1
|
Siozopoulou V, Smits E, Zwaenepoel K, Liu J, Pouliakis A, Pauwels PA, Marcq E. PD-1, PD-L1, IDO, CD70 and microsatellite instability as potential targets to prevent immune evasion in sarcomas. Immunotherapy 2023; 15:1257-1273. [PMID: 37661910 DOI: 10.2217/imt-2022-0049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2023] Open
Abstract
Background: Soft tissue and bone sarcomas are rare entities, hence, standardized therapeutic strategies are difficult to assess. Materials & methods: Immunohistochemistry was performed on 68 sarcoma samples to assess the expression of PD-1, PD-L1, IDO and CD70 in different tumor compartments and molecular analysis was performed to assess microsatellite instability status. Results: PD-1/PD-L1, IDO and CD70 pathways are at play in the immune evasion of sarcomas in general. Soft tissue sarcomas more often show an inflamed phenotype compared with bone sarcomas. Specific histologic sarcoma types show high expression levels of different markers. Finally, this is the first presentation of a microsatellite instability-high Kaposi sarcoma. Discussion/conclusion: Immune evasion occurs in sarcomas. Specific histologic types might benefit from immunotherapy, for which further investigation is needed.
Collapse
Affiliation(s)
- Vasiliki Siozopoulou
- Department of Pathology, Antwerp University Hospital, Edegem, 2650, Belgium
- Center for Oncological Research, Integrated Personalized & Precision Oncology Network, University of Antwerp, Wilrijk, 2610, Belgium
| | - Evelien Smits
- Center for Oncological Research, Integrated Personalized & Precision Oncology Network, University of Antwerp, Wilrijk, 2610, Belgium
- Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, 2650, Belgium
| | - Karen Zwaenepoel
- Department of Pathology, Antwerp University Hospital, Edegem, 2650, Belgium
- Center for Oncological Research, Integrated Personalized & Precision Oncology Network, University of Antwerp, Wilrijk, 2610, Belgium
| | - Jimmy Liu
- Department of Pathology, Antwerp University Hospital, Edegem, 2650, Belgium
| | - Abraham Pouliakis
- Second Department of Pathology, National & Kapodistrian University of Athens, "Attikon" University Hospital, Athens, 12464, Greece
| | - Patrick A Pauwels
- Department of Pathology, Antwerp University Hospital, Edegem, 2650, Belgium
- Center for Oncological Research, Integrated Personalized & Precision Oncology Network, University of Antwerp, Wilrijk, 2610, Belgium
| | - Elly Marcq
- Center for Oncological Research, Integrated Personalized & Precision Oncology Network, University of Antwerp, Wilrijk, 2610, Belgium
| |
Collapse
|
2
|
Sun B, Dong Y, Xu J, Wang Z. Current status and progress in immunotherapy for malignant pleural mesothelioma. Chronic Dis Transl Med 2022; 8:91-99. [PMID: 35774429 PMCID: PMC9215716 DOI: 10.1002/cdt3.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/09/2021] [Indexed: 11/09/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare and aggressive malignant disease. Currently, the platinum doublet of pemetrexed and cisplatin is the standard first-line treatment for unresectable MPM. However, recent promising results of immunotherapy have markedly changed the landscape of MPM treatment. Further, the ongoing innovative therapeutic strategies are expected to expand the range of treatment options; however, several questions remain unanswered. First, establishing predictive biomarkers with high potency is urgently needed to optimize the patient selection process. Second, further exploration of the combination algorithm is expected to unveil more effective and safe regimens. Moreover, other dilemmas, such as the resistance mechanism of immunotherapy and the role of immunotherapy in perioperative settings, still warrant further exploration.
Collapse
Affiliation(s)
- Boyang Sun
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100021China
| | - Yiting Dong
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100021China
| | - Jiachen Xu
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100021China
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing100021China
| |
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
|
Wu M, Huang Q, Xie Y, Wu X, Ma H, Zhang Y, Xia Y. Improvement of the anticancer efficacy of PD-1/PD-L1 blockade via combination therapy and PD-L1 regulation. J Hematol Oncol 2022; 15:24. [PMID: 35279217 PMCID: PMC8917703 DOI: 10.1186/s13045-022-01242-2] [Citation(s) in RCA: 237] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/22/2022] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint molecules are promising anticancer targets, among which therapeutic antibodies targeting the PD-1/PD-L1 pathway have been widely applied to cancer treatment in clinical practice and have great potential. However, this treatment is greatly limited by its low response rates in certain cancers, lack of known biomarkers, immune-related toxicity, innate and acquired drug resistance, etc. Overcoming these limitations would significantly expand the anticancer applications of PD-1/PD-L1 blockade and improve the response rate and survival time of cancer patients. In the present review, we first illustrate the biological mechanisms of the PD-1/PD-L1 immune checkpoints and their role in the healthy immune system as well as in the tumor microenvironment (TME). The PD-1/PD-L1 pathway inhibits the anticancer effect of T cells in the TME, which in turn regulates the expression levels of PD-1 and PD-L1 through multiple mechanisms. Several strategies have been proposed to solve the limitations of anti-PD-1/PD-L1 treatment, including combination therapy with other standard treatments, such as chemotherapy, radiotherapy, targeted therapy, anti-angiogenic therapy, other immunotherapies and even diet control. Downregulation of PD-L1 expression in the TME via pharmacological or gene regulation methods improves the efficacy of anti-PD-1/PD-L1 treatment. Surprisingly, recent preclinical studies have shown that upregulation of PD-L1 in the TME also improves the response and efficacy of immune checkpoint blockade. Immunotherapy is a promising anticancer strategy that provides novel insight into clinical applications. This review aims to guide the development of more effective and less toxic anti-PD-1/PD-L1 immunotherapies.
Collapse
Affiliation(s)
- Mengling Wu
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Qianrui Huang
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yao Xie
- Department of Obstetrics and Gynaecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China.,Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Xuyi Wu
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, 610041, China
| | - Hongbo Ma
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yiwen Zhang
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Yong Xia
- Department of Rehabilitation Medicine, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, China. .,Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, 610041, China.
