451
|
Henriksen JR, Nederby L, Donskov F, Waldstrøm M, Adimi P, Jakobsen A, Dahl Steffensen K. Blood natural killer cells during treatment in recurrent ovarian cancer. Acta Oncol 2020; 59:1365-1373. [PMID: 32692270 DOI: 10.1080/0284186x.2020.1791358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVE Recent research indicated favorable prognostic impact of intratumoral natural killer (NK) cells in ovarian carcinoma (OC). The role of NK cells during chemotherapy in OC is unknown. We investigated impact of NK cells in OC patients treated with palliative chemotherapy. METHODS Participants receiving palliative chemotherapy for recurrent OC (N = 72) had prospectively blood samples at baseline and before cycle 2. NK cell counts were quantified by flow cytometry. NK cell activity was measured by the NK Vue® assay, estimating interferon-gamma production. Overall survival (OS) was the primary endpoint. Cutoffs were predefined, NK numbers (≥184 × 106 cells/L vs. <184 × 106 cells/L) and NK activity (<200 pg/mL vs. ≥200 pg/mL). RESULTS Median OS in patients with low vs. high NK cell count at baseline was 7.1 months vs. 15.6 months (p = .028), respectively, and before cycle 2 was 5.7 vs. 17.3 months, p < .001, respectively. The difference in restricted mean survival (ΔRMST) was 5.7 months (95% CI: 3.3-8.0) at cycle 2 vs. 2.5 months (95% CI: -0.6 to 5.6) at baseline, showing a significant difference with no overlap of confidence intervals. In multivariate analyses, low NK cell count remained significant with a hazard ratio (HR)=2.83, 95% CI: 1.53-5.22, p = .001 (baseline) and HR = 3.34, 95% CI: 1.67-6.71, p = .001 (before cycle 2). Patients with both low NK count and NK activity at baseline (N = 20) had median OS 6.5 months vs. 11.5 months in patients with either high activity, high count or both (p = .007). In parallel, patients with both low NK activity and count at cycle 2 (N = 18) had a median survival of 4.0 months vs. 15.4 months (p < .001). CONCLUSIONS A low blood NK cell count in recurrent metastatic ovarian cancer during chemotherapy is associated with unfavorable prognostic impact. Early increase in survival difference based on NK cell status suggests an association between NK cell count and treatment benefit.
Collapse
Affiliation(s)
- Jon Røikjær Henriksen
- Department of Oncology, Vejle Hospital – University Hospital of Southern Denmark, Vejle, Denmark
- Institute of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Line Nederby
- Department of Biochemistry and Immunology, Vejle Hospital – University Hospital of Southern Denmark, Vejle, Denmark
| | - Frede Donskov
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Marianne Waldstrøm
- Institute of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Vejle Hospital – University Hospital of Southern Denmark, Vejle, Denmark
| | - Parvin Adimi
- Department of Oncology, Vejle Hospital – University Hospital of Southern Denmark, Vejle, Denmark
| | - Anders Jakobsen
- Department of Oncology, Vejle Hospital – University Hospital of Southern Denmark, Vejle, Denmark
- Institute of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Karina Dahl Steffensen
- Department of Oncology, Vejle Hospital – University Hospital of Southern Denmark, Vejle, Denmark
- Institute of Regional Health Research, Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
452
|
Modulation of Immune Infiltration of Ovarian Cancer Tumor Microenvironment by Specific Subpopulations of Fibroblasts. Cancers (Basel) 2020; 12:cancers12113184. [PMID: 33138184 PMCID: PMC7692816 DOI: 10.3390/cancers12113184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/20/2020] [Accepted: 10/24/2020] [Indexed: 12/15/2022] Open
Abstract
Tumor immune infiltration plays a key role in the progression of solid tumors, including ovarian cancer, and immunotherapies are rapidly emerging as effective treatment modalities. However, the role of cancer-associated fibroblasts (CAFs), a predominant stromal constituent, in determining the tumor-immune microenvironment and modulating efficacy of immunotherapies remains poorly understood. We have conducted an extensive bioinformatic analysis of our and other publicly available ovarian cancer datasets (GSE137237, GSE132289 and GSE71340), to determine the correlation of fibroblast subtypes within the tumor microenvironment (TME) with the characteristics of tumor-immune infiltration. We identified (1) four functional modules of CAFs in ovarian cancer that are associated with the TME and metastasis of ovarian cancer, (2) immune-suppressive function of the collagen 1,3,5-expressing CAFs in primary ovarian cancer and omental metastases, and (3) consistent positive correlations between the functional modules of CAFs with anti-immune response genes and negative correlation with pro-immune response genes. Our study identifies a specific fibroblast subtype, fibroblast functional module (FFM)2, in the ovarian cancer tumor microenvironment that can potentially modulate a tumor-promoting immune microenvironment, which may be detrimental toward the effectiveness of ovarian cancer immunotherapies.
Collapse
|
453
|
Yabuno A, Matsushita H, Hamano T, Tan TZ, Shintani D, Fujieda N, Tan DSP, Huang RYJ, Fujiwara K, Kakimi K, Hasegawa K. Identification of serum cytokine clusters associated with outcomes in ovarian clear cell carcinoma. Sci Rep 2020; 10:18503. [PMID: 33116254 PMCID: PMC7595156 DOI: 10.1038/s41598-020-75536-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 10/13/2020] [Indexed: 01/01/2023] Open
Abstract
Serum cytokine and chemokine networks may reflect the complex systemic immunological interactions in cancer patients. Studying groups of cytokines and their networks may help to understand their clinical biology. A total of 178 cases of ovarian cancer were analyzed in this study, including 73 high-grade serous (HGSC), 66 clear cell (CCC) and 39 endometrioid carcinomas. Suspension cytokine arrays were performed with the patients’ sera taken before the primary surgery. Associations between each cytokine and clinicopathological factors were analyzed in all patients using multivariate linear regression models, and cluster analyses were performed for each histotype. In the multivariate analyses, twelve of 27 cytokines were correlated with histotypes. Cluster analyses in each histotype revealed 2 cytokine signatures S1 and S2 in HGSC, and similarly C1 and C2 in CCC. Twenty-two of 27 cytokines were commonly clustered in HGSC and CCC. Signature S1 and C1 included IL-2,6,8,15, chemokines and angiogenic factors, whereas signature S2 and C2 included IL-4,5,9,10,13, TNF-α and G-CSF. Four subgroups based on a high or low level for each signature were identified, and this cluster-based classification demonstrated significantly different progression-free and overall survivals for CCC patients (P = 0.00097 and P = 0.017).
Collapse
Affiliation(s)
- Akira Yabuno
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1298, Japan
| | - Hirokazu Matsushita
- Department of Immunotherapeutics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.
| | - Tetsutaro Hamano
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1298, Japan
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Daisuke Shintani
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1298, Japan
| | - Nao Fujieda
- Department of Immunotherapeutics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - David S P Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,National University Cancer Institute, Singapore, National University Hospital, Singapore, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1298, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, 1397-1 Yamane, Hidaka-shi, Saitama, 350-1298, Japan.
| |
Collapse
|
454
|
Voutsadakis IA. Further Understanding of High-Grade Serous Ovarian Carcinogenesis: Potential Therapeutic Targets. Cancer Manag Res 2020; 12:10423-10437. [PMID: 33116896 PMCID: PMC7585777 DOI: 10.2147/cmar.s249540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/25/2020] [Indexed: 01/09/2023] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the most common type of ovarian cancer and the most lethal gynecologic malignancy due to advanced stage at presentation. Recent years have witnessed progress in the therapy of HGSOC with the introduction of PARP (poly-adenosine diphosphate ribose polymerase) inhibitors and the anti-angiogenic monoclonal antibody bevacizumab to the backbone of chemotherapy or as maintenance therapy after chemotherapy. The improved molecular understanding of ovarian cancer pathogenesis, which has brought these therapies into the clinic, aspires to extend the boundaries of therapies through elucidation of other molecular aspects of ovarian carcinogenesis. This accumulating knowledge has started to be translated to additional targeted therapies that are in various stages of development. These include inhibitors of the function of other proteins involved in homologous recombination deficiency (HRD), such as WEE1 kinase, ATM/ATR kinases and CDK12 inhibitors. Despite disappointing results with immune checkpoint inhibitors monotherapy, harnessing the immune system in HGSOC with combination therapies that promote antigen production and immune cell activation is an avenue being explored. This paper examines arising HGSOC therapies based on molecular understanding of pathogenesis.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, Sault Ste. Marie, Ontario, Canada
- Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, Ontario, Canada
| |
Collapse
|
455
|
Lorusso D, Ceni V, Muratore M, Salutari V, Nero C, Pietragalla A, Ciccarone F, Carbone V, Daniele G, Scambia G. Emerging role of immune checkpoint inhibitors in the treatment of ovarian cancer. Expert Opin Emerg Drugs 2020; 25:445-453. [PMID: 33040627 DOI: 10.1080/14728214.2020.1836155] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION In recent years, ovarian cancer (OC) treatment has been enriched with many new target therapies, most of all antiangiogenic drugs and PARP inhibitors (PARPis), which have literally changed the natural history of the disease. The impressive results of immunotherapy in other malignancies, mainly melanoma and lung cancer, and the good signals of activity in gynecological neoplasms like cervical and microsatellite instable (MSI-H) endometrial cancer, opened the space to the introduction of immune-stimulatory drugs in ovarian cancer. AREA COVERED The goal of this article is to summarize the newest evidence on the use of immune check point inhibitors in OC trying to explain why, at present, this strategy has failed to improve clinical outcome and focusing on the possible strategies to overcome treatment failure. EXPERT OPINION Although numerous trials have been undertaken, only scanty results have been obtained so far with immune check-point inhibitors (ICIs) in OC either when used as single agents or in combination with antiangiogenic therapy and ongoing trials are exploring the association of ICIs with PARPis and other ICIs. A better knowledge of predictive biomarkers of response and mechanisms of immunotherapy resistance, will help in identifying the most appropriate population to treat with ICIs.
Collapse
Affiliation(s)
- Domenica Lorusso
- Scientific Directorate 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
| | - Valentina Ceni
- Department of Gynecology and Obstetrics, University of Parma, Via A. Gramsci , Parma, Italy.,Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli , Rome, Italy
| | - Margherita Muratore
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli , Rome, Italy.,Department of Medical Oncology, University of Naples Federico II, Via S.Pansini , Naples, Italy
| | - Vanda Salutari
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli , Rome, Italy
| | - Camilla Nero
- Scientific Directorate Fondazione Policlinico Universitario A Gemelli IRCCS , Rome, Italy
| | - Antonella Pietragalla
- Scientific Directorate Fondazione Policlinico Universitario A Gemelli IRCCS , Rome, Italy
| | - Francesca Ciccarone
- Scientific Directorate Fondazione Policlinico Universitario A Gemelli IRCCS , Rome, Italy
| | - Vittoria Carbone
- Department of Women and Child Health, Division of Gynecologic Oncology, Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Agostino Gemelli , Rome, Italy
| | - Gennaro Daniele
- Scientific Directorate Fondazione Policlinico Universitario A Gemelli IRCCS , Rome, Italy
| | - Giovanni Scambia
- Scientific Directorate 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
| |
Collapse
|
456
|
Haunschild CE, Tewari KS. The current landscape of molecular profiling in the treatment of epithelial ovarian cancer. Gynecol Oncol 2020; 160:333-345. [PMID: 33055011 DOI: 10.1016/j.ygyno.2020.09.043] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/27/2020] [Indexed: 01/05/2023]
Abstract
Advances in next generation sequencing have allowed for rapid and economical germline and tumor genomic profiling. Targeted therapies based on molecular tumor profiling are now integrated into treatment guidelines for many solid tumors. In epithelial ovarian cancer, 50% of tumors possess damaging mutations in homologous recombination repair genes (aka homologous recombination deficiency or HRD) which includes the BRCA genes. Deleterious BRCA mutations and HRD have recently emerged as predictive biomarkers for the use of PARP inhibitors in ovarian cancer. Every patient with ovarian cancer must be referred for genetic counseling and germline testing for BRCA mutations. Multigene panel genetic testing may be more informative and cost-effective than limited testing of cancer susceptibility genes. Patients without a germline deleterious BRCA mutation must be assessed for a somatic BRCA mutation. Assays for HRD may help guide treatment options in women who do not have a BRCA mutation. Currently, all patients with a germline or somatic BRCA mutation should be offered upfront maintenance therapy with a PARP inhibitor. During May 2020, options for maintenance therapy with a PARP inhibitor were expanded to patients with HRD and HR-proficient tumors.
