1
|
Uyar D, Michener CM, Bishop E, Hopp E, Simpson P, Zhang L, Rader JS, Rose PG, Mahdi HS, Debernardo R, Christian Q, Bradley W. Carboplatin, paclitaxel, and pembrolizumab followed by pembrolizumab maintenance for primary treatment of incompletely resected epithelial ovarian cancer. Front Oncol 2024; 14:1291090. [PMID: 38410102 PMCID: PMC10894939 DOI: 10.3389/fonc.2024.1291090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 01/15/2024] [Indexed: 02/28/2024] Open
Abstract
Objective Incompletely resected epithelial ovarian cancer represents a poor prognostic subset of patients. Novel treatment strategies are needed to improve outcomes for this population. We evaluated a treatment strategy combining platinum-based chemotherapy with pembrolizumab followed by pembrolizumab maintenance therapy in the first-line treatment after incomplete resection of epithelial ovarian cancer patients. Methods This was a single-arm, non-randomized pilot study of carboplatin, taxane, and immune checkpoint inhibitor, pembrolizumab, followed by 12 months of maintenance pembrolizumab in patients with incompletely resected epithelial ovarian cancer (EOC). Results A total of 29 patients were enrolled and evaluated for efficacy and safety. The best response to therapy was complete response in 16 (55%) patients, partial response in 9 (31%) patients, and 3 (10%) patients with progression of disease. The median progression-free survival (PFS) was 13.2 months. Grade 3 and 4 toxicities occurred in 20% of patients. In all, 7 patients discontinued therapy due to adverse events. Quality-of-life scores remained high during therapy. Response to therapy did not correlate with PD-L1 tumor expression. Conclusions Combination platinum-taxane therapy with pembrolizumab did not increase median progression-free survival in this cohort of patients. Key message EOC is an immunogenic disease, but immune checkpoint inhibitor therapy has yet to impact outcomes. The current study utilized pembrolizumab in combination with standard chemotherapy followed by a maintenance treatment strategy in incompletely resected EOC. Progression-free survival was not extended in this poor prognostic group with combined chemotherapy and immunotherapy. Clinical trial registration https://clinicaltrials.gov/, identifier NCT 027766582.
Collapse
Affiliation(s)
- Denise Uyar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Chad M. Michener
- Obstetrics and Gynecology Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Erin Bishop
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Elizabeth Hopp
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Pippa Simpson
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Liyun Zhang
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Janet S. Rader
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Peter G. Rose
- Obstetrics and Gynecology Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Haider S. Mahdi
- Obstetrics and Gynecology Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Robert Debernardo
- Obstetrics and Gynecology Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Qiana Christian
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - William Bradley
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
2
|
Zhang H, Xu W, Zhu H, Chen X, Tsai HI. Overcoming the limitations of immunotherapy in pancreatic ductal adenocarcinoma: Combining radiotherapy and metabolic targeting therapy. J Cancer 2024; 15:2003-2023. [PMID: 38434964 PMCID: PMC10905401 DOI: 10.7150/jca.92502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/20/2024] [Indexed: 03/05/2024] Open
Abstract
As a novel anticancer therapy, immunotherapy has demonstrated robust efficacy against a few solid tumors but poor efficacy against pancreatic ductal adenocarcinoma (PDAC). This poor outcome is primarily attributable to the intrinsic cancer cell resistance and T-cell exhaustion, which is also the reason for the failure of conventional therapy. The present review summarizes the current PDAC immunotherapy avenues and the underlying resistance mechanisms. Then, the review discusses synergistic combination therapies, such as radiotherapy (RT) and metabolic targeting. Research suggests that RT boosts the antigen of PDAC, which facilitates the anti-tumor immune cell infiltration and exerts function. Metabolic reprogramming contributes to restoring the exhausted T cell function. The current review will help in tailoring combination regimens to enhance the efficacy of immunotherapy. In addition, it will help provide new approaches to address the limitations of the immunosuppressive tumor microenvironment (TME) by examining the relationship among immunotherapy, RT, and metabolism targeting therapy in PDAC.
Collapse
Affiliation(s)
- Han Zhang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
| | - Wenjin Xu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
| | - Haitao Zhu
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xuelian Chen
- Department of Radiology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, China
| | - Hsiang-I Tsai
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
3
|
Dergham AP, Vaz de Paula CB, Nagashima S, Olandoski M, de Noronha L, Sotomaior VS. Immunohistochemical Profiling of PD-1, PD-L1, CD8, MSI, and p53 and Prognostic Implications in Advanced Serous Ovarian Carcinoma: A Retrospective Study. J Pers Med 2023; 13:1045. [PMID: 37511658 PMCID: PMC10381449 DOI: 10.3390/jpm13071045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/03/2023] [Accepted: 06/09/2023] [Indexed: 07/30/2023] Open
Abstract
Advanced high-grade serous ovarian carcinoma is a serious malignant neoplasm with a late diagnosis and high mortality rate. Even when treated with standard therapy, such as surgery followed by carboplatin and paclitaxel chemotherapy, the prognosis remains unfavorable. Immunotherapy is a treatment alternative that requires further study. Therefore, we aimed to evaluate the expression of PD-1, PD-L1, CD8, MSI (MLH1, MSH2, MSH6, and PMS2), and p53 in the paraffin samples of high-grade serous ovarian carcinoma. A retrospective study of 28 southern Brazilian patients with advanced serous ovarian carcinoma (EC III or IV) was conducted between 2009 and 2020. The expression of these proteins was evaluated using immunohistochemistry, and the results were correlated with the patients' clinicopathological data. At diagnosis, the mean age was 61 years, and the most common clinical stage (60%) was EC III. Among the cases, 84.6% exhibited p53 overexpression, 14.8% had MSI, 92.0% were sensitive to platinum, and more than 50.0% relapsed after treatment. Patients with MSI had a lower CD8/PD-1 ratio and more relapses (p = 0.03). In conclusion, analysis of immunotherapeutic markers in paraffin-embedded advanced serous ovarian carcinoma samples is feasible and may assist in prognosis.
Collapse
Affiliation(s)
- Ana Paula Dergham
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Brazil
- Neo Oncologia Núcleo de Estudos Oncológicos, Curitiba 80440-210, Brazil
| | - Caroline Busatta Vaz de Paula
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Brazil
| | - Seigo Nagashima
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Brazil
| | - Márcia Olandoski
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Brazil
| | - Lucia de Noronha
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Brazil
| | - Vanessa Santos Sotomaior
- Graduate Program in Health Sciences, School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná (PUCPR), Curitiba 80215-901, Brazil
| |
Collapse
|
4
|
Bandi DSR, Sarvesh S, Farran B, Nagaraju GP, El-Rayes BF. Targeting the metabolism and immune system in pancreatic ductal adenocarcinoma: Insights and future directions. Cytokine Growth Factor Rev 2023; 71-72:26-39. [PMID: 37407355 DOI: 10.1016/j.cytogfr.2023.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/29/2023] [Accepted: 06/29/2023] [Indexed: 07/07/2023]
Abstract
Pancreatic cancer, specifically pancreatic ductal adenocarcinoma (PDAC), presents a challenging landscape due to its complex nature and the highly immunosuppressive tumor microenvironment (TME). This immunosuppression severely limits the effectiveness of immune-based therapies. Studies have revealed the critical role of immunometabolism in shaping the TME and influencing PDAC progression. Genetic alterations, lysosomal dysfunction, gut microbiome dysbiosis, and altered metabolic pathways have been shown to modulate immunometabolism in PDAC. These metabolic alterations can significantly impact immune cell functions, including T-cells, myeloid-derived suppressor cells (MDSCs), and macrophages, evading anti-tumor immunity. Advances in immunotherapy offer promising avenues for overcoming immunosuppressive TME and enhancing patient outcomes. This review highlights the challenges and opportunities for future research in this evolving field. By exploring the connections between immunometabolism, genetic alterations, and the microbiome in PDAC, it is possible to tailor novel approaches capable of improving immunotherapy outcomes and addressing the limitations posed by immunosuppressive TME. Ultimately, these insights may pave the way for improved treatment options and better outcomes for PDAC patients.
Collapse
Affiliation(s)
- Dhana Sekhar Reddy Bandi
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA
| | - Sujith Sarvesh
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA
| | - Batoul Farran
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA.
| | - Bassel F El-Rayes
- Department of Hematology and Oncology, Heersink School of Medicine, University of Alabama, Birmingham, AL 35233, USA.
| |
Collapse
|
5
|
Pankowska KA, Będkowska GE, Chociej-Stypułkowska J, Rusak M, Dąbrowska M, Osada J. Crosstalk of Immune Cells and Platelets in an Ovarian Cancer Microenvironment and Their Prognostic Significance. Int J Mol Sci 2023; 24:ijms24119279. [PMID: 37298230 DOI: 10.3390/ijms24119279] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/17/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Ovarian cancer (OC) is one of the deadliest gynecological cancers, largely due to the fast development of metastasis and drug resistance. The immune system is a critical component of the OC tumor microenvironment (TME) and immune cells such as T cells, NK cells, and dendritic cells (DC) play a key role in anti-tumor immunity. However, OC tumor cells are well known for evading immune surveillance by modulating the immune response through various mechanisms. Recruiting immune-suppressive cells such as regulatory T cells (Treg cells), macrophages, or myeloid-derived suppressor cells (MDSC) inhibit the anti-tumor immune response and promote the development and progression of OC. Platelets are also involved in immune evasion by interaction with tumor cells or through the secretion of a variety of growth factors and cytokines to promote tumor growth and angiogenesis. In this review, we discuss the role and contribution of immune cells and platelets in TME. Furthermore, we discuss their potential prognostic significance to help in the early detection of OC and to predict disease outcome.
Collapse
Affiliation(s)
- Katarzyna Aneta Pankowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Grażyna Ewa Będkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Chociej-Stypułkowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Małgorzata Rusak
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Milena Dąbrowska
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| | - Joanna Osada
- Department of Haematological Diagnostics, Medical University of Bialystok, Waszyngtona 15A Street, 15-269 Bialystok, Poland
| |
Collapse
|
6
|
Pawłowska A, Skiba W, Suszczyk D, Kuryło W, Jakubowicz-Gil J, Paduch R, Wertel I. The Dual Blockade of the TIGIT and PD-1/PD-L1 Pathway as a New Hope for Ovarian Cancer Patients. Cancers (Basel) 2022; 14:5757. [PMID: 36497240 PMCID: PMC9740841 DOI: 10.3390/cancers14235757] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/17/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
The prognosis for ovarian cancer (OC) patients is poor and the five-year survival rate is only 47%. Immune checkpoints (ICPs) appear to be the potential targets in up-and-coming OC treatment. However, the response of OC patients to immunotherapy based on programmed cell death pathway (PD-1/PD-L1) inhibitors totals only 6-15%. The promising approach is a combined therapy, including other ICPs such as the T-cell immunoglobulin and ITIM domain/CD155/DNAX accessory molecule-1 (TIGIT/CD155/DNAM-1) axis. Preclinical studies in a murine model of colorectal cancer showed that the dual blockade of PD-1/PD-L1 and TIGIT led to remission in the whole studied group vs. the regression of the tumors with the blockade of a single pathway. The approach stimulates the effector activity of T cells and NK cells, and redirects the immune system activity against the tumor. The understanding of the synergistic action of the TIGIT and PD-1/PD-L1 blockade is, however, poor. Thus, the aim of this review is to summarize the current knowledge about the mode of action of the dual TIGIT and PD-1/PD-L1 blockade and its potential benefits for OC patients. Considering the positive impact of this combined therapy in malignancies, including lung and colorectal cancer, it appears to be a promising approach in OC treatment.