| |
Collapse
|
5
|
Lu C, Yang D, Klement JD, Colson YL, Oberlies NH, Pearce CJ, Colby AH, Grinstaff MW, Ding HF, Shi H, Liu K. G6PD functions as a metabolic checkpoint to regulate granzyme B expression in tumor-specific cytotoxic T lymphocytes. J Immunother Cancer 2022; 10:jitc-2021-003543. [PMID: 35017152 PMCID: PMC8753452 DOI: 10.1136/jitc-2021-003543] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2021] [Indexed: 12/19/2022] Open
Abstract
Background Granzyme B is a key effector of cytotoxic T lymphocytes (CTLs), and its expression level positively correlates with the response of patients with mesothelioma to immune checkpoint inhibitor immunotherapy. Whether metabolic pathways regulate Gzmb expression in CTLs is incompletely understood. Methods A tumor-specific CTL and tumor coculture model and a tumor-bearing mouse model were used to determine the role of glucose-6-phosphate dehydrogenase (G6PD) in CTL function and tumor immune evasion. A link between granzyme B expression and patient survival was analyzed in human patients with epithelioid mesothelioma. Results Mesothelioma cells alone are sufficient to activate tumor-specific CTLs and to enhance aerobic glycolysis to induce a PD-1hi Gzmblo CTL phenotype. However, inhibition of lactate dehydrogenase A, the key enzyme of the aerobic glycolysis pathway, has no significant effect on tumor-induced CTL activation. Tumor cells induce H3K9me3 deposition at the promoter of G6pd, the gene that encodes the rate-limiting enzyme G6PD in the pentose phosphate pathway, to downregulate G6pd expression in tumor-specific CTLs. G6PD activation increases acetyl-coenzyme A (CoA) production to increase H3K9ac deposition at the Gzmb promoter and to increase Gzmb expression in tumor-specific CTLs converting them from a Gzmblo to a Gzmbhi phenotype, thus increasing CTL tumor lytic activity. Activation of G6PD increases Gzmb+ tumor-specific CTLs and suppresses tumor growth in tumor-bearing mice. Consistent with these findings, GZMB expression level was found to correlate with increased survival in patients with epithelioid mesothelioma. Conclusion G6PD is a metabolic checkpoint in tumor-activated CTLs. The H3K9me3/G6PD/acetyl-CoA/H3K9ac/Gzmb pathway is particularly important in CTL activation and immune evasion in epithelioid mesothelioma.
Collapse
Affiliation(s)
- Chunwan Lu
- School of Life Sciences, Tianjin University, Tianjin, China .,Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, USA
| | - Dafeng Yang
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, USA.,Georgia Cancer Center, Medical College of Georgia, Augusta, GA, USA.,Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - John D Klement
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, USA.,Georgia Cancer Center, Medical College of Georgia, Augusta, GA, USA.,Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Yolonda L Colson
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholas H Oberlies
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, NC, USA
| | | | - Aaron H Colby
- Ionic Pharmaceuticals, Brookline, MA, USA.,Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Mark W Grinstaff
- Ionic Pharmaceuticals, Brookline, MA, USA.,Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Han-Fei Ding
- Georgia Cancer Center, Medical College of Georgia, Augusta, GA, USA
| | - Huidong Shi
- Georgia Cancer Center, Medical College of Georgia, Augusta, GA, USA
| | - Kebin Liu
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA, USA .,Georgia Cancer Center, Medical College of Georgia, Augusta, GA, USA.,Charlie Norwood VA Medical Center, Augusta, GA, USA
| |
Collapse
|
6
|
Terenziani R, Zoppi S, Fumarola C, Alfieri R, Bonelli M. Immunotherapeutic Approaches in Malignant Pleural Mesothelioma. Cancers (Basel) 2021; 13:2793. [PMID: 34199722 PMCID: PMC8200040 DOI: 10.3390/cancers13112793] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023] Open
Abstract
Malignant pleural mesothelioma (MPM) is a rare and aggressive malignant disease affecting the mesothelium, commonly associated to asbestos exposure. The current therapeutic actions, based on cisplatin/pemetrexed treatment, are limited due to the late stage at which most patients are diagnosed and to the intrinsic chemo-resistance of the tumor. Another relevant point is the absence of approved therapies in the second line setting following progression of MPM after chemotherapy. Considering the poor prognosis of the disease and the fact that the incidence of this tumor is expected to increase in the next decade, novel therapeutic approaches are urgently needed. In the last few years, several studies have investigated the efficacy and safety of immune-checkpoint inhibitors (ICIs) in the treatment of unresectable advanced MPM, and a number of trials with immunotherapeutic agents are ongoing in both first line and second line settings. In this review, we describe the most promising emerging immunotherapy treatments for MPM (ICIs, engineered T cells to express chimeric antigen receptors (CARs), dendritic cells (DCs) vaccines), focusing on the biological and immunological features of this tumor as well as on the issues surrounding clinical trial design.
Collapse
Affiliation(s)
| | | | | | - Roberta Alfieri
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (R.T.); (S.Z.); (C.F.)
| | - Mara Bonelli
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy; (R.T.); (S.Z.); (C.F.)
| |
Collapse
|
7
|
Gray SG. Emerging avenues in immunotherapy for the management of malignant pleural mesothelioma. BMC Pulm Med 2021; 21:148. [PMID: 33952230 PMCID: PMC8097826 DOI: 10.1186/s12890-021-01513-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/25/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The role of immunotherapy in cancer is now well-established, and therapeutic options such as checkpoint inhibitors are increasingly being approved in many cancers such as non-small cell lung cancer (NSCLC). Malignant pleural mesothelioma (MPM) is a rare orphan disease associated with prior exposure to asbestos, with a dismal prognosis. Evidence from clinical trials of checkpoint inhibitors in this rare disease, suggest that such therapies may play a role as a treatment option for a proportion of patients with this cancer. MAIN TEXT While the majority of studies currently focus on the established checkpoint inhibitors (CTLA4 and PD1/PDL1), there are many other potential checkpoints that could also be targeted. In this review I provide a synopsis of current clinical trials of immunotherapies in MPM, explore potential candidate new avenues that may become future targets for immunotherapy and discuss aspects of immunotherapy that may affect the clinical outcomes of such therapies in this cancer. CONCLUSIONS The current situation regarding checkpoint inhibitors in the management of MPM whilst encouraging, despite impressive durable responses, immune checkpoint inhibitors do not provide a long-term benefit to the majority of patients with cancer. Additional studies are therefore required to further delineate and improve our understanding of both checkpoint inhibitors and the immune system in MPM. Moreover, many new potential checkpoints have yet to be studied for their therapeutic potential in MPM. All these plus the existing checkpoint inhibitors will require the development of new biomarkers for patient stratification, response and also for predicting or monitoring the emergence of resistance to these agents in MPM patients. Other potential therapeutic avenues such CAR-T therapy or treatments like oncolytic viruses or agents that target the interferon pathway designed to recruit more immune cells to the tumor also hold great promise in this hard to treat cancer.
Collapse
Affiliation(s)
- Steven G Gray
- Thoracic Oncology Research Group, Central Pathology Laboratory, CPL 30, TCDSJ Cancer Institute, St James's Hospital, Dublin, D08 RX0X, Ireland.
- Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland.