Collapse
Affiliation(s)
- Carolyn E Haunschild
- The Division of Gynecologic Oncology, The Chao Family Comprehensive Cancer Center, University of California, Irvine Medical Center, United States of America
| | - Krishnansu S Tewari
- The Division of Gynecologic Oncology, The Chao Family Comprehensive Cancer Center, University of California, Irvine Medical Center, United States of America.
| |
Collapse
|
457
|
Hamilton G. Avelumab: search for combinations of immune checkpoint inhibition with chemotherapy. Expert Opin Biol Ther 2020; 21:311-322. [PMID: 32954871 DOI: 10.1080/14712598.2021.1825679] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Immune checkpoint inhibition (ICI) has proved successful for selected tumors and a subpopulation of patients. The human monoclonal IgG1 antibody (mAB) avelumab capable of mediating antibody-dependent cytotoxicity (ADCC) lysis is directed to programmed death ligand-1 (PD-L1) of tumor cells and is tested in trials aiming to improve ICI in combination with chemotherapeutic drugs. AREAS COVERED This article presents an overview of the current trials to enhance ICI regimens using avelumab in combination with chemotherapeutics, antiangiogenetic drugs, and immunomodulators. Predictive factors for this kind of immunochemotherapy are discussed. EXPERT OPINION Clinical data demonstrate that avelumab shows efficacy in cancer patients against Merkel cell carcinoma (MCC), renal cell carcinoma (RCC), and urothelial cancers as single agent. Furthermore, avelumab in combination with axitinib in RCC increases survival and exhibits activity in combination with docetaxel in urothelial carcinoma. However, several other immunochemotherapy trials for ovarian cancer, gastric cancer, and non-small lung cancer (NSCLC) showed no activity due to factors disfavoring administration of immunotherapy combos.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Department of Vascular Biology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
458
|
Association of PD-L1 expression by immunohistochemistry and gene microarray with molecular subtypes of ovarian tumors. Mod Pathol 2020; 33:2001-2010. [PMID: 32404957 DOI: 10.1038/s41379-020-0567-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/26/2020] [Accepted: 04/26/2020] [Indexed: 12/15/2022]
Abstract
Identifying patients who respond to immune checkpoint blockade (ICB) is a significant challenge in oncology. While PD-L1 expression by immunohistochemistry (IHC) is the current diagnostic gold standard for patient selection, it nevertheless does not capture all patients who may respond to ICB. Recent gene expression studies in high-grade serous ovarian carcinoma have defined an immunoreactive molecular subtype that has a measurable difference in patient survival compared with non-immunoreactive subtypes, but no studies have yet demonstrated its impact on predicting response to ICB. As a step toward establishing the predictive value of gene expression classifiers in ICB, we assessed the relationship between PD-L1 IHC and molecular subtypes of ovarian epithelial cancer. This was done by analyzing a total of 93 tissue specimens from patients with stage III and IV disease, and comparing PD-L1 IHC with gene expression by Agilent microarrays using TCGA-defined subtypes. We showed that ovarian tumors with elevated IHC PD-L1 expression are most strongly associated with immunoreactive subtype as compared with other molecular subtypes, reaching statistical significance against differentiated (Dunn's test, 33.39, p = 0.0003) and mesenchymal (39.63, p < 0.0001) subtypes. Comparing PD-L1 scoring with CPS vs. TPS showed similar trends, but with stronger correlation strength when using CPS (Kruskal-Wallis, H = 27.52, p < 0.0001), as opposed to TPS (H = 25.04, p < 0.0001). Interestingly, while PD-L1 gene expression by microarray was significantly increased in the immunoreactive subtype (H = 20.25, p = 0.0002), it showed a positive but relatively poor correlation to IHC. Overall, the results demonstrate potential value in use of the molecular classifier to select patients for ICB, pending further studies that assess its ability to predict treatment outcomes. In the future, integration of cellular, protein, and genomic biomarkers in the tumor and tumor microenvironment may improve current methods of predicting treatment response.
Collapse
|
459
|
Gou R, Dong H, Lin B. Application and reflection of genomic scar assays in evaluating the efficacy of platinum salts and PARP inhibitors in cancer therapy. Life Sci 2020; 261:118434. [PMID: 32941897 DOI: 10.1016/j.lfs.2020.118434] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/05/2020] [Accepted: 09/11/2020] [Indexed: 12/24/2022]
Abstract
Defective DNA repair is one of the most important features of tumors. BRCA1/2 participates in homologous recombination repair as a key tumor suppressor gene. BRCA1/2 mutation is an important biomarker for predicting the sensitivity of platinum salts and Poly (ADP-ribose) polymerase (PARP) inhibitors in breast cancer, ovarian cancer, and other cancers. However, epigenetic modifications and other mutations in homologous recombination repair (HRR) genes can also cause homologous recombination deficiency (HRD). Patients with no BRCA1/2 mutations, but bearing similar molecular phenotypes (BRCAness) can still obtain clinical benefits from treatment with platinum salts or PARP inhibitors. Therefore, an accurate assessment of HRD is essential for the formulation of personalized treatments. However, methods to identify HRD in tumors vary and are controversial. Currently, genomic scar assays have been used in multiple clinical trials to assess patient clinical benefit. This review summarizes the therapeutic effects of platinum salts and PARP inhibitors in breast and ovarian cancer, clarifies the predictive value of genomic scar assays in evaluating the clinical benefit of different patient groups and treatment options, and proposes the limitations and optimization of current HRD scoring methods. Using and optimizing genomic scar assays can help to accurately screen the population with the most benefit, expand the scope of drug application, and make the most suitable clinical decision based on individual differences.
Collapse
Affiliation(s)
- Rui Gou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Hui Dong
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China
| | - Bei Lin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning, China; Key Laboratory of Maternal-Fetal Medicine of Liaoning Province, Key Laboratory of Obstetrics and Gynecology of Higher Education of Liaoning Province, Liaoning, China.
| |
Collapse
|
460
|
Definition and Independent Validation of a Proteomic-Classifier in Ovarian Cancer. Cancers (Basel) 2020; 12:cancers12092519. [PMID: 32899818 PMCID: PMC7564837 DOI: 10.3390/cancers12092519] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/25/2020] [Accepted: 08/27/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary The heterogeneity of epithelial ovarian cancer and its associated molecular biological characteristics are continuously integrated in the development of therapy guidelines. In a next step, future therapy recommendations might also be able to focus on the patient’s systemic status, not only the tumor’s molecular pattern. Therefore, new methods to identify and validate host-related biomarkers need to be established. Using mass spectrometry, we developed and independently validated a blood-based proteomic classifier, stratifying epithelial ovarian cancer patients into good and poor survival groups. We also determined an age dependence of the prognostic performance of this classifier and its association with important biological processes. This work highlights that, just like molecular markers of the tumor itself, the systemic condition of a patient (partly reflected in proteomic patterns) also influences survival and therapy response and could therefore be integrated into future processes of therapy planning. Abstract Mass-spectrometry-based analyses have identified a variety of candidate protein biomarkers that might be crucial for epithelial ovarian cancer (EOC) development and therapy response. Comprehensive validation studies of the biological and clinical implications of proteomics are needed to advance them toward clinical use. Using the Deep MALDI method of mass spectrometry, we developed and independently validated (development cohort: n = 199, validation cohort: n = 135) a blood-based proteomic classifier, stratifying EOC patients into good and poor survival groups. We also determined an age dependency of the prognostic performance of this classifier, and our protein set enrichment analysis showed that the good and poor proteomic phenotypes were associated with, respectively, lower and higher levels of complement activation, inflammatory response, and acute phase reactants. This work highlights that, just like molecular markers of the tumor itself, the systemic condition of a patient (partly reflected in proteomic patterns) also influences survival and therapy response in a subset of ovarian cancer patients and could therefore be integrated into future processes of therapy planning.
Collapse
|
461
|
Le Saux O, Ray-Coquard I, Labidi-Galy SI. Challenges for immunotherapy for the treatment of platinum resistant ovarian cancer. Semin Cancer Biol 2020; 77:127-143. [DOI: 10.1016/j.semcancer.2020.08.017] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 12/24/2022]
|
462
|
Morphological and molecular heterogeneity of epithelial ovarian cancer: Therapeutic implications. EJC Suppl 2020; 15:1-15. [PMID: 33240438 PMCID: PMC7573476 DOI: 10.1016/j.ejcsup.2020.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 02/03/2020] [Accepted: 02/09/2020] [Indexed: 12/11/2022] Open
Abstract
Ovarian epithelial cancer (OEC) is the most lethal gynecologic malignancy. Despite current chemotherapeutic and surgical options, this high lethality can be attributed to multiple factors, including late-stage presentation. In order to optimize OEC treatment, it is important to highlight that it is composed of five main subtypes: high-grade serous ovarian carcinoma (HGSOC), low-grade serous ovarian carcinoma (LGSOC), endometrioid ovarian carcinoma (EOC), ovarian clear cell carcinoma (CCOC), and mucinous ovarian carcinoma (MOC). These subtypes differ in their precursor lesions, as well as in epidemiological, morphological, molecular and clinical features. OEC is one of the tumours in which most pathogenic germline mutations have been identified. Accordingly, up to 20% OC show alterations in BRCA1/2 genes, and also, although with a lower frequency, in other low penetrance genes associated with homologous recombination deficiency (HRD), mismatch repair genes (Lynch syndrome) and TP53. The most important prognostic factor is the 2014 FIGO staging, while older age is also associated with worse survival. HGSOC in all stages and CCC and MOC in advanced stages have the worse prognosis among histological types. Molecular markers have emerged as prognostic factors, particularly mutations in BRCA1/2, which are associated with a better outcome. Regarding treatment, whereas a proportion of HGSOC is sensible to platinum-based treatment and PARP inhibitors due to HRD, the rest of the histological types are relatively chemoresistant. New treatments based in specific molecular alterations are being tested in different histological types. In addition, immunotherapy could be an option, especially for EOC carrying mismatch repair deficiency or POLE mutations. The five different histological types have different precursor lesions and epidemiological, morphological, genetic, epigenetic and clinical features. Histological type is an important prognostic factor. Drugs targeting homologous recombination deficiency have been approved for treatment. The use of immunotherapy is limited due to lack of predictive biomarkers
Collapse
|
463
|
Combined pembrolizumab and pegylated liposomal doxorubicin in platinum resistant ovarian cancer: A phase 2 clinical trial. Gynecol Oncol 2020; 159:72-78. [PMID: 32771276 DOI: 10.1016/j.ygyno.2020.07.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/18/2020] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Pegylated liposomal doxorubicin (PLD) in vitro may have immunomodulatory abilities and preclinical evidence suggests it synergizes with immune checkpoint blockade. We hypothesized that combining PLD and pembrolizumab would be active in patients with platinum-resistant ovarian cancer (PROC). METHODS This was a single-arm, multi-center phase II trial. Eligible patients had PROC with ≤2 prior lines of cytotoxic therapy for recurrent or persistent disease. Twenty-six patients were enrolled and given pembrolizumab 200 mg intravenously (IV) every 3 weeks and PLD 40 mg/m2 IV every 4 weeks. Patients were assessed radiographically every 8 weeks. The primary endpoint was clinical benefit rate (CBR), defined as complete response (CR) + partial response (PR) + stable disease (SD) ≥24 weeks. The study was powered to detect an improvement in CBR from 25% to 50%, with rejection of the null hypothesis if at least 10 patients achieved clinical benefit. T-cell inflamed gene expression profiles (GEP) and PD-L1 were assessed and correlated with clinical outcome. RESULTS Twenty-three patients were evaluable for best overall response. The study satisfied its primary endpoint, with 12 patients achieving clinical benefit for a CBR of 52.2% (95% CI 30.6-73.2%). There were 5 PRs (21.7%) and 1 CR (4.3%), for an overall response rate (ORR) of 26.1%. Six patients had SD lasting at least 24 weeks. Combination therapy was well tolerated without unexpected toxicities. CONCLUSIONS The combination of pembrolizumab and PLD was manageable, without unexpected toxicities, and showed preliminary evidence of clinical benefit in the treatment of platinum resistant ovarian cancer. ORR and median PFS of combination therapy in this study was higher than historical comparisons of PLD alone or anti-PD-1/PD-L1 agents alone. TRIAL REGISTRATION Clinicaltrials.gov identifier: NCT02865811.
Collapse
|
464
|
Bi F, Chen Y, Yang Q. Significance of tumor mutation burden combined with immune infiltrates in the progression and prognosis of ovarian cancer. Cancer Cell Int 2020; 20:373. [PMID: 32774167 PMCID: PMC7405355 DOI: 10.1186/s12935-020-01472-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
Background Ovarian cancer (OC) is the most malignant tumor in the female reproductive system. About 75% of OC in complete remission of clinical symptoms still develop a recurrence. Therefore, searching for new treatment methods plays an important role in improving the prognosis of OC. Methods We downloaded the MAF files, RNA-seq data and clinical information from the TCGA database. The “maftools” package in R software was used to visualize the OC mutation data. We calculated the tumor mutation burden (TMB) of OC and analyzed its correlation with clinicopathological parameters and prognostic value. Tumor mutation burden related signature model was constructed to predict the overall survival (OS) of OC. Results The results revealed that there was a statistical correlation between TMB and FIGO stage, grade and tumor residual size of ovarian cancer patients. The Kaplan–Meier curve indicated that a high TMB is associated with better clinical outcomes of OC. The difference analysis indicated 24 upregulated genes and 619 downregulated genes in the high-TMB group compared with the low-TMB group. Besides, the TMBRS model based on five hub genes (RBMS3, PLA2G5, CDH2, AMHR2 and ADAMTS8) was constructed to predict the OS of OC. The ROC curve and validation data sets all revealed that the TMBRS model was reliable in predicting recurrence risk. Immune microenvironment analysis indicated the correlations between TMB and infiltrating immune cells. Conclusions Our results suggest that TMB plays an important role in the prognosis and guiding immunotherapy of OC. By detecting the TMB of OC, clinicians can more accurately treat patients with immunotherapy, thereby improving their survival rate.