Collapse
Affiliation(s)
- Anna Pawłowska
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Wiktoria Skiba
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Dorota Suszczyk
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Weronika Kuryło
- Students’ Scientific Association, Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| | - Joanna Jakubowicz-Gil
- Department of Functional Anatomy and Cytobiology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Roman Paduch
- Department of Virology and Immunology, Institute of Microbiology and Biotechnology, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, Medical University of Lublin, Chodźki 4a, 20-093 Lublin, Poland
| |
Collapse
|
7
|
Zhang Y, Wang X, Shi M, Song Y, Yu J, Han S. Programmed death ligand 1 and tumor-infiltrating CD8+ T lymphocytes are associated with the clinical features in meningioma. BMC Cancer 2022; 22:1171. [DOI: 10.1186/s12885-022-10249-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/29/2022] [Indexed: 11/13/2022] Open
Abstract
Abstract
Objective
To investigate the expression of programmed death ligand-1 (PD-L1) and the levels of CD8+ tumor-infiltrating lymphocytes (TILs) in meningioma as well as determine the association between their levels and the clinical outcomes.
Methods
We performed a retrospective case-control study on 93 patients with meningioma. The patients showed tumor recurrence and were matched with the control patients without recurrence in their age, gender, admission time, tumor sites, tumor volume, peritumoral brain edema (PTBE), Simpson grade resection, WHO grade, postoperative radiotherapy, and the follow-up duration. We reviewed the clinical data of patients and performed immunohistochemistry analysis to investigate the PD-L1 expression and the levels of CD8+ TILs. Multivariate logistic regression was performed to analyze the association between clinical features and immune markers. The conditional logistic regression models were used to calculate the odds ratios (ORs) with 95% confidence intervals (CIs), and Kaplan–Meier analysis was performed to analyze tumor recurrence.
Results
Tumor volume was correlated with the PD-L1 expression (P = 0.003, HR = 5.288, 95%CI, 1.786–15.651). PTBE served as an independent predictor of CD8+ TIL levels (P = 0.001, HR = 0.176, 95%CI 0.065–0.477). The levels of CD8+ TILs were associated with tumor recurrence (P = 0.020, OR = 0.325, 95%CI, 0.125–0.840).
Conclusion
Tumor volume was associated with PD-L1 expression, and PTBE was an independent predictor of CD8+ TIL levels in meningioma. CD8+ TIL levels correlated with tumor recurrence in meningioma.
Collapse
|
8
|
Dumitru A, Dobrica EC, Croitoru A, Cretoiu SM, Gaspar BS. Focus on PD-1/PD-L1 as a Therapeutic Target in Ovarian Cancer. Int J Mol Sci 2022; 23:ijms232012067. [PMID: 36292922 PMCID: PMC9603705 DOI: 10.3390/ijms232012067] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/27/2022] [Accepted: 10/06/2022] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is considered one of the most aggressive and deadliest gynecological malignancies worldwide. Unfortunately, the therapeutic methods that are considered the gold standard at this moment are associated with frequent recurrences. Survival in ovarian cancer is associated with the presence of a high number of intra tumor infiltrating lymphocytes (TILs). Therefore, immunomodulation is considered to have an important role in cancer treatment, and immune checkpoint inhibitors may be useful for restoring T cell-mediated antitumor immunity. However, the data presented in the literature until now are not sufficient to allow for the identification and selection of patients who really respond to immunotherapy among those with ovarian cancer. Although there are some studies with favorable results, more prospective trials are needed in this sense. This review focuses on the current and future perspectives of PD-1/L1 blockade in ovarian cancer and analyzes the most important immune checkpoint inhibitors used, with the aim of achieving optimal clinical outcomes. Future studies and trials are needed to maximize the efficacy of immune checkpoint blockade therapy in ovarian cancer, as well as in all cancers, in general.
Collapse
Affiliation(s)
- Adrian Dumitru
- Department of Pathology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Pathology, University Emergency Hospital of Bucharest, 050098 Bucharest, Romania
| | - Elena-Codruta Dobrica
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
- Department of Dermatology, Elias University Hospital, 011461 Bucharest, Romania
| | - Adina Croitoru
- Department of Medical Oncology, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Department of Oncology, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Sanda Maria Cretoiu
- Department of Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Correspondence:
| | - Bogdan Severus Gaspar
- Surgery Department, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
9
|
Fanale D, Dimino A, Pedone E, Brando C, Corsini LR, Filorizzo C, Fiorino A, Lisanti MC, Magrin L, Randazzo U, Bazan Russo TD, Russo A, Bazan V. Prognostic and Predictive Role of Tumor-Infiltrating Lymphocytes (TILs) in Ovarian Cancer. Cancers (Basel) 2022; 14:4344. [PMID: 36139508 PMCID: PMC9497073 DOI: 10.3390/cancers14184344] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 12/12/2022] Open
Abstract
In the last decade, tumor-infiltrating lymphocytes (TILs) have been recognized as clinically relevant prognostic markers for improved survival, providing the immunological basis for the development of new therapeutic strategies and showing a significant prognostic and predictive role in several malignancies, including ovarian cancer (OC). In fact, many OCs show TILs whose typology and degree of infiltration have been shown to be strongly correlated with prognosis and survival. The OC histological subtype with the higher presence of TILs is the high-grade serous carcinoma (HGSC) followed by the endometrioid subtype, whereas mucinous and clear cell OCs seem to contain a lower percentage of TILs. The abundant presence of TILs in OC suggests an immunogenic potential for this tumor. Despite the high immunogenic potential, OC has been described as a highly immunosuppressive tumor with a high expression of PD1 by TILs. Although further studies are needed to better define their role in prognostic stratification and the therapeutic implication, intraepithelial TILs represent a relevant prognostic factor to take into account in OC. In this review, we will discuss the promising role of TILs as markers which are able to reflect the anticancer immune response, describing their potential capability to predict prognosis and therapy response in OC.
Collapse
Affiliation(s)
- Daniele Fanale
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Alessandra Dimino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Erika Pedone
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Chiara Brando
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Lidia Rita Corsini
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Clarissa Filorizzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Alessia Fiorino
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Maria Chiara Lisanti
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Luigi Magrin
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Ugo Randazzo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Tancredi Didier Bazan Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Antonio Russo
- Section of Medical Oncology, Department of Surgical, Oncological and Oral Sciences, University of Palermo, 90127 Palermo, Italy
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
10
|
Zhang CJ, Zhang JY, Li LJ, Xu NW. Refractory lymphoma treated with chimeric antigen receptor T cells combined with programmed cell death-1 inhibitor: A case report. World J Clin Cases 2022; 10:7502-7508. [PMID: 36157987 PMCID: PMC9353931 DOI: 10.12998/wjcc.v10.i21.7502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 04/17/2022] [Accepted: 06/03/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diffuse large B-cell lymphoma (DLBCL) is a common aggressive non-Hodgkin's lymphoma (NHL), accounting for 30%-40% of adult NHL. Primary testicular (PT) lymphoma is an uncommon extranodal disease representing approximately 1%-2% of lymphoma. Approximately 30%–40% of patients are refractory to frontline therapy or relapse after complete remission. Refractory DLBCL responds poorly to other lines of chemotherapy, and experiences short-term survival.
CASE SUMMARY We present a 41-year-old male patient who was diagnosed with PT-DLBCL. Further disease progression was observed after multiline chemotherapy. Chimeric antigen receptor T cells (CAR-T) therapy salvaged the patient. Unfortunately, a new mass was observed in the right adrenal area after six months. The patient was administered programmed cell death protein-1 (PD-1) inhibitor therapy and maintained progression-free survival at more than 17 mo of follow-up.
CONCLUSION Our findings support the potential benefit of CAR-T combined with PD-1 inhibitor therapies in this type of relapsed and refractory PT-DLBCL.
Collapse
Affiliation(s)
- Cang-Jian Zhang
- Department of Hematopathology, Lishui Municipal Central Hospital, Lishui 323000, Zhejiang Province, China
| | - Jun-Yu Zhang
- Department of Hematopathology, Lishui Municipal Central Hospital, Lishui 323000, Zhejiang Province, China
| | - Lin-Jie Li
- Department of Hematopathology, Lishui Municipal Central Hospital, Lishui 323000, Zhejiang Province, China
| | - Neng-Wen Xu
- Department of Hematopathology, Lishui Municipal Central Hospital, Lishui 323000, Zhejiang Province, China
| |
Collapse
|
11
|
Liang HY, Liang QL, Chen M, Wang ZW, Huang J. One case of comprehensive treatment for advanced liver cancer followed up over three years. Arch Med Sci 2022; 18:1112-1117. [PMID: 35832717 PMCID: PMC9266952 DOI: 10.5114/aoms/150440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/26/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Hai-Yan Liang
- Oncology Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qi-Lian Liang
- Oncology Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Man Chen
- Oncology Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhen-Wei Wang
- Oncology Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jie Huang
- Oncology Center, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
12
|
PD-L1 Expression in Different Segments and Histological Types of Ovarian Cancer According to Lymphocytic Infiltrate. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:medicina57121309. [PMID: 34946254 PMCID: PMC8703734 DOI: 10.3390/medicina57121309] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/19/2021] [Accepted: 11/25/2021] [Indexed: 11/17/2022]
Abstract
Background and Objectives: Ovarian cancer is the leading cause of death among gynecological tumors. PD-1/PD-L1 immunoregulatory mechanism is activated in ovarian cancers. Lymphocyte infiltration is a significant factor that affects its expression. We analyzed the correlation between localization of lymphocytic infiltrate and PD-L1 expression in epithelial ovarian tumors. Materials and Methods: PD-L1 expression was analyzed in 328 subjects, 122 with epithelial ovarian carcinoma, 42 with atypical proliferative tumor, and 164 with benign epithelial ovarian tumor. Expression in central and invasive tumor parts in epithelial ovarian carcinoma was combined with the most pronounced lymphocyte reaction. Immunohistochemical analysis was performed using the tissue microarray and correlated with a set of histopathology parameters. Results: PD-L1 expression was most prominent in epithelial ovarian carcinoma with different levels of expression observed between invasive and central tumor segments. A high level of PD-L1 expression on tumor cells was more frequently present in the invasive than in the central tumor parts (p < 0.001) only in high-grade serous ovarian carcinoma (HGSC). There was no significant correlation between peritumoral lymphocytic infiltrate and PD-L1 expression regardless of tumor segment. In the central tumor parts of HGSC, there was a correlation of intratumoral lymphocytic infiltrate with a higher level of PD-L1 expression (p = 0.003). Conclusions: The most prominent PD-L1 expression was observed in the invasive tumor parts of HGSC. Only the central parts of the HGSC exhibited significant PD-L1 expression in association with considerable intratumoral lymphocytic infiltrate.
Collapse
|
13
|
Udumula MP, Sakr S, Dar S, Alvero AB, Ali-Fehmi R, Abdulfatah E, Li J, Jiang J, Tang A, Buekers T, Morris R, Munkarah A, Giri S, Rattan R. Ovarian cancer modulates the immunosuppressive function of CD11b +Gr1 + myeloid cells via glutamine metabolism. Mol Metab 2021; 53:101272. [PMID: 34144215 PMCID: PMC8267600 DOI: 10.1016/j.molmet.2021.101272] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/03/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Immature CD11b + Gr1+ myeloid cells that acquire immunosuppressive capability, also known as myeloid-derived suppressor cells (MDSCs), are a heterogeneous population of cells that regulate immune responses. Our study's objective was to elucidate the role of ovarian cancer microenvironment in regulating the immunosuppressive function of CD11b+Gr1+ myeloid cells. METHODS All studies were performed using the intraperitoneal ID8 syngeneic epithelial ovarian cancer mouse model. Myeloid cell depletion and immunotherapy were carried out using anti-Gr1 mAb, gemcitabine treatments, and/or anti-PD1 mAb. The treatment effect was assessed by a survival curve, in situ luciferase-guided imaging, and histopathologic evaluation. Adoptive transfer assays were carried out between congenic CD45.2 and CD45.1 mice. Immune surface and intracellular markers were assessed by flow cytometry. ELISA, western blot, and RT-PCR techniques were employed to assess the protein and RNA expression of various markers. Bone marrow-derived myeloid cells were used for ex-vivo studies. RESULTS The depletion of Gr1+ immunosuppressive myeloid cells alone and in combination with anti-PD1 immunotherapy inhibited ovarian cancer growth. In addition to the adoptive transfer studies, these findings validate the role of immunosuppressive CD11b+Gr1+ myeloid cells in promoting ovarian cancer. Mechanistic investigations showed that ID8 tumor cells and their microenvironments produced recruitment and regulatory factors for immunosuppressive CD11b+Gr1+ myeloid cells. CD11b+Gr1+ myeloid cells primed by ID8 tumors showed increased immunosuppressive marker expression and acquired an energetic metabolic phenotype promoted primarily by increased oxidative phosphorylation fueled by glutamine. Inhibiting the glutamine metabolic pathway reduced the increased oxidative phosphorylation and decreased immunosuppressive markers' expression and function. Dihydrolipoamide succinyl transferase (DLST), a subunit of α-KGDC in the TCA cycle, was found to be the most significantly elevated gene in tumor-primed myeloid cells. The inhibition of DLST reduced oxidative phosphorylation, immunosuppressive marker expression and function in myeloid cells. CONCLUSION Our study shows that the ovarian cancer microenvironment can regulate the metabolism and function of immunosuppressive CD11b + Gr1+ myeloid cells and modulate its immune microenvironment. Targeting glutamine metabolism via DLST in immunosuppressive myeloid cells decreased their activity, leading to a reduction in the immunosuppressive tumor microenvironment. Thus, targeting glutamine metabolism has the potential to enhance the success of immunotherapy in ovarian cancer.
Collapse
Affiliation(s)
- Mary P Udumula
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA
| | - Sharif Sakr
- Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Sajad Dar
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA
| | - Ayesha B Alvero
- Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Rouba Ali-Fehmi
- Department of Pathology, Wayne State University and Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Eman Abdulfatah
- Department of Pathology, Wayne State University and Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Jing Li
- Metabolomics Core, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Jun Jiang
- Metabolomics Core, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Amy Tang
- Department of Public Health Services, Henry Ford Health System, Detroit, MI, USA
| | - Thomas Buekers
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA; Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Robert Morris
- Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Adnan Munkarah
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
| | - Ramandeep Rattan
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA; Department of Oncology, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
14
|
Pawłowska A, Kwiatkowska A, Suszczyk D, Chudzik A, Tarkowski R, Barczyński B, Kotarski J, Wertel I. Clinical and Prognostic Value of Antigen-Presenting Cells with PD-L1/PD-L2 Expression in Ovarian Cancer Patients. Int J Mol Sci 2021; 22:11563. [PMID: 34768993 PMCID: PMC8583913 DOI: 10.3390/ijms222111563] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/21/2022] Open
Abstract
The latest literature demonstrates the predominant role of the programmed cell death axis (PD-1/PD-L1/PD-L2) in ovarian cancer (OC) pathogenesis. However, data concerning this issue is ambiguous. Our research aimed to evaluate the clinical importance of PD-L1/PD-L2 expression in OC environments. We evaluated the role of PD-L1/PD-L2 in OC patients (n = 53). The analysis was performed via flow cytometry on myeloid (mDCs) and plasmacytoid dendritic cells (pDCs) and monocytes/macrophages (MO/MA) in peripheral blood, peritoneal fluid (PF), and tumor tissue (TT). The data were correlated with clinicopathological characteristics and prognosis of OC patients. The concentration of soluble PD-L1 (sPD-L1) and PD-1 in the plasma and PF were determined by ELISA. We established an accumulation of PD-L1+/PD-L2+ mDCs, pDCs, and MA in the tumor microenvironment. We showed an elevated level of sPD-L1 in the PF of OC patients in comparison to plasma and healthy subjects. sPD-L1 levels in PF showed a positive relationship with Ca125 concentration. Moreover, we established an association between higher sPD-L1 levels in PF and shorter survival of OC patients. An accumulation of PD-L1+/PD-L2+ mDCs, pDCs, and MA in the TT and high sPD-L1 levels in PF could represent the hallmark of immune regulation in OC patients.
Collapse
Affiliation(s)
- Anna Pawłowska
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (D.S.); (A.C.); (I.W.)
| | - Agnieszka Kwiatkowska
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (D.S.); (A.C.); (I.W.)
| | - Dorota Suszczyk
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (D.S.); (A.C.); (I.W.)
| | - Agata Chudzik
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (D.S.); (A.C.); (I.W.)
| | - Rafał Tarkowski
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-081 Lublin, Poland; (R.T.); (B.B.); (J.K.)
| | - Bartłomiej Barczyński
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-081 Lublin, Poland; (R.T.); (B.B.); (J.K.)
| | - Jan Kotarski
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-081 Lublin, Poland; (R.T.); (B.B.); (J.K.)
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (D.S.); (A.C.); (I.W.)
| |
Collapse
|
15
|
Singh D, Dheer D, Samykutty A, Shankar R. Antibody drug conjugates in gastrointestinal cancer: From lab to clinical development. J Control Release 2021; 340:1-34. [PMID: 34673122 DOI: 10.1016/j.jconrel.2021.10.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 10/05/2021] [Accepted: 10/06/2021] [Indexed: 12/15/2022]
Abstract
The antibody-drug conjugates (ADCs) are one the fastest growing biotherapeutics in oncology and are still in their infancy in gastrointestinal (GI) cancer for clinical applications to improve patient survival. The ADC based approach is developed with tumor specific antigen, antibody carrying cytotoxic agents to precisely target and deliver chemotherapeutics at the tumor site. To date, 11 ADCs have been approved by US-FDA, and more than 80 are in the clinical development phase for different oncological indications. However, The ADCs based therapies in GI cancers are still far from having high-efficient clinical outcomes. The limited success of these ADCs and lessons learned from the past are now being used to develop a newer generation of ADC against GI cancers. In this review, we did a comprehensive assessment of the key components of ADCs, including tumor marker, antibody, cytotoxic payload, and linkage strategy, with a focus on technical improvement and some future trends in the pipeline for clinical translation. The various preclinical and clinical ADCs used in gastrointestinal malignancies, their target, composition and bioconjugation, along with preclinical and clinical outcomes, are discussed. The emphasis is also given to new generation ADCs employing novel mAb, payload, linker, and bioconjugation methods are also included.
Collapse
Affiliation(s)
- Davinder Singh
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Divya Dheer
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Abhilash Samykutty
- Stephenson Comprehensive Cancer Center, University of Oklahoma, Oklahoma City, OK 73104, USA.
| | - Ravi Shankar
- Natural Products and Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
16
|
Chen S, Li Y, Qian L, Deng S, Liu L, Xiao W, Zhou Y. A Review of the Clinical Characteristics and Novel Molecular Subtypes of Endometrioid Ovarian Cancer. Front Oncol 2021; 11:668151. [PMID: 34150634 PMCID: PMC8210668 DOI: 10.3389/fonc.2021.668151] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Ovarian cancer is one of the most common gynecologic cancers that has the highest mortality rate. Endometrioid ovarian cancer, a distinct subtype of epithelial ovarian cancer, is associated with endometriosis and Lynch syndrome, and is often accompanied by synchronous endometrial carcinoma. In recent years, dysbiosis of the microbiota within the female reproductive tract has been suggested to be involved in the pathogenesis of endometrial cancer and ovarian cancer, with some specific pathogens exhibiting oncogenic having been found to contribute to cancer development. It has been shown that dysregulation of the microenvironment and accumulation of mutations are stimulatory factors in the progression of endometrioid ovarian carcinoma. This would be a potential therapeutic target in the future. Simultaneously, multiple studies have demonstrated the role of four molecular subtypes of endometrioid ovarian cancer, which are of particular importance in the prediction of prognosis. This literature review aims to compile the potential mechanisms of endometrioid ovarian cancer, molecular characteristics, and molecular pathological types that could potentially play a role in the prediction of prognosis, and the novel therapeutic strategies, providing some guidance for the stratified management of ovarian cancer.
Collapse
Affiliation(s)
- Shuangfeng Chen
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Anhui Medical University, Hefei, China
| | - Yuebo Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lili Qian
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Sisi Deng
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Luwen Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Weihua Xiao
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Institute of Immunology, University of Science and Technology of China, Hefei, China
| | - Ying Zhou
- Department of Obstetrics and Gynecology, Anhui Provincial Hospital, Anhui Medical University, Hefei, China.,Department of Obstetrics and Gynecology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
17
|
Potential Impact of Human Cytomegalovirus Infection on Immunity to Ovarian Tumours and Cancer Progression. Biomedicines 2021; 9:biomedicines9040351. [PMID: 33808294 PMCID: PMC8065684 DOI: 10.3390/biomedicines9040351] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Ovarian cancer (OC) is one of the most common, and life-threatening gynaecological cancer affecting females. Almost 75% of all OC cases are diagnosed at late stages, where the 5-year survival rate is less than 30%. The aetiology of the disease is still unclear, and there are currently no screening method nor effective treatment strategies for the advanced disease. A growing body of evidence shows that human cytomegalovirus (HCMV) infecting more than 50% of the world population, may play a role in inducing carcinogenesis through its immunomodulatory activities. In healthy subjects, the primary HCMV infection is essentially asymptomatic. The virus then establishes a life-long chronic latency primarily in the hematopoietic progenitor cells in the bone marrow, with periodic reactivation from latency that is often characterized by high levels of circulating pro-inflammatory cytokines. Currently, infection-induced chronic inflammation is considered as an essential process for OC progression and metastasis. In line with this observation, few recent studies have identified high expressions of HCMV proteins on OC tissue biopsies that were associated with poor survival outcomes. Active HCMV infection in the OC tumour microenvironment may thus directly contribute to OC progression. In this review, we highlight the potential impact of HCMV infection-induced immunomodulatory effects on host immune responses to OC that may promote OC progression.
Collapse
|
18
|
Baş Y, Koç N, Helvacı K, Koçak C, Akdeniz R, Şahin HHK. Clinical and pathological significance of programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) expression in high grade serous ovarian cancer. Transl Oncol 2021; 14:100994. [PMID: 33333370 PMCID: PMC7736714 DOI: 10.1016/j.tranon.2020.100994] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/18/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022] Open
Abstract
We investigated programmed cell death 1 (PD-1) / programmed cell death ligand 1 (PD-L1) expression in high grade serous ovarian cancer (HGSOC) and its relationship to tumor infiltrating lymphocytes (TIL) and prognosis. Formalin fixed paraffin embedded (FFPE) samples of 94 HGSOC cases were included in the study. Immunohistochemical analysis (CD3, CD4, CD8, PD-1 and PD-L1) was performed. Samples were analyzed for expression of immune proteins in the peritumoral stromal and intratumoral areas, scored, and expression was correlated with overall survival, stage, and age. PD-L1 staining ratio with a score greater than 0 was found to have lower survival. There were two positive staining patterns, patchy/diffuse and patchy/focal patterns, in 24 (25.5%) cases. Considering the threshold value ≥5%, we demonstrated that the PD-L1 positive cancer cell membrane immunoreactivity rate and patchy/diffuse PD-L1 expression were 9.6% (n = 9). There was statistically significant relationship between high PD-1 scores and PD-L1 cases of ≥ 5%. A statistically significant difference was found between PD-L1 staining and survival in patients with a threshold ≥ 5%. However an appropriate rate for treatment was determined in 9.6% cases. There was a statistically significant correlation between PD-1 positive TIL score and intratumoral CD3, peritumoral stromal CD3, intratumoral CD4 and intratumoral CD8 positive cells. Survival was lower in cases with higher PD-L1 positive stromal TIL score.