- School of Biology, Technical University of Dublin, Dublin, Ireland.
| |
Collapse
|
8
|
Siozopoulou V, Domen A, Zwaenepoel K, Van Beeck A, Smits E, Pauwels P, Marcq E. Immune Checkpoint Inhibitory Therapy in Sarcomas: Is There Light at the End of the Tunnel? Cancers (Basel) 2021; 13:360. [PMID: 33478080 PMCID: PMC7835811 DOI: 10.3390/cancers13020360] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/12/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Soft tissue and bone sarcomas are a very heterogeneous group of tumors with many subtypes for which diagnosis and treatment remains a very challenging task. On top of that, the treatment choices are limited, and the prognosis of aggressive sarcomas remains poor. Immune checkpoint inhibitors (ICIs) have drawn a lot of attention last years because of their promising response rates and their durable effects. ICIs are currently widely used in the daily routine practice for the treatment of a different malignancies, such as melanoma, Hodgkin lymphoma, and non-small cell lung carcinoma. Still, ICIs are not included in the standard treatment protocols of the different sarcoma types. However, a plethora of clinical trials investigates the clinical benefit of ICIs in sarcomas. There is clear need to develop predictive biomarkers to determine which sarcoma patients are most likely to benefit from immune checkpoint blockade. This review will focus on (i) the clinical trial results on the use of ICIs in different sarcoma types; and on (ii) possible biomarkers predictive for the effectiveness of these drugs in sarcomas.
Collapse
Affiliation(s)
- Vasiliki Siozopoulou
- Department of Pathology, Antwerp University Hospital, 2650 Edegem, Belgium; (K.Z.); (P.P.)
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium; (A.D.); (E.S.); (E.M.)
| | - Andreas Domen
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium; (A.D.); (E.S.); (E.M.)
- Department of Oncology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Karen Zwaenepoel
- Department of Pathology, Antwerp University Hospital, 2650 Edegem, Belgium; (K.Z.); (P.P.)
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium; (A.D.); (E.S.); (E.M.)
| | - Annelies Van Beeck
- Department of Orthopedics, Antwerp University Hospital, 2650 Edegem, Belgium;
| | - Evelien Smits
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium; (A.D.); (E.S.); (E.M.)
- Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Patrick Pauwels
- Department of Pathology, Antwerp University Hospital, 2650 Edegem, Belgium; (K.Z.); (P.P.)
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium; (A.D.); (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.D.); (E.S.); (E.M.)
| |
Collapse
|
9
|
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
|
10
|
Zou MX, Lv GH, Wang XB, Huang W, Li J, Jiang Y, She XL. Clinical Impact of the Immune Microenvironment in Spinal Chordoma: Immunoscore as an Independent Favorable Prognostic Factor. Neurosurgery 2020; 84:E318-E333. [PMID: 30032257 DOI: 10.1093/neuros/nyy274] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 05/27/2018] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Currently, clinical implications of immune system cells in chordoma remain to be elucidated. OBJECTIVE To characterize in situ immune cell infiltrates, the Immunoscore, and investigate their correlation with clinicopathologic data of spinal chordoma patients and outcome. METHODS Tumor-infiltrating lymphocytes (TILs) subtypes were assessed in 54 tumor specimens using immunohistochemistry for CD3, CD4, CD8, CD20, Foxp3, PD-1, and PD-L1. RESULTS Overall, immune cell infiltrates were present in all samples and there was low or moderate correlation among several TILs subsets. PD-1+ TILs density, CD3+, and CD8+ TILs densities in the tumor interior (TI) subarea were associated with surrounding muscle invasion by tumor, whereas PD-L1+ TILs showed inverse association with tumor pathological grade and stage. The density of PD-1+ TILs, PD-L1+ TILs, CD4+ TILs, and CD3+ TILs both in the TI and combined tumor regions (TI and invasion margin) were significantly associated with local recurrence-free survival and overall survival (OS). However, Foxp3+ TILs (P = .024) and CD8+ TILs evaluated in the TI (P < .001) only correlated with OS. The Immunoscore predicted less aggressive clinical features and favorable outcomes. Patients with an Immunoscore of 4 had a median OS of 128 mo, while I0 (Immunoscore of 0) patients survived only 27 mo. Multivariate analysis demonstrated that the Immunoscore was an independent favorable prognostic factor of both local recurrence-free survival (P = .026) and OS (P = .046). CONCLUSION Our data suggest a clinically relevant role of the immune microenvironment in spinal chordoma and identify the Immunoscore as promising prognostic marker.
Collapse
Affiliation(s)
- Ming-Xiang Zou
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Guo-Hua Lv
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Bin Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Huang
- Institute of Precision Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Jing Li
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yi Jiang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Xiao-Ling She
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
11
|
Cantini L, Hassan R, Sterman DH, Aerts JGJV. Emerging Treatments for Malignant Pleural Mesothelioma: Where Are We Heading? Front Oncol 2020; 10:343. [PMID: 32226777 PMCID: PMC7080957 DOI: 10.3389/fonc.2020.00343] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 02/27/2020] [Indexed: 12/21/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an uncommon but aggressive and treatment resistant neoplasm with low survival rates. In the last years we assisted to an exponential growth in the appreciation of mesothelioma pathobiology, leading several new treatments to be investigated both in the early stage of the disease and in the advanced setting. In particular, expectations are now high that immunotherapy will have a leading role in the next years. However, caution is required as results from phase II studies in MPM were often not replicated in larger, randomized, phase III trials. In this review, we describe the most promising emerging therapies for the treatment of MPM, discussing the biological rationale underlying their development as well as the issues surrounding clinical trial design and proper selection of patients for every treatment.
Collapse
Affiliation(s)
- Luca Cantini
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
- Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
- Clinical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti Ancona, Ancona, Italy
| | - Raffit Hassan
- Thoracic and GI Malignancies Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Daniel H. Sterman
- Division of Pulmonary, Critical Care, and Sleep Medicine, New York University (NYU) School of Medicine/NYU Langone Medical Center, New York, NY, United States
| | - Joachim G. J. V. Aerts
- Department of Pulmonary Medicine, Erasmus MC, Rotterdam, Netherlands
- Erasmus Cancer Institute, Erasmus MC, Rotterdam, Netherlands
| |
Collapse
|
12
|
Salaroglio IC, Kopecka J, Napoli F, Pradotto M, Maletta F, Costardi L, Gagliasso M, Milosevic V, Ananthanarayanan P, Bironzo P, Tabbò F, Cartia CF, Passone E, Comunanza V, Ardissone F, Ruffini E, Bussolino F, Righi L, Novello S, Di Maio M, Papotti M, Scagliotti GV, Riganti C. Potential Diagnostic and Prognostic Role of Microenvironment in Malignant Pleural Mesothelioma. J Thorac Oncol 2019; 14:1458-1471. [PMID: 31078776 DOI: 10.1016/j.jtho.2019.03.029] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 03/12/2019] [Accepted: 03/31/2019] [Indexed: 01/01/2023]
Abstract
INTRODUCTION A comprehensive analysis of the immune cell infiltrate collected from pleural fluid and from biopsy specimens of malignant pleural mesothelioma (MPM) may contribute to understanding the immune-evasion mechanisms related to tumor progression, aiding in differential diagnosis and potential prognostic stratification. Until now such approach has not routinely been verified. METHODS We enrolled 275 patients with an initial clinical diagnosis of pleural effusion. Specimens of pleural fluids and pleural biopsy samples used for the pathologic diagnosis and the immune phenotype analyses were blindly investigated by multiparametric flow cytometry. The results were analyzed using the Kruskal-Wallis test. The Kaplan-Meier and log-rank tests were used to correlate immune phenotype data with patients' outcome. RESULTS The cutoffs of intratumor T-regulatory (>1.1%) cells, M2-macrophages (>36%), granulocytic and monocytic myeloid-derived suppressor cells (MDSC; >5.1% and 4.2%, respectively), CD4 molecule-positive (CD4+) programmed death 1-positive (PD-1+) (>5.2%) and CD8+PD-1+ (6.4%) cells, CD4+ lymphocyte activating 3-positive (LAG-3+) (>2.8% ) and CD8+LAG-3+ (>2.8%) cells, CD4+ T cell immunoglobulin and mucin domain 3-positive (TIM-3+) (>2.5%), and CD8+TIM-3+ (>2.6%) cells discriminated MPM from pleuritis with 100% sensitivity and 89% specificity. The presence of intratumor MDSC contributed to the anergy of tumor-infiltrating lymphocytes. The immune phenotype of pleural fluid cells had no prognostic significance. By contrast, the intratumor T-regulatory and MDSC levels significantly correlated with progression-free and overall survival, the PD-1+/LAG-3+/TIM-3+ CD4+ tumor-infiltrating lymphocytes correlated with overall survival. CONCLUSIONS A clear immune signature of pleural fluids and tissues of MPM patients may contribute to better predict patients' outcome.