Collapse
Affiliation(s)
- Fangfang Bi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, NO. 36 Sanhao Road, Shenyang, 110000 China
| | - Ying Chen
- Department of Ultrasound, Jiangnan Hospital Affiliated to Zhejiang University of Traditional Chinese Medicine, Hangzhou, China
| | - Qing Yang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, NO. 36 Sanhao Road, Shenyang, 110000 China
| |
Collapse
|
465
|
Profound Functional Suppression of Tumor-Infiltrating T-Cells in Ovarian Cancer Patients Can Be Reversed Using PD-1-Blocking Antibodies or DARPin® Proteins. J Immunol Res 2020; 2020:7375947. [PMID: 32832572 PMCID: PMC7424497 DOI: 10.1155/2020/7375947] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 06/30/2020] [Indexed: 12/29/2022] Open
Abstract
PD-1/PD-L1 blockade has revolutionized the field of immunooncology. Despite the relative success, the response rate to anti-PD-1 therapy requires further improvements. Our aim was to explore the enhancement of T-cell function by using novel PD-1-blocking proteins and compare with clinically approved monoclonal antibodies (mAbs). We isolated T-cells from the ascites and tumor of 17 patients with advanced epithelial ovarian cancer (EOC) and analyzed the effects using the mAbs nivolumab and pembrolizumab and two novel engineered ankyrin repeat proteins (DARPin® proteins). PD-1 blockade with either mAb or DARPin® molecule significantly increased the release of IFN-γ, granzyme B, IL-2, and TNF-α, demonstrating successful reinvigoration. The monovalent DARPin® protein was less effective compared to its bivalent equivalent, demonstrating that bivalency brings an additional benefit to PD-1 blockade. Overall, we found a higher fold increase of lymphokine secretion in response to the PD-1 blockade by tumor-derived T-cells; however, the absolute amounts were significantly lower compared to the release from ascites-derived T-cells. Our results demonstrate that PD-1 blockade can only partially reinvigorate functionally suppressed T-cells from EOC patients. This warrants further investigation preferably in combination with other therapeutics. The study provides an early pilot proof-of-concept for the potential use of DARPin® proteins as eligible alternative scaffold proteins to block PD-1.
Collapse
|
466
|
Brain Metastases from Ovarian Cancer: Current Evidence in Diagnosis, Treatment, and Prognosis. Cancers (Basel) 2020; 12:cancers12082156. [PMID: 32759682 PMCID: PMC7464214 DOI: 10.3390/cancers12082156] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 07/28/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
With this review, we provide the state of the art concerning brain metastases (BMs) from ovarian cancer (OC), a rare condition. Clinical, pathological, and molecular features, treatment options, and future perspectives are comprehensively discussed. Overall, a diagnosis of high-grade serous OC and an advanced disease stage are common features among patients who develop brain metastases. BRCA1 and BRCA2 gene mutations, as well as the expression of androgen receptors in the primary tumor, are emerging risk and prognostic factors which could allow one to identify categories of patients at greater risk of BMs, who could benefit from a tailored follow-up. Based on present data, a multidisciplinary approach combining surgery, radiotherapy, and chemotherapy seem to be the best approach for patients with good performance status, although the median overall survival (<1 year) remains largely disappointing. Hopefully, novel therapeutic avenues are being explored, like PARP inhibitors and immunotherapy, based on our improved knowledge regarding tumor biology, but further investigation is warranted.
Collapse
|
467
|
McMullen M, Madariaga A, Lheureux S. New approaches for targeting platinum-resistant ovarian cancer. Semin Cancer Biol 2020; 77:167-181. [DOI: 10.1016/j.semcancer.2020.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/15/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022]
|
468
|
Ngoi NY, Heong V, Ow S, Chay WY, Kim HS, Choi CH, Goss G, Goh JC, Tai BC, Lim DG, Kaliaperumal N, Au VB, Connolly JE, Kim JW, Friedlander M, Kim K, Tan DS. A multicenter phase II randomized trial of durvalumab (MEDI-4736) versus physician's choice chemotherapy in recurrent ovarian clear cell adenocarcinoma (MOCCA). Int J Gynecol Cancer 2020; 30:1239-1242. [PMID: 32591370 PMCID: PMC7418587 DOI: 10.1136/ijgc-2020-001604] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND The optimal treatment of recurrent ovarian clear cell carcinoma remains unknown. There is increasing rationale to support the role of immune checkpoint inhibitors targeting the programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) axis in ovarian clear cell carcinoma. PRIMARY OBJECTIVE To evaluate the efficacy of durvalumab (MEDI-4736) compared with standard chemotherapy in patients with recurrent ovarian clear cell carcinoma. STUDY HYPOTHESIS Patients with recurrent ovarian clear cell carcinoma treated with durvalumab will have improved progression-free survival compared with those treated with chemotherapy of physician's choice. TRIAL DESIGN The MOCCA study is a multicenter, open-label, randomized phase II trial in patients with recurrent ovarian clear cell carcinoma, which recruited from eight sites across Gynecologic Cancer Group Singapore (GCGS), Korean Gynecologic-Oncology Group (KGOG), and Australia New Zealand Gynecological Oncology Group (ANZGOG). Enrolled patients were randomized in a 2:1 ratio to receive durvalumab or physician's choice of chemotherapy until disease progression, intolerable toxicity, or withdrawal of patient consent. MAJOR INCLUSION/EXCLUSION CRITERIA Eligible patients required histologically documented diagnosis of recurrent ovarian clear cell carcinoma, as evidenced by WT1 negativity. All patients must have been of Eastern Cooperative Oncology Group (ECOG) performance status 2 or better, and have had previous treatment with, and progressed or recurred after prior platinum-based chemotherapy. No more than four prior lines of treatment were allowed and prior immune checkpoint inhibitor treatment was not permitted. PRIMARY ENDPOINTS The primary endpoint was the median progression-free survival following treatment with durvalumab, compared with physician's choice of chemotherapy. Progression-free survival was defined as the time from the first day of treatment to the first observation of disease progression, or death due to any cause, or last follow-up. SAMPLE SIZE The target sample size was 46 patients. ESTIMATED DATES FOR COMPLETING ACCRUAL AND PRESENTING RESULTS Accrual has been completed and results are expected to be presented by mid-2021. TRIAL REGISTRATION Clinicaltrials.gov: NCT03405454.
Collapse
Affiliation(s)
- Natalie Yl Ngoi
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Valerie Heong
- Department of Medical Oncology, Tan Tock Seng Hospital, Singapore
| | - Samuel Ow
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Wen Yee Chay
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Hee Seung Kim
- Department of Obstetrics and Gynecology, Seoul National University, Seoul, Republic of Korea
| | - Chel Hun Choi
- Department of Obstetrics and Gynecology, Samsung Medical Center, Seoul, Republic of Korea
| | | | - Jeffrey C Goh
- Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
- The University of Queensland, Saint Lucia, Queensland, Australia
| | - Bee Choo Tai
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Diana Gz Lim
- Department of Pathology, National University Hospital, Singapore
| | - Nivashini Kaliaperumal
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - Veonice B Au
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
| | - John E Connolly
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore
- Institute of Biomedical Studies, Baylor University, Waco, Texas, USA
| | - Jae-Weon Kim
- Department of Obstetrics and Gynecology, Seoul National University, Seoul, Republic of Korea
| | - Michael Friedlander
- The Prince of Wales Hospital, Randwick, New South Wales, Australia
- Prince of Wales Clinical School University of New South Wales, Randwick, New South Wales, Australia
| | - Kidong Kim
- Seoul National University Bundang Hospital, Seoul, Republic of Korea
| | - David Sp Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Cancer Science Institute Singapore, Singapore
| |
Collapse
|
469
|
Wang Q, Peng H, Qi X, Wu M, Zhao X. Targeted therapies in gynecological cancers: a comprehensive review of clinical evidence. Signal Transduct Target Ther 2020; 5:137. [PMID: 32728057 PMCID: PMC7391668 DOI: 10.1038/s41392-020-0199-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/12/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
Advanced and recurrent gynecological cancers are associated with poor prognosis and lack of effective treatment. The developments of the molecular mechanisms on cancer progression provide insight into novel targeted therapies, which are emerging as groundbreaking and promising cancer treatment strategies. In gynecologic malignancies, potential therapeutic targeted agents include antiangiogenic agents, poly (ADP-ribose) polymerase (PARP) inhibitors, tumor-intrinsic signaling pathway inhibitors, selective estrogen receptor downregulators, and immune checkpoint inhibitors. In this article, we provide a comprehensive review of the clinical evidence of targeted agents in gynecological cancers and discuss the future implication.
Collapse
Affiliation(s)
- Qiao Wang
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Hongling Peng
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Xiaorong Qi
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Min Wu
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND, 58203, USA
| | - Xia Zhao
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|
470
|
Li H, Zhou X, Zhang D, Wang G, Cheng X, Xu C, Yao B, Pang L, Chen J. Early Onset Immune-Related Adverse Event to Identify Pseudo-Progression in a Patient With Ovarian Cancer Treated With Nivolumab: A Case Report and Review of the Literature. Front Med (Lausanne) 2020; 7:366. [PMID: 32850889 PMCID: PMC7399050 DOI: 10.3389/fmed.2020.00366] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 06/15/2020] [Indexed: 01/17/2023] Open
Abstract
Background: Immune checkpoint inhibitors (ICIs) have shown clinical benefit in many advanced tumors, however, pseudo-progression is a noted phenomenon of ICIs characterized by radiologic enlargement of the tumor burden, followed by regression. How to differentiate pseudo-progression from progression remains a critical clinical issue. Recent studies have demonstrated the association between immune-related adverse events (irAEs) and efficacy of ICIs. Here we demonstrated an ovarian cancer patient treated with nivolumab in whom the early-onset irAE was identified to differentiate pseudo-progression from progression. Case presentation: Here we present the case of a 47-years-old woman with histopathologically confirmed diagnosis of metastatic ovarian cystadenocarcinoma. Immunohistochemical examination showed 10% of tumor cells to express the programmed cell death receptor ligand 1 (PD-L1) and a 381-gene panel sequencing in a College of American Pathologists (CAP) certificated lab revealed a moderate mutational tumor burden with 5.7 Mutants/Mb. The patient received nivolumab 100 mg every 2 weeks as a fourth line treatment. Within the first 2 months, the tumor volume increased by 23.6%. However, the patient experienced an elevation of Alanine aminotransferase (ALT) and Aspartate aminotransmerase (AST), which was diagnosed as the immune-related hepatitis. Thus, the treatment continued and afterwards, the patient reached a partial response (PR) to nivolumab and the progression-free survival was 9 months. Conclusion: To our knowledge, this is the first case describing early-onset immune-related adverse events to identify pseudo-progression in a patient with ovarian cancer treated with nivolumab, and PD-L1 expression level may be a predictive biomarker in the immunotherapy of ovarian cancer.
Collapse
Affiliation(s)
- Hui Li
- Department of Radiotherapy and Chemotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Xin Zhou
- Department of Radiotherapy and Chemotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Ding Zhang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Guoqiang Wang
- The Medical Department, 3D Medicines Inc., Shanghai, China
| | - Xiaochun Cheng
- Department of Radiotherapy and Chemotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Caihong Xu
- Department of Radiotherapy and Chemotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Bin Yao
- Department of Radiotherapy and Chemotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Linrong Pang
- Department of Radiotherapy and Chemotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Jun Chen
- Department of Radiotherapy and Chemotherapy, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| |
Collapse
|
471
|
Nanki Y, Chiyoda T, Hirasawa A, Ookubo A, Itoh M, Ueno M, Akahane T, Kameyama K, Yamagami W, Kataoka F, Aoki D. Patient-derived ovarian cancer organoids capture the genomic profiles of primary tumours applicable for drug sensitivity and resistance testing. Sci Rep 2020; 10:12581. [PMID: 32724113 PMCID: PMC7387538 DOI: 10.1038/s41598-020-69488-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 07/13/2020] [Indexed: 12/19/2022] Open
Abstract
The use of primary patient-derived organoids for drug sensitivity and resistance testing could play an important role in precision cancer medicine. We developed expandable ovarian cancer organoids in < 3 weeks; these organoids captured the characteristics of histological cancer subtypes and replicated the mutational landscape of the primary tumours. Seven pairs of organoids (3 high-grade serous, 1 clear cell, 3 endometrioid) and original tumours shared 59.5% (36.1–73.1%) of the variants identified. Copy number variations were also similar among organoids and primary tumours. The organoid that harboured the BRCA1 pathogenic variant (p.L63*) showed a higher sensitivity to PARP inhibitor, olaparib, as well as to platinum drugs compared to the other organoids, whereas an organoid derived from clear cell ovarian cancer was resistant to conventional drugs for ovarian cancer, namely platinum drugs, paclitaxel, and olaparib. The overall success rate of primary organoid culture, including those of various histological subtypes, was 80% (28/35). Our data show that patient-derived organoids are suitable physiological ex vivo cancer models that can be used to screen effective personalised ovarian cancer drugs.
