1
|
Xu M, Tu Y, Bi W, Lundberg MZ, Klooster I, Fletcher JA, Ou WB. SETDB1 tumour suppressor roles in near-haploid mesothelioma involve TP53. Br J Cancer 2023; 129:531-540. [PMID: 37369845 PMCID: PMC10403575 DOI: 10.1038/s41416-023-02330-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 05/17/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Mutational inactivation of the SETDB1 histone methyltransferase is found in a subset of mesothelioma, particularly in cases with near-haploidy and TP53 mutations. However, the tumourigenic consequences of SETDB1 inactivation are poorly understood. METHODS In this study, we investigated SETDB1 tumour suppressor functions in mesothelioma and explored biologic relationships between SETDB1 and TP53. RESULTS Immunoblotting of early passage cultures showed that SETDB1 was undetectable in 7 of 8 near-haploid mesotheliomas whereas SETDB1 expression was retained in each of 13 near-diploid mesotheliomas. TP53 aberrations were present in 5 of 8 near-haploid mesotheliomas compared to 2 of 13 near-diploid mesotheliomas, and BAP1 inactivation was demonstrated only in near-diploid mesotheliomas, indicating that near-haploid and near-diploid mesothelioma have distinct molecular and biologic profiles. Lentiviral SETDB1 restoration in near-haploid mesotheliomas (MESO257 and MESO542) reduced cell viability, colony formation, reactive oxygen species levels, proliferative marker cyclin A expression, and inhibited growth of MESO542 xenografts. The combination of SETDB1 restoration with pemetrexed and/or cisplatin treatment additively inhibited tumour growth in vitro and in vivo. Furthermore, SETDB1 restoration upregulated TP53 expression in MESO542 and MESO257, whereas SETDB1 knockdown inhibited mutant TP53 expression in JMN1B near-haploid mesothelioma cells. Likewise, TP53 knockdown inhibited SETDB1 expression. Similarly, immunoblotting evaluations of ten near-diploid mesothelioma biopsies and analysis of TCGA expression profiles showed that SETDB1 expression levels paralleled TP53 expression. CONCLUSION These findings demonstrate that SETDB1 inactivation in near-haploid mesothelioma is generally associated with complete loss of SETDB1 protein expression and dysregulates TP53 expression. Targeting SETDB1 pathways could be an effective therapeutic strategy in these often untreatable tumours.
Collapse
Affiliation(s)
- Mengting Xu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Yuqing Tu
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenhui Bi
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Meijun Z Lundberg
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Isabella Klooster
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jonathan A Fletcher
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Wen-Bin Ou
- Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China.
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Zhang S, Wan X, Lv M, Li C, Chu Q, Wang G. TMEM92 acts as an immune-resistance and prognostic marker in pancreatic cancer from the perspective of predictive, preventive, and personalized medicine. EPMA J 2022; 13:519-534. [PMID: 36061829 PMCID: PMC9437164 DOI: 10.1007/s13167-022-00287-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/13/2022] [Indexed: 10/17/2022]
Abstract
Background Pancreatic cancer presents extremely poor prognosis due to the difficulty of early diagnosis, low resection rate, and high rates of recurrence and metastasis. Immune checkpoint blockades have been widely used in many cancer types but showed limited efficacy in pancreatic cancer. The current study aimed to evaluate the landscape of tumor microenvironment (TME) of pancreatic cancer and identify the potential markers of prognosis and immunotherapy efficacy which might contribute to improve the targeted therapy strategy and efficacy in pancreatic cancer in the context of predictive, preventive, and personalized medicine (PPPM). Methods In the current study, a total of 382 pancreatic samples from the datasets of Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) were selected. LM22 gene signature matrix was applied to quantify the fraction of immune cells based on "CIBERSORT" algorithm. Weighted Gene Co-expression Network Analysis (WGCNA) and Molecular Complex Detection (MCODE) algorithm was applied to confirm the hub-network of immune-resistance phenotype. A nomogram model based on COX and Logistic regression was constructed to evaluate the prognostic value and the predictive value of hub-gene in immune-response. The role of transmembrane protein 92 (TMEM92) in regulating cell proliferation was evaluated by MTS assay. Western blot and Real-time PCR were applied to assess the biological effects of PD-L1 inhibition by TMEM92. Moreover, the