Collapse
Affiliation(s)
| | - Joanna Kopecka
- Department of Oncology, University of Torino, Torino, Italy
| | - Francesca Napoli
- Pathology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Monica Pradotto
- Thoracic Oncology Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino Regione Gonzole 10, Orbassano, Italy
| | - Francesca Maletta
- Department of Oncology, University of Torino, Torino, Italy; Pathology Unit, Department of Oncology at AOU Città della Salute e della Scienza, Torino, Italy
| | - Lorena Costardi
- Thoracic Surgery Unit, Department of Surgery, AOU Città della Salute e Della Scienza, University of Torino, Torino, Italy
| | - Matteo Gagliasso
- Thoracic Surgery Unit, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | | | | | - Paolo Bironzo
- Department of Oncology, University of Torino, Torino, Italy; Thoracic Oncology Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino Regione Gonzole 10, Orbassano, Italy
| | - Fabrizio Tabbò
- Department of Oncology, University of Torino, Torino, Italy; Thoracic Oncology Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino Regione Gonzole 10, Orbassano, Italy
| | - Carlotta F Cartia
- Thoracic Surgery Unit, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Erika Passone
- Thoracic Surgery Unit, Department of Surgery, AOU Città della Salute e Della Scienza, University of Torino, Torino, Italy
| | - Valentina Comunanza
- Department of Oncology, University of Torino, Torino, Italy; Candiolo Cancer Institute - FPO IRCCS, Candiolo, Department of Oncology, University of Torino, Candiolo, Italy
| | - Francesco Ardissone
- Thoracic Surgery Unit, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Enrico Ruffini
- Thoracic Surgery Unit, Department of Surgery, AOU Città della Salute e Della Scienza, University of Torino, Torino, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, Torino, Italy; Candiolo Cancer Institute - FPO IRCCS, Candiolo, Department of Oncology, University of Torino, Candiolo, Italy
| | - Luisella Righi
- Department of Oncology, University of Torino, Torino, Italy; Pathology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Silvia Novello
- Department of Oncology, University of Torino, Torino, Italy; Thoracic Oncology Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino Regione Gonzole 10, Orbassano, Italy
| | - Massimo Di Maio
- Department of Oncology, University of Torino, Torino, Italy; Medical Oncology Division, Department of Oncology at AOU Ordine Mauriziano di Torino, Torino, Italy
| | - Mauro Papotti
- Department of Oncology, University of Torino, Torino, Italy; Pathology Unit, Department of Oncology at AOU Città della Salute e della Scienza, Torino, Italy
| | - Giorgio V Scagliotti
- Department of Oncology, University of Torino, Torino, Italy; Thoracic Oncology Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino Regione Gonzole 10, Orbassano, Italy
| | - Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy; Interdepartmental Center "G. Scansetti" for the Study of Asbestos and Other Toxic Particulates, University of Torino, Torino, Italy.
| |
Collapse
|
13
|
Brusselmans L, Arnouts L, Millevert C, Vandersnickt J, van Meerbeeck JP, Lamote K. Breath analysis as a diagnostic and screening tool for malignant pleural mesothelioma: a systematic review. Transl Lung Cancer Res 2018; 7:520-536. [PMID: 30450290 PMCID: PMC6204411 DOI: 10.21037/tlcr.2018.04.09] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/10/2018] [Indexed: 12/14/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a tumour related to a historical exposure to asbestos fibres. Currently, the definite diagnosis is made only by the histological examination of a biopsy obtained through an invasive thoracoscopy. However, diagnosis is made too late for curative treatment because of non-specific symptoms mainly appearing at advanced stage disease. Hence, due to its biologic aggressiveness and the late diagnosis, survival rate is low and the patients' outcome poor. In addition, radiological imaging, like computed tomographic scans, and blood biomarkers are found not to be sensitive enough to be used as an early diagnostic tool. Detection in an early stage is assumed to improve the patients' outcome but is hampered due to non-specific and late symptomology. Hence, there is a need for a new screening and diagnostic test which could improve the patients' outcome. Despite extensive research has focused on blood biomarkers, not a single has been shown clinically useful, and therefore research recently shifted to "breathomics" techniques to recognize specific volatile organic compounds (VOCs) in the breath of the patient as potential non-invasive biomarkers for disease. In this review, we summarize the acquired knowledge about using breath analysis for diagnosing and monitoring MPM and asbestos-related disorders (ARD). Gas chromatography-mass spectrometry (GC-MS), the gold standard of breath analysis, appears to be the method with the highest accuracy (97%) to differentiate MPM patients from at risk asbestos-exposed subjects. There have already been found some interesting biomarkers that are significantly elevated in asbestosis (NO, 8-isoprostane, leukotriene B4, α-Pinene…) and MPM (cyclohexane) patients. Regrettably, the different techniques and the plethora of studies suffer some limitations. Most studies are pilot studies with the inclusion of a limited number of patients. Nevertheless, given the promising results and easy sampling methods, we can conclude that breath analysis may become a useful tool in the future to screen for MPM, but further research is warranted.