Collapse
Affiliation(s)
- Yılmaz Baş
- Department of Pathology, Hitit University Faculty of Medicine, Çorum Turkey.
| | - Nermin Koç
- Department of Pathology, Zeynep Kamil Maternity and Pediatric Research and Training Hospital, İstanbul, Turkey.
| | - Kaan Helvacı
- Department of Oncology, Hitit University Faculty of Medicine, Çorum, Turkey.
| | - Cem Koçak
- Department of Statistics, Hitit University Faculty of Health Sciences/Nursing, Çorum Turkey.
| | - Raşit Akdeniz
- Department of Pathology, Hitit University Erol Olçok Education and Research Hospital, Çorum, Turkey.
| | | |
Collapse
|
19
|
CXCR4 inhibition modulates the tumor microenvironment and retards the growth of B16-OVA melanoma and Renca tumors. Melanoma Res 2020; 30:14-25. [PMID: 31524789 DOI: 10.1097/cmr.0000000000000639] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To determine whether blockade of the chemokine receptor CXCR4 might alter the tumor microenvironment and inhibit tumor growth, we tested the efficacy of the CXCR4 antagonist X4-136 as a single agent and in combination with various immune checkpoint inhibitors in the syngeneic murine melanoma model B16-OVA. We also tested its activity alone and in combination with axitinib in the renal cancer model Renca. We found that X4-136 exhibited potent single agent antitumor activity in the B16-OVA model that was additive to that of an anti-PDL1 antibody. The antitumor activities were associated with a reduction in the number of immunosuppressive regulatory T cells and myeloid-derived suppressor cells and an increase in the number of tumor-specific CD8/perforin cells in the tumor-microenvironment. Apart from these immune effects, X4-136 alone and in combination with checkpoint inhibitors inhibited the Akt/FOXO-3a cell survival pathway in vitro and in vivo, suggesting that it might have antitumor activity independent of its effects on immune cell trafficking. Similar effects on tumor growth and cytotoxic T lymphocytes infiltration were observed in the Renca model. These studies show that the effects of CXCR4 blockade on immune cell trafficking might serve as a useful adjunct to immune checkpoint inhibitors and other therapies in the treatment of cancer.
Collapse
|
20
|
Pawłowska A, Suszczyk D, Tarkowski R, Paduch R, Kotarski J, Wertel I. Programmed Death-1 Receptor (PD-1) as a Potential Prognosis Biomarker for Ovarian Cancer Patients. Cancer Manag Res 2020; 12:9691-9709. [PMID: 33116828 PMCID: PMC7548235 DOI: 10.2147/cmar.s263010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/03/2020] [Indexed: 12/25/2022] Open
Abstract
Aim Ovarian cancer (OC) is one of the most lethal gynecological malignancies. Recent studies suggest a crucial role of the PD-1/PD-L1 pathway in OC pathogenesis. Therefore, our study aimed at evaluation of the clinical importance of PD-1 expression in ovarian cancer patients. Patients and Methods In this study, we investigated the role of PD-1 in OC patients (n=50) by analyzing its expression on CD4+ and CD8+ T cells in three OC environments: peripheral blood (PB), peritoneal fluid (PF), and tumor (TT) as well as soluble PD-1 (sPD-1) in plasma and PF in terms of their clinical and prognostic significance. T cells with PD-1 expression were analyzed using flow cytometry. The concentration of sPD-1 was determined with the use of ELISA. Our research demonstrated differences in PD-1 expression on CD4+ and CD8+ T cells in the OC environments. Results We found an elevated level of CD4+PD-1+ T cells in tumor and PF, compared to PB. Additionally, we found the highest percentage of CD8+ PD-1+ in tumor, compared to PB and PF. The levels of sPD-1 were higher (p<0.0001) in plasma than in PF. For the first time, we discovered that the higher level of CD4+PD-1+ T cells in the circulation and the higher sPD-1 level in plasma predict poor survival of OC patients. Conclusion We suggest that PD-1 could be a predictive biomarker for OC patients and successful immunotherapy.
Collapse
Affiliation(s)
- Anna Pawłowska
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin 20-081, Poland
| | - Dorota Suszczyk
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin 20-081, Poland
| | - Rafał Tarkowski
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin 20-081, Poland
| | - Roman Paduch
- Department of Virology and Immunology, Maria Curie-Sklodowska University, Lublin 20-033, Poland
| | - Jan Kotarski
- I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin 20-081, Poland
| | - Iwona Wertel
- Independent Laboratory of Cancer Diagnostics and Immunology, I Chair and Department of Oncological Gynaecology and Gynaecology, Medical University of Lublin, Lublin 20-081, Poland
| |
Collapse
|
21
|
Understanding and addressing barriers to successful adenovirus-based virotherapy for ovarian cancer. Cancer Gene Ther 2020; 28:375-389. [PMID: 32951021 DOI: 10.1038/s41417-020-00227-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/09/2020] [Indexed: 01/17/2023]
Abstract
Ovarian cancer is the leading cause of death among women with gynecological cancer, with an overall 5-year survival rate below 50% due to a lack of specific symptoms, late stage at time of diagnosis and a high rate of recurrence after standard therapy. A better understanding of heterogeneity, genetic mutations, biological behavior and immunosuppression in the tumor microenvironment have allowed the development of more effective therapies based on anti-angiogenic treatments, PARP and immune checkpoint inhibitors, adoptive cell therapies and oncolytic vectors. Oncolytic adenoviruses are commonly used platforms in cancer gene therapy that selectively replicate in tumor cells and at the same time are able to stimulate the immune system. In addition, they can be genetically modified to enhance their potency and overcome physical and immunological barriers. In this review we highlight the challenges of adenovirus-based oncolytic therapies targeting ovarian cancer and outline recent advances to improve their potential in combination with immunotherapies.
Collapse
|
22
|
James NE, Woodman M, DiSilvestro PA, Ribeiro JR. The Perfect Combination: Enhancing Patient Response to PD-1-Based Therapies in Epithelial Ovarian Cancer. Cancers (Basel) 2020; 12:E2150. [PMID: 32756436 PMCID: PMC7466102 DOI: 10.3390/cancers12082150] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/28/2020] [Accepted: 08/01/2020] [Indexed: 12/17/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecologic malignancy, with an overall 5-year survival of only 47%. As the development of novel targeted therapies is drastically necessary in order to improve patient survival, current EOC clinical trials have heavily focused on immunotherapeutic approaches, centered upon programmed cell death 1 (PD-1) inhibitors. While PD-1 monotherapies have only exhibited modest responses for patients, it has been theorized that in order to enhance EOC patient response to immunotherapy, combinatorial regimens must be investigated. In this review, unique challenges to EOC PD-1 response will be discussed, along with a comprehensive description of both preclinical and clinical studies evaluating PD-1-based combinatorial therapies. Promising aspects of PD-1-based combinatorial approaches are highlighted, while also discussing specific preclinical and clinical areas of research that need to be addressed, in order to optimize EOC patient immunotherapy response.
Collapse
Affiliation(s)
- Nicole E. James
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
| | - Morgan Woodman
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
| | - Paul A. DiSilvestro
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
- Department of Obstetrics and Gynecology, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA
| | - Jennifer R. Ribeiro
- Program in Women’s Oncology, Department of Obstetrics and Gynecology, Women and Infants Hospital, Providence, RI 02905, USA; (N.E.J.); (M.W.); (P.A.D.)
- Department of Obstetrics and Gynecology, Warren Alpert School of Medicine, Brown University, Providence, RI 02903, USA
| |
Collapse
|
23
|
Chen F, Xu Y, Chen Y, Shan S. TIGIT enhances CD4 + regulatory T-cell response and mediates immune suppression in a murine ovarian cancer model. Cancer Med 2020; 9:3584-3591. [PMID: 32212317 PMCID: PMC7221438 DOI: 10.1002/cam4.2976] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 02/24/2020] [Accepted: 02/25/2020] [Indexed: 12/27/2022] Open
Abstract
Ovarian cancer (OC) is the fifth-leading cause of cancer-related death in women with a pathogenesis involving activation of regulatory T cells (Tregs). The T-cell immunoglobulin and ITIM domain (TIGIT) is a well-known immune checkpoint molecule that inhibits T-cell responses. However, the role of TIGIT in OC is not comprehensively understood. In this study, we revealed crucial functions of TIGIT in the development and progression of OC. ID8 cells were used to establish a murine OC model. TIGIT expression was increased in immune cells of OC mice, particularly in CD4+ Tregs. Anti-TIGIT monoclonal antibodies (mAb) were used to block the function of TIGIT in OC mice, and we found that the anti-TIGIT treatment reduced the proportion of CD4+ Tregs, but did not affect CD4+ and CD8+ T cells or natural killer cells. Splenic CD4+ Tregs from OC mice were isolated after the anti-TIGIT treatment, and their functioning was examined. Inhibition of TIGIT lowered the degree of immunosuppression induced by CD4+ Tregs. A survival curve suggested that anti-TIGIT treatment can improve the survival rate of OC in mice. We conclude that TIGIT enhanced CD4+ Tregs response and mediated immunosuppression in the OC model. Our data suggest that inhibition of TIGIT is a potential therapeutic target in OC patients.
Collapse
Affiliation(s)
- Fengzhen Chen
- Department of Gynecology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yanying Xu
- Department of Gynecology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yulong Chen
- Department of Lung Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Shu Shan
- Department of Gynecology and Obstetrics, Affiliated Tongji Hospital, Tongji University, Shanghai, China
| |
Collapse
|
24
|
Xue C, Xu Y, Ye W, Xie Q, Gao H, Xu B, Zhang D, Jiang J. Expression of PD-L1 in ovarian cancer and its synergistic antitumor effect with PARP inhibitor. Gynecol Oncol 2020; 157:222-233. [PMID: 31987601 DOI: 10.1016/j.ygyno.2019.12.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/01/2019] [Accepted: 12/08/2019] [Indexed: 01/09/2023]
Abstract
BACKGROUND Ovarian cancer samples were studied to determine the expression of programmed death ligand-1 (PD-L1) and its relationship with prognosis, and to explore the effect and potential mechanism of a PARP inhibitor combined with PD-L1 monoclonal antibody for the treatment of ovarian cancer. MATERIALS AND METHODS PD-L1 expression in paraffin-embedded tissues of ovarian cancer was detected by immunohistochemistry (IHC). Flow cytometry was used to detect PD-L1 expression in TILs. Furthermore, we investigated the mechanism of the upregulation of PD-L1 expression by PARP inhibitors in vitro and verified the combined effect in vivo. RESULTS Our study demonstrated that PD-L1 expression in ovarian cancer tissues was associated with the FIGO stage (P = 0.026). OS was significantly lower in high PD-L1 expression group than in the low expression group (P = 0.0005, HR = 2.689), PD-L1 high expression (P = 0.023, HR = 2.275) and FIGO stage (P = 0.024, HR = 11.229) were independent risk factors affecting the survival and prognosis of ovarian cancer patients. Flow cytometry test suggested that PD-L1+ expression was negatively correlated with CD8+ T cell count in ovarian cancer cells (P = 0.054, r = -0.624). In vitro experiments revealed that PD-L1 expression of ovarian cancer cell lines was upregulated after intervention with PARP inhibitors through the Chk1 pathway. The results of in vivo experiments suggested that the growth volume and quality of tumors in the combination group were significantly lower than those in control group (P < 0.05). CONCLUSIONS PARP inhibitors could induce upregulation of PD-L1 expression by promoting phosphorylation of chk1. Antagonistic PD-L1 could reverse the inhibitory effect of PARP inhibitors on CD8+T cells, and had synergistic antitumor effect with PARP inhibitors.
Collapse
Affiliation(s)
- Chunyan Xue
- Department of Tumor Biological Treatment, Soochow University, Jiangsu, Changzhou 213003, China; Department of Obstetrics and Gynecology, Soochow University, Jiangsu, Changzhou 213003, China
| | - Yun Xu
- Department of Tumor Biological Treatment, Soochow University, Jiangsu, Changzhou 213003, China; Department of Obstetrics and Gynecology, Soochow University, Jiangsu, Changzhou 213003, China
| | - Wenfeng Ye
- Department of Tumor Biological Treatment, Soochow University, Jiangsu, Changzhou 213003, China; Department of Obstetrics and Gynecology, Soochow University, Jiangsu, Changzhou 213003, China
| | - Quanqin Xie
- Department of Tumor Biological Treatment, Soochow University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Soochow University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Jiangsu, Changzhou 213003, China
| | - Hongyan Gao
- Department of Tumor Biological Treatment, Soochow University, Jiangsu, Changzhou 213003, China; Department of Obstetrics and Gynecology, Soochow University, Jiangsu, Changzhou 213003, China
| | - Bin Xu
- Department of Tumor Biological Treatment, Soochow University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Soochow University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Jiangsu, Changzhou 213003, China
| | - Dachuan Zhang
- Department of Tumor Biological Treatment, Soochow University, Jiangsu, Changzhou 213003, China; Pathology Department, The Third Affiliated Hospital of Soochow University, Soochow University, Jiangsu, Changzhou 213003, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, Soochow University, Jiangsu, Changzhou 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, Soochow University, Jiangsu, Changzhou 213003, China; Institute of Cell Therapy, Soochow University, Jiangsu, Changzhou 213003, China.
| |
Collapse
|
25
|
Jeong YIL, Yoo SY, Heo J, Kang DH. Chlorin e6-Conjugated and PEGylated Immune Checkpoint Inhibitor Nanocomposites for Pulmonary Metastatic Colorectal Cancer. ACS OMEGA 2019; 4:18593-18599. [PMID: 31737818 PMCID: PMC6854570 DOI: 10.1021/acsomega.9b02386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 10/23/2019] [Indexed: 05/04/2023]
Abstract
Here we demonstrate theranostic immune checkpoint inhibitor nanocomposites (ICI NC) having an improved tumor targeting ability in pulmonary metastatic colon cancer model. Atezolizumab, a PD-L1 antibody, was conjugated with methoxy poly(ethylene glycol) (MePEG) and chlorin e6 (Ce6) via cathepsin-B-sensitive peptide as a linkage (named as ICI nanocomposites, ICI NC). This ICI NC is delivered to tumor sites enriched with tumor-specific enzymes such as cathepsin B, whereas undesired ICI exposure to normal tissue is avoided. When ICI NC were incubated with cathepsin B, Ce6 was released from ICI NC with increased fluorescence intensity in cathepsin B dose-dependent manner, which was by degradation of the peptide and then liberated Ce6 was activated in the aqueous solution. In animal pulmonary metastasis model using CT26 cells, ICI NC showed superior tumor targetability, i.e., fluorescence intensity was significantly strong in the mouse lung having metastatic tumor. On the contrary, cathepsin-B-deficient carriers such as atezolizumab-Ce6 conjugates or atezolizumab-Ce6/MePEG conjugates showed strong fluorescence intensity in the liver as well as lung. Our proposed ICI NC may be used for theranostic cancer therapy with superior tumor specificity of releasing ICI and Ce6 into tumor microenvironment, thereby showing an efficient inhibitory effect on pulmonary metastasis of CT26 cells.
Collapse
Affiliation(s)
- Young-IL Jeong
- Research
Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Gyeongnam 50612, Republic of Korea
| | - So Young Yoo
- Research
Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Gyeongnam 50612, Republic of Korea
- BIO-IT
Foundry Technology Institute, Pusan National
University, Gumjeong-gu, Busan 46241, Republic
of Korea
| | - Jeong Heo
- Department
of Internal Medicine and Biomedical Research Institute, Pusan National University Hospital, Seo-gu, Busan 49241, Republic of Korea
| | - Dae Hwan Kang
- Research
Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Gyeongnam 50612, Republic of Korea
| |
Collapse
|
26
|
MicroRNA 155 Contributes to Host Immunity against Leishmania donovani but Is Not Essential for Resolution of Infection. Infect Immun 2019; 87:IAI.00307-19. [PMID: 31182615 DOI: 10.1128/iai.00307-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 05/27/2019] [Indexed: 02/07/2023] Open
Abstract
CD4+ T helper 1 (Th1) cells producing interferon gamma (IFN-γ) are critical for the resolution of visceral leishmaniasis (VL). MicroRNA 155 (miR155) promotes CD4+ Th1 responses and IFN-γ production by targeting suppressor of cytokine signaling-1 (SOCS1) and Src homology-2 domain-containing inositol 5-phosphatase 1 (SHIP-1) and therefore could play a role in the resolution of VL. To determine the role of miR155 in VL, we monitored the course of Leishmania donovani infection in miR155 knockout (miR155KO) and wild-type (WT) C57BL/6 mice. miR155KO mice displayed significantly higher liver and spleen parasite loads than WT controls and showed impaired hepatic granuloma formation. However, parasite growth eventually declined in miR155KO mice, suggesting the induction of a compensatory miR155-independent antileishmanial pathway. Leishmania antigen-stimulated splenocytes from miR155KO mice produced significantly lower levels of Th1-associated IFN-γ than controls. Interestingly, at later time points, levels of Th2-associated interleukin-4 (IL-4) and IL-10 were also lower in miR155KO splenocyte supernatants than in WT mice. On the other hand, miR155KO mice displayed significantly higher levels of IFN-γ, iNOS, and TNF-α gene transcripts in their livers than WT mice, indicating that distinct organ-specific antiparasitic mechanisms were involved in control of L. donovani infection in miR155KO mice. Throughout the course of infection, organs of miR155KO mice showed significantly more PDL1-expressing Ly6Chi inflammatory monocytes than WT mice. Conversely, blockade of Ly6Chi inflammatory monocyte recruitment in miR155KO mice significantly reduced parasitic loads, indicating that these cells contributed to disease susceptibility. In conclusion, we found that miR155 contributes to the control of L. donovani but is not essential for infection resolution.
Collapse
|
27
|
Toner K, Bollard CM, Dave H. T-cell therapies for T-cell lymphoma. Cytotherapy 2019; 21:935-942. [PMID: 31320195 DOI: 10.1016/j.jcyt.2019.04.058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 04/21/2019] [Accepted: 04/23/2019] [Indexed: 01/05/2023]
Abstract
T-cell lymphomas represent a subpopulation of non-Hodgkin lymphomas with poor outcomes when treated with conventional chemotherapy. A variety of novel agents have been introduced as new treatment strategies either as first-line treatment or in conjunction with chemotherapy. Immunotherapy has been demonstrated to be a promising area for new therapeutics, including monoclonal antibodies and adoptive cellular therapeutics. T-cell therapeutics have been shown to have significant success in the treatment of B-cell malignancies and are rapidly expanding as potential treatment options for other cancers including T-cell lymphomas. Although treating T-cell lymphomas with T-cell therapeutics has unique challenges, multiple targets are currently being studied both preclinically and in clinical trials.
Collapse
Affiliation(s)
- Keri Toner
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, USA; The George Washington School of Medicine and Health Sciences, Washington, DC, USA
| | - Hema Dave
- Center for Cancer and Immunology Research, Children's National Health System, Washington, DC, USA; The George Washington School of Medicine and Health Sciences, Washington, DC, USA.
| |
Collapse
|
28
|
Zhu X, Yang H, Lang J, Zhang Y. Ras association domain family protein 1a hypermethylation and PD-L1 expression in ovarian cancer: A retrospective study of 112 cases. Eur J Obstet Gynecol Reprod Biol 2019; 240:103-108. [PMID: 31242460 DOI: 10.1016/j.ejogrb.2019.06.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 06/05/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study assessed the interrelationships between Ras association domain family protein 1a (RASSF1A) gene hypermethylation, PD-L1 protein expression, and the clinicopathological characteristics of 112 ovarian cancer (OC) samples. METHODS Formalin-fixed paraffin-embedded OC tissue samples from surgical resection were assessed. Bisulfite pyrosequencing and immunohistochemistry were applied to detect RASSF1A gene methylation and PD-L1 protein expression in tumor cells, respectively. RASSF1A gene methylation and PD-L1 protein expression levels were analyzed against clinicopathological features and prognosis through standard statistical methods. RESULTS Of the 112 OC samples, 49.1% (55/112) exhibited RASSF1A gene hypermethylation. The frequency of RASSF1A hypermethylation was significantly higher in nonserous subtype (73.0%), early stage (66.7%), and nonrecurrent OC (62.9%, p < 0.05). Among all samples, 61.6% (69/112) were positive for PD-L1 protein expression in tumor cells. No significant differences in PD-L1 expression were identified for age, menstrual status, histological type, tumor location, grade, stage, lymph node metastasis, or prognosis (p > 0.05). RASSF1A methylation and PD-L1 expression were not correlated (p > 0.05). CONCLUSIONS This was the first study linking RASSF1A hypermethylation variability to PD-L1 expression and clinicopathological characteristics of OC. Epigenetic alteration of RASSF1A was closely associated with nonserous subtype, early stage, and nonrecurrent OC, indicating that RASSF1A hypermethylation may play a role in early detection of OC. Expression of PD-L1 had no relationship with the studied clinicopathological characteristics or RASSF1A hypermethylation in the 112 OC samples.
Collapse
Affiliation(s)
- Xinxin Zhu
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Huixia Yang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China
| | - Jinghe Lang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yan Zhang
- Department of Obstetrics and Gynecology, Peking University First Hospital, Beijing, China.
| |
Collapse
|
29
|
Abstract
Immunotherapy has significantly advanced the field of oncology in recent decades. Understanding normal immunosurveillance, as well as the ways in which tumor cells have evolved to evade it, has provided the knowledge for development of drugs that allow one's own immune system to target and destroy malignant cells (immunotherapy). Cutaneous malignancies are particularly sensitive to this class of drugs. In a very sensitive anatomic region such as the periocular tissue, where surgical excision may come with significant morbidity, this technology has had a strong impact in the successful treatment of historically challenging tumors.
Collapse
Affiliation(s)
- Larissa A Habib
- a Ophthalmic Plastic Surgery , Massachusetts Eye and Ear Infirmary , Boston , MA , USA.,b Department of Ophthalmology , Harvard Medical School , Boston , MA , USA
| | - Natalie Wolkow
- a Ophthalmic Plastic Surgery , Massachusetts Eye and Ear Infirmary , Boston , MA , USA.,b Department of Ophthalmology , Harvard Medical School , Boston , MA , USA
| | - Suzanne K Freitag
- a Ophthalmic Plastic Surgery , Massachusetts Eye and Ear Infirmary , Boston , MA , USA.,b Department of Ophthalmology , Harvard Medical School , Boston , MA , USA
| | - Michael K Yoon
- a Ophthalmic Plastic Surgery , Massachusetts Eye and Ear Infirmary , Boston , MA , USA.,b Department of Ophthalmology , Harvard Medical School , Boston , MA , USA
| |
Collapse
|
30
|
Soluble PD-1 ligands regulate T-cell function in Waldenstrom macroglobulinemia. Blood Adv 2019; 2:1985-1997. [PMID: 30104397 DOI: 10.1182/bloodadvances.2018021113] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/18/2018] [Indexed: 01/11/2023] Open
Abstract
Although immune checkpoint molecules regulate the progression of certain cancers, their significance in malignant development of Waldenstrom macroglobulinemia (WM), an incurable low-grade B-cell lymphoma, remains unknown. Recently, cytokines in the bone marrow (BM) microenvironment are shown to contribute to the pathobiology of WM. Here, we investigated the impact of cytokines, including interleukin-6 (IL-6) and IL-21, on immune regulation and particularly on the programmed death-1 (PD-1) and its ligands PD-L1 and PD-L2. We showed that IL-21, interferon γ, and IL-6 significantly induced PD-L1 and PD-L2 gene expression in WM cell lines. Increased PD-L1 and PD-L2 messenger RNA was also detected in patients' BM cells. Patients' nonmalignant BM cells, including T cells and monocytes, showed increased PD-L1, but minimal or undetectable PD-L2 surface expression. There was also very modest PD-L1 and PD-L2 surface expression by malignant WM cells, suggesting that ligands are cleaved from the cell surface. Levels of soluble ligands were higher in patients' BM plasma and blood serum than controls. Furthermore, IL-21 and IL-6 increased secreted PD-L1 in the culture media of WM cell lines, implying that elevated levels of soluble PD-1 ligands are cytokine mediated. Soluble PD-1 ligands reduced T-cell proliferation, phosphorylated extracellular signal-regulated kinase and cyclin A levels, mitochondrial adenosine triphosphate production, and spare respiratory capacity. In conclusion, we identify that soluble PD-1 ligands are elevated in WM patients and, in addition to surface-bound ligands in WM BM, could regulate T-cell function. Given the capability of secreted forms to be bioactive at distant sites, soluble PD-1 ligands have the potential to promote disease progression in WM.
Collapse
|
31
|
Pawłowska A, Suszczyk D, Okła K, Barczyński B, Kotarski J, Wertel I. Immunotherapies based on PD-1/PD-L1 pathway inhibitors in ovarian cancer treatment. Clin Exp Immunol 2019; 195:334-344. [PMID: 30582756 PMCID: PMC6378380 DOI: 10.1111/cei.13255] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2018] [Indexed: 12/12/2022] Open
Abstract
Immunotherapies based on anti-programmed death 1/programmed death ligand 1 (PD-1/PD-L1) pathway inhibitors may turn out effective in ovarian cancer (OC) treatment. They can be used in combination with standard therapy and are especially promising in recurrent and platinum-resistant OC. There is growing evidence that the mechanism of the PD-1/PD-L1 pathway can be specific for a particular histological cancer type. Interestingly, the data have shown that the PD-1/PD-L1 pathway blockade may be effective, especially in the endometrioid type of OC. It is important to identify the cause of anti-tumor immune response suppression and exclude its other mechanisms in OC patients. It is also necessary to conduct subsequent studies to confirm in which OC cases the treatment is effective and how to select patients and combine drugs to improve patient survival.
Collapse
Affiliation(s)
- A. Pawłowska
- Tumor Immunology Laboratory, 1st Chair and Department of Oncological Gynaecology and GynaecologyMedical University of LublinLublinPoland
| | - D. Suszczyk
- Tumor Immunology Laboratory, 1st Chair and Department of Oncological Gynaecology and GynaecologyMedical University of LublinLublinPoland
| | - K. Okła
- Tumor Immunology Laboratory, 1st Chair and Department of Oncological Gynaecology and GynaecologyMedical University of LublinLublinPoland
| | - B. Barczyński
- Tumor Immunology Laboratory, 1st Chair and Department of Oncological Gynaecology and GynaecologyMedical University of LublinLublinPoland
| | - J. Kotarski
- Tumor Immunology Laboratory, 1st Chair and Department of Oncological Gynaecology and GynaecologyMedical University of LublinLublinPoland
| | - I. Wertel
- Tumor Immunology Laboratory, 1st Chair and Department of Oncological Gynaecology and GynaecologyMedical University of LublinLublinPoland
| |
Collapse
|
32
|
Reverse signaling via PD-L1 supports malignant cell growth and survival in classical Hodgkin lymphoma. Blood Cancer J 2019; 9:22. [PMID: 30783096 PMCID: PMC6381098 DOI: 10.1038/s41408-019-0185-9] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 12/11/2018] [Accepted: 12/21/2018] [Indexed: 02/07/2023] Open
Abstract
Treatment with programmed death-1 (PD-1) blocking antibodies results in high overall response rates in refractory and relapsed classical Hodgkin lymphoma (cHL) patients, indicating that PD-1/PD-1 ligand interactions are integral to progression of this disease. Given the genetically driven increased PD-L1/2 expression in HL, we hypothesized that reverse signaling through PD-1 ligands may be a potential mechanism contributing to the growth and survival of Hodgkin Reed–Sternberg (HRS) cells in cHL. Our data show that engagement of PD-L1 using an agonistic monoclonal antibody increases cell survival and proliferation and reduces apoptosis in HL cell lines. We show that HL patients have significantly higher serum levels of soluble PD-1 than healthy controls, and find that both membrane-bound and soluble forms of PD-1 are able to induce PD-L1 reverse signaling in HL cell lines. PD-L1 signaling, which is associated with activation of the MAPK pathway and increased mitochondrial oxygen consumption, is reversed by PD-1 blockade. In summary, our data identify inhibition of reverse signaling through PD-L1 as an additional mechanism that accounts for clinical responses to PD-1 blockade in cHL.
Collapse
|
33
|
Xue C, Zhu D, Chen L, Xu Y, Xu B, Zhang D, Jiang J. Expression and prognostic value of PD-L1 and PD-L2 in ovarian cancer. Transl Cancer Res 2019; 8:111-119. [PMID: 35116740 PMCID: PMC8797717 DOI: 10.21037/tcr.2019.01.09] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/05/2019] [Indexed: 12/05/2022]
Abstract
Background In the present study, we aimed to investigate the expression and prognostic value of co-stimulatory molecules, programmed death ligand-1 (PD-L1) and PD-L2, in ovarian cancer (OC). Methods Immunohistochemical (IHC) staining was used to assess the expressions of PD-L1 and PD-L2 in 77 cases of OC, and 10 cases of benign ovarian cyst were employed as negative controls. Moreover, χ2 test was used to analyze the correlation between the PD-L1/PD-L2 expression and clinicopathological parameters. Kaplan-Meier method was used to compare the effects of PD-L1/PD-L2 expression level on the overall survival (OS) of OC patients. Results PD-L1 and PD-L2 were mainly expressed on membrane and in cytoplasm of OC cells. The high-expression rate of PD-L1 and PD-L2 in OC tissues was 44.16% (34/77) and 22.08% (17/77), respectively. The expression of PD-L1 in OC cells was significantly correlated with FIGO stage (P=0.026), while its expression was not significantly correlated with other clinicopathological parameters. There was no significant correlation between PD-L2 and any clinicopathological parameters. Kaplan-Meier survival analysis showed that the OS of high PD-L1 expression group was significantly shorter compared with the low PD-L1 expression group (HR =2.689, 95% CI: 1.400–5.163). Patients with high PD-L2 expression also exhibited significantly shorter OS (HR =2.204, 95% CI: 1.037–4.682). Multivariable analysis displayed that high expression of PD-L1 (HR =2.275, 95% CI: 1.120–4.169), high expression of PD-L2 (HR =2.314, 95% CI: 1.136–4.714) and FIGO stage (HR =11.229, 95% CI: 1.373–91.865) were independent prognostic factors of OC. When negative expressions of both PD-L1 and PD-L2 were used as a combined prognostic factor, the OS was significantly prolonged (HR =3.396, 95% CI: 1.858–6.029). According to our previous studies, patients with negative PD-L1 expression and high T-bet+ TIL infiltration have higher OS than other patients. Patients with positive PD-L1 expression and low T-bet+ TIL infiltration exhibit the shortest OS. Collectively, our findings provided the basis for PD-1/PD-L1 or PD-1/PD-L2 blockade therapy for OC patients. Conclusions Co-stimulatory molecules, PD-L1 and PD-L2, were highly expressed in OC tissues, and their expression levels were correlated with FIGO stage, age and prognosis. These results suggested that PD-L1 and PD-L2 were involved in the occurrence and development of malignant OC, indicating their potential value in clinical diagnosis and prognosis of OC.
Collapse
Affiliation(s)
- Chunyan Xue
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Department of Gynaecology, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Institute of Cell Therapy, Soochow University, Suzhou 215021, China
| | - Dawei Zhu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Institute of Cell Therapy, Soochow University, Suzhou 215021, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Institute of Cell Therapy, Soochow University, Suzhou 215021, China
| | - Yun Xu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Department of Gynaecology, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Institute of Cell Therapy, Soochow University, Suzhou 215021, China
| | - Bin Xu
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Institute of Cell Therapy, Soochow University, Suzhou 215021, China
| | - Dachuan Zhang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Institute of Cell Therapy, Soochow University, Suzhou 215021, China.,Department of Pathology, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, the Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Institute of Cell Therapy, Soochow University, Suzhou 215021, China
| |
Collapse
|
34
|
Wan H, Ma H, Zhu S, Wang F, Tian Y, Ma R, Yang Q, Hu Z, Zhu T, Wang W, Ma Z, Zhang M, Zhong Y, Sun H, Liang Y, Dai H. Developing a Bright NIR-II Fluorophore with Fast Renal Excretion and Its Application in Molecular Imaging of Immune Checkpoint PD-L1. ADVANCED FUNCTIONAL MATERIALS 2018; 28:1804956. [PMID: 31832053 PMCID: PMC6907024 DOI: 10.1002/adfm.201804956] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Indexed: 05/22/2023]
Abstract
Fluorescence imaging in the second near-infrared (NIR-II) window holds impressive advantages of enhanced penetration depth and improved signal-to-noise ratio. Bright NIR-II fluorophores with renal excretion ability and low tissue accumulation are favorable for in vivo molecular imaging applications as they can render the target-mediated molecular imaging process easily distinguishable. Here, a probe (anti-PD-L1-BGP6) comprising a fluorophore (IR-BGP6) covalently bonded to the programmed cell death ligand-1 monoclonal antibody (PD-L1 mAb) for molecular imaging of immune checkpoint PD-L1 (a targeting site upregulated in various tumors for cancer imaging) in the NIR-II window is reported. Through molecular optimization, the bright NIR-II fluorophore IR-BGP6 with fast renal excretion (≈91% excretion in general through urine within the first 10 h postinjection) is developed. The conjugate anti-PD-L1-BGP6 succeeds in profiling PD-L1 expression and realizes efficient noninvasive molecular imaging in vivo, achieving a tumor to normal tissue (T/NT) signal ratio as high as ≈9.5. Compared with the NIR-II fluorophore with high nonspecific tissue accumulation, IR-BGP6 derived PD-L1 imaging significantly enhances the molecular imaging performance, serving as a strong tool for potentially studying underlying mechanism of immunotherapy. The work provides rationales to design renal-excreted NIR-II fluorophores and illustrate their advantages for in vivo molecular imaging.
Collapse
Affiliation(s)
- Hao Wan
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Huilong Ma
- Department of Materials Science and Engineering, South University of Science and Technology of China, Shenzhen 518055, China
| | - Shoujun Zhu
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - FeiFei Wang
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Ye Tian
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Rui Ma
- Department of Materials Science and Engineering, South University of Science and Technology of China, Shenzhen 518055, China
| | - Qinglai Yang
- Department of Materials Science and Engineering, South University of Science and Technology of China, Shenzhen 518055, China
| | - Zhubin Hu
- State Key Laboratory of Precision Spectroscopy, School of Physics and Materials Science, East China Normal University Shanghai 200062, China
| | - Tong Zhu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Weizhi Wang
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Zhuoran Ma
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Mingxi Zhang
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Yeteng Zhong
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Haitao Sun
- State Key Laboratory of Precision Spectroscopy, School of Physics and Materials Science, East China Normal University Shanghai 200062, China
| | - Yongye Liang
- Department of Materials Science and Engineering, South University of Science and Technology of China, Shenzhen 518055, China
| | - Hongjie Dai
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
35
|
Zhang Y, Liu YD, Luo YL, Liu BL, Huang QT, Wang F, Zhong Q. Prognostic Value of Lymphocyte Activation Gene-3 (LAG-3) Expression in Esophageal Squamous Cell Carcinoma. J Cancer 2018; 9:4287-4293. [PMID: 30519331 PMCID: PMC6277627 DOI: 10.7150/jca.26949] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 08/26/2018] [Indexed: 01/01/2023] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a malignant epithelial tumor with a high incidence in East Asia and the Middle East. The outcomes for ESCC patients are usually not optimal due to the recurrence and metastasis. This study is aim to examine the expression and the prognostic value of LAG-3 in ESCC. We applied immunohistochemistry analysis to examine the expression of LAG-3, CD4 and CD8 in 287 ESCC cohorts. Our study demonstrated that the decreased LAG-3 expression was significantly associated with CD4 tumor-infiltrated lymphocytes (TILs) (p=0.000), CD8 TILs (p=0.000), and the advanced clinical stages (p=0.041) by Chi-square analysis. Kaplan-Meier survival analysis revealed that higher LAG-3 expression were positively correlated with a better overall survival (OS) (p=0.010) and better progression free survival (PFS) (p=0.006), especially in the patients at stages T1-2 status (p=0.001, OS; p=0.001, PFS), N0 status (p=0.036, OS; p=0.050, PFS), and early stages (I-II) (p=0.006, OS; p=0.008, PFS). Both high of CD4 TIL /CD8 TIL ratio and LAG-3 expression were correlated with longer OS and PFS. Cox proportional hazards regression analysis showed that LAG-3 is an independent biomarker of survival (HR, 0.724; 95% CI 0.526-0.995; p = 0.047) (p=0.036). Taken together, we found that high expression of LAG-3 was correlated with an improved survival and LAG-3 is an independent predictor of survival, suggesting that LAG-3 may serve as a useful immune marker for the prognosis of ESCC.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pathology, Sun Yat-sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Yong-Dong Liu
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, No. 58th Zhongshan 2nd Road, Guangzhou 510080, China
| | - Yi-Ling Luo
- State Key Laboratory of Oncology in South China, Department of Experimental Research, Sun Yat-sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Bin-Liu Liu
- State Key Laboratory of Oncology in South China, Department of Experimental Research, Sun Yat-sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Qi-Tao Huang
- Department of Pathology, Sun Yat-sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Fang Wang
- Department of Molecular Diagnosis, Sun Yat-sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou 510060, China
| | - Qian Zhong
- State Key Laboratory of Oncology in South China, Department of Experimental Research, Sun Yat-sen University Cancer Center, No. 651 Dongfeng East Road, Guangzhou 510060, China
| |
Collapse
|
36
|
Smith M, García-Martínez E, Pitter MR, Fucikova J, Spisek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Toll-like receptor agonists in cancer immunotherapy. Oncoimmunology 2018; 7:e1526250. [PMID: 30524908 PMCID: PMC6279325 DOI: 10.1080/2162402x.2018.1526250] [Citation(s) in RCA: 177] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Indexed: 12/14/2022] Open
Abstract
Toll-like receptor (TLR) agonists demonstrate therapeutic promise as immunological adjuvants for anticancer immunotherapy. To date, three TLR agonists have been approved by US regulatory agencies for use in cancer patients. Additionally, the potential of hitherto experimental TLR ligands to mediate clinically useful immunostimulatory effects has been extensively investigated over the past few years. Here, we summarize recent preclinical and clinical advances in the development of TLR agonists for cancer therapy.
Collapse
Affiliation(s)
- Melody Smith
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elena García-Martínez
- Hematology and Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Michael R. Pitter
- Department of Medicine and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jitka Fucikova
- Sotio a.c., Prague, Czech Republic
- Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic
- Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- INSERM, U1015, Villejuif, France
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/ Paris V, Paris, France
- Université Pierre et Marie Curie/Paris VI, Paris, France
- INSERM, U1138, Paris, France
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Karolinska Institute, Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Université Paris Descartes/ Paris V, Paris, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
37
|
Zhuang Y, Li S, Wang H, Pi J, Xing Y, Li G. PD-1 blockade enhances radio-immunotherapy efficacy in murine tumor models. J Cancer Res Clin Oncol 2018; 144:1909-1920. [PMID: 30074066 DOI: 10.1007/s00432-018-2723-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/30/2018] [Indexed: 12/23/2022]
Abstract
PURPOSE It has become increasingly clear in cancer treatment that radiotherapy can be enhanced by immunotherapy. In the present study, we evaluated a novel triple combination therapy consisting of local radiotherapy, intratumoral CpG, and systemic PD-1 blockade in lung cancer models. METHODS The efficacy of a novel triple therapy was examined by recording tumor volume and survival time. The immunologic effects of this novel triple therapy were evaluated by the frequency and percentage of immune cells and cytokines using flow cytometry. RESULTS This triple combination proved more effective than its subcomponents and its positive antitumor effects included reducing tumor growth and improving host survival. The antitumor effect was not only observed in directly irradiated tumors but also in at distant tumor sites in a CD8+ T-cell-dependent fashion. Phenotypic analyses of CD8+ T cells revealed that the triple combination therapy increased the percentage of effector memory T cells in the spleen. Furthermore, the combination therapy significantly increased the frequency of IFN-γ and TNF-α-positive-CD8+ tumor-infiltrating lymphocytes (TIL) and mature-activated dendritic cells (DCs) within treated tumors, indicating that the antitumor effects likely depend on the activation of a DC subset specialized in antigen crosspriming to induce cytotoxic lymphocyte (CTLs). In addition, the triple therapy reduced immunosuppressive factors, like regulatory T cells (Tregs) in the spleen and tumor microenvironment while inducing the robust systemic antitumor effect. Finally, the triple treatment was, indeed, well tolerated and had a little effect on the hemogram and lung. CONCLUSIONS These results suggest that this triple therapy promotes a local antitumor immune response with systemic consequences. The efficacy and limited toxicity of this strategy are attractive for clinical translation.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/pharmacology
- Apoptosis
- CD8-Positive T-Lymphocytes/immunology
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/metabolism
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Lewis Lung/therapy
- Cell Proliferation
- Cytokines/metabolism
- Cytotoxicity, Immunologic
- Female
- Interferon-gamma/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Male
- Mice
- Mice, Inbred C57BL
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Radioimmunotherapy
- T-Lymphocytes, Regulatory/immunology
- Tumor Cells, Cultured
- Tumor Microenvironment
Collapse
Affiliation(s)
- Yuan Zhuang
- Department of Radiation Oncology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Sihan Li
- Department of Radiation Oncology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Huihui Wang
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Jingbo Pi
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Yuhui Xing
- Department of Radiation Oncology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China
| | - Guang Li
- Department of Radiation Oncology, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Heping District, Shenyang, 110001, Liaoning, China.
| |
Collapse
|
38
|
Gao Q, Xiang SD, Wilson K, Madondo M, Stephens AN, Plebanski M. Sperm Protein 17 Expression by Murine Epithelial Ovarian Cancer Cells and Its Impact on Tumor Progression. Cancers (Basel) 2018; 10:cancers10080276. [PMID: 30127274 PMCID: PMC6115966 DOI: 10.3390/cancers10080276] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/13/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
The cancer testis antigen sperm protein 17 (Sp17) is a promising antigenic target in epithelial ovarian cancer (EOC) vaccine development. However, its role in ovarian cancer is unclear. We isolated and expanded Sp17+ and Sp17− clones from the murine EOC cell line ID8, and compared their in-vitro cell growth characteristics and in-vivo tumorigenicity. We also examined the potential co-expression of molecules that may influence cancer cell survival and interaction with immune cells. These include stimulatory and immunosuppressive molecules, such as major histocompatibility class I molecules (MHC I), MHC II, cytotoxic T lymphocyte associated antigen-4 (CTLA-4), CD73, CD39, tumor necrosis factor receptor II (TNFRII), signal transducer and activator of transcription 3 (STAT3) and programmed death-ligand 1 (PD-L1). Whilst the presence of Sp17 was not correlated with the ID8 cell proliferation/growth capacity in vitro, it was critical to enable progressive tumor formation in vivo. Flow cytometry revealed that Sp17+ ID8 cells displayed higher expression of both STAT3 and PD-L1, whilst MHC II expression was lower. Moreover, Sp17high (PD-L1+MHCII−) cell populations showed significantly enhanced resistance to Paclitaxel-induced cell death in vitro compared to Sp17low (PD-L1−MHCII+) cells, which was associated in turn with increased STAT3 expression. Together, the data support Sp17 as a factor associated with in-vivo tumor progression and chemo-resistance, validating it as a suitable target for vaccine development.
Collapse
Affiliation(s)
- Qian Gao
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
- Department of Clinical Laboratory, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Sue D Xiang
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria 3168, Australia.
| | - Kirsty Wilson
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
| | - Mutsa Madondo
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
| | - Andrew N Stephens
- Centre for Cancer Research, Hudson Institute of Medical Research, Clayton, Victoria 3168, Australia.
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria 3168, Australia.
| | - Magdalena Plebanski
- Department of Immunology and Pathology, Central Clinical School, Faculty of Medicine, Nursing and Health Science, Monash University, Melbourne, Victoria 3004, Australia.
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria 3083, Australia.
| |
Collapse
|
39
|
Wanderley CW, Colon DF, Luiz JPM, Oliveira FF, Viacava PR, Leite CA, Pereira JA, Silva CM, Silva CR, Silva RL, Speck-Hernandez CA, Mota JM, Alves-Filho JC, Lima-Júnior RC, Cunha TM, Cunha FQ. Paclitaxel reduces tumor growth by reprogramming tumor-associated macrophages to an M1- profile in a TLR4-dependent manner. Cancer Res 2018; 78:5891-5900. [DOI: 10.1158/0008-5472.can-17-3480] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 05/23/2018] [Accepted: 06/18/2018] [Indexed: 12/18/2022]
|
40
|
Li A, Sun S, Song T, Li X, Cheng W, Yao R, Zhang D, Cai Z, Zhang J, Zhai D, Yu C. Targeted therapy and immunotherapy for platinum-refractory advanced ovarian adenosquamous carcinoma: a case report. Onco Targets Ther 2018; 11:3705-3711. [PMID: 29983579 PMCID: PMC6027823 DOI: 10.2147/ott.s162985] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background Ovarian adenosquamous carcinoma is an extremely rare type of ovarian histology. Platinum-refractory disease is also uncommon, but can be fatal because of the lack of available treatment options. To date, there is no study or case report on platinum-refractory ovarian adenosquamous carcinoma or its relevant treatment. Case presentation Herein, we report the case of a 38-year-old Chinese woman with platinum-refractory advanced ovarian adenosquamous carcinoma who received clinical benefit from poly adenosine diphosphate ([ADP] ribose) polymerase and programmed death-1 inhibitors after failure of prior multiline chemotherapies and antiangiogenic agents. The targeted therapy and immunotherapy-controlled disease deterioration and improved performance status. Thus far, the patient has survived longer than 15 months, and she is taking nivolumab as maintenance treatment. Conclusion Targeted therapy and immunotherapy may be options for rare categories of ovarian cancer, but this warrants more clinical evidence of efficacy and toxicity.
Collapse
Affiliation(s)
- Anji Li
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, People's Republic of China.,Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China, ;
| | - Shuai Sun
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China, ;
| | - Tao Song
- Department of Radiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Xi Li
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China, ;
| | - Wen Cheng
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China, ;
| | - Ruipin Yao
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China, ;
| | - Danying Zhang
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China, ;
| | - Zailong Cai
- Department of Biochemistry and Molecular Biology, Second Military Medical University, Shanghai 200433, People's Republic of China
| | - Jie Zhang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, People's Republic of China
| | - Dongxia Zhai
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China, ;
| | - Chaoqin Yu
- Department of Gynecology of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, People's Republic of China, ;
| |
Collapse
|
41
|
Drakes ML, Mehrotra S, Aldulescu M, Potkul RK, Liu Y, Grisoli A, Joyce C, O'Brien TE, Stack MS, Stiff PJ. Stratification of ovarian tumor pathology by expression of programmed cell death-1 (PD-1) and PD-ligand- 1 (PD-L1) in ovarian cancer. J Ovarian Res 2018. [PMID: 29843813 DOI: 10.1186/s13048-018-0414-z] [] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Ovarian cancer is the major cause of death among gynecologic cancers with 75% of patients diagnosed with advanced disease, and only 20% of these patients having a survival duration of five years. Treatments blocking immune checkpoint molecules, programmed cell death (PD-1) or its ligand PD-ligand- I (PD-L1) have produced a beneficial and prolonged effect in a subgroup of these patients. However, there is debate in the literature concerning the prognostic value of the expression of these molecules in tumors, with immunotherapy responsiveness, and survival. We evaluated the immune landscape of the ovarian tumor microenvironment of patients, by measuring the impact of the expression of tumor PD-1, PD-L1 and infiltrating lymphocytes on stage and grade of tumors and survival, in a cohort of 55 patients with gynecologic malignancies. Most patients under study were diagnosed with advanced disease ovarian cancer. RESULTS Our studies revealed that a low density of PD-1 and of PD-L1 expressing cells in tumor tissue were significantly associated with advanced disease (P = 0.028 and P = 0.033, respectively). Moreover, PD-L1 was expressed significantly more often in high grade tumors (41.5%) than in low grade tumors of patients (7.7%) (P = 0.040). The presence of CD3 or of FoxP3 infiltrating cells with PD-L1 in patient tumors did not impact the significance of the association of PD-L1 with high grade tumors (P = 0.040), and our analyses did not show an association between the presence of PD-1 or PD-L1 and survival. CONCLUSIONS We conclude that a subgroup of advanced disease ovarian cancer patients with high grade tumors, expressing PD-L1, may be prime candidates for immunotherapy targeting PD-1 signaling.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Oncology Research Institute, Department of Medicine, Loyola University Chicago, Bldg. 112, Room 232, 2160 South First Avenue, Maywood, IL, 60153, USA.
| | - Swati Mehrotra
- Department of Pathology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Monica Aldulescu
- Department of Pathology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Ronald K Potkul
- Department of Obstetrics and Gynecology, Loyola University Chicago, Maywood, IL, USA
| | - Yueying Liu
- Department of Chemistry & Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Anne Grisoli
- Department of Chemistry & Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Cara Joyce
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL, USA
| | - Timothy E O'Brien
- Department of Mathematics & Statistics, and Institute of Environmental Sustainability, Loyola University Chicago, Chicago, IL, USA
| | - M Sharon Stack
- Department of Chemistry & Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Oncology Research Institute, Department of Medicine, Loyola University Chicago, Bldg. 112, Room 232, 2160 South First Avenue, Maywood, IL, 60153, USA
| |
Collapse
|
42
|
Drakes ML, Mehrotra S, Aldulescu M, Potkul RK, Liu Y, Grisoli A, Joyce C, O'Brien TE, Stack MS, Stiff PJ. Stratification of ovarian tumor pathology by expression of programmed cell death-1 (PD-1) and PD-ligand- 1 (PD-L1) in ovarian cancer. J Ovarian Res 2018; 11:43. [PMID: 29843813 PMCID: PMC5975524 DOI: 10.1186/s13048-018-0414-z] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 05/09/2018] [Indexed: 01/06/2023] Open
Abstract
Background Ovarian cancer is the major cause of death among gynecologic cancers with 75% of patients diagnosed with advanced disease, and only 20% of these patients having a survival duration of five years. Treatments blocking immune checkpoint molecules, programmed cell death (PD-1) or its ligand PD-ligand- I (PD-L1) have produced a beneficial and prolonged effect in a subgroup of these patients. However, there is debate in the literature concerning the prognostic value of the expression of these molecules in tumors, with immunotherapy responsiveness, and survival. We evaluated the immune landscape of the ovarian tumor microenvironment of patients, by measuring the impact of the expression of tumor PD-1, PD-L1 and infiltrating lymphocytes on stage and grade of tumors and survival, in a cohort of 55 patients with gynecologic malignancies. Most patients under study were diagnosed with advanced disease ovarian cancer. Results Our studies revealed that a low density of PD-1 and of PD-L1 expressing cells in tumor tissue were significantly associated with advanced disease (P = 0.028 and P = 0.033, respectively). Moreover, PD-L1 was expressed significantly more often in high grade tumors (41.5%) than in low grade tumors of patients (7.7%) (P = 0.040). The presence of CD3 or of FoxP3 infiltrating cells with PD-L1 in patient tumors did not impact the significance of the association of PD-L1 with high grade tumors (P = 0.040), and our analyses did not show an association between the presence of PD-1 or PD-L1 and survival. Conclusions We conclude that a subgroup of advanced disease ovarian cancer patients with high grade tumors, expressing PD-L1, may be prime candidates for immunotherapy targeting PD-1 signaling. Electronic supplementary material The online version of this article (10.1186/s13048-018-0414-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Oncology Research Institute, Department of Medicine, Loyola University Chicago, Bldg. 112, Room 232, 2160 South First Avenue, Maywood, IL, 60153, USA.
| | - Swati Mehrotra
- Department of Pathology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Monica Aldulescu
- Department of Pathology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Ronald K Potkul
- Department of Obstetrics and Gynecology, Loyola University Chicago, Maywood, IL, USA
| | - Yueying Liu
- Department of Chemistry & Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Anne Grisoli
- Department of Chemistry & Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Cara Joyce
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL, USA
| | - Timothy E O'Brien
- Department of Mathematics & Statistics, and Institute of Environmental Sustainability, Loyola University Chicago, Chicago, IL, USA
| | - M Sharon Stack
- Department of Chemistry & Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Oncology Research Institute, Department of Medicine, Loyola University Chicago, Bldg. 112, Room 232, 2160 South First Avenue, Maywood, IL, 60153, USA
| |
Collapse
|
43
|
Drakes ML, Mehrotra S, Aldulescu M, Potkul RK, Liu Y, Grisoli A, Joyce C, O'Brien TE, Stack MS, Stiff PJ. Stratification of ovarian tumor pathology by expression of programmed cell death-1 (PD-1) and PD-ligand- 1 (PD-L1) in ovarian cancer. J Ovarian Res 2018. [PMID: 29843813 DOI: 10.1186/s13048-018-0414-z]+[] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ovarian cancer is the major cause of death among gynecologic cancers with 75% of patients diagnosed with advanced disease, and only 20% of these patients having a survival duration of five years. Treatments blocking immune checkpoint molecules, programmed cell death (PD-1) or its ligand PD-ligand- I (PD-L1) have produced a beneficial and prolonged effect in a subgroup of these patients. However, there is debate in the literature concerning the prognostic value of the expression of these molecules in tumors, with immunotherapy responsiveness, and survival. We evaluated the immune landscape of the ovarian tumor microenvironment of patients, by measuring the impact of the expression of tumor PD-1, PD-L1 and infiltrating lymphocytes on stage and grade of tumors and survival, in a cohort of 55 patients with gynecologic malignancies. Most patients under study were diagnosed with advanced disease ovarian cancer. RESULTS Our studies revealed that a low density of PD-1 and of PD-L1 expressing cells in tumor tissue were significantly associated with advanced disease (P = 0.028 and P = 0.033, respectively). Moreover, PD-L1 was expressed significantly more often in high grade tumors (41.5%) than in low grade tumors of patients (7.7%) (P = 0.040). The presence of CD3 or of FoxP3 infiltrating cells with PD-L1 in patient tumors did not impact the significance of the association of PD-L1 with high grade tumors (P = 0.040), and our analyses did not show an association between the presence of PD-1 or PD-L1 and survival. CONCLUSIONS We conclude that a subgroup of advanced disease ovarian cancer patients with high grade tumors, expressing PD-L1, may be prime candidates for immunotherapy targeting PD-1 signaling.
Collapse
Affiliation(s)
- Maureen L Drakes
- Cardinal Bernardin Cancer Center, Oncology Research Institute, Department of Medicine, Loyola University Chicago, Bldg. 112, Room 232, 2160 South First Avenue, Maywood, IL, 60153, USA.
| | - Swati Mehrotra
- Department of Pathology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Monica Aldulescu
- Department of Pathology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, USA
| | - Ronald K Potkul
- Department of Obstetrics and Gynecology, Loyola University Chicago, Maywood, IL, USA
| | - Yueying Liu
- Department of Chemistry & Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Anne Grisoli
- Department of Chemistry & Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Cara Joyce
- Department of Public Health Sciences, Loyola University Chicago, Maywood, IL, USA
| | - Timothy E O'Brien
- Department of Mathematics & Statistics, and Institute of Environmental Sustainability, Loyola University Chicago, Chicago, IL, USA
| | - M Sharon Stack
- Department of Chemistry & Biochemistry, Harper Cancer Research Institute, University of Notre Dame, South Bend, IN, USA
| | - Patrick J Stiff
- Cardinal Bernardin Cancer Center, Oncology Research Institute, Department of Medicine, Loyola University Chicago, Bldg. 112, Room 232, 2160 South First Avenue, Maywood, IL, 60153, USA
| |
Collapse
|
44
|
Zöller M. Janus-Faced Myeloid-Derived Suppressor Cell Exosomes for the Good and the Bad in Cancer and Autoimmune Disease. Front Immunol 2018; 9:137. [PMID: 29456536 PMCID: PMC5801414 DOI: 10.3389/fimmu.2018.00137] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 01/16/2018] [Indexed: 12/22/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells originally described to hamper immune responses in chronic infections. Meanwhile, they are known to be a major obstacle in cancer immunotherapy. On the other hand, MDSC can interfere with allogeneic transplant rejection and may dampen autoreactive T cell activity. Whether MDSC-Exosomes (Exo) can cope with the dangerous and potentially therapeutic activities of MDSC is not yet fully explored. After introducing MDSC and Exo, it will be discussed, whether a blockade of MDSC-Exo could foster the efficacy of immunotherapy in cancer and mitigate tumor progression supporting activities of MDSC. It also will be outlined, whether application of native or tailored MDSC-Exo might prohibit autoimmune disease progression. These considerations are based on the steadily increasing knowledge on Exo composition, their capacity to distribute throughout the organism combined with selectivity of targeting, and the ease to tailor Exo and includes open questions that answers will facilitate optimizing protocols for a MDSC-Exo blockade in cancer as well as for strengthening their therapeutic efficacy in autoimmune disease.
Collapse
Affiliation(s)
- Margot Zöller
- Tumor Cell Biology, University Hospital of Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
45
|
Emerging Therapeutics to Overcome Chemoresistance in Epithelial Ovarian Cancer: A Mini-Review. Int J Mol Sci 2017; 18:ijms18102171. [PMID: 29057791 PMCID: PMC5666852 DOI: 10.3390/ijms18102171] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 10/13/2017] [Accepted: 10/16/2017] [Indexed: 12/14/2022] Open
Abstract
Ovarian cancer is the fifth leading cause of cancer death among women and the most lethal gynecologic malignancy. One of the leading causes of death in high-grade serous ovarian cancer (HGSOC) is chemoresistant disease, which may present as intrinsic or acquired resistance to therapies. Here we discuss some of the known molecular mechanisms of chemoresistance that have been exhaustively investigated in chemoresistant ovarian cancer, including drug efflux pump multidrug resistance protein 1 (MDR1), the epithelial–mesenchymal transition, DNA damage and repair capacity. We also discuss novel therapeutics that may address some of the challenges in bringing approaches that target chemoresistant processes from bench to bedside. Some of these new therapies include novel drug delivery systems, targets that may halt adaptive changes in the tumor, exploitation of tumor mutations that leave cancer cells vulnerable to irreversible damage, and novel drugs that target ribosomal biogenesis, a process that may be uniquely different in cancer versus non-cancerous cells. Each of these approaches, or a combination of them, may provide a greater number of positive outcomes for a broader population of HGSOC patients.
Collapse
|