effect of TMEM92 in immunotherapy was evaluated with PBMC co-culture and by MTS assay. Results Two tumor-infiltrating immune cell (TIIC) phenotypes were identified and a weighted gene co-expression network was constructed to confirm the 167 gene signatures correlated with immune-resistance TIIC subtype. TMEM92 was further identified as a core gene of 167 gene signature network based on MCODE algorithm. High TMEM92 expression was significantly correlated with unfavorable prognosis, characterizing by immune resistance. A nomogram model and external validation confirmed that TMEM92 was an independent prognostic factor in pancreatic cancer. An elevated tumor mutation burden (TMB), mostly is consistent with commonly mutations of KRAS and TP53, was found in the high TMEM92 group. The predictive role of TMEM92 in immunotherapeutic response was also confirmed by IMvigor210 datasets. In addition, the specific biological roles of TMEM92 in cancer was explored in vitro. The results showed that abnormal overexpression of TMEM92 was significantly associated with the poor survival rate of pancreatic cancer. Moreover, we demonstrated that TMEM92 inhibit tumour immune responses of the anti-PD-1 antibody with PBMC co-culture. Conclusion The current study explored for the first time the immune-resistance phenotype of pancreatic cancer and identified TMEM92 as an innovative marker in predicting clinical outcomes and immunotherapeutic efficacy. These findings not only help to recognize high-risk and immune-resistance population which could be supplied targeted prevention, but also provide personalized medical services by intervening TMEM92 function to improve the prognosis of pancreatic cancer. In addition, the biological role of TMEM92 might reveal the potential molecular mechanisms of pancreatic cancer and lead to a novel sight for development of a PPPM approach for pancreatic cancer management. Supplementary Information The online version contains supplementary material available at 10.1007/s13167-022-00287-0.
Collapse
Affiliation(s)
- Simeng Zhang
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001 China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001 China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, 110001 China
| | - Xing Wan
- Department of Medical Oncology, the First Hospital of Dalian Medical University, Dalian, 116000 China
| | - Mengzhu Lv
- Department of Plastic Surgery, the First Hospital of China Medical University, Shenyang, 110001 China
| | - Ce Li
- Department of Medical Oncology, the First Hospital of China Medical University, Shenyang, 110001 China
- Key Laboratory of Anticancer Drugs and Biotherapy of Liaoning Province, the First Hospital of China Medical University, Shenyang, 110001 China
- Liaoning Province Clinical Research Center for Cancer, Shenyang, 110001 China
| | - Qiaoyun Chu
- School of Basic Medicine, Capital Medical University, Beijing, 100069 China
| | - Guan Wang
- Department of Radiology, the First Affiliated Hospital of China Medical University, Shenyang, 110001 China
| |
Collapse
|
3
|
Rovers S, Janssens A, Raskin J, Pauwels P, van Meerbeeck JP, Smits E, Marcq E. Recent Advances of Immune Checkpoint Inhibition and Potential for (Combined) TIGIT Blockade as a New Strategy for Malignant Pleural Mesothelioma. Biomedicines 2022; 10:673. [PMID: 35327475 PMCID: PMC8945074 DOI: 10.3390/biomedicines10030673] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 12/13/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a fatal cancer type that affects the membranes lining the lungs, and is causally associated with asbestos exposure. Until recently, the first-line treatment consisted of a combination of chemotherapeutics that only had a limited impact on survival, and had not been improved in decades. With the recent approval of combined immune checkpoint inhibition for MPM, promising new immunotherapeutic strategies are now emerging for this disease. In this review, we describe the current preclinical and clinical evidence of various immune checkpoint inhibitors in MPM. We will consider the advantages of combined immune checkpoint blockade in comparison with single agent checkpoint inhibitor drugs. Furthermore, recent evidence suggests a role for T cell immunoglobulin and ITIM domain (TIGIT), an inhibitory immunoreceptor, as a novel target for immunotherapy. As this novel immune checkpoint remains largely unexplored in mesothelioma, we will discuss the potential of TIGIT blockade as an alternative therapeutic approach for MPM. This review will emphasize the necessity for new and improved treatments for MPM, while highlighting the recent advances and future perspectives of combined immune checkpoint blockade, particularly aimed at PD-L1 and TIGIT.