Collapse
Affiliation(s)
- Lisa Brusselmans
- Laboratory of Experimental Medicine and Paediatrics, Antwerp University, Wilrijk, Belgium
| | - Lieselot Arnouts
- Laboratory of Experimental Medicine and Paediatrics, Antwerp University, Wilrijk, Belgium
| | - Charissa Millevert
- Laboratory of Experimental Medicine and Paediatrics, Antwerp University, Wilrijk, Belgium
| | - Joyce Vandersnickt
- Laboratory of Experimental Medicine and Paediatrics, Antwerp University, Wilrijk, Belgium
| | - Jan P. van Meerbeeck
- Laboratory of Experimental Medicine and Paediatrics, Antwerp University, Wilrijk, Belgium
- Internal Medicine, Ghent University, Ghent, Belgium
- Department of Pneumology, Antwerp University Hospital, Edegem, Belgium
| | - Kevin Lamote
- Laboratory of Experimental Medicine and Paediatrics, Antwerp University, Wilrijk, Belgium
- Internal Medicine, Ghent University, Ghent, Belgium
| |
Collapse
|
14
|
Jones RG, Karthik F, Dugar A, Kanagarajan K, Desai K, Bhandari M. Nivolumab Immunotherapy in Malignant Mesothelioma: A Case Report Highlighting a New Opportunity for Exceptional Outcomes. AMERICAN JOURNAL OF CASE REPORTS 2018; 19:783-789. [PMID: 29970876 PMCID: PMC6061454 DOI: 10.12659/ajcr.909584] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Malignant pleural mesothelioma (MPM) is a highly lethal cancer with a median survival of ~12 months even with aggressive intervention. Frontline therapy relies on systemic cisplatin and pemetrexed chemotherapy and has a response rate of ~35-41%; currently, there are no US Food and Drug Administration approved second-line therapies for MPM. Herein, we present a patient with MPM who experienced rapid disease progression after standard therapy but who had an exceptional and sustained response to immune checkpoint inhibition with single agent nivolumab. CASE REPORT A 68-year-old male with a history of work-related asbestos exposure was diagnosed with MPM. He was treated with primary resection followed by systemic chemotherapy with cisplatin and pemetrexed. When chemotherapy failed, he was switched to immunotherapy with nivolumab and achieved an exceptional response. CONCLUSIONS We report the first case of a patient with MPM who experienced rapid disease progression after standard therapy but had an exceptional and sustained response to immune checkpoint inhibition with single agent nivolumab. As outcomes with traditional chemotherapy regimens remain disappointing, there is a substantial need for new approaches to MPM; our case highlights a new therapeutic opportunity even in the face of aggressive disease. Indeed, a new era of investigation utilizing immunotherapy for mesothelioma is beginning, with much anticipation.
Collapse
Affiliation(s)
- Riley G Jones
- Department of Internal Medicine, University of Florida, Gainesville, FL, USA.,Department of Internal Medicine, The Christ Hospital, Cincinnati, OH, USA
| | - Felix Karthik
- Department of Pulmonary Diseases, The Christ Hosptial, Cincinnati, OH, USA
| | - Anushree Dugar
- Department of hematology/oncology, The Christ Hosptial, Cincinnati, OH, USA
| | | | - Kalpan Desai
- Department of Internal Medicine, The Christ Hosptial, Cincinnati, OH, USA
| | - Manish Bhandari
- Department of Hematology/Oncology, The Christ Hosptial, Cincinnati, OH, USA
| |
Collapse
|
15
|
FDG PET-derived parameters as prognostic tool in progressive malignant pleural mesothelioma treated patients. Eur J Nucl Med Mol Imaging 2018; 45:2071-2078. [DOI: 10.1007/s00259-018-4056-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/23/2018] [Indexed: 12/17/2022]
|
16
|
Watanabe T, Okuda K, Murase T, Moriyama S, Haneda H, Kawano O, Yokota K, Sakane T, Oda R, Inagaki H, Nakanishi R. Four immunohistochemical assays to measure the PD-L1 expression in malignant pleural mesothelioma. Oncotarget 2018; 9:20769-20780. [PMID: 29755688 PMCID: PMC5945532 DOI: 10.18632/oncotarget.25100] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/24/2018] [Indexed: 12/29/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 pathway are expected to be a novel therapy for combating future increases in numbers of malignant pleural mesothelioma (MPM) patients. However, the PD-L1 expression, which is a predictor of the response to ICIs, is unclear in MPM. We studied the PD-L1 expression using four immunohistochemical assays (SP142, SP263, 28-8 and 22C3) in 32 MPM patients. The PD-L1 expression in tumor cells and immune cells was evaluated to clarify the rate of PD-L1 expression and the concordance among the four assays in MPM. The positivity rate of PD-L1 expression was 53.1% for SP142, 28.1% for SP263, 53.1% for 28-8, and 56.3% for 22C3. Nine cases were positive and 10 were negative for all assays. Discordance among the four assays was found in 13 cases. The concordance rates between SP142 and 22C3 and between 28-8 and 22C3 were the highest (84.4%). The concordance rates between SP263 and the other three assays were low (71.9% to 75.0%). The PD-L1 expression in MPM was almost equivalent for three of the assays. Given the cut-off values set in our study, these findings suggested that these assays, except for SP263, can be used for accurate PD-L1 immunostaining in MPM.
Collapse
Affiliation(s)
- Takuya Watanabe
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Katsuhiro Okuda
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Takayuki Murase
- Department of Pathology and Molecular Diagnostics, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Satoru Moriyama
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Hiroshi Haneda
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Osamu Kawano
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Keisuke Yokota
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Tadashi Sakane
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Risa Oda
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Hiroshi Inagaki
- Department of Pathology and Molecular Diagnostics, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| | - Ryoichi Nakanishi
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Mizuho-cho, Mizuho-ku, Nagoya 467-8601, Japan
| |
Collapse
|
17
|
Zou MX, Guo KM, Lv GH, Huang W, Li J, Wang XB, Jiang Y, She XL. Clinicopathologic implications of CD8 +/Foxp3 + ratio and miR-574-3p/PD-L1 axis in spinal chordoma patients. Cancer Immunol Immunother 2018; 67:209-224. [PMID: 29051990 PMCID: PMC11028121 DOI: 10.1007/s00262-017-2080-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/14/2017] [Indexed: 12/11/2022]
Abstract
Currently, little is known about the interactions between microRNAs (miRNAs) and the PD-1/PD-L1 signaling pathway in chordoma, and data discussing the role of the immune milieu in chordoma prognosis are limited. We aimed to analyze the relationship between PD-L1, miR-574-3p, microenvironmental tumor-infiltrating lymphocytes (TILs) and clinicopathological features of spinal chordoma patients. PD-L1 expression and TILs (including Foxp3+, CD8+, PD-1+ and PD-L1+) were assessed by immunohistochemistry in tumor specimens of 54 spinal chordoma patients. MiRNAs microarray and bioinformatical analysis were used to identify miRNAs potentially regulating PD-L1 expression, which were further validated by quantitative RT-PCR. miR-574-3p was identified to potentially regulate PD-L1 expression in chordoma, which inversely correlated with PD-L1. Positive PD-L1 expression on tumor cells was associated with advanced stages (P = 0.041) and TILs infiltration (P = 0.005), whereas decreased miR-574-3p level correlated with higher muscle invasion (P = 0.012), more severe tumor necrosis (P = 0.022) and poor patient survival. Importantly, a patient subgroup with PD-L1+/miR-574-3plow chordoma phenotype was significantly associated with worse local recurrence-free survival (LRFS) (P = 0.026). PD-1+ TILs density was associated with surrounding muscle invasion (P = 0.014), and independently portended poor LRFS (P = 0.040), while PD-L1+ TILs showed tendencies of less aggressive clinical outcomes. Multivariate analysis of OS only found CD8+/Foxp3+ ratio to be independent prognostic factor (P = 0.022). These findings may be useful to stratify patients into prognostic groups and provide a rationale for the use of checkpoint blockade therapy, possibly by administering miR-574-3p mimics, in spinal chordoma.
Collapse
Affiliation(s)
- Ming-Xiang Zou
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renminzhong Road, Changsha, Hunan, 410011, China
| | - Ke-Miao Guo
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renminzhong Road, Changsha, Hunan, 410011, China
| | - Guo-Hua Lv
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renminzhong Road, Changsha, Hunan, 410011, China
| | - Wei Huang
- Institute of Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jing Li
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renminzhong Road, Changsha, Hunan, 410011, China.
| | - Xiao-Bin Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, 139 Renminzhong Road, Changsha, Hunan, 410011, China.
| | - Yi Jiang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Xiao-Ling She
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| |
Collapse
|
18
|
Molecular and Histopathological Characterization of the Tumor Immune Microenvironment in Advanced Stage of Malignant Pleural Mesothelioma. J Thorac Oncol 2018; 13:124-133. [DOI: 10.1016/j.jtho.2017.09.1968] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/21/2017] [Accepted: 09/22/2017] [Indexed: 01/21/2023]
|
19
|
De Waele J, Marcq E, Van Audenaerde JR, Van Loenhout J, Deben C, Zwaenepoel K, Van de Kelft E, Van der Planken D, Menovsky T, Van den Bergh JM, Willemen Y, Pauwels P, Berneman ZN, Lardon F, Peeters M, Wouters A, Smits EL. Poly(I:C) primes primary human glioblastoma cells for an immune response invigorated by PD-L1 blockade. Oncoimmunology 2017; 7:e1407899. [PMID: 29399410 DOI: 10.1080/2162402x.2017.1407899] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 10/31/2017] [Accepted: 11/17/2017] [Indexed: 02/08/2023] Open
Abstract
Prognosis of glioblastoma remains dismal, underscoring the need for novel therapies. Immunotherapy is generating promising results, but requires combination strategies to unlock its full potential. We investigated the immunomodulatory capacities of poly(I:C) on primary human glioblastoma cells and its combinatorial potential with programmed death ligand (PD-L) blockade. In our experiments, poly(I:C) stimulated expression of both PD-L1 and PD-L2 on glioblastoma cells, and a pro-inflammatory secretome, including type I interferons (IFN) and chemokines CXCL9, CXCL10, CCL4 and CCL5. IFN-β was partially responsible for the elevated PD-1 ligand expression on these cells. Moreover, real-time PCR and chloroquine-mediated blocking experiments indicated that poly(I:C) triggered Toll-like receptor 3 to elicit its effect. Cocultures of poly(I:C)-treated glioblastoma cells with peripheral blood mononuclear cells enhanced lymphocytic activation (CD69, IFN-γ) and cytotoxic capacity (CD107a, granzyme B). Additional PD-L1 blockade further propagated immune activation. Besides activating immunity, poly(I:C)-treated glioblastoma cells also doubled the attraction of CD8+ T cells, and to a lesser extent CD4+ T cells, via a mechanism which included CXCR3 and CCR5 ligands. Our results indicate that by triggering glioblastoma cells, poly(I:C) primes the tumor microenvironment for an immune response. Secreted cytokines allow for immune activation while chemokines attract CD8+ T cells to the front, which are postulated as a prerequisite for effective PD-1/PD-L1 blockade. Accordingly, additional blockade of the concurrently elevated tumoral PD-L1 further reinforces the immune activation. In conclusion, our data proposes poly(I:C) treatment combined with PD-L1 blockade to invigorate the immune checkpoint inhibition response in glioblastoma.
Collapse
Affiliation(s)
- Jorrit De Waele
- Center for Oncological Research, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Elly Marcq
- Center for Oncological Research, University of Antwerp, Wilrijk, Antwerp, Belgium
| | | | - Jinthe Van Loenhout
- Center for Oncological Research, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Christophe Deben
- Center for Oncological Research, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Karen Zwaenepoel
- Department of Pathology, Antwerp University Hospital, Edegem, Antwerp, Belgium
| | - Erik Van de Kelft
- Department of Neurosurgery, AZ Nikolaas, Sint-Niklaas, East Flanders, Belgium
| | | | - Tomas Menovsky
- Department of Neurosurgery, Antwerp University Hospital, Edegem, Antwerp, Belgium
| | | | - Yannick Willemen
- Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Patrick Pauwels
- Center for Oncological Research, University of Antwerp, Wilrijk, Antwerp, Belgium.,Department of Pathology, Antwerp University Hospital, Edegem, Antwerp, Belgium
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Antwerp, Belgium.,Department of Hematology, Antwerp University Hospital, Edegem, Antwerp, Belgium
| | - Filip Lardon
- Center for Oncological Research, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Marc Peeters
- Center for Oncological Research, University of Antwerp, Wilrijk, Antwerp, Belgium.,Department of Oncology, Multidisciplinary Oncological Center Antwerp, Antwerp University Hospital, Antwerp, Edegem, Belgium
| | - An Wouters
- Center for Oncological Research, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Evelien Lj Smits
- Center for Oncological Research, University of Antwerp, Wilrijk, Antwerp, Belgium.,Laboratory of Experimental Hematology, University of Antwerp, Wilrijk, Antwerp, Belgium
| |
Collapse
|
20
|
Riganti C, Lingua MF, Salaroglio IC, Falcomatà C, Righi L, Morena D, Picca F, Oddo D, Kopecka J, Pradotto M, Libener R, Orecchia S, Bironzo P, Comunanza V, Bussolino F, Novello S, Scagliotti GV, Di Nicolantonio F, Taulli R. Bromodomain inhibition exerts its therapeutic potential in malignant pleural mesothelioma by promoting immunogenic cell death and changing the tumor immune-environment. Oncoimmunology 2017; 7:e1398874. [PMID: 29399399 DOI: 10.1080/2162402x.2017.1398874] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 12/28/2022] Open
Abstract
Systemic treatment of malignant pleural mesothelioma (MPM) is moderately active for the intrinsic pharmacological resistance of MPM cell and its ability to induce an immune suppressive environment. Here we showed that the expression of bromodomain (BRD) proteins BRD2, BRD4 and BRD9 was significantly higher in human primary MPM cells compared to normal mesothelial cells (HMC). Nanomolar concentrations of bromodomain inhibitors (BBIs) JQ1 or OTX015 impaired patient-derived MPM cell proliferation and induced cell-cycle arrest without affecting apoptosis. Importantly, BBIs primed MPM cells for immunogenic cell death, by increasing extracellular release of ATP and HMGB1, and by promoting membrane exposure of calreticulin and ERp57. Accordingly, BBIs activated dendritic cell (DC)-mediated phagocytosis and expansion of CD8+ T-lymphocyte clones endorsed with antitumor cytotoxic activity. BBIs reduced the expression of the immune checkpoint ligand PD-L1 in MPM cells; while both CD8+ and CD4+ T-lymphocytes co-cultured with JQ1-treated MPM cells decreased PD-1 expression, suggesting a disruption of the immune-suppressive PD-L1/PD-1 axis. Additionally, BBIs reduced the expansion of myeloid-derived suppressor cells (MDSC) induced by MPM cells. Finally, a preclinical model of MPM confirmed that the anti-tumor efficacy of JQ1 was largely due to its ability to restore an immune-active environment, by increasing intra-tumor DC and CD8+ T-lymphocytes, and decreasing MDSC. Thereby, we propose that, among novel drugs, BBIs should be investigated for MPM treatment for their combined activity on both tumor cells and surrounding immune-environment.
Collapse
Affiliation(s)
- Chiara Riganti
- Department of Oncology, University of Torino, Torino, Italy
| | | | | | - Chiara Falcomatà
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Italy
| | - Luisella Righi
- Pathology Unit, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Deborah Morena
- Department of Oncology, University of Torino, Torino, Italy
| | | | - Daniele Oddo
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Italy
| | - Joanna Kopecka
- Department of Oncology, University of Torino, Torino, Italy
| | - Monica Pradotto
- Thoracic Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Roberta Libener
- Pathology Division, S. Antonio and Biagio Hospital, Alessandria, Italy
| | - Sara Orecchia
- Pathology Division, S. Antonio and Biagio Hospital, Alessandria, Italy
| | - Paolo Bironzo
- Thoracic Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Valentina Comunanza
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Italy
| | - Federico Bussolino
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Italy
| | - Silvia Novello
- Thoracic Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Giorgio Vittorio Scagliotti
- Thoracic Unit and Medical Oncology Division, Department of Oncology at San Luigi Hospital, University of Torino, Orbassano, Italy
| | - Federica Di Nicolantonio
- Department of Oncology, University of Torino, Torino, Italy.,Candiolo Cancer Institute - FPO, IRCCS, Candiolo, Italy
| | | |
Collapse
|
21
|
Marcq E, Waele JD, Audenaerde JV, Lion E, Santermans E, Hens N, Pauwels P, van Meerbeeck JP, Smits ELJ. Abundant expression of TIM-3, LAG-3, PD-1 and PD-L1 as immunotherapy checkpoint targets in effusions of mesothelioma patients. Oncotarget 2017; 8:89722-89735. [PMID: 29163783 PMCID: PMC5685704 DOI: 10.18632/oncotarget.21113] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 09/01/2017] [Indexed: 12/20/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer with an increasing incidence, poor prognosis and limited effective treatment options. Hence, new treatment strategies are warranted which include immune checkpoint blockade approaches with encouraging preliminary data. Research on the immunological aspects of the easily accessible mesothelioma microenvironment could identify prognostic and/or predictive biomarkers and provide useful insights for developing effective immunotherapy. In this context, we investigated the immune cell composition of effusions (pleural and ascites fluids) from 11 different chemotherapy-treated MPM patients. We used multicolor flow cytometry to describe different subsets of immune cells and their expression of immune checkpoint molecules TIM-3, LAG-3, PD-1 and PD-L1. We demonstrate a patient-dependent inter- and intraspecific variation comparing pleural and ascites fluids in immune cell composition and immune checkpoint expression. We found CD4+ and CD8+ T cells, B cells, macrophages, natural killer cells, dendritic cells and tumor cells in the fluids. To the best of our knowledge, we are the first to report TIM-3 and LAG-3 expression and we confirm PD-1 and PD-L1 expression on different MPM effusion-resident immune cells. Moreover, we identified two MPM effusion-related factors with clinical value: CD4+ T cells were significantly correlated with better response to chemotherapy, while the percentage of PD-L1+ podoplanin (PDPN)+ tumor cells is a significant prognostic factor for worse outcome. Our data provide a basis for more elaborate research on MPM effusion material in the context of treatment follow-up and prognostic biomarkers and the development of immune checkpoint-targeted immunotherapy.
Collapse
Affiliation(s)
- Elly Marcq
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium
| | - Jorrit De Waele
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium
| | | | - Eva Lion
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
| | - Eva Santermans
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Diepenbeek, Belgium
| | - Niel Hens
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Diepenbeek, Belgium.,Center for Health Economics Research and Modelling Infectious Diseases, University of Antwerp, Antwerp, Belgium
| | - Patrick Pauwels
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium.,Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
| | - Jan P van Meerbeeck
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium.,Thoracic Oncology/MOCA, Antwerp University Hospital, Antwerp, Belgium
| | - Evelien L J Smits
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium.,Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
22
|
Dumoulin DW, Aerts JG, Cornelissen R. Is immunotherapy a viable option in treating mesothelioma? Future Oncol 2017; 13:1747-1750. [PMID: 28774186 DOI: 10.2217/fon-2017-0234] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Affiliation(s)
- Daphne W Dumoulin
- Erasmus MC Cancer Institute, Department of Pulmonary Medicine, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| | - Joachim Gjv Aerts
- Erasmus MC Cancer Institute, Department of Pulmonary Medicine, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| | - Robin Cornelissen
- Erasmus MC Cancer Institute, Department of Pulmonary Medicine, 's-Gravendijkwal 230, 3015 CE Rotterdam, The Netherlands
| |
Collapse
|
23
|
Abstract
Malignant mesothelioma is a universally lethal cancer that is increasing in incidence worldwide. There is a dearth of effective therapies, with only one treatment (pemetrexed and cisplatin combination chemotherapy) approved in the past 13 years. However, the past 5 years have witnessed an exponential growth in our understanding of mesothelioma pathobiology, which is set to revolutionize therapeutic strategies. From a genomic standpoint, mesothelioma is characterized by a preponderance of tumour suppressor alterations, for which novel therapies are currently in development. Other promising antitumour agents include inhibitors against angiogenesis, mesothelin and immune checkpoints, which are at various phases of clinical trial testing.
Collapse
Affiliation(s)
- Timothy A Yap
- The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Joachim G Aerts
- Erasmus MC Cancer Institute, 3015 CE Rotterdam, The Netherlands
| | - Sanjay Popat
- Royal Marsden Hospital, London SW3 6JJ, UK
- National Heart and Lung Institute, Imperial College London SW3 6NP, UK
| | | |
Collapse
|
24
|
De La Maza L, Wu M, Wu L, Yun H, Zhao Y, Cattral M, McCart A, Cho BJ, de Perrot M. In Situ Vaccination after Accelerated Hypofractionated Radiation and Surgery in a Mesothelioma Mouse Model. Clin Cancer Res 2017; 23:5502-5513. [PMID: 28606922 DOI: 10.1158/1078-0432.ccr-17-0438] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/27/2017] [Accepted: 06/05/2017] [Indexed: 11/16/2022]
Abstract
Purpose: How best to sequence and integrate immunotherapy into standard of care is currently unknown. Clinical protocols with accelerated nonablative hypofractionated radiation followed by surgery could provide an opportunity to implement immune checkpoint blockade.Experimental Design: We therefore assessed the impact of nonablative hypofractionated radiation on the immune system in combination with surgery in a mouse mesothelioma model. Blunt surgery (R1 resection) was used to analyze the short-term effect, and radical surgery (R0 resection) was used to analyze the long-term effect of this radiation protocol before surgery.Results: Nonablative hypofractionated radiation led to a specific immune activation against the tumor associated with significant upregulation of CD8+ T cells, limiting the negative effect of an incomplete resection. The same radiation protocol performed 7 days before radical surgery led to a long-term antitumor immune protection that was primarily driven by CD4+ T cells. Radical surgery alone or with a short course of nonablative radiation completed 24 hours before radical surgery did not provide this vaccination effect. Combining this radiation protocol with CTLA-4 blockade provided better results than radiation alone. The effect of PD-1 or PD-L1 blockade with this radiation protocol was less effective than the combination with CTLA-4 blockade.Conclusions: A specific activation of the immune system against the tumor contributes to the benefit of accelerated, hypofractionated radiation before surgery. Nonablative hypofractionated radiation combined with surgery provides an opportunity to introduce immune checkpoint blockades in the clinical setting. Clin Cancer Res; 23(18); 5502-13. ©2017 AACR.
Collapse
Affiliation(s)
- Luis De La Maza
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Matthew Wu
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Licun Wu
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Hana Yun
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Yidan Zhao
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Mark Cattral
- Department of General Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Andrea McCart
- Department of General Surgery, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada
| | - Bc John Cho
- Department of Radiation Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Marc de Perrot
- Latner Thoracic Surgery Research Laboratories, Toronto General Research Institute, University of Toronto, Toronto, Ontario, Canada. .,Division of Thoracic Surgery, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
25
|
Bakker E, Guazzelli A, Krstic-Demonacos M, Lisanti M, Sotgia F, Mutti L. Current and prospective pharmacotherapies for the treatment of pleural mesothelioma. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1325358] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Emyr Bakker
- Biomedical Research Centre, School of Environment and Life Sciences, University of Salford, Salford, UK
| | - Alice Guazzelli
- Biomedical Research Centre, School of Environment and Life Sciences, University of Salford, Salford, UK
| | - Marija Krstic-Demonacos
- Biomedical Research Centre, School of Environment and Life Sciences, University of Salford, Salford, UK
| | - Michael Lisanti
- Biomedical Research Centre, School of Environment and Life Sciences, University of Salford, Salford, UK
| | - Federica Sotgia
- Biomedical Research Centre, School of Environment and Life Sciences, University of Salford, Salford, UK
| | - Luciano Mutti
- Biomedical Research Centre, School of Environment and Life Sciences, University of Salford, Salford, UK
| |
Collapse
|
26
|
Lantuejoul S, Le Stang N, Damiola F, Scherpereel A, Galateau-Sallé F. PD-L1 Testing for Immune Checkpoint Inhibitors in Mesothelioma: For Want of Anything Better? J Thorac Oncol 2017; 12:778-781. [DOI: 10.1016/j.jtho.2017.03.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 03/20/2017] [Accepted: 03/20/2017] [Indexed: 02/07/2023]
|
27
|
Goričar K, Kovač V, Dolžan V. Clinical-pharmacogenetic models for personalized cancer treatment: application to malignant mesothelioma. Sci Rep 2017; 7:46537. [PMID: 28422153 PMCID: PMC5396189 DOI: 10.1038/srep46537] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 03/22/2017] [Indexed: 12/29/2022] Open
Abstract
Large interindividual differences in treatment outcome are observed in cancer patients undergoing chemotherapy. Our aim was to develop and validate clinical-pharmacogenetic prediction models of gemcitabine/cisplatin or pemetrexed/cisplatin treatment outcome and develop an algorithm for genotype-based treatment recommendations in malignant mesothelioma (MM). We genotyped 189 MM patients for polymorphisms in gemcitabine, pemetrexed and cisplatin metabolism, transport and drug target genes and DNA repair pathways. To build respective clinical-pharmacogenetic models, pharmacogenetic scores were assigned by rounding regression coefficients. Gemcitabine/cisplatin model was based on training group of 71 patients and included CRP, histological type, performance status, RRM1 rs1042927, ERCC2 rs13181, ERCC1 rs3212986, and XRCC1 rs25487. Patients with higher score had shorter progression-free (PFS) and overall survival (P < 0.001). This model’s sensitivity was 0.615 and specificity 0.812. In independent validation group of 66 patients the sensitivity and specificity were 0.667 and 0.641, respectively. Pemetrexed/cisplatin model was based on 57 patients and included CRP, MTHFD1 rs2236225, and ABCC2 rs2273697. Patients with higher score had worse response and shorter PFS (P < 0.001). This model’s sensitivity was 0.750 and specificity 0.607. In independent validation group of 20 patients the sensitivity and specificity were 0.889 and 0.500, respectively. The proposed algorithm based on these models could enable the choice of the most effective chemotherapy for 85.5% of patients and lead to improved treatment outcome in MM.
Collapse
Affiliation(s)
- Katja Goričar
- Pharmacogenetics Laboratory, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Viljem Kovač
- Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Vita Dolžan
- Pharmacogenetics Laboratory, Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
28
|
Does selective pleural irradiation of malignant pleural mesothelioma allow radiation dose escalation? Strahlenther Onkol 2017; 193:285-294. [DOI: 10.1007/s00066-017-1108-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/27/2017] [Indexed: 12/16/2022]
|
29
|
Kiyotani K, Park JH, Inoue H, Husain A, Olugbile S, Zewde M, Nakamura Y, Vigneswaran WT. Integrated analysis of somatic mutations and immune microenvironment in malignant pleural mesothelioma. Oncoimmunology 2017; 6:e1278330. [PMID: 28344893 DOI: 10.1080/2162402x.2016.1278330] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 12/27/2016] [Accepted: 12/28/2016] [Indexed: 12/29/2022] Open
Abstract
To investigate the link between the genomic landscape of cancer cells and immune microenvironment in tumor tissues, we characterized somatic mutations and tumor-infiltrating lymphocytes (TILs) in malignant pleural mesothelioma (MPM), including mutation/neoantigen load, spatial heterogeneity of somatic mutations of cancer cells and TILs (T-cell receptor β (TCRβ) repertoire), and expression profiles of immune-related genes using specimens of three different tumor sites (anterior, posterior, and diaphragm) obtained from six MPM patients. Integrated analysis identified the distinct patterns of somatic mutations and the immune microenvironment signatures both intratumorally and interindividually. MPM cases showed intratumoral heterogeneity in somatic mutations with unique TCRβ clonotypes of TILs that were restricted to each tumor site, suggesting the presence of a neoantigen-related immune response. Correlation analyses showed that higher neoantigen load was significantly correlated with stronger clonal expansion of TILs (p = 0.048) and a higher expression level of an immune-associated cytolytic factor (PRF1 (p = 0.0041) in tumor tissues), suggesting that high neoantigen loads in tumor cells might promote expansion of functional tumor-specific T cells in the tumor bed. