how it impacts the efficacy of current treatments, including immunotherapies, needs effective in vivo models that are fully characterized. In particular, understanding the role of immune cells within the tumor and ascitic fluid could provide important insights into why OC fails to respond to immunotherapies. In this work, we comprehensively described the immune cell infiltrates in tumor nodules and the ascitic fluid within an optimized preclinical model of advanced ovarian cancer. METHODS Green Fluorescent Protein (GFP)-ID8 OC cells were injected intraperitoneally into C57BL/6 mice and the development of advanced stage OC monitored. Nine weeks after tumor injection, mice were sacrificed and tumor nodules analyzed to identify specific immune infiltrates by immunohistochemistry. Ascites, developed in tumor bearing mice over a 10-week period, was characterized by mass cytometry (CyTOF) to qualitatively and quantitatively assess the distribution of the immune cell subsets, and their relationship to ascites from ovarian cancer patients. RESULTS Tumor nodules in the peritoneal cavity proved to be enriched in T cells, antigen presenting cells and macrophages, demonstrating an active immune environment and cell-mediated immunity. Assessment of the immune landscape in the ascites showed the predominance of CD8+ , CD4+ , B- , and memory T cells, among others, and the coexistance of different immune cell types within the same tumor microenvironment. CONCLUSIONS We performed, for the first time, a multiparametric analysis of the ascitic fluid and specifically identify immune cell populations in the peritoneal cavity of mice with advanced OC. Data obtained highlights the impact of CytOF as a diagnostic tool for this malignancy, with the opportunity to concomitantly identify novel targets, and define personalized therapeutic options.
Collapse
Affiliation(s)
- Simone Pisano
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
- Center for NanoHealthSwansea University Medical SchoolSwanseaUK
| | - Stefania Lenna
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
| | | | | | - Lucille Meeks
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
| | - Yajaira S. Jimenez
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
- Texas A&M Health Science CenterCollege of MedicineBryanTexas
| | - Oscar S Velazquez
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
| | | | - Robert Steven Conlan
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
- Center for NanoHealthSwansea University Medical SchoolSwanseaUK
| | - Bruna Corradetti
- Department of NanomedicineHouston Methodist Research InstituteHoustonTexas
- Center for NanoHealthSwansea University Medical SchoolSwanseaUK
- Texas A&M Health Science CenterCollege of MedicineBryanTexas
| |
Collapse
|
358
|
Glickman A, Paredes P, Carreras-Diéguez N, Niñerola-Baizán A, Gaba L, Pahisa J, Fusté P, Del Pino M, Díaz-Feijóo B, González-Bosquet E, Agustí N, Sánchez-Izquierdo N, Fuster D, Perissinotti A, Romero I, Fernández-Galán E, Carrasco JL, Gil-Ibáñez B, Torné A. Evaluation of patients with advanced epithelial ovarian cancer before primary treatment: correlation between tumour burden assessed by [ 18F]FDG PET/CT volumetric parameters and tumour markers HE4 and CA125. Eur Radiol 2021; 32:2200-2208. [PMID: 34586465 DOI: 10.1007/s00330-021-08305-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 08/11/2021] [Accepted: 08/25/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVES Accurate assessment of disease extent is required to select the best primary treatment for advanced epithelial ovarian cancer patients. Estimation of tumour burden is challenging and it is usually performed by means of a surgical procedure. Imaging techniques and tumour markers can help to estimate tumour burden non-invasively. 2-[18F]FDG PET/CT allows the evaluation of the whole-body disease. This study aimed to correlate HE4 and CA125 serum concentrations with tumour burden evaluated by volumetric 2-[18F]FDG PET/CT parameters in advanced high-grade epithelial ovarian cancer. METHODS We included 66 patients who underwent 2-[18F]FDG PET/CT and serum tumour markers determination before primary treatment. Volumes of interest were delimited in every pathological uptake. Whole-body metabolic tumour volume (wb_MTV) and total lesion glycolysis (wb_TLG) were calculated summing up every VOI's MTV value. SUVmax thresholds were set at 40% (MTV40 and TLG40) and 50% (MTV50 and TLG50). In addition, four VOI subgroups were defined: peritoneal carcinomatosis, retroperitoneal nodes, supradiaphragmatic nodes, and distant metastases. MTV and TLG were calculated for each group by adding up the corresponding MTV values. TLG was calculated likewise. RESULTS wb_MTV and wb_TLG were found to be significantly correlated with serum CA125 and HE4 concentrations. The strongest correlation was observed between HE4 and wb_MTV40 (r = 0.62, p < 0.001). Pearson's correlation coefficients between peritoneal carcinomatosis MTV40 and tumour markers were 0.61 (p < 0.0001) and 0.29 (p = 0.02) for HE4 and CA125 respectively. None of these tumour markers showed a positive correlation with tumour load outside the abdominal cavity assessed by volumetric parameters. CONCLUSION HE4 performs better than CA125 to predict metabolic tumour burden in high-grade epithelial ovarian cancer before primary treatment. 2-[18F]FDG PET/CT volumetric parameters arise as feasible tools for the objective assessment of tumour load and its anatomical distribution. These results support the usefulness of HE4 and PET/CT to improve the stratification of these patients in clinical practice. KEY POINTS • In patients with high-grade advanced ovarian epithelial carcinoma, both CA125 and HE4 correlate to whole-body tumour burden assessed by PET/CT before primary treatment. • HE4 estimates peritoneal disease much better than CA125. • PET/CT volumetric parameters arise as feasible tools for the objective assessment of tumour load and its anatomical distribution.
Collapse
Affiliation(s)
- Ariel Glickman
- Gynaecologic Oncology Unit, Institut Clínic de GinecologiaObstetrícia i Neonatologia, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Pilar Paredes
- Department of Nuclear Medicine, Hospital Clínic Barcelona, Barcelona, Spain.
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.
- Faculty of Medicine - University of Barcelona, Barcelona, Spain.
| | - Núria Carreras-Diéguez
- Gynaecologic Oncology Unit, Institut Clínic de GinecologiaObstetrícia i Neonatologia, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Aida Niñerola-Baizán
- Department of Nuclear Medicine, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Faculty of Medicine - University of Barcelona, Barcelona, Spain
- Biomedical Research Networking Center of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Lydia Gaba
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Department of Medical Oncology, Hospital Clínic Barcelona, Barcelona, Spain
| | - Jaume Pahisa
- Gynaecologic Oncology Unit, Institut Clínic de GinecologiaObstetrícia i Neonatologia, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Pere Fusté
- Gynaecologic Oncology Unit, Institut Clínic de GinecologiaObstetrícia i Neonatologia, Hospital Clínic de Barcelona, Barcelona, Spain
- Faculty of Medicine - University of Barcelona, Barcelona, Spain
| | - Marta Del Pino
- Gynaecologic Oncology Unit, Institut Clínic de GinecologiaObstetrícia i Neonatologia, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Faculty of Medicine - University of Barcelona, Barcelona, Spain
| | - Berta Díaz-Feijóo
- Gynaecologic Oncology Unit, Institut Clínic de GinecologiaObstetrícia i Neonatologia, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Faculty of Medicine - University of Barcelona, Barcelona, Spain
| | - Eduardo González-Bosquet
- Gynaecologic Oncology Unit, Institut Clínic de GinecologiaObstetrícia i Neonatologia, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Núria Agustí
- Gynaecologic Oncology Unit, Institut Clínic de GinecologiaObstetrícia i Neonatologia, Hospital Clínic de Barcelona, Barcelona, Spain
| | | | - David Fuster
- Department of Nuclear Medicine, Hospital Clínic Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Faculty of Medicine - University of Barcelona, Barcelona, Spain
| | - Andrés Perissinotti
- Department of Nuclear Medicine, Hospital Clínic Barcelona, Barcelona, Spain
- Biomedical Research Networking Center of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Barcelona, Spain
| | - Inmaculada Romero
- Department of Nuclear Medicine, Hospital Clínic Barcelona, Barcelona, Spain
| | - Esther Fernández-Galán
- Department of Biochemistry and Molecular Genetics, Biomedical Diagnostic Centre, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Josep Lluís Carrasco
- Biostatistics, Department of Basic Clinical Practice, University of Barcelona, Barcelona, Spain
| | - Blanca Gil-Ibáñez
- Gynecologic Oncology and Minimally Invasive Gynecologic Surgery Unit, Department of Obstetrics and Gynecology, 12 de Octubre University Hospital, Madrid, Spain
| | - Aureli Torné
- Gynaecologic Oncology Unit, Institut Clínic de GinecologiaObstetrícia i Neonatologia, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Faculty of Medicine - University of Barcelona, Barcelona, Spain
| |
Collapse
|
359
|
Esmail S, Danter WR. Stem-cell based, machine learning approach for optimizing natural killer cell-based personalized immunotherapy for high-grade ovarian cancer. FEBS J 2021; 289:985-998. [PMID: 34582617 DOI: 10.1111/febs.16214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/01/2021] [Accepted: 09/27/2021] [Indexed: 12/18/2022]
Abstract
Advanced high-grade serous ovarian cancer continues to be a therapeutic challenge for those affected using the current therapeutic interventions. There is an increasing interest in personalized cancer immunotherapy using activated natural killer (NK) cells. NK cells account for approximately 15% of circulating white blood cells. They are also an important element of the tumor microenvironment (TME) and the body's immune response to cancers. In the present study, DeepNEU-C2Rx, a machine learning platform, was first used to create validated artificially induced pluripotent stem cell simulations. These simulations were then used to generate wild-type artificially induced NK cells (aiNK-WT) and TME simulations. Once validated, the aiNK-WT simulations were exposed to artificially induced high-grade serous ovarian cancer represented by aiOVCAR3. Cytolytic activity of aiNK was evaluated in presence and absence of aiOVCAR3 and data were compared with the literature for validation. The TME simulations suggested 26 factors that could be evaluated based on their ability to enhance aiNK-WT cytolytic activity in the presence of aiOVCAR3. The addition of programmed cell death-1 inhibitor leads to significant reinvigoration of aiNK cytolytic activity. The combination of programmed cell death-1 and glycogen synthase kinase 3 inhibitors showed further improvement. Further addition of ascitic fluid factor inhibitors leads to optimal aiNK activation. Our data showed that NK cell simulations could be used not only to pinpoint novel immunotherapeutic targets to reinvigorate the activity of NK cells against cancers, but also to predict the outcome of targeting tumors with specific genetic expression and mutation profiles.
Collapse
|
360
|
Liao JB, Gwin WR, Urban RR, Hitchcock-Bernhardt KM, Coveler AL, Higgins DM, Childs JS, Shakalia HN, Swensen RE, Stanton SE, Tinker AV, Wahl TA, Ancheta RG, McGonigle KF, Dai JY, Disis ML, Goff BA. Pembrolizumab with low-dose carboplatin for recurrent platinum-resistant ovarian, fallopian tube, and primary peritoneal cancer: survival and immune correlates. J Immunother Cancer 2021; 9:jitc-2021-003122. [PMID: 34531249 PMCID: PMC8449961 DOI: 10.1136/jitc-2021-003122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2021] [Indexed: 11/10/2022] Open
Abstract
Background Anti-programmed death 1 (PD1)/programmed cell death ligand 1 (PD-L1) therapies have shown modest activity as monotherapy in recurrent ovarian cancer. Platinum chemotherapies induce T-cell proliferation and enhance tumor recognition. We assessed activity and safety of pembrolizumab with carboplatin in recurrent platinum-resistant ovarian cancer. Patients and methods This phase I/II, single-arm clinical trial studied concurrent carboplatin and pembrolizumab in recurrent platinum-resistant ovarian, fallopian tube, and primary peritoneal cancer. Primary platinum refractory patients were excluded. Patients were treated after progression on subsequent non-platinum systemic therapy after becoming platinum resistant or refractory. Pembrolizumab 200 mg was given on day 1 and carboplatin area under the curve 2 on days 8 and 15 of a 3-week cycle until progression. Imaging was assessed by blinded independent review. PD-L1 expression was assessed by immunohistochemistry. Flow cytometry on peripheral blood mononuclear cells was performed for CD3, CD4, CD8, PD1, CTLA4 and Ki67. Results The most common treatment-related adverse events were lymphopenia (18%) and anemia (9%) with most being grade 1 or 2 (93%). Of 29 patients treated, 23 patients were evaluable for best objective response: 10.3% (95% CI 2.2 to 27.4) had partial response (PR), 51.7% (95% CI 32.5 to 70.6) had stable disease (SD). 56.5% of patients had decreases in target lesions from baseline. All PD-L1-positive patients achieved PR (3/7, 42.8%) or SD (4/7, 57.2%). Median progression-free survival was 4.63 months (95% CI 4.3 to 4.96). Median OS was 11.3 months (95% CI 6.094 to 16.506). Peripheral CD8+PD1+Ki67+ T cells expanded after 3 (p=0.0015) and 5 (p=0.0023) cycles. CTLA4+PD1+CD8+ T cells decreased through the course of treatment up to the 12th cycle (p=0.004). When stratified by ratio of peripheral CD8+PD1+Ki67+ T cells to tumor burden at baseline, patients with a ratio ≥0.0375 who had a significantly longer median OS of 18.37 months compared with those with a ratio <0.0375 who had a median OS of 8.72 months (p=0.0099). No survival advantage was seen with stratification by tumor burden alone (p=0.24) or by CD8+PD1+Ki67+ T cells alone (p=0.53). Conclusions Pembrolizumab with carboplatin was well-tolerated and active in recurrent platinum-resistant ovarian cancer. A ratio of peripheral T-cell exhaustion to radiographic tumor burden may identify patients more likely to benefit from this chemoimmunotherapy. Trial registration number NCT03029598.
Collapse
Affiliation(s)
- John B Liao
- University of Washington School of Medicine, Seattle, Washington, USA
| | - William R Gwin
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Renata R Urban
- University of Washington School of Medicine, Seattle, Washington, USA
| | | | - Andrew L Coveler
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Doreen M Higgins
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Jennifer S Childs
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Hania N Shakalia
- University of Washington School of Medicine, Seattle, Washington, USA
| | | | | | - Anna V Tinker
- BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Tanya A Wahl
- Swedish Medical Center, Seattle, Washington, USA
| | | | | | - James Y Dai
- Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Mary L Disis
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Barbara A Goff
- University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
361
|
Cao K, Zhang G, Zhang X, Yang M, Wang Y, He M, Lu J, Liu H. Stromal infiltrating mast cells identify immunoevasive subtype high-grade serous ovarian cancer with poor prognosis and inferior immunotherapeutic response. Oncoimmunology 2021; 10:1969075. [PMID: 34527431 PMCID: PMC8437532 DOI: 10.1080/2162402x.2021.1969075] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Tumor infiltrating mast cells (TIMs), with pro- or anti-tumorigenic role in different types of malignancies, have been implicated in resistance to anti-PD1 therapy. Here, we aimed to identify the relevance of TIMs with the prognosis, immune contexture, and immunotherapy in high-grade serous ovarian cancer (HGSOC). Tissue microarrays containing 197 HGSOC patients were assessed by immunohistochemistry (IHC) for detecting the expression of mast cell tryptase and other immune markers. Kaplan-Meier curve, log-rank test, and Cox regression model were applied to perform survival analysis. Single-cell RNA-seq analysis and flow cytometric analysis were selected to characterize TIMs. Furthermore, short-term HGSOC organoids were employed to validate the effect of TIMs on anti-PD1 therapy. Abundance of stromal TIMs (sTIMs) predicted dismal prognosis and linked to immunoevasive subtype of HGSOC, characterized by increased infiltration of pro-tumor cells (Treg cells, M2-polarized macrophages, and neutrophils) and impaired anti-tumor immune functions. Intensive inter-cell interactions between TIMs and other immune cells were identified, suggesting potential cross-talks to foster an immunosuppressive microenvironment. Organoids derived from sTIMs-low patients were associated with increased response to anti-PD-1 treatment other than the presence of high sTIMs infiltration. A nomogram, constructed by combining FIGO stage, sTIMs, and PD-L1, with an area under the curve (AUC) for predicting 5-year overall survival of 0.771 was better than that of FIGO staging system of 0.619. sTIMs/PD-L1-based classifier has potential clinical application in predicting prognosis of patients with HGSOC. sTIMs-high tumors correlate with immunosuppressive tumor microenvironment (TME) and possess potential insensitivity to immunotherapy.
Collapse
Affiliation(s)
- Kankan Cao
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Guodong Zhang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Xiangyun Zhang
- Department of Gynecology, Suzhou Municipal Hospital, Suzhou, Jiangsu Province, China
| | - Moran Yang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yiying Wang
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Mengdi He
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jiaqi Lu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Haiou Liu
- Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| |
Collapse
|
362
|
Wood GE, Ledermann JA. Adjuvant and post-surgical treatment in high-grade epithelial ovarian cancer. Best Pract Res Clin Obstet Gynaecol 2021; 78:64-73. [PMID: 34607745 DOI: 10.1016/j.bpobgyn.2021.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/06/2021] [Indexed: 12/21/2022]
Abstract
Cytoreductive surgery is the mainstay of treatment for high-grade epithelial ovarian cancer. Although for early stage disease outcomes following surgery alone are good, the risk of recurrence necessitates adjuvant chemotherapy for the majority of patients. Post-operative chemotherapy in advanced-stage disease, or neoadjuvant chemotherapy followed by surgery has improved progression-free survival (PFS) and overall survival (OS). However, despite the use of chemotherapy, the rate of recurrence remains high. In recent years, there has been considerable increase in knowledge regarding the biology of ovarian cancer, which has led to a journey of drug discovery, facilitating the use of novel targeted agents such as VEGF inhibitors and, more recently, PARP inhibitors in the first-line treatment of ovarian cancer. Here, we outline the current evidence-based guidance for systemic therapies in ovarian cancer and highlight the ongoing research to improve patient outcome.
Collapse
Affiliation(s)
- Georgina E Wood
- Department of Oncology, UCL Hospitals, 250 Euston Road, London, NW1 2BU, UK
| | - Jonathan A Ledermann
- Department of Oncology, UCL Hospitals, 250 Euston Road, London, NW1 2BU, UK; UCL Cancer Institute, University College London, UK.
| |
Collapse
|
363
|
Chirshev E, Suzuki T, Wang H, Nguyen A, Hojo N, Sanderman L, Mirshahidi S, Ioffe YJ, Unternaehrer JJ. Let-7i Reduces Aggressive Phenotype and Induces BRCAness in Ovarian Cancer Cells. Cancers (Basel) 2021; 13:cancers13184617. [PMID: 34572843 PMCID: PMC8468164 DOI: 10.3390/cancers13184617] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/05/2021] [Accepted: 09/09/2021] [Indexed: 12/24/2022] Open
Abstract
High-grade serous carcinoma of the ovary is a deadly gynecological cancer with poor long-term survival. Dysregulation of microRNAs has been shown to contribute to the formation of cancer stem cells (CSCs), an important part of oncogenesis and tumor progression. The let-7 family of microRNAs has previously been shown to regulate stemness and has tumor suppressive actions in a variety of cancers, including ovarian. Here, we demonstrate tumor suppressor actions of let-7i: repression of cancer cell stemness, inhibition of migration and invasion, and promotion of apoptosis, features important for cancer progression, relapse, and metastasis. Let-7i over-expression results in increased sensitivity to the PARP inhibitor olaparib in samples without BRCA mutations, consistent with induction of BRCAness phenotype. We also show that let-7i inhibits the expression of several factors involved in the homologous recombination repair (HRR) pathway, providing potential mechanisms by which the BRCAness phenotype could be induced. These actions of let-7i add to the rationale for use of this miRNA as a treatment for ovarian cancer patients, including those without mutations in the HRR pathway.
Collapse
Affiliation(s)
- Evgeny Chirshev
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
- Department of Pathology and Human Anatomy, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Tise Suzuki
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
| | - Hanmin Wang
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
| | - Anthony Nguyen
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
| | - Nozomi Hojo
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
| | - Linda Sanderman
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
- Biology Department, California State University San Bernardino, San Bernardino, CA 92407, USA
| | - Saied Mirshahidi
- Biospecimen Laboratory, Loma Linda University Cancer Center, Department of Basic Sciences, Division of Microbiology & Molecular Genetics, Loma Linda University, Loma Linda, CA 92354, USA;
| | - Yevgeniya J. Ioffe
- Department of Gynecology and Obstetrics, Division of Gynecologic Oncology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA;
| | - Juli J. Unternaehrer
- Department of Basic Sciences, Division of Biochemistry, Loma Linda University, Loma Linda, CA 92354, USA; (E.C.); (T.S.); (H.W.); (A.N.); (N.H.); (L.S.)
- Department of Gynecology and Obstetrics, Loma Linda University, Loma Linda, CA 92354, USA
- Center for Health Disparities and Molecular Medicine, Loma Linda University, Loma Linda, CA 92354, USA
- Correspondence:
| |
Collapse
|
364
|
Rochefort P, Desseigne F, Bonadona V, Dussart S, Coutzac C, Sarabi M, la Fouchardiere CD. Immune checkpoint inhibitor sensitivity of DNA repair deficient tumors. Immunotherapy 2021; 13:1205-1213. [PMID: 34494466 DOI: 10.2217/imt-2021-0024] [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: 11/21/2022] Open
Abstract
Faithful DNA replication is necessary to maintain genome stability and implicates a complex network with several pathways depending on DNA damage type: homologous repair, nonhomologous end joining, base excision repair, nucleotide excision repair and mismatch repair. Alteration in components of DNA repair machinery led to DNA damage accumulation and potentially carcinogenesis. Preclinical data suggest sensitivity to immune checkpoint inhibitors in tumors with DNA repair deficiency. Here, we review clinical studies that explored the use of immune checkpoint inhibitor in patient harboring tumor with DNA repair deficiency.
Collapse
Affiliation(s)
- Pauline Rochefort
- Department of Medical Oncology, Centre Léon Bérard, 69008, Lyon, France
| | | | - Valérie Bonadona
- Unit of Genetic Epidemiology & Prevention, Centre Léon Bérard, 69008, Lyon, France
| | - Sophie Dussart
- Unit of Genetic Epidemiology & Prevention, Centre Léon Bérard, 69008, Lyon, France
| | - Clélia Coutzac
- Department of Medical Oncology, Centre Léon Bérard, 69008, Lyon, France
| | - Matthieu Sarabi
- Department of Medical Oncology, Centre Léon Bérard, 69008, Lyon, France
| | | |
Collapse
|
365
|
Lu Y, Jin J, Du Q, Hu M, Wei Y, Wang M, Li H, Li Q. Multi-Omics Analysis of the Anti-tumor Synergistic Mechanism and Potential Application of Immune Checkpoint Blockade Combined With Lenvatinib. Front Cell Dev Biol 2021; 9:730240. [PMID: 34568339 PMCID: PMC8458708 DOI: 10.3389/fcell.2021.730240] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 08/16/2021] [Indexed: 12/12/2022] Open
Abstract
The combination of immune-checkpoint blockade (ICB) and lenvatinib has demonstrated robust clinical effects that are superior to those of monotherapies, but the synergistic anti-tumor mechanisms remain unclear. Exploring the synergistic molecular mechanisms and early identifying potential application have key importance for clinical therapeutics. We firstly systematically reviewed published data of ICB in combination with lenvatinib for the treatment of cancer by meta-analysis. A subsequent bioinformatics analysis explored the mechanism of combined ICB and lenvatinib therapy in 33 cancer types. Transcriptomic analysis was conducted by RNA-seq, and genomic analysis was performed on gene mutations and copy-number alteration data. Tumor-related pathways and tumor immune micro-environment (TIME) were also investigated. The meta-analysis showed a 38.0% objective response rate (ORR) and 79% disease control rate (DCR) for ICB combined with lenvatinib. Multi-omics analysis revealed that ICB and lenvatinib target genes were highly expressed and showed driving alterations in six specific malignancies. Pathway-enrichment analysis found target genes were implicated in tumor development, angiogenesis, and immunoregulatory associated pathways. This study verified the potential synergistic mechanisms of ICB combined with lenvatinib at transcriptomics, genomics, protein, and cellular levels and recognized nine tumor types had ≥ 2 positive treatment-related molecular characteristics, which might benefit particularly from this combined strategy. The findings would help to provide clinical insights and theoretical basis for optimizing of targeted therapy-immunotherapy combinations, and for guiding individualized precision-medicine approaches for cancer treatment.
Collapse
Affiliation(s)
- Yuting Lu
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jiangtao Jin
- Department of Intervention Therapy, Zezhou People’s Hospital, Jincheng, China
| | - Qi Du
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Min Hu
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, China
| | - Yuhan Wei
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Miao Wang
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hongzhong Li
- Chongqing Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qin Li
- Department of Oncology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
366
|
Maiorano BA, Maiorano MFP, Lorusso D, Maiello E. Ovarian Cancer in the Era of Immune Checkpoint Inhibitors: State of the Art and Future Perspectives. Cancers (Basel) 2021; 13:4438. [PMID: 34503248 PMCID: PMC8430975 DOI: 10.3390/cancers13174438] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/31/2021] [Accepted: 09/01/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Ovarian cancer (OC) represents the eighth most common cancer and the fifth leading cause of cancer-related deaths among the female population. In an advanced setting, chemotherapy represents the first-choice treatment, despite a high recurrence rate. In the last ten years, immunotherapy based on immune checkpoint inhibitors (ICIs) has profoundly modified the therapeutic scenario of many solid tumors. We sought to summarize the main findings regarding the clinical use of ICIs in OC. METHODS We searched PubMed, Embase, and Cochrane Databases, and conference abstracts from international congresses (such as ASCO, ESMO, SGO) for clinical trials, focusing on ICIs both as monotherapy and as combinations in the advanced OC. RESULTS 20 studies were identified, of which 16 were phase I or II and 4 phase III trials. These trials used ICIs targeting PD1 (nivolumab, pembrolizumab), PD-L1 (avelumab, aterolizumab, durvalumab), and CTLA4 (ipilimumab, tremelimumab). There was no reported improvement in survival, and some trials were terminated early due to toxicity or lack of response. Combining ICIs with chemotherapy, anti-VEGF therapy, or PARP inhibitors improved response rates and survival in spite of a worse safety profile. CONCLUSIONS The identification of biomarkers with a predictive role for ICIs' efficacy is mandatory. Moreover, genomic and immune profiling of OC might lead to better treatment options and facilitate the design of tailored trials.
Collapse
Affiliation(s)
- Brigida Anna Maiorano
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 71013 San Giovanni Rotondo, Italy;
- Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | - Mauro Francesco Pio Maiorano
- Division of Obstetrics and Gynecology, Biomedical and Human Oncological Science, University of Bari “Aldo Moro”, 70121 Bari, Italy;
| | - Domenica Lorusso
- Gynecologic Oncology Unit, Catholic University of the Sacred Heart, 00168 Rome, Italy;
- Scientific Directorate, Fondazione Policlinico “A.Gemelli” IRCCS, 00168 Rome, Italy
| | - Evaristo Maiello
- Oncology Unit, Foundation Casa Sollievo della Sofferenza IRCCS, 71013 San Giovanni Rotondo, Italy;
| |
Collapse
|
367
|
Marchetti C, De Felice F, Romito A, Iacobelli V, Sassu CM, Corrado G, Ricci C, Scambia G, Fagotti A. Chemotherapy resistance in epithelial ovarian cancer: Mechanisms and emerging treatments. Semin Cancer Biol 2021; 77:144-166. [PMID: 34464704 DOI: 10.1016/j.semcancer.2021.08.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 12/12/2022]
Abstract
Ovarian cancer (OC) remains a fatal malignancy because most patients experience recurrent disease, which is resistant to chemotherapy. The outcomes for patients with platinum-resistant OC are poor, response rates to further chemotherapy are low and median survival is lower than 12 months. The complexity of platinum-resistant OC, which comprises a heterogeneous spectrum of diseases, is indeed far from being completely understood. Therefore, comprehending tumors' biological behaviour to identify reliable biomarkers, which may predict responses to therapies, is a demanding challenge to improve OC management. In the age of precision medicine, efforts to overcome platinum resistance in OC represent a dynamic and vast field in which innovative drugs and clinical trials rapidly develop. This review will present the exceptional biochemical environment implicated in OC and highlights mechanisms of chemoresistance. Furthermore, innovative molecules and new therapeutic opportunities are presented, along with currently available therapies and ongoing clinical trials.
Collapse
Affiliation(s)
- Claudia Marchetti
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy.
| | - Francesca De Felice
- Division of Radiotherapy and Oncology, Policlinico Umberto I, Roma, Italy; Università La Sapienza, Roma, Italy
| | - Alessia Romito
- Gynecology and Breast Care Center, Mater Olbia Hospital, Olbia, Italy
| | - Valentina Iacobelli
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Department Woman and Child Health Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Carolina Maria Sassu
- Department of Maternal and Child Health and Urological Sciences, "Sapienza" University of Rome, Polyclinic Umberto I, Rome, Italy
| | - Giacomo Corrado
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Caterina Ricci
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy
| | - Giovanni Scambia
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Department Woman and Child Health Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Anna Fagotti
- Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy; Department Woman and Child Health Sciences, Catholic University of the Sacred Heart, Rome, Italy
| |
Collapse
|
368
|
Abstract
PURPOSE OF REVIEW Ovarian cancer (OC) is a heterogeneous disease and a mounting body of evidence shows that a 'one-size-fits-all' approach is obsolete. Differences in epidemiology, tumor biology, genetic profiles and treatment responses of these different types necessitate a tumor and patient-specific approach. Ninety percentage consists of epithelial OC with 70% being high-grade serous OC. The other rarer subtypes are low-grade serous (5%), clear cell (12%), endometrioid (11%) and mucinous carcinoma (3%). The remaining 10% are nonepithelial rare OCs: germ cell (3%) and sex-cord stromal tumors (7%). RECENT FINDINGS Over the past few decades, the 5-year survival rates have only improved modestly, therefore novel therapies are urgently needed. Recently, immunotherapy has been introduced into clinical practice in a number of solid tumors. Although preclinical data confirm the presence of an immunogenic microenvironment in a number of ovarian tumor types, no single-agent immune checkpoint inhibitor has been approved hitherto. Identifying suitable treatment combinations, adequate patient selection and thus correct implementation of immunotherapy remain major challenges. SUMMARY In this review, we focus on the rationale of incorporating immune therapy in rare OC, we summarize the recent developments with preclinical data and results of clinical trials, with particular focus on rare ovarian histological subtypes.
Collapse
Affiliation(s)
- Tina Laga
- Division of Gynecological Oncology, Department of Gynecology and Obstetrics and Leuven Cancer Institute, University Hospitals Leuven, Leuven, Belgium
| | | | | |
Collapse
|
369
|
Su R, Jin C, Zhou L, Cao Y, Kuang M, Li L, Xiang J. Construction of a ceRNA network of hub genes affecting immune infiltration in ovarian cancer identified by WGCNA. BMC Cancer 2021; 21:970. [PMID: 34461858 PMCID: PMC8404317 DOI: 10.1186/s12885-021-08711-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 08/19/2021] [Indexed: 12/22/2022] Open
Abstract
Background Ovarian cancer is the leading cause of death among gynecological malignancies. Immunotherapy has demonstrated potential effects in ovarian cancer. However, few studies on immune-related prognostic signatures in ovarian cancer have been reported. This study aimed to identify hub genes associated with immune infiltrates to provide insight into the immune regulatory mechanisms in ovarian cancer. Methods Raw data and clinical information were downloaded from The Cancer Genome Atlas (TCGA) and University of California, Santa Cruz (UCSC) Xena websites. Single-sample gene set enrichment analysis (ssGSEA) and weighted gene co-expression network analysis (WGCNA) were used to identify hub genes. Kaplan-Meier analysis and differential expression analysis were applied to explore the real hub genes. Results Through ssGSEA and WGCNA, 7 hub genes (LY9, CD5, CXCL9, IL2RG, SLAMF1, SLAMF6, and SLAMF7) were identified. Finally, LY9 and SLAMF1 were recognized as the real hub genes in immune infiltrates of ovarian cancer. LY9 and SLAMF1 are classified as SLAM family receptors involved in the activation of hematopoietic cells and the pathogenesis of multiple malignancies. Furthermore, 12 lncRNAs and 43 miRNAs significantly related to the 2 hub genes were applied to construct a lncRNA-miRNA-mRNA ceRNA network. The lncRNA-miRNA-mRNA ceRNA network shows upstream regulatory sites of the 2 hub genes. Conclusions These findings improve our understanding of the regulatory mechanism of and reveal potential immune checkpoints for immunotherapy for ovarian cancer.
Collapse
Affiliation(s)
- Rongjia Su
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Chengjuan Jin
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Lina Zhou
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Yannan Cao
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Menghua Kuang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Linxia Li
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xin Songjiang Road, Shanghai, 201620, China
| | - Jiangdong Xiang
- Department of Obstetrics and Gynecology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xin Songjiang Road, Shanghai, 201620, China.
| |
Collapse
|
370
|
Zhu J, Yan L, Wang Q. Efficacy of PD-1/PD-L1 inhibitors in ovarian cancer: a single-arm meta-analysis. J Ovarian Res 2021; 14:112. [PMID: 34454562 PMCID: PMC8403414 DOI: 10.1186/s13048-021-00862-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 08/18/2021] [Indexed: 01/15/2023] Open
Abstract
Several studies have evaluated the efficacy of PD-1/PD-L1 inhibitors in ovarian cancer; however, the response rate varies. This study aims to explore the efficacy of anti-PD-1/PD-L1 therapy in ovarian cancer. A quantitative meta-analysis was performed through a systematic search in PubMed, Web of Science, and the Cochrane Library. The pooled ORR was calculated and compared. Fifteen trials were included in this meta-analysis. Our analyses showed that the pooled ORR of all included studies was 19% (95% CI: 13%, 27%). Single PD-1/PD-L1 inhibitors had the lowest ORR of 9% (95% CI: 7%, 12%), while the combination of PD-1/PD-L1 inhibitors and chemotherapy had the highest ORR of 36% (95% CI: 24%, 51%). This study showed that PD-1/PD-L1 inhibitors alone have limited efficacy for ovarian cancer. The combination of PD-1/PD-L1 inhibitors and chemotherapy could be chosen as the recommended modality for further study.
Collapse
Affiliation(s)
- Jue Zhu
- Department of Gynecology, Ningbo Women & Children's Hospital, Ningbo, 315211, China
| | - Lifeng Yan
- Department of Gynecology, Ningbo Women & Children's Hospital, Ningbo, 315211, China
| | - Qiming Wang
- Department of Gynecology, Ningbo Women & Children's Hospital, Ningbo, 315211, China.
| |
Collapse
|
371
|
Leary A, Tan D, Ledermann J. Immune checkpoint inhibitors in ovarian cancer: where do we stand? Ther Adv Med Oncol 2021; 13:17588359211039899. [PMID: 34422119 PMCID: PMC8377306 DOI: 10.1177/17588359211039899] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/29/2021] [Indexed: 11/16/2022] Open
Abstract
Numerous retrospective studies have demonstrated that the density of intra-tumoral immune cell infiltration is prognostic in epithelial ovarian cancer (OC). These observations together with reports of programmed death ligand-1 (PD-L1) expression in advanced OC provided the rationale for investigating the benefit of programmed death-1 (PD1) or PD-L1 inhibition in OC. Unfortunately clinical trials to date evaluating PD1/PD-L1 inhibition in patients with relapsed OC have been disappointing. In this review we will discuss early results from single agent PD1/PD-L1 inhibitors and the strategies to enhance benefit from immune-oncology agents in OC, including proposing anti-PD-L1 in combination with other agents (cytotoxics, anti-angiogenics, poly(ADP-ribose) polymerase. (PARP) inhibitors, targeted therapies or other immunotherapies), as well as evaluating these agents earlier in the disease course, or in biomarker selected patients.
Collapse
Affiliation(s)
- Alexandra Leary
- Institut Gustave Roussy, 114 rue Edouard Vaillant, Villejuif 94805, France, Université Paris-Saclay, INSERM U981, Villejuif, France
| | - David Tan
- Department of Haematology–Oncology, National University Cancer Institute, Singapore, Cancer Science Institute, National University of Singapore, Singapore
| | - Jonathan Ledermann
- UCL Cancer Institute, Cancer Research UK and UCL Trials Centre, London, UK
| |
Collapse
|
372
|
De Pauw A, Naert E, Van de Vijver K, Philippe T, Vandecasteele K, Denys H. A CLEARER VIEW ON OVARIAN CLEAR CELL CARCINOMA. Acta Clin Belg 2021; 77:792-804. [PMID: 34404331 DOI: 10.1080/17843286.2021.1964051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Introduction: Ovarian clear cell carcinoma (OCCC) is a less common subtype accounting for approximately 5% of all epithelial ovarian cancers (EOCs). Clinical experience and research findings confirm the remarkable differences in clinical behavior, molecular alterations and pathogenesis of OCCC. The diagnosis of OCCC is typically set at a younger age, and earlier stage and in a background of endometriosis.Results: Molecularly, OCCCs rarely harbor BRCA1/BRCA2 mutations and have fewer copy number variants (CNVs). The most common molecular changes occur in the SWI/SNF chromatin remodeling complex genes, the PI3K/AKT signaling pathway and the receptor tyrosine kinase (RTK)/Ras signaling pathway.Five-year disease-specific survival of patients with OCCC is worse compared to high grade serous carcinomas (HGSOC). The current treatment options for OCCC are based on studies that included patients with predominantly HGSOC and only a minor proportion of cancers with clear cell histology. In order to improve outcomes for patients with OCCC, research should be specific for this subtype.Discussion: As the available information about the specific characteristics of OCCC is increasing, especially at a molecular level, it should be possible to continuously improve the specific diagnostics and treatment. Since OCCC is so rare, it is essential to collect new evidence at an international level. To avoid extrapolation from EOC trials with possible erroneous conclusions, patients should always be encouraged to participate in specific histological trials and basket trials, while paying extra attention to OCCC-like subtypes.
Collapse
Affiliation(s)
- Aglaja De Pauw
- Department of Internal Medicine and Pediatrics, Medical Oncology, Ghent University Hospital, Ghent, Belgium
| | - Eline Naert
- Department of Internal Medicine and Pediatrics, Medical Oncology, Ghent University Hospital, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Koen Van de Vijver
- Department of Diagnostic Sciences, Pathology, Ghent University Hospital, Ghent, Belgium
| | - Tummers Philippe
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Human Structure and Repair, Gynecology, Gent University Hospital, Ghent, Belgium
| | - Katrien Vandecasteele
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
- Department of Human Structure and Repair, Radiation Therapy, Ghent University Hospital, Ghent, Belgium
| | - Hannelore Denys
- Department of Internal Medicine and Pediatrics, Medical Oncology, Ghent University Hospital, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
373
|
Rocconi RP, Stanbery L, Madeira da Silva L, Barrington RA, Aaron P, Manning L, Horvath S, Wallraven G, Bognar E, Walter A, Nemunaitis J. Long-Term Follow-Up of Gemogenovatucel-T (Vigil) Survival and Molecular Signals of Immune Response in Recurrent Ovarian Cancer. Vaccines (Basel) 2021; 9:vaccines9080894. [PMID: 34452019 PMCID: PMC8402348 DOI: 10.3390/vaccines9080894] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/03/2021] [Accepted: 08/06/2021] [Indexed: 12/20/2022] Open
Abstract
Aim: To determine the relationship between gene expression profile (GEP) and overall survival (OS) by NanoString following treatment with Vigil. Patients and Methods: Recurrent ovarian cancer patients (n = 21) enrolled in prior clinical trials. Results: GEP stratified by TISHIGH vs. TISLOW demonstrated OS benefit (NR vs. 5.8 months HR 0.23; p = 0.0379), and in particular, MHC-II elevated baseline expression was correlated with OS advantage (p = 0.038). Moreover, 1-year OS was 75% in TISHIGH patients vs. 25% in TISLOW (p = 0.03795). OS was also correlated with positive γ-IFN ELISPOT response, 36.8 vs. 23.0 months (HR 0.19, p = 0.0098). Conclusion: Vigil demonstrates OS benefit in correlation with TISHIGH score, elevated MHC-II expression and positive γ-IFN ELISPOT in recurrent ovarian cancer patients.
Collapse
Affiliation(s)
- Rodney P. Rocconi
- Mitchell Cancer Institute, Division of Gynecologic Oncology, University of South Alabama, Mobile, AL 36604, USA;
| | - Laura Stanbery
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
| | - Luciana Madeira da Silva
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688, USA; (L.M.d.S.); (R.A.B.)
| | - Robert A. Barrington
- Department of Microbiology and Immunology, University of South Alabama, Mobile, AL 36688, USA; (L.M.d.S.); (R.A.B.)
| | - Phylicia Aaron
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
| | - Luisa Manning
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
| | - Staci Horvath
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
| | - Gladice Wallraven
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
| | - Ernest Bognar
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
| | | | - John Nemunaitis
- Gradalis, Inc., 2545 Golden Bear Drive, Suite 110, Carrollton, TX 75006, USA; (L.S.); (P.A.); (L.M.); (S.H.); (G.W.); (E.B.)
- Correspondence:
| |
Collapse
|
374
|
Monk BJ, Colombo N, Oza AM, Fujiwara K, Birrer MJ, Randall L, Poddubskaya EV, Scambia G, Shparyk YV, Lim MC, Bhoola SM, Sohn J, Yonemori K, Stewart RA, Zhang X, Perkins Smith J, Linn C, Ledermann JA. Chemotherapy with or without avelumab followed by avelumab maintenance versus chemotherapy alone in patients with previously untreated epithelial ovarian cancer (JAVELIN Ovarian 100): an open-label, randomised, phase 3 trial. Lancet Oncol 2021; 22:1275-1289. [PMID: 34363762 DOI: 10.1016/s1470-2045(21)00342-9] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/04/2021] [Accepted: 06/08/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Although most patients with epithelial ovarian cancer respond to frontline platinum-based chemotherapy, around 70% will relapse within 3 years. The phase 3 JAVELIN Ovarian 100 trial compared avelumab (anti-PD-L1 monoclonal antibody) in combination with chemotherapy followed by avelumab maintenance, or chemotherapy followed by avelumab maintenance, versus chemotherapy alone in patients with treatment-naive epithelial ovarian cancer. METHODS JAVELIN Ovarian 100 was a global, open-label, three-arm, parallel, randomised, phase 3 trial run at 159 hospitals and cancer treatment centres in 25 countries. Eligible women were aged 18 years and older with stage III-IV epithelial ovarian, fallopian tube, or peritoneal cancer (following debulking surgery, or candidates for neoadjuvant chemotherapy), and had an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients were randomly assigned (1:1:1) via interactive response technology to receive chemotherapy (six cycles; carboplatin dosed at an area under the serum-concentration-time curve of 5 or 6 intravenously every 3 weeks plus paclitaxel 175 mg/m2 every 3 weeks or 80 mg/m2 once a week [investigators' choice]) followed by avelumab maintenance (10 mg/kg intravenously every 2 weeks; avelumab maintenance group); chemotherapy plus avelumab (10 mg/kg intravenously every 3 weeks) followed by avelumab maintenance (avelumab combination group); or chemotherapy followed by observation (control group). Randomisation was in permuted blocks of size six and stratified by paclitaxel regimen and resection status. Patients and investigators were masked to assignment to the two chemotherapy groups without avelumab at the time of randomisation until completion of the chemotherapy phase. The primary endpoint was progression-free survival assessed by blinded independent central review in all randomly assigned patients (analysed by intention to treat). Safety was analysed in all patients who received at least one dose of study treatment. This trial is registered with ClinicalTrials.gov, NCT02718417. The trial was fully enrolled and terminated at interim analysis due to futility, and efficacy is no longer being assessed. FINDINGS Between May 19, 2016 and Jan 23, 2018, 998 patients were randomly assigned (avelumab maintenance n=332, avelumab combination n=331, and control n=335). At the planned interim analysis (data cutoff Sept 7, 2018), prespecified futility boundaries were crossed for the progression-free survival analysis, and the trial was stopped as recommended by the independent data monitoring committee and endorsed by the protocol steering committee. Median follow-up for progression-free survival for all patients was 10·8 months (IQR 7·1-14·9); 11·1 months (7·0-15·3) for the avelumab maintenance group, 11·0 months (7·4-14·5) for the avelumab combination group, and 10·2 months (6·7-14·0) for the control group. Median progression-free survival was 16·8 months (95% CI 13·5-not estimable [NE]) with avelumab maintenance, 18·1 months (14·8-NE) with avelumab combination treatment, and NE (18·2 months-NE) with control treatment. The stratified hazard ratio for progression-free survival was 1·43 (95% CI 1·05-1·95; one-sided p=0·99) with the avelumab maintenance regimen and 1·14 (0·83-1·56; one-sided p=0·79) with the avelumab combination regimen, versus control treatment. The most common grade 3-4 adverse events were anaemia (69 [21%] patients in the avelumab maintenance group, 63 [19%] in the avelumab combination group, and 53 [16%] in the control group), neutropenia (91 [28%], 99 [30%], and 88 [26%]), and neutrophil count decrease (49 [15%], 45 [14%], and 59 [18%]). Serious adverse events of any grade occurred in 92 (28%) patients in the avelumab maintenance group, 118 (36%) in the avelumab combination group, and 64 (19%) in the control group. Treatment-related deaths occurred in one (<1%) patient in the avelumab maintenance group (due to atrial fibrillation) and one (<1%) patient in the avelumab combination group (due to disease progression). INTERPRETATION Although no new safety signals were observed, results do not support the use of avelumab in the frontline treatment setting. Alternative treatment regimens are needed to improve outcomes in patients with advanced epithelial ovarian cancer. FUNDING Pfizer and Merck KGaA, Darmstadt, Germany.
Collapse
Affiliation(s)
- Bradley J Monk
- Arizona Oncology (US Oncology Network), Phoenix, AZ, USA; Department of Obstetrics and Gynecology, University of Arizona College of Medicine, Phoenix, AZ, USA; Department of Obstetrics and Gynecology, Creighton University School of Medicine at Dignity Health St Joseph's Hospital and Medical Center, Phoenix, AZ, USA.
| | - Nicoletta Colombo
- University of Milan-Bicocca, Milan, Italy; Istituto Europeo di Oncologia, IRCCS, Milan, Italy
| | - Amit M Oza
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Keiichi Fujiwara
- Saitama Medical University International Medical Center, Hidaka-City, Saitama, Japan
| | | | - Leslie Randall
- Virginia Commonwealth University, Massey Cancer Center, Richmond, VA, USA
| | - Elena V Poddubskaya
- I M Sechenov First Moscow State Medical University, Moscow, Russia; Clinical Center Vitamed, Moscow, Russia
| | - Giovanni Scambia
- Gynecologic Oncology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Yaroslav V Shparyk
- Lviv State Oncological Regional Treatment and Diagnostic Center, Lviv, Ukraine
| | - Myong Cheol Lim
- Research Institute and Hospital, National Cancer Center, Goyang, South Korea
| | - Snehalkumar M Bhoola
- Department of Obstetrics and Gynecology, University of Arizona College of Medicine, Phoenix, AZ, USA; Arizona Oncology Associates PC-HAL, Tempe, AZ, USA; Gynecologic Oncology, Cancer and Blood Specialists of Arizona, Gilbert, AZ, USA
| | - Joohyuk Sohn
- Severance Hospital, Yonsei University Health System, Seoul, South Korea
| | - Kan Yonemori
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Ross A Stewart
- Pfizer Oncology, Pfizer, San Diego, CA, USA; Oncology Research and Development, AstraZeneca, Cambridge, UK
| | | | | | - Carlos Linn
- Global Product Development, Pfizer, Taipei, Taiwan
| | | |
Collapse
|
375
|
Khalique S, Nash S, Mansfield D, Wampfler J, Attygale A, Vroobel K, Kemp H, Buus R, Cottom H, Roxanis I, Jones T, von Loga K, Begum D, Guppy N, Ramagiri P, Fenwick K, Matthews N, Hubank MJF, Lord CJ, Haider S, Melcher A, Banerjee S, Natrajan R. Quantitative Assessment and Prognostic Associations of the Immune Landscape in Ovarian Clear Cell Carcinoma. Cancers (Basel) 2021; 13:3854. [PMID: 34359755 PMCID: PMC8345766 DOI: 10.3390/cancers13153854] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 07/22/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
Ovarian clear cell carcinoma (OCCC) is a rare subtype of epithelial ovarian cancer characterised by a high frequency of loss-of-function ARID1A mutations and a poor response to chemotherapy. Despite their generally low mutational burden, an intratumoural T cell response has been reported in a subset of OCCC, with ARID1A purported to be a biomarker for the response to the immune checkpoint blockade independent of micro-satellite instability (MSI). However, assessment of the different immune cell types and spatial distribution specifically within OCCC patients has not been described to date. Here, we characterised the immune landscape of OCCC by profiling a cohort of 33 microsatellite stable OCCCs at the genomic, gene expression and histological level using targeted sequencing, gene expression profiling using the NanoString targeted immune panel, and multiplex immunofluorescence to assess the spatial distribution and abundance of immune cell populations at the protein level. Analysis of these tumours and subsequent independent validation identified an immune-related gene expression signature associated with risk of recurrence of OCCC. Whilst histological quantification of tumour-infiltrating lymphocytes (TIL, Salgado scoring) showed no association with the risk of recurrence or ARID1A mutational status, the characterisation of TILs via multiplexed immunofluorescence identified spatial differences in immunosuppressive cell populations in OCCC. Tumour-associated macrophages (TAM) and regulatory T cells were excluded from the vicinity of tumour cells in low-risk patients, suggesting that high-risk patients have a more immunosuppressive microenvironment. We also found that TAMs and cytotoxic T cells were also excluded from the vicinity of tumour cells in ARID1A-mutated OCCCs compared to ARID1A wild-type tumours, suggesting that the exclusion of these immune effectors could determine the host response of ARID1A-mutant OCCCs to therapy. Overall, our study has provided new insights into the immune landscape and prognostic associations in OCCC and suggest that tailored immunotherapeutic approaches may be warranted for different subgroups of OCCC patients.
Collapse
Affiliation(s)
- Saira Khalique
- Division of Brest Cancer, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.K.); (S.N.); (H.K.); (R.B.); (H.C.); (I.R.); (N.G.); (C.J.L.); (S.H.)
| | - Sarah Nash
- Division of Brest Cancer, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.K.); (S.N.); (H.K.); (R.B.); (H.C.); (I.R.); (N.G.); (C.J.L.); (S.H.)
| | - David Mansfield
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK; (D.M.); (A.M.)
| | - Julian Wampfler
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (J.W.); (A.A.); (K.V.)
| | - Ayoma Attygale
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (J.W.); (A.A.); (K.V.)
- Department of Histopathology, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK
| | - Katherine Vroobel
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (J.W.); (A.A.); (K.V.)
- Department of Histopathology, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK
| | - Harriet Kemp
- Division of Brest Cancer, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.K.); (S.N.); (H.K.); (R.B.); (H.C.); (I.R.); (N.G.); (C.J.L.); (S.H.)
| | - Richard Buus
- Division of Brest Cancer, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.K.); (S.N.); (H.K.); (R.B.); (H.C.); (I.R.); (N.G.); (C.J.L.); (S.H.)
| | - Hannah Cottom
- Division of Brest Cancer, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.K.); (S.N.); (H.K.); (R.B.); (H.C.); (I.R.); (N.G.); (C.J.L.); (S.H.)
| | - Ioannis Roxanis
- Division of Brest Cancer, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.K.); (S.N.); (H.K.); (R.B.); (H.C.); (I.R.); (N.G.); (C.J.L.); (S.H.)
| | - Thomas Jones
- Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK; (T.J.); (M.J.F.H.)
| | - Katharina von Loga
- Biomedical Research Centre, The Royal Marsden NHS Foundation Trust, London SM2 5PT, UK; (K.v.L.); (D.B.)
| | - Dipa Begum
- Biomedical Research Centre, The Royal Marsden NHS Foundation Trust, London SM2 5PT, UK; (K.v.L.); (D.B.)
| | - Naomi Guppy
- Division of Brest Cancer, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.K.); (S.N.); (H.K.); (R.B.); (H.C.); (I.R.); (N.G.); (C.J.L.); (S.H.)
| | - Pradeep Ramagiri
- Tumour Profiling Unit, The Institute of Cancer Research, London SW3 6JB, UK; (P.R.); (K.F.); (N.M.)
| | - Kerry Fenwick
- Tumour Profiling Unit, The Institute of Cancer Research, London SW3 6JB, UK; (P.R.); (K.F.); (N.M.)
| | - Nik Matthews
- Tumour Profiling Unit, The Institute of Cancer Research, London SW3 6JB, UK; (P.R.); (K.F.); (N.M.)
| | - Michael J. F. Hubank
- Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK; (T.J.); (M.J.F.H.)
| | - Christopher J. Lord
- Division of Brest Cancer, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.K.); (S.N.); (H.K.); (R.B.); (H.C.); (I.R.); (N.G.); (C.J.L.); (S.H.)
- The CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Syed Haider
- Division of Brest Cancer, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.K.); (S.N.); (H.K.); (R.B.); (H.C.); (I.R.); (N.G.); (C.J.L.); (S.H.)
| | - Alan Melcher
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW3 6JB, UK; (D.M.); (A.M.)
| | - Susana Banerjee
- Gynaecology Unit, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (J.W.); (A.A.); (K.V.)
- Division of Clinical Studies, The Institute of Cancer Research, London SM2 5NG, UK
| | - Rachael Natrajan
- Division of Brest Cancer, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK; (S.K.); (S.N.); (H.K.); (R.B.); (H.C.); (I.R.); (N.G.); (C.J.L.); (S.H.)
| |
Collapse
|
376
|
Heong V, Tan TZ, Miwa M, Ye J, Lim D, Herrington CS, Iida Y, Yano M, Yasuda M, Ngoi NY, Wong SJ, Okamoto A, Gourley C, Hasegawa K, Tan DS, Huang RY. A multi-ethnic analysis of immune-related gene expression signatures in patients with ovarian clear cell carcinoma. J Pathol 2021; 255:285-295. [PMID: 34322886 PMCID: PMC9539643 DOI: 10.1002/path.5769] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/07/2021] [Accepted: 07/23/2021] [Indexed: 11/23/2022]
Abstract
Little is known about the immune environment of ovarian clear cell carcinoma (OCCC) and its impact on various ethnic backgrounds. The aim of this OCCC immune‐related gene expression signatures (irGES) study was to address the interaction between tumour and immune environment of ethnically‐diverse Asian and Caucasian populations and to identify relevant molecular subsets of biological and clinical importance. Our study included 264 women from three different countries (Singapore, Japan, and the UK) and identified four novel immune subtypes (PD1‐high, CTLA4‐high, antigen‐presentation, and pro‐angiogenic subtype) with differentially expressed pathways, and gene ontologies using the NanoString nCounter PanCancer Immune Profiling Panel. The PD1‐high and CTLA4‐high subtypes demonstrated significantly higher PD1, PDL1, and CTLA4 expression, and were associated with poorer clinical outcomes. Mismatch repair (MMR) protein expression, assessed by immunohistochemistry, revealed that about 5% of OCCCs had deficient MMR expression. The prevalence was similar across the three countries and appeared to cluster in the CTLA4‐high subtype. Our results suggest that OCCC from women of Asian and Caucasian descent shares significant clinical and molecular similarities. To our knowledge, our study is the first study to include both Asian and Caucasian women with OCCC and helps to shine light on the impact of ethnic differences on the immune microenvironment of OCCC. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Valerie Heong
- Department of Medical Oncology, Tan Tock Seng Hospital, Singapore
| | - Tuan Z Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Maiko Miwa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Jieru Ye
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Diana Lim
- Department of Pathology, National University Hospital, Singapore
| | - C Simon Herrington
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC IGMM, University of Edinburgh, Edinburgh, UK
| | - Yasushi Iida
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC IGMM, University of Edinburgh, Edinburgh, UK
| | - Mitsutake Yano
- Department of Pathology, Saitama Medical University International Medical Center, Hidaka-shi, Japan
| | - Masanori Yasuda
- Department of Pathology, Saitama Medical University International Medical Center, Hidaka-shi, Japan
| | - Natalie Yl Ngoi
- Department of Haematology and Oncology, National University Cancer Institute Singapore, Singapore
| | - Sb Justin Wong
- Department of Pathology, National University Hospital, Singapore
| | - Aikou Okamoto
- Department of Obstetrics and Gynecology, The Jikei University School of Medicine, Tokyo, Japan
| | - Charlie Gourley
- Nicola Murray Centre for Ovarian Cancer Research, Cancer Research UK Edinburgh Centre, MRC IGMM, University of Edinburgh, Edinburgh, UK
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - David Sp Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Haematology and Oncology, National University Cancer Institute Singapore, Singapore
| | - Ruby Yj Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
377
|
Kang Y, Flores L, Ngai HW, Cornejo YR, Haber T, McDonald M, Moreira DF, Gonzaga JM, Abidi W, Zhang Y, Hammad M, Kortylewski M, Aboody KS, Berlin JM. Large, Anionic Liposomes Enable Targeted Intraperitoneal Delivery of a TLR 7/8 Agonist To Repolarize Ovarian Tumors' Microenvironment. Bioconjug Chem 2021; 32:1581-1592. [PMID: 34289694 DOI: 10.1021/acs.bioconjchem.1c00139] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Ovarian cancer is the most lethal gynecological malignancy in the United States. Current standard of treatment includes surgical debulking and chemotherapy, such as cisplatin and paclitaxel. However, the patients' response rate for chemotherapy in ovarian cancer is not optimal, and they often develop chemoresistance and suffer from side effects. Current clinical trials make extensive use of immune checkpoint blockade (ICB) as a novel cancer immunotherapeutic strategy against ovarian tumors. However, the response rates for ICB antibodies remain limited to 10-20% of treated ovarian cancer patients despite the success of this approach in melanoma, renal, head and neck, and nonsmall cell lung cancers. This lack of efficacy is often attributed to the "cold" immune status of ovarian tumors, as these tumors often have a low number of tumor-infiltrating lymphocytes (TILs) but a high number of suppressive immune cells, including tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), or regulatory T cells (Tregs). Repolarizing TAMs could be a promising strategy to reshape the tumor immune microenvironment and promote antitumor activity when combined with ICBs. Toll-like receptor (TLR) 7 and 8 agonists, such as imiquimod and resiquimod, are potent immunostimulatory molecules with potential to repolarize macrophages. However, these small molecules have poor pharmacokinetic profiles and can induce severe side effects when administered systemically. Previously, our group demonstrated that various large, anionic nanomaterials (silica, PLGA, and polystyrene) specifically target TAMs when administered intraperitoneally (IP) to ovarian tumor-bearing mice. In the present study, we demonstrate that large, anionic liposomes administered IP also efficiently localize to TAMs and can be used to target the delivery of resiquimod. Resiquimod delivered in this targeted fashion promoted activation of M1 macrophages and T cell infiltration, while reducing the percentage of Tregs in the tumor microenvironment. Finally, liposome-formulated resiquimod significantly enhanced the efficacy of PD1 blockade against syngeneic ovarian tumors. We anticipate that further optimization of our liposomal delivery strategy can generate a clinically relevant strategy for more effective and safer immunotherapy for ovarian cancer patients.
Collapse
|
378
|
Chen X, Lan H, He D, Xu R, Zhang Y, Cheng Y, Chen H, Xiao S, Cao K. Multi-Omics Profiling Identifies Risk Hypoxia-Related Signatures for Ovarian Cancer Prognosis. Front Immunol 2021; 12:645839. [PMID: 34349753 PMCID: PMC8327177 DOI: 10.3389/fimmu.2021.645839] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 07/01/2021] [Indexed: 12/23/2022] Open
Abstract
Background Ovarian cancer (OC) has the highest mortality rate among gynecologic malignancy. Hypoxia is a driver of the malignant progression in OC, which results in poor prognosis. We herein aimed to develop a validated model that was based on the hypoxia genes to systematically evaluate its prognosis in tumor immune microenvironment (TIM). Results We identified 395 hypoxia-immune genes using weighted gene co-expression network analysis (WGCNA). We then established a nine hypoxia-related genes risk model using least absolute shrinkage and selection operator (LASSO) Cox regression, which efficiently distinguished high-risk patients from low-risk ones. We found that high-risk patients were significantly related to poor prognosis. The high-risk group showed unique immunosuppressive microenvironment, lower antigen presentation, and higher levels of inhibitory cytokines. There were also significant differences in somatic copy number alterations (SCNAs) and mutations between the high- and low-risk groups, indicating immune escape in the high-risk group. Tumor immune dysfunction and exclusion (TIDE) and SubMap algorithms showed that low-risk patients are significantly responsive to programmed cell death protein-1 (PD-1) inhibitors. Conclusions In this study, we highlighted the clinical significance of hypoxia in OC and established a hypoxia-related model for predicting prognosis and providing potential immunotherapy strategies.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Hua Lan
- Department of Obstetrics and Gynecology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Dong He
- Department of Respiration, The Second People's Hospital of Hunan Province of Hunan University of Chinese Medicine, Changsha, China
| | - Runshi Xu
- Medical school, Hunan University of Chinese Medicine, Changsha, China
| | - Yao Zhang
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Yaxin Cheng
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Haotian Chen
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Songshu Xiao
- Department of Obstetrics and Gynecology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Ke Cao
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
379
|
Rasmussen M, Lim K, Rambech E, Andersen MH, Svane IM, Andersen O, Jensen LH, Nilbert M, Therkildsen C. Lynch syndrome-associated epithelial ovarian cancer and its immunological profile. Gynecol Oncol 2021; 162:686-693. [PMID: 34275654 DOI: 10.1016/j.ygyno.2021.07.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/29/2021] [Accepted: 07/01/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Lynch syndrome is a multi-tumor syndrome characterized by mismatch repair deficiency (MMR-d), microsatellite instability (MSI), and increased tumor-infiltrating lymphocytes (TILs) making these tumors candidates for treatment with immune checkpoint inhibitors. However, response may depend on tumor-induced immune evasion mechanisms, e.g. loss of Beta-2-Microglobulin (B2M) or upregulation of programmed death protein ligand 1 (PD-L1). We investigated the immune response and B2M and PD-L1 expression in Lynch syndrome-associated ovarian cancers. METHODS We successfully analyzed 30 Lynch syndrome-associated epithelial ovarian cancers collected through the Danish Hereditary Non-Polyposis Colorectal Cancer (HNPCC) register. MMR-d, MSI, immune response (CD3, CD8, and CD68), and immune evasion mechanisms (B2M and PD-L1) were investigated. Statistical associations between these markers were evaluated in addition to survival in relation to B2M/PD-L1. RESULTS Of the 29 evaluable tumors, 27 were MMR-d (93.1%). Likewise of 26 evaluable tumors, 14 were MSI (53.8%). MMR-d/MMR-proficiency associated with MSI/MSS in 60.0%. Half of the ovarian tumors presented with high levels of TILs. Loss of B2M expression was observed in 46.7% of the tumors, while expression of PD-L1 was seen in 28.0% of the cases. There was no association between B2M/PD-L1 and MSI/TILs/survival. Loss of B2M was often seen in tumors with low TILs (p = 0.056 or p = 0.059 for CD3 and CD8 positive cells, respectively). CONCLUSION MMR-d, MSI, and TILs are also seen in Lynch syndrome-associated ovarian cancers making these potential candidates for checkpoint-based immunotherapy. The clinical impact from immune evasion through loss of B2M needs to be investigated further in larger cohorts.
Collapse
Affiliation(s)
- Maria Rasmussen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark.
| | - Kevin Lim
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Eva Rambech
- Institute of Clinical Sciences, Division of Oncology and Pathology, Lund University, Sweden
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital - Herlev and Gentofte, Copenhagen, Denmark
| | - Ove Andersen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Lars Henrik Jensen
- Department of Oncology, University Hospital of Southern Denmark, Vejle, Denmark
| | - Mef Nilbert
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark; Institute of Clinical Sciences, Division of Oncology and Pathology, Lund University, Sweden; Danish Cancer Society Research Center, The Danish Cancer Society, Copenhagen, Denmark
| | - Christina Therkildsen
- Department of Clinical Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark; The Danish HNPCC Register, Gastro Unit, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| |
Collapse
|
380
|
Vanacker H, Harter P, Labidi-Galy SI, Banerjee S, Oaknin A, Lorusso D, Ray-Coquard I. PARP-inhibitors in epithelial ovarian cancer: Actual positioning and future expectations. Cancer Treat Rev 2021; 99:102255. [PMID: 34332292 DOI: 10.1016/j.ctrv.2021.102255] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 12/11/2022]
Abstract
Poly-(ADP)-ribose polymerase inhibitors (PARPi) are a class of oral anticancer drugs first developed as "synthetically lethal" in cancers harboring BRCA1/BRCA2 inactivating mutations. In high-grade serous or endometrioid ovarian cancers (HGOC), PARPi demonstrated benefit as maintenance therapy in relapsing BRCA-mutated and non-mutated tumors. Recently, they extended their indications to frontline maintenance therapy. This review summarizes the current place of PARPi (i) as maintenance or single agent in recurrent disease and (ii) frontline maintenance with different settings. We reviewed the course of biomarker identification, the challenge of overcoming resistance to PARPi and future combinations with targeted therapies, including anti-angiogenic, immune checkpoint inhibitors and DNA damage response inhibitors.
Collapse
Affiliation(s)
- Hélène Vanacker
- Centre Léon Bérard, Lyon, France; University Claude Bernard Lyon 1, France.
| | - Philipp Harter
- Department of Gynecology & Gynecologic Oncology, Ev. Kliniken Essen-Mitte, Essen, Germany.
| | - Sana Intidhar Labidi-Galy
- Department of Oncology, Hôpitaux Universitaires de Genève, Switzerland; Faculty of Medicine, Swiss Cancer Center Leman, Geneva, Switzerland.
| | - Susana Banerjee
- Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, United Kingdom.
| | - Ana Oaknin
- Vall d'Hebron Institute of Oncology, Barcelona, Spain.
| | | | | |
Collapse
|
381
|
ARID1A mutation/ARID1A loss is associated with a high immunogenic profile in clear cell ovarian cancer. Gynecol Oncol 2021; 162:679-685. [PMID: 34272091 DOI: 10.1016/j.ygyno.2021.07.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/10/2021] [Accepted: 07/04/2021] [Indexed: 11/23/2022]
Abstract
OBJECTIVES ARID1A mutation is frequently found in clear cell ovarian cancer (CCC) and endometrioid ovarian cancer (EC). Anti-PD-1 monotherapy has been found to have limited efficacy in epithelial ovarian cancer; however, anti-PD-1 therapy showed significant clinical benefit in some CCC. We sought to define the relationship of ARID1A mutation/ARID1A expression to the immunogenic profile of different histologic subtypes of ovarian cancer. METHODS We performed next-generation sequencing of 160 cancer-related genes. Also, we analyzed the immunohistochemical status of ARID1A, PD-L1, and CD8 with survival in different histologic subtypes of ovarian cancer in a total of 103 cases. RESULTS ARID1A mutation was found in 0% of the high-grade serous ovarian cancer (HGSC) (n = 36), 41.5% of the CCC (n = 41), 45.0% of the EC (n = 20), and 33.3% of the mucinous ovarian cancer (MC) (n = 6) cases. ARID1A loss was found in 19.4% of the HGSC, 75.6% of the CCC, 60.0% of the EC and 0% of the MC cases. ARID1A mutation was found to be associated with high PD-L1 (p < 0.001) or CD8 levels (p < 0.001) in CCC but not in other histologic subtypes. Meanwhile, ARID1A loss was associated with high PD-L1 or CD8 levels in CCC (p < 0.001) and HGSC (p < 0.001) but not in EC and MC. In addition, ARID1A mutation was associated with high tumor mutation burden in CCC (p = 0.006). CONCLUSIONS ARID1A mutation/ARID1A expression is associated with immune microenvironmental factors in CCC but not in EC. ARID1A status can be a biomarker for selecting candidates for immune checkpoint blockade in CCC.
Collapse
|
382
|
Development of MRI-Based Radiomics Model to Predict the Risk of Recurrence in Patients With Advanced High-Grade Serous Ovarian Carcinoma. AJR Am J Roentgenol 2021; 217:664-675. [PMID: 34259544 DOI: 10.2214/ajr.20.23195] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE. The purpose of our study was to develop a radiomics model based on preoperative MRI and clinical information for predicting recurrence-free survival (RFS) in patients with advanced high-grade serous ovarian carcinoma (HGSOC). MATERIALS AND METHODS. This retrospective study enrolled 117 patients with HGSOC, including 90 patients with recurrence and 27 without recurrence; 1046 radiomics features were extracted from T2-weighted images and contrast-enhanced T1-weighted images using a manual segmentation method. L1 regularization-based least absolute shrinkage and selection operator (LASSO) regression was performed to select features, and the synthetic minority oversampling technique (SMOTE) was used to balance our dataset. A support vector machine (SVM) classifier was used to build the classification model. To validate the performance of the proposed models, we applied a leave-one-out cross-validation method to train and test the classifier. Cox proportional hazards regression, Harrell concordance index (C-index), and Kaplan-Meier plots analysis were used to evaluate the associations between radiomics signatures and RFS. RESULTS. The fusion radiomics-based model yielded a significantly higher AUC value of 0.85 in evaluating RFS than the model using contrast-enhanced T1-weighted imaging features alone or T2-weighted imaging features alone (AUC = 0.79 and 0.74 and p = .02 and .01, respectively). Kaplan-Meier survival curves showed significant differences between high and low recurrence risk in patients with HGSOC by different models. The fusion model combining radiomics features and clinical information showed higher performance than the clinical model (C-index = 0.62 and 0.60, respectively). CONCLUSION. The proposed MRI-based radiomics signatures may provide a potential way to develop a prediction model and can help identify patients with advanced HGSOC who have a high risk of recurrence.
Collapse
|
383
|
Farrag MS, Abdelwahab K, Farrag NS, Elrefaie WE, Emarah Z. Programmed death ligand-1 and CD8 tumor-infiltrating lymphocytes (TILs) as prognostic predictors in ovarian high-grade serous carcinoma (HGSC). J Egypt Natl Canc Inst 2021; 33:16. [PMID: 34241710 DOI: 10.1186/s43046-021-00073-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 06/22/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND P D-L1 is expressed in tumor cells and plays a crucial role in tumor immune escape. Tumor-infiltrating lymphocytes (TILs) as CD8 T cells contribute to reduced tumor growth. Few studies investigated the prognostic effect of PD-L1 and CD8 TILs in ovarian high-grade serous carcinoma (HGSC). In the present study, we analyzed the expression of PD-L1 and CD8 TILs in HGSC by immunohistochemistry, and results were correlated to prognosis. It was carried on 54 cases of ovarian HGSC who attended the Oncology Centre, Mansoura University, Egypt, from 2012 till 2019. RESULTS Nearly 60% of cases showed positive PD-L1 expression in tumor cells. Regarding the clinicopathological characteristics, higher PD-L1 expression was found among patients with residual tumor (82.4%) compared to patients with no residual tumor (54.5%), with marginal statistical significance (p 0.07). PD-L1 was significantly associated with CD8 TILs expression. Higher PD-L1 expression was found among tumors with low expression of CD8 TILs with statistically significant difference (p≤0.001). Disease-free survival (DFS) was significantly lower among the group with positive expression of PD-L1 compared to the group with negative expression of PD-L1 (p 0.01), while overall survival (OS) was not associated with PD-L1 expression. On the other hand, the overall survival (OS) in patients with high CD8 expression was significantly higher than patients with low CD8 expression (p 0.043), while DFS was not significantly different among both CD8 TILS groups. CONCLUSIONS PD-L1 and CD8 TILs may become a promising therapeutic target for patients with ovarian HGSC. More studies are needed to further validate their prognostic effect. Precise identification of patients who will benefit from PD-L1 checkpoint blockade and TILs adaptive immunotherapy is mandatory.
Collapse
Affiliation(s)
- Mayada Saad Farrag
- Department of Pathology, Faculty of Medicine, Port Said University, Port Said, Egypt.
| | - Khaled Abdelwahab
- Department of Surgical Oncology, Mansoura Oncology Center, Mansoura University, Mansoura, Egypt
| | - Nesrine Saad Farrag
- Department of Community Medicine and Public Health, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Waleed Elsayed Elrefaie
- Department of Obstetrics and Gynecology, Port Said Faculty of Medicine, Port Said University, Port Said, Egypt
| | - Ziad Emarah
- Department of Medical Oncology, Mansoura Oncology Center, Mansoura University, Mansoura, Egypt
| |
Collapse
|
384
|
Rzhevskiy A, Kapitannikova A, Malinina P, Volovetsky A, Aboulkheyr Es H, Kulasinghe A, Thiery JP, Maslennikova A, Zvyagin AV, Ebrahimi Warkiani M. Emerging role of circulating tumor cells in immunotherapy. Theranostics 2021; 11:8057-8075. [PMID: 34335980 PMCID: PMC8315079 DOI: 10.7150/thno.59677] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 06/17/2021] [Indexed: 12/24/2022] Open
Abstract
Over the last few years, immunotherapy, in particular, immune checkpoint inhibitor therapy, has revolutionized the treatment of several types of cancer. At the same time, the uptake in clinical oncology has been slow owing to the high cost of treatment, associated toxicity profiles and variability of the response to treatment between patients. In response, personalized approaches based on predictive biomarkers have emerged as new tools for patient stratification to achieve effective immunotherapy. Recently, the enumeration and molecular analysis of circulating tumor cells (CTCs) have been highlighted as prognostic biomarkers for the management of cancer patients during chemotherapy and for targeted therapy in a personalized manner. The expression of immune checkpoints on CTCs has been reported in a number of solid tumor types and has provided new insight into cancer immunotherapy management. In this review, we discuss recent advances in the identification of immune checkpoints using CTCs and shed light on the potential applications of CTCs towards the identification of predictive biomarkers for immunotherapy.
Collapse
Affiliation(s)
- Alexey Rzhevskiy
- ARC Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW 2109, Australia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Institute for Urology and Reproductive Health, Sechenov University, Moscow 119991, Russia
| | - Alina Kapitannikova
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Polina Malinina
- Privolzhsky Research Medical University, 10/1, Minini Pozharsky Square, Nizhny Novgorod 603005, Russia
| | - Arthur Volovetsky
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russia
| | | | - Arutha Kulasinghe
- Queensland University of Technology, Centre for Genomics and Personalised Health, School of Biomedical Sciences, Faculty of Health, Woolloongabba, QLD 4102, Australia
- Translational Research Institute, Woolloongabba, QLD 4102 Australia
| | - Jean Paul Thiery
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- Guangzhou Institutes of Biomedicine and Health, Guangzhou, People's Republic of China
| | - Anna Maslennikova
- Lobachevsky State University of Nizhny Novgorod, Gagarina Avenue 23, Nizhny Novgorod 603950, Russia
- The Chair of Cancer, Radiotherapy and Radiologic Diagnostics, Privolzhsky Research Medical University, Nizhniy Novgorod. Russia 603005
| | - Andrei V. Zvyagin
- ARC Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW 2109, Australia
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- IBCh - Shemyakin Ovchinnikov Institute of BioOrganic Chemistry of the Russian Academy of Sciences, Miklukho Maklai Street, 16, Moscow, Russia
| | - Majid Ebrahimi Warkiani
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, 119991 Moscow, Russia
- School of Biomedical Engineering, University of Technology Sydney, 2007 Sydney, Australia
| |
Collapse
|
385
|
Gonzalez-Junca A, Liu FD, Nagaraja AS, Mullenix A, Lee CT, Gordley RM, Frimannsson DO, Maller O, Garrison BS, Iyer D, Benabbas A, Truong TA, Quach A, Tian M, Martinez R, Savur R, Perry-McNamara A, Nguyen D, Almudhfar N, Blanco C, Huynh C, Nand A, Lay YAE, Magal A, Mangalampalli S, Lee PJ, Lu TK, Lee G. SENTI-101, a Preparation of Mesenchymal Stromal Cells Engineered to Express IL12 and IL21, Induces Localized and Durable Antitumor Immunity in Preclinical Models of Peritoneal Solid Tumors. Mol Cancer Ther 2021; 20:1508-1520. [PMID: 34210826 DOI: 10.1158/1535-7163.mct-21-0030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/15/2021] [Accepted: 06/23/2021] [Indexed: 11/16/2022]
Abstract
Advanced peritoneal carcinomatosis including high-grade ovarian cancer has poor prognoses and a poor response rate to current checkpoint inhibitor immunotherapies; thus, there is an unmet need for effective therapeutics that would provide benefit to these patients. Here we present the preclinical development of SENTI-101, a cell preparation of bone marrow-derived mesenchymal stromal (also known as stem) cells (MSC), which are engineered to express two potent immune-modulatory cytokines, IL12 and IL21. Intraperitoneal administration of SENTI-101 results in selective tumor-homing and localized and sustained cytokine production in murine models of peritoneal cancer. SENTI-101 has extended half-life, reduced systemic distribution, and improved antitumor activity when compared with recombinant cytokines, suggesting that it is more effective and has lower risk of systemic immunotoxicities. Treatment of tumor-bearing immune-competent mice with a murine surrogate of SENTI-101 (mSENTI-101) results in a potent and localized immune response consistent with increased number and activation of antigen presenting cells, T cells and B cells, which leads to antitumor response and memory-induced long-term immunity. Consistent with this mechanism of action, co-administration of mSENTI-101 with checkpoint inhibitors leads to synergistic improvement in antitumor response. Collectively, these data warrant potential clinical development of SENTI-101 for patients with peritoneal carcinomatosis and high-grade ovarian cancer.Graphical abstract: SENTI-101 schematic and mechanism of actionSENTI-101 is a novel cell-based immunotherapeutic consisting of bone marrow-derived mesenchymal stromal cells (BM-MSC) engineered to express IL12 and IL21 intended for the treatment of peritoneal carcinomatosis including high-grade serous ovarian cancer. Upon intraperitoneal administration, SENTI-101 homes to peritoneal solid tumors and secretes IL12 and IL21 in a localized and sustained fashion. The expression of these two potent cytokines drives tumor infiltration and engagement of multiple components of the immune system: antigen-presenting cells, T cells, and B cells, resulting in durable antitumor immunity in preclinical models of cancer.
Collapse
Affiliation(s)
- Alba Gonzalez-Junca
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California.
| | - Frances D Liu
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Archana S Nagaraja
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Alyssa Mullenix
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Chen-Ting Lee
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Russell M Gordley
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Daniel O Frimannsson
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Ori Maller
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Brian S Garrison
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Dharini Iyer
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California.,Department of Technology and Operations, Senti Biosciences, Inc., South San Francisco, California
| | - Anissa Benabbas
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Tiffany A Truong
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Allison Quach
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Mengxi Tian
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Rowena Martinez
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Rishi Savur
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Alyssa Perry-McNamara
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Denny Nguyen
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Niran Almudhfar
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Carmina Blanco
- Department of Technology and Operations, Senti Biosciences, Inc., South San Francisco, California
| | - Christina Huynh
- Department of Technology and Operations, Senti Biosciences, Inc., South San Francisco, California
| | - Asish Nand
- Department of Technology and Operations, Senti Biosciences, Inc., South San Francisco, California
| | - Yu-An E Lay
- Department of Technology and Operations, Senti Biosciences, Inc., South San Francisco, California
| | - Ashita Magal
- Department of Technology and Operations, Senti Biosciences, Inc., South San Francisco, California
| | - Sravani Mangalampalli
- Department of Technology and Operations, Senti Biosciences, Inc., South San Francisco, California
| | - Philip J Lee
- Department of Technology and Operations, Senti Biosciences, Inc., South San Francisco, California
| | - Timothy K Lu
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| | - Gary Lee
- Department of Research and Development, Senti Biosciences, Inc., South San Francisco, California
| |
Collapse
|
386
|
Morand S, Devanaboyina M, Staats H, Stanbery L, Nemunaitis J. Ovarian Cancer Immunotherapy and Personalized Medicine. Int J Mol Sci 2021; 22:6532. [PMID: 34207103 PMCID: PMC8234871 DOI: 10.3390/ijms22126532] [Citation(s) in RCA: 210] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/11/2021] [Accepted: 06/13/2021] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer response to immunotherapy is limited; however, the evaluation of sensitive/resistant target treatment subpopulations based on stratification by tumor biomarkers may improve the predictiveness of response to immunotherapy. These markers include tumor mutation burden, PD-L1, tumor-infiltrating lymphocytes, homologous recombination deficiency, and neoantigen intratumoral heterogeneity. Future directions in the treatment of ovarian cancer include the utilization of these biomarkers to select ideal candidates. This paper reviews the role of immunotherapy in ovarian cancer as well as novel therapeutics and study designs involving tumor biomarkers that increase the likelihood of success with immunotherapy in ovarian cancer.
Collapse
Affiliation(s)
- Susan Morand
- Department of Medicine, University of Toledo, Toledo, OH 43614, USA; (S.M.); (M.D.); (H.S.)
| | - Monika Devanaboyina
- Department of Medicine, University of Toledo, Toledo, OH 43614, USA; (S.M.); (M.D.); (H.S.)
| | - Hannah Staats
- Department of Medicine, University of Toledo, Toledo, OH 43614, USA; (S.M.); (M.D.); (H.S.)
| | | | | |
Collapse
|
387
|
Shen J, Liu T, Bei Q, Xu S. Comprehensive Landscape of Ovarian Cancer Immune Microenvironment Based on Integrated Multi-Omics Analysis. Front Oncol 2021; 11:685065. [PMID: 34222009 PMCID: PMC8247482 DOI: 10.3389/fonc.2021.685065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/21/2021] [Indexed: 12/15/2022] Open
Abstract
Epithelial ovarian cancer has a low response rate to immunotherapy and a complex immune microenvironment that regulates its treatment outcomes. Understanding the immune microenvironment and its molecular basis is of great clinical significance in the effort to improve immunotherapy response and outcomes. To determine the characteristics of the immune microenvironment in ovarian cancer, we stratified ovarian cancer patients into three immune subtypes (C1, C2, and C3) using immune-related genes based on gene expression data from The Cancer Genome Atlas and found that these three subtypes had significant differences in immune characteristics and prognosis. Methylation and copy number variant analysis showed that the immune checkpoint genes that influenced immune response were significantly hypermethylated and highly deleted in the immunosuppressive C3 subtype, suggesting that epigenetic therapy may be able to reverse the efficacy of immunotherapy. In addition, the mutation frequencies of BRCA2 and CDK12 were significantly higher in the C2 subtype than in the other two subtypes, suggesting that mutation of DNA repair-related genes significantly affects the prognosis of ovarian cancer patients. Our study further elucidated the molecular characteristics of the immune microenvironment of ovarian cancer, which providing an effective hierarchical method for the immunotherapy of ovarian cancer patients, and has clinical relevance to the design of new immunotherapies and a reasonable combination strategies.
Collapse
Affiliation(s)
- Jiacheng Shen
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Tingwei Liu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Qiaoli Bei
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shaohua Xu
- Department of Gynecology, Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
388
|
Leung SOA, Konstantinopoulos PA. Advances in the treatment of platinum resistant epithelial ovarian cancer: an update on standard and experimental therapies. Expert Opin Investig Drugs 2021; 30:695-707. [PMID: 34082614 DOI: 10.1080/13543784.2021.1939305] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Platinum-resistant ovarian cancer (PROC) is broadly defined as disease recurrence within 6 months of completing platinum-based chemotherapy, either in the primary or recurrent setting. Although there is significant heterogeneity, PROC is generally associated with poor outcomes and low response rates to standard chemotherapy. There have been novel developments in therapeutics for PROC based on biomarkers and a more nuanced understanding of DNA repair and immunologic pathways.Areas covered: This review provides a summary of standard of care and experimental therapies for patients with PROC. Recent advances in our understanding of the DNA damage response and immunobiology of ovarian cancer have paved the way for single agent and combinatorial strategies involving PARP inhibitors, cell cycle checkpoint inhibitors, and immune checkpoint inhibitors to overcome PARP resistance, capitalize on high replication stress, and promote effective anti-tumor immunity, respectively. Furthermore, novel agents including antibody drug conjugates, bispecific antibodies, and recombinant fusion proteins show promise as experimental treatment options.Expert opinion: Standard and experimental treatment options available to patients with PROC have expanded. Testing for BRCA status, tumor mutational burden, and mismatch repair deficiency is recommended to guide therapy. Clinical trial participation is strongly encouraged with a focus on biomarker-driven trials targeting specific patient populations. Novel approaches such as ADCs, bispecific antibodies, targeting the GAS6/AXL and Notch pathways, and oncolytic virotherapy show considerable promise as emerging therapies.
Collapse
Affiliation(s)
- Shuk On Annie Leung
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Brigham and Women's Hospital, Harvard Medical School, Dana-Farber Cancer Institute, Boston, MA, US
| | | |
Collapse
|
389
|
Pujade-Lauraine E, Fujiwara K, Ledermann JA, Oza AM, Kristeleit R, Ray-Coquard IL, Richardson GE, Sessa C, Yonemori K, Banerjee S, Leary A, Tinker AV, Jung KH, Madry R, Park SY, Anderson CK, Zohren F, Stewart RA, Wei C, Dychter SS, Monk BJ. Avelumab alone or in combination with chemotherapy versus chemotherapy alone in platinum-resistant or platinum-refractory ovarian cancer (JAVELIN Ovarian 200): an open-label, three-arm, randomised, phase 3 study. Lancet Oncol 2021; 22:1034-1046. [PMID: 34143970 DOI: 10.1016/s1470-2045(21)00216-3] [Citation(s) in RCA: 232] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/25/2022]
Abstract
BACKGROUND Most patients with ovarian cancer will relapse after receiving frontline platinum-based chemotherapy and eventually develop platinum-resistant or platinum-refractory disease. We report results of avelumab alone or avelumab plus pegylated liposomal doxorubicin (PLD) compared with PLD alone in patients with platinum-resistant or platinum-refractory ovarian cancer. METHODS JAVELIN Ovarian 200 was an open-label, parallel-group, three-arm, randomised, phase 3 trial, done at 149 hospitals and cancer treatment centres in 24 countries. Eligible patients were aged 18 years or older with epithelial ovarian, fallopian tube, or peritoneal cancer (maximum of three previous lines for platinum-sensitive disease, none for platinum-resistant disease) and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients were randomly assigned (1:1:1) via interactive response technology to avelumab (10 mg/kg intravenously every 2 weeks), avelumab plus PLD (40 mg/m2 intravenously every 4 weeks), or PLD and stratified by disease platinum status, number of previous anticancer regimens, and bulky disease. Primary endpoints were progression-free survival by blinded independent central review and overall survival in all randomly assigned patients, with the objective to show whether avelumab alone or avelumab plus PLD is superior to PLD. Safety was assessed in all patients who received at least one dose of study treatment. This trial is registered with ClinicalTrials.gov, NCT02580058. The trial is no longer enrolling patients and this is the final analysis of both primary endpoints. FINDINGS Between Jan 5, 2016, and May 16, 2017, 566 patients were enrolled and randomly assigned (combination n=188; PLD n=190, avelumab n=188). At data cutoff (Sept 19, 2018), median duration of follow-up for overall survival was 18·4 months (IQR 15·6-21·9) for the combination group, 17·4 months (15·2-21·3) for the PLD group, and 18·2 months (15·8-21·2) for the avelumab group. Median progression-free survival by blinded independent central review was 3·7 months (95% CI 3·3-5·1) in the combination group, 3·5 months (2·1-4·0) in the PLD group, and 1·9 months (1·8-1·9) in the avelumab group (combination vs PLD: stratified HR 0·78 [repeated 93·1% CI 0·59-1·24], one-sided p=0·030; avelumab vs PLD: 1·68 [1·32-2·60], one-sided p>0·99). Median overall survival was 15·7 months (95% CI 12·7-18·7) in the combination group, 13·1 months (11·8-15·5) in the PLD group, and 11·8 months (8·9-14·1) in the avelumab group (combination vs PLD: stratified HR 0·89 [repeated 88·85% CI 0·74-1·24], one-sided p=0·21; avelumab vs PLD: 1·14 [0·95-1·58], one-sided p=0·83]). The most common grade 3 or worse treatment-related adverse events were palmar-plantar erythrodysesthesia syndrome (18 [10%] in the combination group vs nine [5%] in the PLD group vs none in the avelumab group), rash (11 [6%] vs three [2%] vs none), fatigue (ten [5%] vs three [2%] vs none), stomatitis (ten [5%] vs five [3%] vs none), anaemia (six [3%] vs nine [5%] vs three [2%]), neutropenia (nine [5%] vs nine [5%] vs none), and neutrophil count decreased (eight [5%] vs seven [4%] vs none). Serious treatment-related adverse events occurred in 32 (18%) patients in the combination group, 19 (11%) in the PLD group, and 14 (7%) in the avelumab group. Treatment-related adverse events resulted in death in one patient each in the PLD group (sepsis) and avelumab group (intestinal obstruction). INTERPRETATION Neither avelumab plus PLD nor avelumab alone significantly improved progression-free survival or overall survival versus PLD. These results provide insights for patient selection in future studies of immune checkpoint inhibitors in platinum-resistant or platinum-refractory ovarian cancer. FUNDING Pfizer and Merck KGaA, Darmstadt, Germany.
Collapse
Affiliation(s)
| | - Keiichi Fujiwara
- Saitama Medical University International Medical Center, Hidaka, Saitama, Japan
| | - Jonathan A Ledermann
- University College London Cancer Institute, London, UK; University College London Hospitals, London, UK
| | - Amit M Oza
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Rebecca Kristeleit
- University College London Cancer Institute, London, UK; University College London Hospitals, London, UK
| | - Isabelle-Laure Ray-Coquard
- Centre Léon Bérard, Service de Cancérologie Médicale, Université Claude Bernard Lyon 1, Lyon, France; Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Paris, France
| | - Gary E Richardson
- Cabrini Hospital, Department of Medical Oncology, Malvern, VIC, Australia
| | - Cristiana Sessa
- Oncology Institute of Southern Switzerland, Ospedale San Giovanni, Bellinzona, Switzerland
| | - Kan Yonemori
- Department of Breast and Medical Oncology, National Cancer Center Hospital, Chuo-ku, Tokyo, Japan
| | - Susana Banerjee
- The Royal Marsden NHS Foundation Trust, London, UK; The Institute of Cancer Research, London, UK
| | - Alexandra Leary
- Groupe d'Investigateurs Nationaux pour l'Etude des Cancers Ovariens (GINECO), Paris, France; Gustave Roussy Cancer Campus, Villejuif, France
| | | | - Kyung Hae Jung
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Songpa-gu, Seoul, South Korea
| | - Radoslaw Madry
- Poznan University of Medical Sciences, Department of Oncology, Poznan, Poland
| | - Sang-Yoon Park
- Center for Uterine Cancer, National Cancer Center, Ilsandong-gu, Goyang-si, Gyeonggi-do, South Korea
| | | | | | | | - Caimiao Wei
- Pfizer, Global Biometrics and Data Management, Groton, CT, USA
| | | | - Bradley J Monk
- Arizona Oncology (US Oncology Network), University of Arizona College of Medicine, Creighton University School of Medicine, Phoenix, AZ, USA
| |
Collapse
|
390
|
Abstract
Purpose of Review Advanced epithelial ovarian cancer remains the most lethal gynaecological cancer. Most patients with advanced disease will relapse within 3 years after primary treatment with surgery and chemotherapy. Recurrences become increasing difficult to treat due to the emergence of drug resistance and 5-year survival has changed little over the last decade. Maintenance treatment, here defined as treatment given beyond primary chemotherapy, can both consolidate the response and prolong the control of disease which is an approach to improve survival. Recent Findings Here we review maintenance strategies such as targeting angiogenesis, interference of DNA repair through inhibition of PARP, combinations of targeting agents, and immunotherapy and hormonal therapy. Summary Much has been learnt from the success and challenges of these treatments that have in the last few years which led to significant reduction in disease recurrence, changed the guidelines for treatment, and established a new paradigm for the treatment of ovarian cancer.
Collapse
|
391
|
Konstantinopoulos PA, Cannistra SA. Immune Checkpoint Inhibitors in Ovarian Cancer: Can We Bridge the Gap Between IMagynation and Reality? J Clin Oncol 2021; 39:1833-1838. [PMID: 33891471 DOI: 10.1200/jco.21.00571] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
392
|
Paijens ST, Vledder A, Loiero D, Duiker EW, Bart J, Hendriks AM, Jalving M, Workel HH, Hollema H, Werner N, Plat A, Wisman GBA, Yigit R, Arts H, Kruse AJ, de Lange N, Koelzer VH, de Bruyn M, Nijman HW. Prognostic image-based quantification of CD8CD103 T cell subsets in high-grade serous ovarian cancer patients. Oncoimmunology 2021; 10:1935104. [PMID: 34123576 PMCID: PMC8183551 DOI: 10.1080/2162402x.2021.1935104] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/20/2021] [Indexed: 11/06/2022] Open
Abstract
CD103-positive tissue resident memory-like CD8+ T cells (CD8CD103 TRM) are associated with improved prognosis across malignancies, including high-grade serous ovarian cancer (HGSOC). However, whether quantification of CD8, CD103 or both is required to improve existing survival prediction and whether all HGSOC patients or only specific subgroups of patients benefit from infiltration, remains unclear. To address this question, we applied image-based quantification of CD8 and CD103 multiplex immunohistochemistry in the intratumoral and stromal compartments of 268 advanced-stage HGSOC patients from two independent clinical institutions. Infiltration of CD8CD103 immune cell subsets was independent of clinicopathological factors. Our results suggest CD8CD103 TRM quantification as a superior method for prognostication compared to single CD8 or CD103 quantification. A survival benefit of CD8CD103 TRM was observed only in patients treated with primary cytoreductive surgery. Moreover, survival benefit in this group was limited to patients with no macroscopic tumor lesions after surgery. This approach provides novel insights into prognostic stratification of HGSOC patients and may contribute to personalized treatment strategies in the future.
Collapse
Affiliation(s)
- S. T. Paijens
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A. Vledder
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - D. Loiero
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - E. W. Duiker
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - J. Bart
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A. M. Hendriks
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - M. Jalving
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H. H. Workel
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H. Hollema
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - N. Werner
- Department of Pathology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A. Plat
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G. B. A. Wisman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - R. Yigit
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H. Arts
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A. J. Kruse
- Department of Obstetrics and Gynecology, Isala Hospital Zwolle, Zwolle, The Netherlands
| | - N.M. de Lange
- Department of Obstetrics and Gynecology, Isala Hospital Zwolle, Zwolle, The Netherlands
| | - V. H. Koelzer
- Department of Pathology and Molecular Pathology, University Hospital and University of Zurich, Zurich, Switzerland
| | - M. de Bruyn
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - H. W. Nijman
- Department of Obstetrics and Gynecology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
393
|
Walsh CS, Kamrava M, Rogatko A, Kim S, Li A, Cass I, Karlan B, Rimel BJ. Phase II trial of cisplatin, gemcitabine and pembrolizumab for platinum-resistant ovarian cancer. PLoS One 2021; 16:e0252665. [PMID: 34081738 PMCID: PMC8174738 DOI: 10.1371/journal.pone.0252665] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 04/26/2021] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE To evaluate the combination of pembrolizumab, cisplatin and gemcitabine in recurrent platinum-resistant ovarian cancer. METHODS Patients received six cycles of chemotherapy with gemcitabine and cisplatin on day 1 and day 8 of a 21-day treatment cycle. Pembrolizumab was administered on day 1 of cycles 3-6 and as maintenance monotherapy in cycles 7-34. Palliative radiation to a non-target symptomatic lesion was allowed. The primary objective was overall response rate by RECIST 1.1 criteria. Secondary objectives included safety, progression-free survival, time to progression, duration of response and overall survival. RESULTS An interim analysis for futility was performed at 18 evaluable patients. Overall response rate was 60%, duration of response was 4.9 months and time to progression was 5.2 months. Progression-free survival at 6 and 12 months was 43% and 5%. Median progression-free survival was 6.2 months and median overall survival was 11.3 months. In all patients, CA125 levels reflected response and progression. There were no pseudoprogression events. After receiving palliative radiation during pembrolizumab maintenance, a patient with recurrent ovarian clear cell carcinoma had an exceptional and durable response that is ongoing for greater than 2 years. After consultation with the sponsor, based on the modest duration of response observed at the interim analysis for futility, the decision was made to close the trial to further accrual. CONCLUSIONS The addition of pembrolizumab to cisplatin and gemcitabine did not appear to provide benefit beyond chemotherapy alone in patients with recurrent platinum-resistant ovarian cancer.
Collapse
Affiliation(s)
- Christine S. Walsh
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- * E-mail:
| | - Mitchell Kamrava
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Andre Rogatko
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Sungjin Kim
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Andrew Li
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Ilana Cass
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Beth Karlan
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| | - Bobbie J. Rimel
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
- Cedars-Sinai Cancer, Cedars-Sinai Medical Center, Los Angeles, California, United States of America
| |
Collapse
|
394
|
Royfman R, Whiteley E, Noe O, Morand S, Creeden J, Stanbery L, Hamouda D, Nemunaitis J. BRCA1/2 signaling and homologous recombination deficiency in breast and ovarian cancer. Future Oncol 2021; 17:2817-2830. [PMID: 34058833 DOI: 10.2217/fon-2021-0072] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Patients who have mutations of the genes BRCA1 or BRCA2 are at an increased risk for developing breast and ovarian cancer. BRCA1/2 function as tumor suppressor genes, responsible for regulating DNA repair, and play an essential role in homologous recombination. Mutation of BRCA1/2 results in homologous recombination deficiency and genomic instability which drives oncogenesis and cancer proliferation. Recently, BRCA1/2 gene expression has been implicated in regulating immune response. Here we discuss the signaling pathway of BRCA1/2 in relation to breast and ovarian cancer, with emphasis on how dysregulation facilitates the path to malignancy and current treatment options.
Collapse
Affiliation(s)
- Rachel Royfman
- University of Toledo Medical Center, Department of Internal Medicine, Toledo, OH 43614, USA
| | - Emma Whiteley
- University of Toledo Medical Center, Department of Internal Medicine, Toledo, OH 43614, USA
| | - Olivia Noe
- University of Toledo Medical Center, Department of Internal Medicine, Toledo, OH 43614, USA
| | - Susan Morand
- University of Toledo Medical Center, Department of Internal Medicine, Toledo, OH 43614, USA
| | - Justin Creeden
- University of Toledo Medical Center, Department of Internal Medicine, Toledo, OH 43614, USA
| | - Laura Stanbery
- Gradalis, Inc., Carrollton, Department of Medical Affairs, Carrollton, TX 75006, USA
| | - Danae Hamouda
- University of Toledo Medical Center, Department of Internal Medicine, Toledo, OH 43614, USA
| | - John Nemunaitis
- Gradalis, Inc., Carrollton, Department of Medical Affairs, Carrollton, TX 75006, USA
| |
Collapse
|
395
|
Ngoi NYL, Tan DSP. The role of homologous recombination deficiency testing in ovarian cancer and its clinical implications: do we need it? ESMO Open 2021; 6:100144. [PMID: 34015643 PMCID: PMC8141874 DOI: 10.1016/j.esmoop.2021.100144] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/19/2021] [Accepted: 04/09/2021] [Indexed: 12/20/2022] Open
Abstract
The recognition of homologous recombination deficiency (HRD) as a frequent feature of high-grade serous ovarian cancer (HGSOC) has transformed treatment paradigms. Poly(ADP-ribose) polymerase inhibitors (PARPis), developed based on the rationale of synthetic lethality that predicates antitumor efficacy in tumors harboring underlying HRD, now represents an important class of therapy for HGSOC. Recent data have drawn attention to the assessment of homologous recombination DNA repair (HRR) as a prognostic and predictive biomarker in HGSOC, leading to increasing debate on the optimal means of defining and evaluating HRD, both genotypically and phenotypically. At present, clinical-grade assays such as myChoice CDx and FoundationOne CDx are approved companion diagnostics which can identify patients with HRD-positive HGSOC by diagnosing a 'genomic scar' reflecting underlying genomic instability. Yet despite the rapid maturation of this field, tumoral HRD status has been recognized to be dynamic over time and with treatment pressure. In practice, this means that restoration of HRR through mechanisms of platinum and PARPi resistance are not adequately represented by genomic scar assays, and contribute toward discordance with clinical PARPi response, or lack-thereof. It is thus critical that HRD testing is optimized to address the controversies of diverse HRD testing methodology, appropriate thresholds for HRD identification, and relevant timepoints for HRD testing, in order to realize the potential for PARPis to maximally benefit patients with HGSOC. Here, we discuss the premise of HRD testing in HGSOC, current methodologies for HRD identification and their performance in the clinic, highlight upcoming strategies, and discuss the challenges faced in moving this field forward.
Collapse
Affiliation(s)
- N Y L Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - D S P Tan
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore; Cancer Science Institute, National University of Singapore, Singapore, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
396
|
Russo L, Avesani G, Gui B, Trombadori CML, Salutari V, Perri MT, Di Paola V, Rodolfino E, Scambia G, Manfredi R. Immunotherapy-Related Imaging Findings in Patients with Gynecological Malignancies: What Radiologists Need to Know. Korean J Radiol 2021; 22:1310-1322. [PMID: 34047505 PMCID: PMC8316780 DOI: 10.3348/kjr.2020.1299] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 01/26/2021] [Accepted: 03/05/2021] [Indexed: 01/15/2023] Open
Abstract
Immunotherapy is an effective treatment option for gynecological malignancies. Radiologists dealing with gynecological patients undergoing treatment with immune checkpoint inhibitors should be aware of unconventional immune-related imaging features for the evaluation of tumor response and immune-related adverse events. In this paper, immune checkpoint inhibitors used for gynecological malignancies and their mechanisms of action are briefly presented. In the second part, patterns of pseudoprogression are illustrated, and different forms of immune-related adverse events are discussed.
Collapse
Affiliation(s)
- Luca Russo
- UOC Radiologia Generale ed Interventistica Generale, Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Area Diagnostica per Immagini, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giacomo Avesani
- UOC Radiologia Generale ed Interventistica Generale, Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Area Diagnostica per Immagini, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Benedetta Gui
- UOC Radiologia Generale ed Interventistica Generale, Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Area Diagnostica per Immagini, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.
| | | | - Vanda Salutari
- UOC Ginecologia Oncologica, Dipartimento per la Salute della Donna e del Bambino e della Salute Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Maria Teresa Perri
- Istituto di Ginecologia e Ostetricia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Valerio Di Paola
- UOC Radiologia Generale ed Interventistica Generale, Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Area Diagnostica per Immagini, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elena Rodolfino
- UOC Radiologia Generale ed Interventistica Generale, Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Area Diagnostica per Immagini, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Giovanni Scambia
- UOC Ginecologia Oncologica, Dipartimento per la Salute della Donna e del Bambino e della Salute Pubblica, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Istituto di Ginecologia e Ostetricia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Riccardo Manfredi
- UOC Radiologia Generale ed Interventistica Generale, Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Area Diagnostica per Immagini, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy.,Dipartimento Universitario di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
397
|
Chen X, Lan H, He D, Wang Z, Xu R, Yuan J, Xiao M, Zhang Y, Gong L, Xiao S, Cao K. Analysis of Autophagy-Related Signatures Identified Two Distinct Subtypes for Evaluating the Tumor Immune Microenvironment and Predicting Prognosis in Ovarian Cancer. Front Oncol 2021; 11:616133. [PMID: 34041016 PMCID: PMC8141647 DOI: 10.3389/fonc.2021.616133] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 04/19/2021] [Indexed: 12/26/2022] Open
Abstract
Ovarian cancer (OC) is one of the most lethal gynecologic malignant tumors. The interaction between autophagy and the tumor immune microenvironment has clinical importance. Hence, it is necessary to explore reliable biomarkers associated with autophagy-related genes (ARGs) for risk stratification in OC. Here, we obtained ARGs from the MSigDB database and downloaded the expression profile of OC from TCGA database. The k-means unsupervised clustering method was used for clustering, and two subclasses of OC (cluster A and cluster B) were identified. SsGSEA method was used to quantify the levels of infiltration of 24 subtypes of immune cells. Metascape and GSEA were performed to reveal the differential gene enrichment in signaling pathways and cellular processes of the subtypes. We found that patients in cluster A were significantly associated with higher immune infiltration and immune-associated signaling pathways. Then, we established a risk model by LASSO Cox regression. ROC analysis and Kaplan-Meier analysis were applied for evaluating the efficiency of the risk signature, patients with low-risk got better outcomes than those with high-risk in overall survival. Finally, ULK2 and GABARAPL1 expression was further validated in clinical samples. In conclusion, Our study constructed an autophagy-related prognostic indicator, and identified two promising targets in OC.
Collapse
Affiliation(s)
- Xingyu Chen
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Hua Lan
- Department of Obstetrics and Gynecology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Dong He
- The Second People's Hospital of Hunan Province, Hunan University of Chinese Medicine, Changsha, China
| | - Zhanwang Wang
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Runshi Xu
- Medical School, Hunan University of Chinese Medicine, Changsha, China
| | - Jing Yuan
- Department of Obstetrics and Gynecology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Mengqing Xiao
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Yao Zhang
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Lian Gong
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Songshu Xiao
- Department of Obstetrics and Gynecology, Third Xiangya Hospital of Central South University, Changsha, China
| | - Ke Cao
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
398
|
van Wilpe S, Tolmeijer SH, Koornstra RHT, de Vries IJM, Gerritsen WR, Ligtenberg M, Mehra N. Homologous Recombination Repair Deficiency and Implications for Tumor Immunogenicity. Cancers (Basel) 2021; 13:cancers13092249. [PMID: 34067105 PMCID: PMC8124836 DOI: 10.3390/cancers13092249] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 01/06/2023] Open
Abstract
Homologous recombination repair deficiency (HRD) can be observed in virtually all cancer types. Although HRD sensitizes tumors to DNA-damaging chemotherapy and poly(ADP-ribose) polymerase (PARP) inhibitors, all patients ultimately develop resistance to these therapies. Therefore, it is necessary to identify therapeutic regimens with a more durable efficacy. HRD tumors have been suggested to be more immunogenic and, therefore, more susceptible to treatment with checkpoint inhibitors. In this review, we describe how HRD might mechanistically affect antitumor immunity and summarize the available translational evidence for an association between HRD and antitumor immunity across multiple tumor types. In addition, we give an overview of all available clinical data on the efficacy of checkpoint inhibitors in HRD tumors and describe the evidence for using treatment strategies that combine checkpoint inhibitors with PARP inhibitors.
Collapse
Affiliation(s)
- Sandra van Wilpe
- Department of Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (S.v.W.); (S.H.T.); (W.R.G.)
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Sofie H. Tolmeijer
- Department of Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (S.v.W.); (S.H.T.); (W.R.G.)
| | | | - I. Jolanda M. de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Winald R. Gerritsen
- Department of Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (S.v.W.); (S.H.T.); (W.R.G.)
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Marjolijn Ligtenberg
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (S.v.W.); (S.H.T.); (W.R.G.)
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
- Correspondence:
| |
Collapse
|
399
|
Razumova Z, Bizzarri N, Kacperczyk-Bartnik J, Pletnev A, Gonzalez Martin A, Persson J. Report from the European Society of Gynaecological Oncology (ESGO) 2020 State-of-the-Art Virtual Meeting. Int J Gynecol Cancer 2021; 31:658-669. [PMID: 33811109 DOI: 10.1136/ijgc-2021-002577] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/10/2021] [Indexed: 11/04/2022] Open
Abstract
This is a report from the European Society of Gynaecological Oncology State-of-the-Art Virtual Meeting held December 14-16, 2020. The unique 3-day conference offered comprehensive state-of-the-art summaries on the major advances in the treatment of different types of gynecological cancers. Sessions opened with a case presentation followed by a keynote lecture and interactive debates with opinion leaders in the field. The speakers also presented scientific reviews on the clinical trial landscape in collaboration with the European Network of Gynecological Oncological Trial (ENGOT) groups. In addition, the new ESGO-ESRTO-ESP endometrial cancer guidelines were officially presented in public. This paper describes the key information and latest studies that were presented for the first time at the conference.
Collapse
Affiliation(s)
- Zoia Razumova
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Nicolò Bizzarri
- Dipartimento per la salute della Donna e del Bambino e della Salute Pubblica, UOC Ginecologia Oncologica, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | | | - Andrei Pletnev
- Department of Gynaecological Oncology, N.N. Alexandrov National Cancer Centre of Belarus, Minsk, Belarus
| | | | - Jan Persson
- Department of Obstetrics and Gynaecology, Skåne University Hospital, Lund, Sweden
- Department of Clinical Sciences, Faculty of Medicine at Lund University, Lund, Sweden
| |
Collapse
|
400
|
Majidpoor J, Mortezaee K. The efficacy of PD-1/PD-L1 blockade in cold cancers and future perspectives. Clin Immunol 2021; 226:108707. [DOI: 10.1016/j.clim.2021.108707] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/15/2021] [Accepted: 02/25/2021] [Indexed: 12/30/2022]
|