Collapse
Affiliation(s)
- Yoshiko Nanki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan.,JSR-Keio University Medical and Chemical Innovation Center (JKiC), Keio University School of Medicine, Tokyo, Japan
| | - Tatsuyuki Chiyoda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan. .,JSR-Keio University Medical and Chemical Innovation Center (JKiC), Keio University School of Medicine, Tokyo, Japan.
| | - Akira Hirasawa
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan. .,JSR-Keio University Medical and Chemical Innovation Center (JKiC), Keio University School of Medicine, Tokyo, Japan. .,Department of Clinical Genomic Medicine, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
| | - Aki Ookubo
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corp., Tokyo, Japan
| | - Manabu Itoh
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corp., Tokyo, Japan
| | - Masaru Ueno
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corp., Tokyo, Japan
| | - Tomoko Akahane
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), Keio University School of Medicine, Tokyo, Japan.,Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Kaori Kameyama
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan.,Department of Pathology, Showa University Northern Yokohama Hospital, Yokohama, Japan
| | - Wataru Yamagami
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Fumio Kataoka
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan.,Department of Obstetrics and Gynecology, International University of Health and Welfare School of Medicine, Narita, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
472
|
Zhao B, Zhao H, Zhao J. Efficacy of PD-1/PD-L1 blockade monotherapy in clinical trials. Ther Adv Med Oncol 2020; 12:1758835920937612. [PMID: 32728392 PMCID: PMC7366397 DOI: 10.1177/1758835920937612] [Citation(s) in RCA: 108] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 06/05/2020] [Indexed: 12/23/2022] Open
Abstract
Background: Inhibitors targeting programmed cell death 1 (PD-1) and programmed
death-ligand 1 (PD-L1) have unprecedented effects in cancer treatment.
However, the objective response rates (ORRs), progression-free survival
(PFS), and overall survival (OS) of PD-1/PD-L1 blockade monotherapy have not
been systematically evaluated. Methods: We searched Embase, PubMed, and Cochrane database from inception to July 2019
for prospective clinical trials on single-agent PD-1/PD-L1 antibodies
(avelumab, atezolizumab, durvalumab, cemiplimab, pembrolizumab, and
nivolumab) with information regarding ORR, PFS, and OS. Results: Totally, 28,304 patients from 160 perspective trials were included. Overall,
4747 responses occurred in 22,165 patients treated with PD-1/PD-L1
monotherapy [ORR, 20.21%; 95% confidence interval (CI), 18.34–22.15%].
Compared with conventional therapy, PD-1/PD-L1 blockade immunotherapy was
associated with more tumor responses (odds ratio, 1.98; 95% CI, 1.52–2.57)
and better OS [hazard ratio (HR), 0.75; 95% CI, 0.67–0.83]. The ORRs varied
significantly across cancer types and PD-L1 expression status. Line of
treatment, clinical phase and drug target also impacted the response rates
in some tumors. A total of 2313 of 9494 PD-L1 positive patients (ORR,
24.39%; 95% CI, 22.29–26.54%) and 456 of 4215 PD-L1 negative patients (ORR,
10.34%; 95% CI, 8.67–12.14%) achieved responses. For PD-L1 negative
patients, the ORR (odds ratio, 0.92; 95% CI, 0.70–1.20) and PFS (HR, 1.15;
95% CI, 0.87–1.51) associated with immunotherapy and conventional treatment
were similar. However, PD-1/PD-L1 blockade monotherapy decreased the risk of
death in both PD-L1 positive (HR, 0.66; 95% CI, 0.60–0.72) and PD-L1
negative (HR, 0.86; 95% CI, 0.74–0.99) patients compared with conventional
therapy. Conclusion: The efficacies associated with PD-1/PD-L1 monotherapy vary significantly
across cancer types and PD-L1 expression. This comprehensive summary of
clinical benefit from immunotherapy in cancer patients provides an important
guide for clinicians.
Collapse
Affiliation(s)
- Bin Zhao
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, 109 Xueyuan West Rd, Wenzhou, 325035, China
| | - Hong Zhao
- The Cancer Center of the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Jiaxin Zhao
- The Second Affiliated Hospital & Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
473
|
Kurnit KC, Reid P, Moroney JW, Fleming GF. Immune checkpoint inhibitors in women with gynecologic cancers: Practical considerations. Gynecol Oncol 2020; 158:531-537. [PMID: 32641238 DOI: 10.1016/j.ygyno.2020.06.499] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 06/21/2020] [Indexed: 12/17/2022]
Abstract
Immune checkpoint inhibitors are an exciting new class of cancer therapeutics. Recently, a PD-1 inhibitor has been approved by the Food and Drug Administration for several indications that are relevant to patients with gynecologic malignancies. In this review, we explore the clinical considerations for the use of checkpoint inhibitor therapy in this population. Specifically, we will discuss the approved indications, recommended dosing, clinical monitoring while on treatment, common adverse events, and treatment of adverse events should they arise. Additionally, we will review mechanisms of resistance and other challenges associated with the use of checkpoint inhibitors. We will conclude with a discussion of possible future directions for immunotherapy in women with gynecologic cancers.
Collapse
Affiliation(s)
- Katherine C Kurnit
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, The University of Chicago, Chicago, IL, United States.
| | - Pankti Reid
- Department of Medicine, Section of Rheumatology, The University of Chicago, Chicago, IL, United States
| | - John W Moroney
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, The University of Chicago, Chicago, IL, United States
| | - Gini F Fleming
- Department of Medicine, Section of Hematology-Oncology, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
474
|
Martinez A, Delord JP, Ayyoub M, Devaud C. Preclinical and Clinical Immunotherapeutic Strategies in Epithelial Ovarian Cancer. Cancers (Basel) 2020; 12:E1761. [PMID: 32630708 PMCID: PMC7409311 DOI: 10.3390/cancers12071761] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 06/24/2020] [Accepted: 06/26/2020] [Indexed: 12/25/2022] Open
Abstract
In the past 20 years, the immune system has increasingly been recognized as a major player in tumor cell control, leading to considerable advances in cancer treatment. While promising with regards to melanoma, renal cancer and non-small cell lung cancer, immunotherapy provides, for the time being, limited success in other cancers, including ovarian cancer, potentially due to insufficient immunogenicity or to a particularly immunosuppressive microenvironment. In this review, we provide a global description of the immune context of ovarian cancer, in particular epithelial ovarian cancer (EOC). We describe the adaptive and innate components involved in the EOC immune response, including infiltrating tumor-specific T lymphocytes, B lymphocytes, and natural killer and myeloid cells. In addition, we highlight the rationale behind the use of EOC preclinical mouse models to assess resistance to immunotherapy, and we summarize the main preclinical studies that yielded anti-EOC immunotherapeutic strategies. Finally, we focus on major published or ongoing immunotherapy clinical trials concerning EOC.
Collapse
Affiliation(s)
- Alejandra Martinez
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé Et de la Recherche Médicale (INSERM) Unité 1037, 31037 Toulouse, France; (A.M.); (J.-P.D.); (M.A.)
- Department of Surgery, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse (IUCT), 31037 Toulouse, France
| | - Jean-Pierre Delord
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé Et de la Recherche Médicale (INSERM) Unité 1037, 31037 Toulouse, France; (A.M.); (J.-P.D.); (M.A.)
- Department of Medical Oncology, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 31037 Toulouse, France
- Université Toulouse III Paul Sabatier, 31037 Toulouse, France
| | - Maha Ayyoub
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé Et de la Recherche Médicale (INSERM) Unité 1037, 31037 Toulouse, France; (A.M.); (J.-P.D.); (M.A.)
- Université Toulouse III Paul Sabatier, 31037 Toulouse, France
- Immune Monitoring Core Facility, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 31037 Toulouse, France
| | - Christel Devaud
- Cancer Research Center of Toulouse (CRCT), Institut National de la Santé Et de la Recherche Médicale (INSERM) Unité 1037, 31037 Toulouse, France; (A.M.); (J.-P.D.); (M.A.)
- Immune Monitoring Core Facility, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, 31037 Toulouse, France
| |
Collapse
|
475
|
Hastings V, McEachron J, Kanis MJ. Cutaneous metastasis of PD-L1 positive ovarian carcinoma. Gynecol Oncol Rep 2020; 33:100607. [PMID: 32671171 PMCID: PMC7339030 DOI: 10.1016/j.gore.2020.100607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/17/2020] [Accepted: 06/27/2020] [Indexed: 12/02/2022] Open
Abstract
Ovarian cancer (OC) metastasizes cutaneous in up to 6% of cases. Cutaneous metastasis to the vulva is rare; with only one prior report in the literature. Resection of cutaneous metastasis offers symptom relief as well as a potential survival benefit. Molecular tumor profiling should be employed to expand therapy options.
Collapse
Affiliation(s)
- Victoria Hastings
- Division of Gynecologic Oncology, New York Presbyterian - Brooklyn Methodist Hospital, Brooklyn, NY, United States
| | - Jennifer McEachron
- Division of Gynecologic Oncology, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Marguax J Kanis
- Division of Gynecologic Oncology, New York Presbyterian - Brooklyn Methodist Hospital, Brooklyn, NY, United States
| |
Collapse
|
476
|
Murakami K, Kotani Y, Nakai H, Matsumura N. Endometriosis-Associated Ovarian Cancer: The Origin and Targeted Therapy. Cancers (Basel) 2020; 12:cancers12061676. [PMID: 32599890 PMCID: PMC7352633 DOI: 10.3390/cancers12061676] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
Endometrial cysts (ECs) are thought to be the origin of endometriosis-associated ovarian cancer (EAOC). A hypothesis that the oxidative stress of iron in cysts causes “malignant transformation of ECs” has been proposed, but this has not been verified. Several population-based studies showed that endometriosis was a risk factor but did not reflect the “malignant transformation of ECs”. A review showed that most patients were diagnosed with EAOC early in monitoring following detection of ECs, and that these cases might have been cancer from the start. Epidemiologically, EAOC was reduced by hysterectomy rather than by cystectomy of ECs. Gene mutation analyses identified oncogenic mutations in endometriosis and normal endometrium and revealed that the same mutations were present at different endometriotic lesions. It was also shown that most of the gene mutations found in endometriosis occurred in normal endometrium. Taking together, EAOC might be caused by eutopic endometrial glandular epithelial cells with oncogenic mutations that have undergone menstrual blood reflux and engrafted in the ovary, rather than by low-risk ECs acquiring oncogenic mutations and becoming malignant. This review discusses the mechanisms of EAOC development and targeted therapy based on genetic variation in EAOC with a focus on eutopic endometrium.
Collapse
|
477
|
Ogasawara A, Sato S, Hasegawa K. Current and future strategies for treatment of ovarian clear cell carcinoma. J Obstet Gynaecol Res 2020; 46:1678-1689. [PMID: 32578333 DOI: 10.1111/jog.14350] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 05/23/2020] [Indexed: 01/04/2023]
Abstract
Ovarian clear cell carcinoma (OCCC) is one of the five histological types of epithelial ovarian cancer (EOC). OCCC comprises 23% of all EOC cases in Japan, whereas the rate of OCCC in North America and Europe is much lower. OCCC is generally categorized as a rare gynecologic malignancy, and there is limited evidence for specific treatment. The clinical basis for treatment of OCCC is mostly based on retrospective studies, many of which were performed in Japan. Until recently, most randomized clinical trials for EOC have included OCCC; therefore, current treatment for OCCC is basically the same as that for other histologic types of EOC. However, the clinical characteristics of OCCC differ from those of high-grade serous carcinoma, particularly for chemosensitivity, and there is a need to develop new treatment for OCCC. The molecular background of OCCC has unique features: tumors are usually negative for p53 mutations and positive for ARID1A and/or PIK3CA mutations, whereas p53 mutations are common in high-grade serous or endometrioid carcinomas. These features may help in development of new treatment for OCCC. In this review, we described the current evidence for treatment of OCCC, including surgery, radiotherapy, chemotherapy, molecular targeted therapy and immunotherapy, and we discuss ongoing clinical trials and preclinical studies of new treatment approaches for OCCC.
Collapse
Affiliation(s)
- Aiko Ogasawara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Sho Sato
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Saitama, Japan
| |
Collapse
|
478
|
Liu YL, Selenica P, Zhou Q, Iasonos A, Callahan M, Feit NZ, Boland J, Vazquez-Garcia I, Mandelker D, Zehir A, Burger RA, Powell DJ, Friedman C, Cadoo K, Grisham R, Konner JA, O'Cearbhaill RE, Aghajanian C, Reis-Filho JS, Weigelt B, Zamarin D. BRCA Mutations, Homologous DNA Repair Deficiency, Tumor Mutational Burden, and Response to Immune Checkpoint Inhibition in Recurrent Ovarian Cancer. JCO Precis Oncol 2020; 4:2000069. [PMID: 32923884 DOI: 10.1200/po.20.00069] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2020] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Homologous DNA repair-deficient (HRD) ovarian cancers (OCs), including those with BRCA1/2 mutations, have higher levels of genetic instability, potentially resulting in higher immunogenicity, and have been suggested to respond better to immune checkpoint inhibitors (ICIs) than homologous DNA repair-proficient OCs. However, clinical evidence is lacking. The study aimed to evaluate the associations between BRCA1/2 mutations, HRD, and other genomic parameters and response to ICIs and survival in OC. METHODS This is a single-institution retrospective analysis of women with recurrent OC treated with ICIs. BRCA1/2 mutation status and clinicopathologic variables were abstracted from the medical records. Targeted and whole-exome sequencing data available for a subset of patients were used to assess tumor mutational burden (TMB), HRD, and fraction of genome altered (FGA). ICI response was defined as lack of disease progression for ≥ 24 weeks. Associations of BRCA1/2 status and genomic alterations with progression-free survival (PFS) and overall survival (OS) were determined using Cox proportional hazards models. RESULTS Of the 143 women treated with ICIs, 134 had known BRCA1/2 mutation status. Deleterious germline or somatic BRCA1/2 mutations were present in 31 women (24%). There was no association between presence of BRCA1/2 mutations and response (P = .796) or survival. Genomic analysis in 73 women found no association between TMB (P = .344) or HRD (P = .222) and response, PFS, or OS. There were also no significant differences in somatic genetic alterations between responders and nonresponders. High FGA was associated with an improvement in PFS (P = .014) and OS (P = .01). CONCLUSION TMB, BRCA1/2 mutations, and HRD are not associated with response or survival, cautioning against their use as selection criteria for ICI in recurrent OC. FGA should be investigated further as a biomarker of response to immunotherapy in OC.
Collapse
Affiliation(s)
- Ying L Liu
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Qin Zhou
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexia Iasonos
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Margaret Callahan
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY.,Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Noah Z Feit
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Julia Boland
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Diana Mandelker
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ahmet Zehir
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Robert A Burger
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA
| | - Daniel J Powell
- Department of Pathology, University of Pennsylvania, Philadelphia, PA
| | - Claire Friedman
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Karen Cadoo
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Rachel Grisham
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Jason A Konner
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Roisin E O'Cearbhaill
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Carol Aghajanian
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Britta Weigelt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Dmitriy Zamarin
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY.,Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
479
|
Santangelo G, Caruso G, Palaia I, Tomao F, Perniola G, Di Donato V, Fischetti M, Muzii L, Benedetti Panici P. The emerging role of precision medicine in the treatment of ovarian cancer. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020. [DOI: 10.1080/23808993.2020.1777854] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Giusi Santangelo
- Department of Maternal and Child Health and Urological Sciences, Sapienza University, Rome, Italy
| | - Giuseppe Caruso
- Department of Maternal and Child Health and Urological Sciences, Sapienza University, Rome, Italy
| | - Innocenza Palaia
- Department of Maternal and Child Health and Urological Sciences, Sapienza University, Rome, Italy
| | - Federica Tomao
- Department of Maternal and Child Health and Urological Sciences, Sapienza University, Rome, Italy
| | - Giorgia Perniola
- Department of Maternal and Child Health and Urological Sciences, Sapienza University, Rome, Italy
| | - Violante Di Donato
- Department of Maternal and Child Health and Urological Sciences, Sapienza University, Rome, Italy
| | - Margherita Fischetti
- Department of Maternal and Child Health and Urological Sciences, Sapienza University, Rome, Italy
| | - Ludovico Muzii
- Department of Maternal and Child Health and Urological Sciences, Sapienza University, Rome, Italy
| | | |
Collapse
|
480
|
Terzic J, Seipel A, Dubuisson J, Tille JC, Tsantoulis P, Addeo A, Labidi-Galy SI. Sustained response to pembrolizumab without prior chemotherapy in high-grade serous ovarian carcinoma with CSMD3 mutation. Gynecol Oncol Rep 2020; 33:100600. [PMID: 32613071 PMCID: PMC7322246 DOI: 10.1016/j.gore.2020.100600] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/06/2020] [Accepted: 06/09/2020] [Indexed: 02/07/2023] Open
Abstract
•Metastatic CSMD3 mutated HGSOC showed objective and sustained response to pembrolizumab.•The tumor was massively infiltrated by CD8+ T cells while PD-L1 TPS was at 10%.•CSMD3 mutated HGSOC showed up-regulation of CCL5 and CXCL9.
Collapse
Affiliation(s)
- Julie Terzic
- Department of Oncology, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - Amanda Seipel
- Department of Diagnosis, Division of Clinical Pathology, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - Jean Dubuisson
- Department of Woman, Child and Adolescent, Division of Gynecology, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - Jean-Christophe Tille
- Department of Diagnosis, Division of Clinical Pathology, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - Petros Tsantoulis
- Department of Oncology, Hôpitaux Universitaires de Genève, Genève, Switzerland.,Department of Medicine, Faculty of Medicine, University of Geneva, Genève, Switzerland
| | - Alfredo Addeo
- Department of Oncology, Hôpitaux Universitaires de Genève, Genève, Switzerland
| | - S Intidhar Labidi-Galy
- Department of Oncology, Hôpitaux Universitaires de Genève, Genève, Switzerland.,Department of Medicine, Faculty of Medicine, University of Geneva, Genève, Switzerland
| |
Collapse
|
481
|
Boland JL, Zhou Q, Iasonos AE, O'Cearbhaill RE, Konner J, Callahan M, Friedman C, Aghajanian C, Sabbatini P, Zamarin D, Cadoo KA. Utility of serum CA-125 monitoring in patients with ovarian cancer undergoing immune checkpoint inhibitor therapy. Gynecol Oncol 2020; 158:303-308. [PMID: 32507515 DOI: 10.1016/j.ygyno.2020.04.710] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 04/30/2020] [Indexed: 11/25/2022]
Abstract
OBJECTIVE This study aimed to evaluate the utility of serum cancer antigen-125 (CA-125) levels to monitor patients with epithelial ovarian cancer (EOC) undergoing immune checkpoint inhibitor (ICI) therapy. METHOD This was a single-center retrospective review of all patients with EOC who were treated with ICI therapy from January 2013 to May 2017. This study compared the percentage change in baseline CA-125 in patients who had clinical benefit, defined as complete response, partial response, or stable disease by RECIST 1.1, with duration ≥24 weeks, versus those who did not. The groups were compared by Wilcoxon rank-sum test. RESULTS Fifty-nine (66%) of 89 patients who underwent ICI therapy had CA-125 data at baseline and during treatment. Of those who derived clinical benefit, 11/15 (73%) experienced an increase in CA-125 from baseline to end of treatment. Of those who did not derive clinical benefit, 36/44 (82%) experienced a CA-125 increase (p = 0.48). The average % increase from baseline to within 12 weeks of treatment initiation for patients with and without clinical benefit was 34% and 195%, respectively (p = 0.008). CONCLUSION Our analysis demonstrates a statistically significant difference in the magnitude of increase in CA-125 levels within the first 12 weeks of treatment between patients who achieved clinical benefit and those who did not. However, both groups of patients were equally likely to experience an increase in CA-125 within 12 weeks. These findings suggest that physicians should apply caution when using early CA-125 data to guide treatment decisions for patients with EOC undergoing ICI therapy.
Collapse
Affiliation(s)
- Julia L Boland
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Qin Zhou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alexia E Iasonos
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roisin E O'Cearbhaill
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weil Cornell Medical College, New York, NY, USA
| | - Jason Konner
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weil Cornell Medical College, New York, NY, USA
| | - Margaret Callahan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weil Cornell Medical College, New York, NY, USA
| | - Claire Friedman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weil Cornell Medical College, New York, NY, USA
| | - Carol Aghajanian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weil Cornell Medical College, New York, NY, USA
| | - Paul Sabbatini
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weil Cornell Medical College, New York, NY, USA
| | - Dmitriy Zamarin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weil Cornell Medical College, New York, NY, USA
| | - Karen A Cadoo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weil Cornell Medical College, New York, NY, USA.
| |
Collapse
|
482
|
Bogani G, Lopez S, Mantiero M, Ducceschi M, Bosio S, Ruisi S, Sarpietro G, Guerrisi R, Brusadelli C, Dell'Acqua A, Di Donato V, Raspagliesi F. Immunotherapy for platinum-resistant ovarian cancer. Gynecol Oncol 2020; 158:484-488. [PMID: 32518015 DOI: 10.1016/j.ygyno.2020.05.681] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 05/28/2020] [Indexed: 12/11/2022]
Abstract
Ovarian cancer is characterized by a high mortality on incidence ratio. Although the majority of patients achieve complete response after primary treatment, approximately 65-80% of patients recur with the first 5 years. Platinum-free interval is one of the main prognostic factors. Patients recurring with 6 months within the end of platinum-based chemotherapy are characterized by poor prognosis. To date no effective treatment modality are identified for those patients. The mainstay of treatment for platinum-resistant ovarian cancer is single agent chemotherapy. Other treatment modalities have tested in this setting with discouraging results. Growing evidence suggested that immunotherapy would improve outcomes of patients with various types of solid tumors including melanoma, non-small cell lung cancer as well as uterine malignancies. Here, we reviewed current evidence on the adoption of immunotherapy in platinum-resistant ovarian cancer. To date no mature evidence supports the routine adoption of immunotherapy in ovarian cancer patients. Further strategies have to be explored.
Collapse
Affiliation(s)
- Giorgio Bogani
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Italy
| | - Salvatore Lopez
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Italy
| | - Mara Mantiero
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Italy.
| | | | - Sara Bosio
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Italy; San Paolo Hospital, Università degli Studi di Milano, 20142 Milan, Italy
| | - Simona Ruisi
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Italy; San Paolo Hospital, Università degli Studi di Milano, 20142 Milan, Italy
| | - Giuseppe Sarpietro
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Italy; Department of General Surgery and Medical Surgical Specialties, Gynecological Clinic University of Catania, Via S. Sofia 78, 95124 Catania, Italy
| | - Rocco Guerrisi
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Italy; Obstetrics and Gynecology Department of the University of Insubria, Varese, Italy
| | - Claudia Brusadelli
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Italy; Obstetrics and Gynecology Department of the University of Insubria, Varese, Italy
| | - Andrea Dell'Acqua
- Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Italy; Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Violante Di Donato
- Department of Maternal and Child Health and Urological Sciences, "Sapienza" University of Rome, Rome, Italy
| | | |
Collapse
|
483
|
Hamilton E, O'Malley DM, O'Cearbhaill R, Cristea M, Fleming GF, Tariq B, Fong A, French D, Rossi M, Brickman D, Moore K. Tamrintamab pamozirine (SC-003) in patients with platinum-resistant/refractory ovarian cancer: Findings of a phase 1 study. Gynecol Oncol 2020; 158:640-645. [PMID: 32513564 DOI: 10.1016/j.ygyno.2020.05.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 05/23/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Epithelial ovarian carcinoma (EOC) is diagnosed at advanced stage in the majority of women and, despite being initially chemosensitive, eventually recurs and develops resistance to known therapies. SC-003 is a pyrrolobenzodiazepine-based antibody-drug conjugate targeting dipeptidase 3 (DPEP3), a membrane-bound dipeptidase associated with tumor-initiating cells in patient-derived EOC xenograft models. This first-in-human phase 1a/1b study evaluated the safety/tolerability, pharmacokinetics, and preliminary antitumor activity of SC-003 alone or in combination with budigalimab (formerly ABBV-181), an antibody targeting PD-1, in patients with platinum-resistant/refractory EOC (NCT02539719). METHODS Patients received SC-003 at 1 of 6 dose levels (0.025-0.4 mg/kg) every 3 weeks (Q3W), utilizing a standard 3 + 3 design (dose-limiting toxicity [DLT] period: 21 days). Patients with DPEP3-positive tumors were enrolled in the dose-expansion phase of the study and treated with SC-003 monotherapy or in combination with budigalimab. RESULTS Seventy-four patients (n = 29, dose escalation; n = 45, dose expansion; n = 3 budigalimab combination) were enrolled and received ≥1 dose of study drug. One DLT occurred (grade 3 ileus) but was considered unrelated to study drug. The MTD for the Q3W schedule was 0.3 mg/kg and the SC-003 doses selected for the dose-expansion phase of the study were 0.3 mg/kg and 0.2 mg/kg. The most common treatment-emergent adverse events were fatigue, nausea, decreased appetite, pleural effusion, abdominal pain, and peripheral edema. The overall response rate was low (4%), and responses were not durable. Post-hoc examination of antitumor activity suggested a higher response rate in patients with higher DPEP3 expression. CONCLUSIONS SC-003 lacked the requisite safety profile and antitumor activity to warrant further development.
Collapse
Affiliation(s)
- Erika Hamilton
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA.
| | - David M O'Malley
- The Ohio State University Wexner Medical Center, James CCC, Columbus, OH, USA.
| | - Roisin O'Cearbhaill
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, NY, USA.
| | | | | | | | | | | | | | | | - Kathleen Moore
- Oklahoma Cancer Center/Sarah Cannon Research Institute, University of Oklahoma, Oklahoma City, OK, USA.
| |
Collapse
|
484
|
Mahmood RD, Morgan RD, Edmondson RJ, Clamp AR, Jayson GC. First-Line Management of Advanced High-Grade Serous Ovarian Cancer. Curr Oncol Rep 2020; 22:64. [PMID: 32494876 PMCID: PMC7270049 DOI: 10.1007/s11912-020-00933-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Epithelial ovarian cancer is a disease that encompasses a number of histologically and molecularly distinct entities; the most prevalent subtype being high-grade serous (HGS) carcinoma. Standard first-line treatment of advanced HGS carcinoma includes cytoreductive surgery plus intravenous paclitaxel/platinum-based chemotherapy. Despite excellent responses to initial treatment, the majority of patients develop recurrent disease within 3 years. The introduction of the vascular endothelial growth factor (VEGF) inhibitor, bevacizumab, and poly(ADP-ribose) polymerase (PARP) inhibitors into first-line management has changed the outlook for this lethal disease. In this review, we summarise the most recent clinical trials that determine current primary therapy of advanced HGS carcinoma and the ongoing trials that aim to change management in the future. RECENT FINDINGS Recent phase III clinical trials have shown that delayed primary surgery after completing neo-adjuvant chemotherapy is non-inferior to immediate primary surgery, but could provide a survival benefit in FIGO (International Federation of Gynecology and Obstetrics) stage IV disease. The use of weekly intravenous chemotherapy regimens has not been proven to be more effective than standard 3-weekly regimens in Western patient populations, and the use of intraperitoneal chemotherapy remains controversial in the first-line setting. In contrast, newer systemic anti-cancer therapies targeting angiogenesis and/or HR-deficient tumours have been successfully incorporated into front-line therapeutic regimens to treat HGS carcinoma. Recent results from randomised trials investigating the use of PARP inhibitors as monotherapy and in combination with the anti-angiogenic agent, bevacizumab, have demonstrated highly impressive efficacy when combined with traditional first-line multi-modality therapy. Management of HGS carcinoma is evolving, but further work is still required to optimise and integrate tumour and plasma biomarkers to exploit the potential of these highly efficacious targeted agents.
Collapse
Affiliation(s)
- Reem D Mahmood
- Department of Medical Oncology, Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| | - Robert D Morgan
- Department of Medical Oncology, Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Richard J Edmondson
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Department of Gynaecological Oncology Surgery, Saint Mary's Hospital, Manchester University NHS Foundation Trust, Oxford Road, Manchester, UK
| | - Andrew R Clamp
- Department of Medical Oncology, Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK
| | - Gordon C Jayson
- Department of Medical Oncology, Christie NHS Foundation Trust, Wilmslow Road, Withington, Manchester, M20 4BX, UK.
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
485
|
Kverneland AH, Pedersen M, Westergaard MCW, Nielsen M, Borch TH, Olsen LR, Aasbjerg G, Santegoets SJ, van der Burg SH, Milne K, Nelson BH, Met Ö, Donia M, Svane IM. Adoptive cell therapy in combination with checkpoint inhibitors in ovarian cancer. Oncotarget 2020; 11:2092-2105. [PMID: 32547707 PMCID: PMC7275789 DOI: 10.18632/oncotarget.27604] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Immune therapy is a promising field within oncology but has been unsuccessful in ovarian cancer (OC). Still, there is rationale and evidence supporting immune therapy in OC. We investigated the potential for adoptive cell therapy (ACT) from in vitro expanded tumor-infiltrating lymphocytes (TILs) in combination with checkpoint inhibitors (ICI) and conducted immunological testing of ex vivo expanded TILs (REP-TILs). Six patients with late-stage metastatic high-grade serous OC were treated with immune therapy consisting of ipilimumab followed by surgery to obtain TILs and infusion of REP-TILs, low-dose IL-2 and nivolumab. One patient achieved a partial response and 5 others experienced disease stabilization for up to 12 months. Analysis of the REP-TILs with flow- and mass-cytometry show primarily activated and differentiated effector memory T cells. REP-TILs showed in vitro reactivity and expression of inhibitory receptors, such as LAG-3 and PD-1. Furthermore, our data indicate that addition of ipilimumab therapy improves the T cell fold expansion during production, increase the level of CD8 T cell tumor reactivity, and favorably affect the T cell phenotype. We show that the combination of ICI and ACT is feasible and safe. With one partial response and one long-lasting SD, we demonstrated the potential of ACT in OC.
Collapse
Affiliation(s)
- Anders Handrup Kverneland
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Magnus Pedersen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | | | - Morten Nielsen
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Troels Holz Borch
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Lars Rønn Olsen
- Section for Bioinformatics, DTU Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark.,Center for Genomic Medicine, Copenhagen University Hospital, Copenhagen, Denmark
| | - Gitte Aasbjerg
- Section for Bioinformatics, DTU Health Technology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Saskia J Santegoets
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Katy Milne
- Deeley Research Centre, BC Cancer, Victoria, Canada
| | - Brad H Nelson
- Deeley Research Centre, BC Cancer, Victoria, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| | - Özcan Met
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Marco Donia
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
486
|
Zamarin D, Walderich S, Holland A, Zhou Q, Iasonos AE, Torrisi JM, Merghoub T, Chesebrough LF, Mcdonnell AS, Gallagher JM, Li Y, Hollmann TJ, Grisham RN, Erskine CL, Block MS, Knutson KL, O'Cearbhaill RE, Aghajanian C, Konner JA. Safety, immunogenicity, and clinical efficacy of durvalumab in combination with folate receptor alpha vaccine TPIV200 in patients with advanced ovarian cancer: a phase II trial. J Immunother Cancer 2020; 8:e000829. [PMID: 32503949 PMCID: PMC7279674 DOI: 10.1136/jitc-2020-000829] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2020] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) to date have demonstrated limited activity in advanced ovarian cancer (OC). Folate receptor alpha (FRα) is overexpressed in the majority of OCs and presents an attractive target for a combination immunotherapy to potentially overcome resistance to ICI in OCs. The current study sought to examine clinical and immunologic responses to TPIV200, a multiepitope FRα vaccine administered with programmed death ligand 1 (PD-L1) inhibitor durvalumab in patients with advanced platinum-resistant OC. METHODS Following Simon two-stage phase II trial design, 27 patients were enrolled. Treatment was administered in 28-day cycles (intradermal TPIV200 and granulocyte-macrophage colony-stimulating factor (GM-CSF) for 6 cycles and intravenous durvalumab for 12 cycles). Primary endpoints included overall response rate and progression-free survival at 24 weeks. Translational parameters focused on tumor microenvironment, PD-L1 and FRα expression, and peripheral vaccine-specific immune responses. RESULTS Treatment was well tolerated, with related grade 3 toxicity rate of 18.5%. Increased T cell responses to the majority of peptides were observed in all patients at 6 weeks (p<0.0001). There was one unconfirmed partial response (3.7%) and nine patients had stable disease (33.3%). Clinical benefit was not associated with baseline FRα or PD-L1 expression. One patient with prolonged clinical benefit demonstrated loss of FRα expression and upregulation of PD-L1 in a progressing lesion. Despite the low overall response rate, the median overall survival was 21 months (13.5-∞), with evidence of benefit from postimmunotherapy regimens. CONCLUSIONS Combination of TPIV200 and durvalumab was safe and elicited robust FRα-specific T cell responses in all patients. Unexpectedly durable survival in this heavily pretreated population highlights the need to investigate the impact of FRα vaccination on the OC biology post-treatment.
Collapse
MESH Headings
- Adenocarcinoma, Clear Cell/drug therapy
- Adenocarcinoma, Clear Cell/immunology
- Adenocarcinoma, Clear Cell/pathology
- Adult
- Aged
- Antibodies, Monoclonal/therapeutic use
- Antineoplastic Agents, Immunological/therapeutic use
- Biomarkers, Tumor/immunology
- Cancer Vaccines/therapeutic use
- Cystadenocarcinoma, Serous/drug therapy
- Cystadenocarcinoma, Serous/immunology
- Cystadenocarcinoma, Serous/pathology
- Drug Therapy, Combination
- Endometrial Neoplasms/drug therapy
- Endometrial Neoplasms/immunology
- Endometrial Neoplasms/pathology
- Female
- Folate Receptor 1/immunology
- Follow-Up Studies
- Gene Expression Regulation, Neoplastic
- Humans
- Middle Aged
- Non-Randomized Controlled Trials as Topic
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/pathology
- Prognosis
- Survival Rate
- Treatment Outcome
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Dmitriy Zamarin
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill-Cornell Medical College, New York, NY, United States
- Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Sven Walderich
- Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Aliya Holland
- Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Qin Zhou
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Alexia E Iasonos
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jean M Torrisi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Taha Merghoub
- Ludwig Collaborative Laboratory, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lewis F Chesebrough
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Autumn S Mcdonnell
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Jacqueline M Gallagher
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Yanyun Li
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Travis J Hollmann
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Rachel N Grisham
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill-Cornell Medical College, New York, NY, United States
| | | | - Mathew S Block
- Department of Oncology, Mayo Clinic, Rochester, MN, United States
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic, Rochester, MN, United States
| | - Roisin E O'Cearbhaill
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill-Cornell Medical College, New York, NY, United States
| | - Carol Aghajanian
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill-Cornell Medical College, New York, NY, United States
| | - Jason A Konner
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Department of Medicine, Weill-Cornell Medical College, New York, NY, United States
| |
Collapse
|
487
|
Clinical Implications of DNA Repair Defects in High-Grade Serous Ovarian Carcinomas. Cancers (Basel) 2020; 12:cancers12051315. [PMID: 32455819 PMCID: PMC7281678 DOI: 10.3390/cancers12051315] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/15/2020] [Accepted: 05/19/2020] [Indexed: 12/21/2022] Open
Abstract
Despite significant improvements in surgical and medical management, high grade serous ovarian cancer (HGSOC) still represents the deadliest gynecologic malignancy and the fifth most frequent cause of cancer-related mortality in women in the USA. Since DNA repair alterations are regarded as the “the Achille’s heel” of HGSOC, both DNA homologous recombination and DNA mismatch repair deficiencies have been explored and targeted in epithelial ovarian cancers in the latest years. In this review, we aim at focusing on the therapeutic issues deriving from a faulty DNA repair machinery in epithelial ovarian cancers, starting from existing and well-established treatments and investigating new therapeutic approaches which could possibly improve ovarian cancer patients’ survival outcomes in the near future. In particular, we concentrate on the role of both Poly (ADP-ribose) Polymerase (PARP) inhibitors (PARPis) and immune checkpoint inhibitors in HGSOC, highlighting their activity in relation to BRCA1/2 mutational status and homologous recombination deficiency (HRD). We investigate the biological rationale supporting their use in the clinical setting, pointing at tracking their route from the laboratory bench to the patient’s bedside. Finally, we deal with the onset of mechanisms of primary and acquired resistance to PARPis, reporting the pioneering strategies aimed at converting homologous-recombination (HR) proficient tumors into homologous recombination (HR)-deficient HGSOC.
Collapse
|
488
|
Henriksen JR, Nederby L, Donskov F, Waldstrøm M, Adimi P, Jakobsen A, Steffensen KD. Prognostic significance of baseline T cells, B cells and neutrophil-lymphocyte ratio (NLR) in recurrent ovarian cancer treated with chemotherapy. J Ovarian Res 2020; 13:59. [PMID: 32414391 PMCID: PMC7229632 DOI: 10.1186/s13048-020-00661-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 05/06/2020] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Biomarkers are needed to guide treatment decisions in recurrent ovarian cancer, as a high proportion of patients do not benefit from treatments. Data on immune subsets in patients receiving chemotherapy are scarce. We investigated the impact of T cells, B cells, neutrophils and the neutrophil-lymphocyte ratio (NLR) in ovarian cancer patients receiving palliative chemotherapy. METHODS Blood samples were collected prospectively at baseline in recurrent ovarian cancer (N = 72) receiving chemotherapy. T cells, B cells, neutrophils, and NLR were analyzed. Primary and secondary endpoints were overall survival (OS) and treatment response, respectively. Cut-offs for T and B cells were predefined. RESULTS In patients with low vs. high T and B cells counts, OS was 6.1 months vs 12.0 months (P = 0.017) and 6.1 months vs 12.0 months (P = 0.011, respectively. Low T and B cells analyzed as continuous variables were also associated with unfavorable OS, P = 0.011 and P = 0.007, respectively. Neutrophils had no significant prognostic impact. Median NLR was 4.1. High vs. low NLR was associated with poor survival, 7.4 months vs. 15.9 months (P = 0.012). In multivariate analysis including platinum sensitivity, number of prior lines of chemotherapy, and performance status, high NLR remained an independent poor prognostic factor HR: 2.17 (95% CI 1.21-3.88) (P = 0.009). High NLR was also significantly associated with lack of response, OR 0.15 (95% CI: 0.04-0.51) (P = 0.002). CONCLUSION In recurrent ovarian cancer patients undergoing palliative chemotherapy, low T and B lymphocyte counts had an unfavorable prognostic impact. High NLR was associated with lack of response and a poor prognosis, and the parameter may be used in patient counselling and treatment decisions.
Collapse
Affiliation(s)
- Jon Røikjær Henriksen
- Department of Oncology, Vejle Hospital - University Hospital of Southern Denmark, Vejle, Denmark.
- Faculty of Health Sciences, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark.
| | - Line Nederby
- Biochemistry and Immunology, Vejle Hospital - University Hospital of Southern Denmark, Vejle, Denmark
| | - Frede Donskov
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Marianne Waldstrøm
- Faculty of Health Sciences, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Vejle Hospital - University Hospital of Southern Denmark, Vejle, Denmark
| | - Parvin Adimi
- Department of Oncology, Vejle Hospital - University Hospital of Southern Denmark, Vejle, Denmark
| | - Anders Jakobsen
- Department of Oncology, Vejle Hospital - University Hospital of Southern Denmark, Vejle, Denmark
- Faculty of Health Sciences, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| | - Karina Dahl Steffensen
- Department of Oncology, Vejle Hospital - University Hospital of Southern Denmark, Vejle, Denmark
- Faculty of Health Sciences, Institute of Regional Health Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
489
|
Mlynska A, Vaišnorė R, Rafanavičius V, Jocys S, Janeiko J, Petrauskytė M, Bijeikis S, Cimmperman P, Intaitė B, Žilionytė K, Barakauskienė A, Meškauskas R, Paberalė E, Pašukonienė V. A gene signature for immune subtyping of desert, excluded, and inflamed ovarian tumors. Am J Reprod Immunol 2020; 84:e13244. [PMID: 32294293 DOI: 10.1111/aji.13244] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 03/24/2020] [Accepted: 04/07/2020] [Indexed: 12/19/2022] Open
Abstract
PROBLEM The current tumor immunology paradigm emphasizes the role of the immune tumor microenvironment and distinguishes several histologically and transcriptionally different immune tumor subtypes. However, the experimental validation of such classification is so far limited to selected cancer types. Here, we aimed to explore the existence of inflamed, excluded, and desert immune subtypes in ovarian cancer, as well as investigate their association with the disease outcome. METHOD OF STUDY We used the publicly available ovarian cancer dataset from The Cancer Genome Atlas for developing subtype assignment algorithm, which was next verified in a cohort of 32 real-world patients of a known tumor subtype. RESULTS Using clinical and gene expression data of 489 ovarian cancer patients in the publicly available dataset, we identified three transcriptionally distinct clusters, representing inflamed, excluded, and desert subtypes. We developed a two-step subtyping algorithm with COL5A2 serving as a marker for separating excluded tumors, and CD2, TAP1, and ICOS for distinguishing between inflamed and desert tumors. The accuracy of gene expression-based subtyping algorithm in a real-world cohort was 75%. Additionally, we confirmed that patients bearing inflamed tumors are more likely to survive longer. CONCLUSION Our results highlight the presence of transcriptionally and histologically distinct immune subtypes among ovarian tumors and emphasize the potential benefit of immune subtyping as a clinical tool for treatment tailoring.
Collapse
Affiliation(s)
| | | | | | - Simonas Jocys
- Baltic Institute of Advanced Technology, Vilnius, Lithuania
| | - Julija Janeiko
- Baltic Institute of Advanced Technology, Vilnius, Lithuania
| | | | - Simas Bijeikis
- Baltic Institute of Advanced Technology, Vilnius, Lithuania
| | | | | | | | - Aušrinė Barakauskienė
- Vilnius University, Vilnius, Lithuania.,Ltd Patologijos Diagnostika, Vilnius, Lithuania
| | | | | | | |
Collapse
|
490
|
Cui J, Li X, Wang S, Su Y, Chen X, Cao L, Zhi X, Qiu Z, Wang Y, Jiang H, Huang B, Ji F, Su J. Triptolide prevents bone loss via suppressing osteoclastogenesis through inhibiting PI3K-AKT-NFATc1 pathway. J Cell Mol Med 2020; 24:6149-6161. [PMID: 32347017 PMCID: PMC7294126 DOI: 10.1111/jcmm.15229] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/14/2020] [Accepted: 02/06/2020] [Indexed: 12/22/2022] Open
Abstract
Bone loss (osteopenia) is a common complication in human solid tumour. In addition, after surgical treatment of gynaecological tumour, osteoporosis often occurs due to the withdrawal of oestrogen. The major characteristic of osteoporosis is the low bone mass with micro-architectural deteriorated bone tissue. And the main cause is the overactivation of osteoclastogenesis, which is one of the most important therapeutic targets. Inflammation could induce the interaction of RANKL/RANK, which is the promoter of osteoclastogenesis. Triptolide is derived from the traditional Chinese herb lei gong teng, presented multiple biological effects, including anti-cancer, anti-inflammation and immunosuppression. We hypothesized that triptolide could inhibits osteoclastogenesis by suppressing inflammation activation. In this study, we confirmed that triptolide could suppress RANKL-induced osteoclastogenesis in bone marrow mononuclear cells (BMMCs) and RAW264.7 cells and inhibited the osteoclast bone resorption functions. PI3K-AKT-NFATc1 pathway is one of the most important downstream pathways of RANKL-induced osteogenesis. The experiments in vitro indicated that triptolide suppresses the activation of PI3K-AKT-NFATc1 pathway and the target point located at the upstream of AKT because both NFATc1 overexpression and AKT phosphorylation could ameliorate the triptolide suppression effects. The expression of MDM2 was elevated, which demonstrated the MDM-p53-induced cell death might contribute to the osteoclastogenesis suppression. Ovariectomy-induced bone loss and inflammation activation were also found to be ameliorated in the experiments in vivo. In summary, the new effect of anti-cancer drug triptolide was demonstrated to be anti-osteoclastogenesis, and we demonstrated triptolide might be a promising therapy for bone loss caused by tumour.
Collapse
Affiliation(s)
- Jin Cui
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China.,China-South Korea Bioengineering Center, Shanghai, China
| | - Xiaoqun Li
- Graduate Management Unit, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | | | - Yiming Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Xiao Chen
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China.,China-South Korea Bioengineering Center, Shanghai, China
| | - Liehu Cao
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xin Zhi
- Graduate Management Unit, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zili Qiu
- Jinling High School, Nanjing, China
| | - Yao Wang
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hao Jiang
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Biaotong Huang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Fang Ji
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jiacan Su
- Department of Orthopedics, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China.,China-South Korea Bioengineering Center, Shanghai, China
| |
Collapse
|
491
|
Zamarin D, Burger RA, Sill MW, Powell DJ, Lankes HA, Feldman MD, Zivanovic O, Gunderson C, Ko E, Mathews C, Sharma S, Hagemann AR, Khleif S, Aghajanian C. Randomized Phase II Trial of Nivolumab Versus Nivolumab and Ipilimumab for Recurrent or Persistent Ovarian Cancer: An NRG Oncology Study. J Clin Oncol 2020; 38:1814-1823. [PMID: 32275468 DOI: 10.1200/jco.19.02059] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Single-agent PD-1 blockade exhibits limited efficacy in epithelial ovarian cancer (EOC). We evaluated ipilimumab plus nivolumab compared with nivolumab alone in women with persistent or recurrent EOC. METHODS Eligibility criteria included measurable disease, 1-3 prior regimens, and platinum-free interval (PFI) < 12 months. Participants were randomly allocated to intravenous nivolumab (every 2 weeks) or induction with nivolumab plus ipilimumab for 4 doses (every 3 weeks), followed by every-2-week maintenance nivolumab for a maximum of 42 doses. The primary null hypothesis was equal probability of objective response within 6 months of random allocation in each arm. RESULTS One hundred patients were allocated to receive either nivolumab (n = 49), or nivolumab plus ipilimumab (n = 51), with PFI of < 6 months in 62%. Six (12.2%) responses occurred within 6 months in the nivolumab group and 16 (31.4%) in the nivolumab plus ipilimumab group (odds ratio, 3.28; 85% CI, 1.54 to infinity; P = .034). The median progression-free survival (PFS) was 2 and 3.9 months in the nivolumab and nivolumab plus ipilimumab groups, respectively, with a PFI-stratified hazard ratio of 0.53 (95% CI, 0.34 to 0.82); the respective hazard ratio for death was 0.79 (95% CI, 0.44 to 1.42). Grade ≥ 3 related adverse events occurred in 33% of patients in the nivolumab group and 49% in the combination group, with no treatment-related deaths. PD-L1 expression was not significantly associated with response in either treatment group. CONCLUSION Compared with nivolumab alone, the combination of nivolumab and ipilimumab in EOC resulted in superior response rate and longer, albeit limited, PFS, with toxicity of the combination regimen comparable to prior reports. Additional combination studies to enhance durability of the dual regimen are warranted.
Collapse
Affiliation(s)
- Dmitriy Zamarin
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, NY
| | - Robert A Burger
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA
| | - Michael W Sill
- Biostatistics and Bioinformatics, Clinical Trial Development Division, NRG Oncology, Roswell Park, Buffalo, NY
| | - Daniel J Powell
- Department of Pathology, University of Pennsylvania, Philadelphia, PA
| | - Heather A Lankes
- NRG Oncology Biospecimen Bank-Columbus, Biopathology Center, The Research Institute at Nationwide Children's Hospital, Columbus, OH
| | - Michael D Feldman
- Department of Pathology, University of Pennsylvania, Philadelphia, PA
| | - Oliver Zivanovic
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, NY
| | - Camille Gunderson
- Stephenson Cancer Center, Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Oklahoma; Oklahoma City, OK
| | - Emily Ko
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, PA
| | - Cara Mathews
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI
| | | | - Andrea R Hagemann
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Washington University, Saint Louis, MO
| | - Samir Khleif
- The Loop Immuno-oncology Laboratory, Lombardi Comprehensive Cancer Center, Georgetown University Medical School, Washington DC
| | - Carol Aghajanian
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, NY
| |
Collapse
|
492
|
Huang HJ, Yang LY, Tung HJ, Ku FC, Wu RC, Tang YH, Chang WY, Jung SM, Wang CC, Lin CT, Liu FY, Lin G, Chen MY, Chou HH, Chang TC, Chao A, Lai CH. Management and clinical outcomes of patients with recurrent/progressive ovarian clear cell carcinoma. J Formos Med Assoc 2020; 119:793-804. [DOI: 10.1016/j.jfma.2019.11.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/26/2019] [Accepted: 11/20/2019] [Indexed: 01/13/2023] Open
|
493
|
Nishio S, Matsumoto K, Takehara K, Kawamura N, Hasegawa K, Takeshima N, Aoki D, Kamiura S, Arakawa A, Kondo E, Hirakawa T, Yamamoto K, Aoki M, Stein K, Keefe S, Fujiwara K, Ushijima K. Pembrolizumab monotherapy in Japanese patients with advanced ovarian cancer: Subgroup analysis from the KEYNOTE-100. Cancer Sci 2020; 111:1324-1332. [PMID: 32012411 PMCID: PMC7156846 DOI: 10.1111/cas.14340] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/15/2020] [Accepted: 01/16/2020] [Indexed: 12/24/2022] Open
Abstract
Interim results from the two-cohort, phase 2 KEYNOTE-100 study (NCT02674061) of 376 patients with previously treated advanced recurrent ovarian cancer (ROC) showed that pembrolizumab monotherapy was associated with an objective response rate (ORR) of 8.0% (95% CI, 5.4-11.2). We present outcomes for the Japanese patients (n = 21) enrolled in KEYNOTE-100. Patients with epithelial ROC had received either 1-3 prior chemotherapy lines and had platinum-free interval or treatment-free interval (PFI; TFI) of 3-12 months (cohort A) or 4-6 prior chemotherapy lines and had PFI/TFI of ≥3 months (cohort B). All patients received pembrolizumab 200 mg every 3 weeks as monotherapy for 2 years or until progression, death, unacceptable toxicity or consent withdrawal. Primary objectives were ORR per RECIST v1.1 for each cohort and higher programmed death ligand-1 (PD-L1) tumor expression. The relationship between PD-L1 expression (measured as combined positive score [CPS]) and ORR was assessed. Twenty-one Japanese patients (cohort A, n = 19; cohort B, n = 2) were treated. The median (range) age was 57 (37-78) years; 19 (90.5%) patients had ECOG status of 0 and 16 (76.2%) patients had stage III-IV disease. ORR was 19.0% (95% CI, 5.4-41.9) and seemed to increase with increasing PD-L1 expression. A total of 13 (61.9%) patients had treatment-related adverse events (TRAE), and 5 (23.8%) had grade 3-4 TRAE. There were no treatment-related deaths in this subpopulation. Pembrolizumab monotherapy was associated with antitumor activity in Japanese patients with ROC, with no new safety signals identified in this subpopulation. The data suggested a trend toward higher PD-L1 expression among some patients with higher ORR.
Collapse
Affiliation(s)
- Shin Nishio
- Kurume University School of MedicineKurumeJapan
| | | | | | | | - Kosei Hasegawa
- Saitama Medical University International Medical CenterHidakaJapan
| | | | | | | | | | | | | | | | | | | | | | - Keiichi Fujiwara
- Saitama Medical University International Medical CenterHidakaJapan
| | | |
Collapse
|
494
|
Lin LL, Lakomy DS, Ning MS, Simpkins F, Jhingran A. Combining novel agents with radiotherapy for gynecologic malignancies: beyond the era of cisplatin. Int J Gynecol Cancer 2020; 30:409-423. [PMID: 32193219 DOI: 10.1136/ijgc-2020-001227] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
Therapeutic strategies combining radiation therapy with novel agents have become an area of intense research focus in oncology and are actively being investigated for a wide range of solid tumors. The mechanism of action of these systemic agents can be stratified into three general categories: (1) enhancement or alteration of the immune system; (2) disruption of DNA damage response mechanisms; and (3) impediment of cellular signaling pathways involving growth, angiogenesis, and hypoxia. Pre-clinical data suggest that radiation therapy has immunogenic qualities and may optimize response to immuno-oncology therapies by priming the immune system, whereas other novel systemic agents can enhance radiosensitivity through augmentation of genomic instability and alteration of central signaling pathways related to growth and survival. Gynecologic cancers in particular have the potential for synergistic response to combination approaches incorporating radiation therapy and novel systemic therapies. Several clinical trials have been proposed to elucidate the efficacy and safety of such approaches. Here we discuss the mechanisms of novel therapies and the rationale for these combination strategies, reviewing the relevant pre-clinical and clinical data. We explore their optimal use with respect to indications, interactions, and potential synergy in combination with radiation therapy and review ongoing trials and active areas of investigation.
Collapse
Affiliation(s)
- Lilie L Lin
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - David S Lakomy
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Dartmouth College Geisel School of Medicine, Hanover, New Hampshire, USA
| | - Matthew S Ning
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Fiona Simpkins
- Department of Obstetrics and Gynecology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Anuja Jhingran
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
495
|
Lymphocytic infiltration and Chemotherapy Response Score as prognostic markers in ovarian cancer patients treated with Neoadjuvant chemotherapy. Gynecol Oncol 2020; 157:599-605. [PMID: 32173048 DOI: 10.1016/j.ygyno.2020.03.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/02/2020] [Indexed: 11/22/2022]
Abstract
Neoadjuvant Chemotherapy (NACT) followed by Interval Debulking Surgery (IDS) is an accepted frontline treatment in patients with advanced Epithelial Ovarian Cancer (EOC). Histopathologic assessment of tumor post NACT may provide a surrogate for response to treatment. The present study aims to characterize the pathological response and to examine its prognostic significance in these patients. Medical records of women with EOC treated in our institution from 2011 to 2016 were retrospectively identified. IDS specimens were reviewed by study pathologist and Chemotherapy Response Score (CRS), lymphocytic infiltration, necrosis and mitosis were assessed. 55 patients with EOC treated with NACT were identified and 48 had complete clinical and pathological data. Median age was 63 years. CRS assessed at omentum predicted PFS when adjusted for age, stage, debulking status (complete, optimal, suboptimal) and post IDS bevacizumab administration (mPFS CRS 1 vs 2 vs 3: 10.3-14-18.7 months 95% CI [7.4-15.7], [12.2-22.9], [13.5-31.3]). Presence of lymphocytic infiltration was associated with improved OS (log-rank test P = 0.015). Post IDS bevacizumab was associated with shorter PFS in patients with lymphocytic infiltration. BRCA status was known for 25 patients and presence of BRCA1/2 mutations was strongly correlated with lymphocytic infiltration (P = 0.011) but not CRS omentum (P = 0.926). Our study confirms the predictive value of CRS in EOC patients treated with NACT and IDS, but also demonstrates the prognostic significance of lymphocytic infiltration as well as its possible interaction with bevacizumab treatment.
Collapse
|
496
|
Immune Checkpoint Inhibitors in Epithelial Ovarian Cancer: An Overview on Efficacy and Future Perspectives. Diagnostics (Basel) 2020; 10:diagnostics10030146. [PMID: 32156035 PMCID: PMC7151145 DOI: 10.3390/diagnostics10030146] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/21/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the leading cause of death among gynecological cancers. Despite improvements in medical treatments, the prognosis for EOC remains poor, and there is an urgent need for new therapeutic strategies. Immune checkpoint inhibitors (CPIs) have dramatically improved survival of several cancers and are under evaluation in OC. Unfortunately, CPIs have shown globally unsatisfactory results. The aim of this manuscript is to critically review the results from early-phase trials with CPIs in terms of safety and activity, discuss the possible reasons for disappointing results and the new therapeutic approaches to improve patient outcomes.
Collapse
|
497
|
Lynam S, Lugade AA, Odunsi K. Immunotherapy for Gynecologic Cancer: Current Applications and Future Directions. Clin Obstet Gynecol 2020; 63:48-63. [PMID: 31833846 PMCID: PMC7298668 DOI: 10.1097/grf.0000000000000513] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The role of the immune system in the development of cancer has been a subject of ongoing clinical investigation in recent years. Emerging data demonstrate that tumorigenesis resulting in ovarian, uterine, and cervical cancers is a consequence of impaired host immune responses to cancerous cells. Leveraging the immune system through the use of immune checkpoint inhibitors, therapeutic vaccine therapy, and adoptive cell transfer presents a profound opportunity to revolutionize cancer treatment. This review will encompass the role of the immune system in development of gynecologic cancers and highlight recent data regarding immunotherapy applications in ovarian, uterine, and cervical cancers.
Collapse
Affiliation(s)
| | - Amit A Lugade
- Center for Immunotherapy Roswell Park Cancer Institute, Buffalo, New York
| | - Kunle Odunsi
- Department of Gynecologic Oncology
- Center for Immunotherapy Roswell Park Cancer Institute, Buffalo, New York
| |
Collapse
|
498
|
Moufarrij S, Srivastava A, Gomez S, Hadley M, Palmer E, Austin PT, Chisholm S, Diab N, Roche K, Yu A, Li J, Zhu W, Lopez-Acevedo M, Villagra A, Chiappinelli KB. Combining DNMT and HDAC6 inhibitors increases anti-tumor immune signaling and decreases tumor burden in ovarian cancer. Sci Rep 2020; 10:3470. [PMID: 32103105 PMCID: PMC7044433 DOI: 10.1038/s41598-020-60409-4] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 02/12/2020] [Indexed: 12/12/2022] Open
Abstract
Novel therapies are urgently needed for ovarian cancer, the deadliest gynecologic malignancy. Ovarian cancer has thus far been refractory to immunotherapies that stimulate the host immune system to recognize and kill cancer cells. This may be because of a suppressive tumor immune microenvironment and lack of recruitment and activation of immune cells that kill cancer cells. Our previous work showed that epigenetic drugs including DNA methyltransferase inhibitors and histone deacetylase 6 inhibitors (DNMTis and HDAC6is) individually increase immune signaling in cancer cells. We find that combining DNMTi and HDAC6i results in an amplified type I interferon response, leading to increased cytokine and chemokine expression and higher expression of the MHC I antigen presentation complex in human and mouse ovarian cancer cell lines. Treating mice bearing ID8 Trp53-/- ovarian cancer with HDAC6i/DNMTi led to an increase in tumor-killing cells such as IFNg+ CD8, NK, and NKT cells and a reversal of the immunosuppressive tumor microenvironment with a decrease in MDSCs and PD-1hi CD4 T cells, corresponding with an increase in survival. Thus combining the epigenetic modulators DNMTi and HDAC6i increases anti-tumor immune signaling from cancer cells and has beneficial effects on the ovarian tumor immune microenvironment.
Collapse
Affiliation(s)
- Sara Moufarrij
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
- The Department of Obstetrics & Gynecology, The George Washington University, Washington, DC, USA
| | - Aneil Srivastava
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
- The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Stephanie Gomez
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
- The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, USA
- The Institute for Biomedical Sciences, The George Washington University, Washington, DC, 20052, USA
| | - Melissa Hadley
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
- The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Erica Palmer
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
- The Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, USA
| | - Paul Tran Austin
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
- The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Sarah Chisholm
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
- The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Noor Diab
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
| | - Kyle Roche
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
- The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Angela Yu
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
- The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, USA
| | - Jing Li
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
- The Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, USA
| | - Wenge Zhu
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
- The Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, USA
| | - Micael Lopez-Acevedo
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA
- The Department of Obstetrics & Gynecology, The George Washington University, Washington, DC, USA
| | - Alejandro Villagra
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA.
- The Department of Biochemistry and Molecular Medicine, The George Washington University, Washington, DC, USA.
| | - Katherine B Chiappinelli
- The George Washington University Cancer Center, The George Washington University, Washington, DC, USA.
- The Department of Microbiology, Immunology, & Tropical Medicine, The George Washington University, Washington, DC, USA.
| |
Collapse
|
499
|
Martin JD, Cabral H, Stylianopoulos T, Jain RK. Improving cancer immunotherapy using nanomedicines: progress, opportunities and challenges. Nat Rev Clin Oncol 2020; 17:251-266. [PMID: 32034288 DOI: 10.1038/s41571-019-0308-z] [Citation(s) in RCA: 438] [Impact Index Per Article: 87.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/25/2019] [Indexed: 02/08/2023]
Abstract
Multiple nanotherapeutics have been approved for patients with cancer, but their effects on survival have been modest and, in some examples, less than those of other approved therapies. At the same time, the clinical successes achieved with immunotherapy have revolutionized the treatment of multiple advanced-stage malignancies. However, the majority of patients do not benefit from the currently available immunotherapies and many develop immune-related adverse events. By contrast, nanomedicines can reduce - but do not eliminate - the risk of certain life-threatening toxicities. Thus, the combination of these therapeutic classes is of intense research interest. The tumour microenvironment (TME) is a major cause of the failure of both nanomedicines and immunotherapies that not only limits delivery, but also can compromise efficacy, even when agents accumulate in the TME. Coincidentally, the same TME features that impair nanomedicine delivery can also cause immunosuppression. In this Perspective, we describe TME normalization strategies that have the potential to simultaneously promote the delivery of nanomedicines and reduce immunosuppression in the TME. Then, we discuss the potential of a combined nanomedicine-based TME normalization and immunotherapeutic strategy designed to overcome each step of the cancer-immunity cycle and propose a broadly applicable 'minimal combination' of therapies designed to increase the number of patients with cancer who are able to benefit from immunotherapy.
Collapse
Affiliation(s)
- John D Martin
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
500
|
Abstract
PURPOSE OF REVIEW This review will provide an update on the most recent clinical developments in immuno-oncology in advanced gynecologic cancers and will also highlight ongoing studies in this field. RECENT FINDINGS Although immune checkpoint blockade (ICB) therapy is rapidly altering the treatment landscape in a myriad of solid tumors, the efficacy of ICB therapy with antibodies directed against CTLA-4, PD-1, and PD-L1 in advanced gynecologic cancers has been limited. The exception has been the PD-1 inhibitor pembrolizumab in microsatellite instability high (MSI-H) or mismatch repair-deficient (dMMR) advanced endometrial cancers, highlighted by the recent conditional approval of pembrolizumab in recurrent/metastatic PD-L1-positive cervical cancers and the accelerated approval of pembrolizumab and lenvatinib in microsatellite stable (MSS) or mismatch repair-proficient (pMMR) advanced endometrial cancer. The discovery of novel, rational ICB combinatorial approaches in advanced gynecologic cancers is highly warranted. SUMMARY Recent advances in the genomic characterization of gynecologic malignancies have informed clinical trial design. However, improved molecular and immunophenotypic biomarkers to more accurately identify patients who will most benefit from immunotherapeutic approaches are urgently needed. This is especially critical as we attempt to integrate immune-oncology agents, chemotherapy, targeted therapy, and radiation therapy in the management of gynecologic cancers.
Collapse
|