Collapse
Affiliation(s)
- Sophie Rovers
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium; (A.J.); (P.P.); (J.P.v.M.); (E.S.); (E.M.)
| | - Annelies Janssens
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium; (A.J.); (P.P.); (J.P.v.M.); (E.S.); (E.M.)
- Department of Thoracic Oncology, Antwerp University Hospital (UZA), 2650 Edegem, Belgium;
| | - Jo Raskin
- Department of Thoracic Oncology, Antwerp University Hospital (UZA), 2650 Edegem, Belgium;
| | - Patrick Pauwels
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium; (A.J.); (P.P.); (J.P.v.M.); (E.S.); (E.M.)
- Department of Pathology, Antwerp University Hospital (UZA), 2650 Edegem, Belgium
| | - Jan P. van Meerbeeck
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium; (A.J.); (P.P.); (J.P.v.M.); (E.S.); (E.M.)
- Department of Thoracic Oncology, Antwerp University Hospital (UZA), 2650 Edegem, Belgium;
| | - Evelien Smits
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium; (A.J.); (P.P.); (J.P.v.M.); (E.S.); (E.M.)
| | - Elly Marcq
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium; (A.J.); (P.P.); (J.P.v.M.); (E.S.); (E.M.)
| |
Collapse
|
4
|
Štrbac D, Dolžan V. Novel and Future Treatment Options in Mesothelioma: A Systematic Review. Int J Mol Sci 2022; 23:1975. [PMID: 35216091 PMCID: PMC8874564 DOI: 10.3390/ijms23041975] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 01/30/2022] [Accepted: 02/03/2022] [Indexed: 12/23/2022] Open
Abstract
Mesothelioma is a rare tumor, frequently associated with asbestos exposure, arising from pleura and peritoneum. Traditionally, diagnosis and treatment have been difficult in a clinical setting. The treatment is based on a trimodal approach involving surgery, chemotherapy, and radiotherapy. The introduction of chemotherapy improved the overall survival. However, the regimen of pemetrexed/cisplatin doublet has not been changed as a standard treatment since 2004. Novel combinations of ipilimumab and nivolumab have only been approved for clinical use in late 2020. The aim of this review was to systematically summarize findings on novel treatment options in mesothelioma. We searched available medical databases online, such as PubMed and Clinicaltrials.gov, to systematically review the literature on novel approaches in immunotherapy, vaccines, and Chimeric Antigen Receptor (CAR)-T cell therapy in mesothelioma. We manually screened 1127 articles on PubMed and 450 trials on ClinicalTrials.gov, and 24 papers and 12 clinical trials published in the last ten years were included in this review. Immunotherapy that was swiftly introduced to treat other thoracic malignancies was slow to reach desirable survival endpoints in mesothelioma, possibly due to limited patient numbers. Novel treatment approaches, such as CAR-T cell therapy, are being investigated. As the incidence of mesothelioma is still rising globally, novel treatment options based on a better understanding of the tumor microenvironment and the genetic drivers that modulate it are needed to support future precision-based therapies.
Collapse
Affiliation(s)
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, Slovenia
| |
Collapse
|
5
|
Désage AL, Karpathiou G, Peoc’h M, Froudarakis ME. The Immune Microenvironment of Malignant Pleural Mesothelioma: A Literature Review. Cancers (Basel) 2021; 13:3205. [PMID: 34206956 PMCID: PMC8269097 DOI: 10.3390/cancers13133205] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/16/2021] [Accepted: 06/22/2021] [Indexed: 12/13/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare and aggressive tumour with a poor prognosis, associated with asbestos exposure. Nowadays, treatment is based on chemotherapy with a median overall survival of less than two years. This review highlights the main characteristics of the immune microenvironment in MPM with special emphasis on recent biological advances. The MPM microenvironment is highly infiltrated by tumour-associated macrophages, mainly M2-macrophages. In line with infiltration by M2-macrophages, which contribute to immune suppression, other effectors of innate immune response are deficient in MPM, such as dendritic cells or natural killer cells. On the other hand, tumour infiltrating lymphocytes (TILs) are also found in MPM, but CD4+ and CD8+ TILs might have decreased cytotoxic effects through T-regulators and high expression of immune checkpoints. Taken together, the immune microenvironment is particularly heterogeneous and can be considered as mainly immunotolerant or immunosuppressive. Therefore, identifying molecular vulnerabilities is particularly relevant to the improvement of patient outcomes and the assessment of promising treatment approaches.
Collapse
Affiliation(s)
- Anne-Laure Désage
- Department of Pulmonology and Thoracic Oncology, North Hospital, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France;
| | - Georgia Karpathiou
- Pathology, North Hospital, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France; (G.K.); (M.P.)
| | - Michel Peoc’h
- Pathology, North Hospital, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France; (G.K.); (M.P.)
| | - Marios E. Froudarakis
- Department of Pulmonology and Thoracic Oncology, North Hospital, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France;
| |
Collapse
|
6
|
Cantini L, Pecci F, Merloni F, Lanese A, Lenci E, Paoloni F, Aerts JG, Berardi R. Old but gold: the role of drug combinations in improving response to immune check-point inhibitors in thoracic malignancies beyond NSCLC. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:1-25. [PMID: 36046087 PMCID: PMC9400728 DOI: 10.37349/etat.2021.00030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 12/08/2020] [Indexed: 12/02/2022] Open
Abstract
The introduction of immune checkpoint inhibitors (ICIs) in non-oncogene addicted non-small cell lung cancer (NSCLC) has revolutionized the treatment scenario and led to a meaningful improvement in patient prognosis. Disappointingly, the success of ICI therapy in NSCLC has not been fully replicated in other thoracic malignancies as small cell lung cancer (SCLC), malignant pleural mesothelioma (MPM), and thymic epithelial tumors (TETs), due to the peculiar biological features of these disease and to the difficulties in the conduction of well-designed, biomarker-driven clinical trials. Therefore, combination strategies of ICIs plus conventional therapies (either chemotherapy, alternative ICIs or targeted agents) have been implemented. Although first approvals of ICI therapy have been recently granted in SCLC and MPM (in combination with chemotherapy and different ICIs), results remain somewhat modest and limited to a small proportion of patients. This work reviews the trial results of ICI therapy in mesothelioma, SCLC, and TETs and discusses the potential of combining ICIs with old drugs.
Collapse
Affiliation(s)
- Luca Cantini
- Clinical Oncology, Università Politecnica delle Marche, A.O.U. Ospedali Riuniti, 60126 Ancona, Italy
| | - Federica Pecci
- Clinical Oncology, Università Politecnica delle Marche, A.O.U. Ospedali Riuniti, 60126 Ancona, Italy
| | - Filippo Merloni
- Clinical Oncology, Università Politecnica delle Marche, A.O.U. Ospedali Riuniti, 60126 Ancona, Italy
| | - Andrea Lanese
- Clinical Oncology, Università Politecnica delle Marche, A.O.U. Ospedali Riuniti, 60126 Ancona, Italy
| | - Edoardo Lenci
- Clinical Oncology, Università Politecnica delle Marche, A.O.U. Ospedali Riuniti, 60126 Ancona, Italy
| | - Francesco Paoloni
- Clinical Oncology, Università Politecnica delle Marche, A.O.U. Ospedali Riuniti, 60126 Ancona, Italy
| | - Joachim G.J.V. Aerts
- Department of Pulmonary Medicine, Erasmus MC, 3015 CE Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, Erasmus MC, 3015 CE Rotterdam, The Netherlands
| | - Rossana Berardi
- Clinical Oncology, Università Politecnica delle Marche, A.O.U. Ospedali Riuniti, 60126 Ancona, Italy
| |
Collapse
|
7
|
The Search for an Interesting Partner to Combine with PD-L1 Blockade in Mesothelioma: Focus on TIM-3 and LAG-3. Cancers (Basel) 2021; 13:cancers13020282. [PMID: 33466653 PMCID: PMC7838786 DOI: 10.3390/cancers13020282] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/06/2021] [Accepted: 01/11/2021] [Indexed: 12/29/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer that is causally associated with previous asbestos exposure in most afflicted patients. The prognosis of patients remains dismal, with a median overall survival of only 9-12 months, due to the limited effectiveness of any conventional anti-cancer treatment. New therapeutic strategies are needed to complement the limited armamentarium against MPM. We decided to focus on the combination of different immune checkpoint (IC) blocking antibodies (Abs). Programmed death-1 (PD-1), programmed death ligand-1 (PD-L1), T-cell immunoglobulin mucin-3 (TIM-3), and lymphocyte activation gene-3 (LAG-3) blocking Abs were tested as monotherapies, and as part of a combination strategy with a second IC inhibitor. We investigated their effect in vitro by examining the changes in the immune-related cytokine secretion profile of supernatant collected from treated allogeneic MPM-peripheral blood mononuclear cell (PBMC) co-cultures. Based on our in vitro results of cytokine secretion, and flow cytometry data that showed a significant upregulation of PD-L1 on PBMC after co-culture, we chose to further investigate the combinations of anti PD-L1 + anti TIM-3 versus anti PD-L1 + anti LAG-3 therapies in vivo in the AB1-HA BALB/cJ mesothelioma mouse model. PD-L1 monotherapy, as well as its combination with LAG-3 blockade, resulted in in-vivo delayed tumor growth and significant survival benefit.
Collapse
|
8
|
Yang Y, Wang X, Bai Y, Feng D, Li A, Tang Y, Wei X, Han P. Programmed death-ligand 2 (PD-L2) expression in bladder cancer. Urol Oncol 2020; 38:603.e9-603.e15. [PMID: 32151519 DOI: 10.1016/j.urolonc.2020.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/05/2020] [Accepted: 01/08/2020] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Immune checkpoint blockade is an optional and effective therapy for bladder cancer. The present study was aimed to investigate the expression of programmed death ligand-2 (PD-L2) in patients with bladder cancer. MATERIALS AND METHODS Paraffin-embedded tissues of 92 patients with bladder cancer were obtained. Then immunohistochemistry of PD-L2 was performed. The expression intensity of PD-L2 was defined with score 0, 1, 2 and 3, in compliance with negative, weak, moderate, and strong, respectively. The association of PD-L2 expression with clinical characteristics was analyzed. A P < 0.05 was considered as significantly different. RESULTS By defining the expression intensity of PD-L2 with score 0 to 3, 73.9% of patients (68/92) had a positive expression of PD-L2, and 43.5% (40/92) had a mediate or strong expression. Furthermore, high expression PD-L2 (mediate or strong expression) was more common among patients ≤ 70 y (P = 0.038) and those with smoke history (P = 0.045). The univariate Kaplan-Meier analysis indicated that high expression of PD-L2 was associated with both shorter overall survival (OS) (78.3 vs. 60.3 months; P = 0.037) and shorter disease-free survival (44.3 vs. 22.5 months; P = 0.004). The multivariate COX regression model showed that high expression of PD-L2 was a poor factor of disease-free survival (hazard ratio = 0.537, 95%CI 0.322-0.898; P = 0.018), but not OS (hazard ratio = 0.565, 95%CI 0.253-1.262; P = 0.164). CONCLUSION Bladder cancer had a high expression of PD-L2. And high expression of PD-L2 may indicate worse prognosis. It may be a potential immunotherapeutic target of immune checkpoint blockade for bladder cancer.
Collapse
Affiliation(s)
- Yubo Yang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiaoming Wang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yunjin Bai
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Dechao Feng
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ao Li
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yin Tang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xin Wei
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Ping Han
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
9
|
Bailly C, Thuru X, Quesnel B. Combined cytotoxic chemotherapy and immunotherapy of cancer: modern times. NAR Cancer 2020; 2:zcaa002. [PMID: 34316682 PMCID: PMC8209987 DOI: 10.1093/narcan/zcaa002] [Citation(s) in RCA: 151] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/24/2020] [Accepted: 01/29/2020] [Indexed: 12/15/2022] Open
Abstract
Monoclonal antibodies targeting programmed cell death 1/programmed cell death ligand 1 (PD-1/PD-L1) immune checkpoints have improved the treatments of cancers. However, not all patients equally benefit from immunotherapy. The use of cytotoxic drugs is practically inevitable to treat advanced cancers and metastases. The repertoire of cytotoxics includes 80 products that principally target nucleic acids or the microtubule network in rapidly proliferating tumor cells. Paradoxically, many of these compounds tend to become essential to promote the activity of immunotherapy and to offer a sustained therapeutic effect. We have analyzed each cytotoxic drug with respect to effect on expression and function of PD-(L)1. The major cytotoxic drugs—carboplatin, cisplatin, cytarabine, dacarbazine, docetaxel, doxorubicin, ecteinascidin, etoposide, fluorouracil, gemcitabine, irinotecan, oxaliplatin, paclitaxel and pemetrexed—all have the capacity to upregulate PD-L1 expression on cancer cells (via the generation of danger signals) and to promote antitumor immunogenicity, via activation of cytotoxic T lymphocytes, maturation of antigen-presenting cells, depletion of immunosuppressive regulatory T cells and/or expansion of myeloid-derived suppressor cells. The use of ‘immunocompatible’ cytotoxic drugs combined with anti-PD-(L)1 antibodies is a modern approach, not only for increasing the direct killing of cancer cells, but also as a strategy to minimize the activation of immunosuppressive and cancer cell prosurvival program responses.
Collapse
Affiliation(s)
| | - Xavier Thuru
- Centre de Recherche Jean-Pierre Aubert, INSERM, University of Lille, UMR-S 1172, CHU Lille, 59045 Lille, France
| | - Bruno Quesnel
- Centre de Recherche Jean-Pierre Aubert, INSERM, University of Lille, UMR-S 1172, CHU Lille, 59045 Lille, France
| |
Collapse
|
10
|
Rébé C, Demontoux L, Pilot T, Ghiringhelli F. Platinum Derivatives Effects on Anticancer Immune Response. Biomolecules 2019; 10:E13. [PMID: 31861811 PMCID: PMC7022223 DOI: 10.3390/biom10010013] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/16/2019] [Accepted: 12/16/2019] [Indexed: 12/12/2022] Open
Abstract
Along with surgery and radiotherapy, chemotherapeutic agents belong to the therapeutic arsenal in cancer treatment. In addition to their direct cytotoxic effects, these agents also impact the host immune system, which might enhance or counteract their antitumor activity. The platinum derivative compounds family, mainly composed of carboplatin, cisplatin and oxaliplatin, belongs to the chemotherapeutical arsenal used in numerous cancer types. Here, we will focus on the effects of these molecules on antitumor immune response. These compounds can induce or not immunogenic cell death (ICD), and some strategies have been found to induce or further enhance it. They also regulate immune cells' fate. Platinum derivatives can lead to their activation. Additionally, they can also dampen immune cells by selective killing or inhibiting their activity, particularly by modulating immune checkpoints' expression.
Collapse
Affiliation(s)
- Cédric Rébé
- Platform of Transfer in Cancer Biology, Centre Georges-François Leclerc, F-21000 Dijon, France
- University of Bourgogne-Franche-Comté, F-21000 Dijon, France; (L.D.); (T.P.); (F.G.)
- INSERM LNC-UMR1231, F-21000 Dijon, France
| | - Lucie Demontoux
- University of Bourgogne-Franche-Comté, F-21000 Dijon, France; (L.D.); (T.P.); (F.G.)
- INSERM LNC-UMR1231, F-21000 Dijon, France
| | - Thomas Pilot
- Platform of Transfer in Cancer Biology, Centre Georges-François Leclerc, F-21000 Dijon, France
- University of Bourgogne-Franche-Comté, F-21000 Dijon, France; (L.D.); (T.P.); (F.G.)
- INSERM LNC-UMR1231, F-21000 Dijon, France
| | - François Ghiringhelli
- Platform of Transfer in Cancer Biology, Centre Georges-François Leclerc, F-21000 Dijon, France
- University of Bourgogne-Franche-Comté, F-21000 Dijon, France; (L.D.); (T.P.); (F.G.)
- INSERM LNC-UMR1231, F-21000 Dijon, France
| |
Collapse
|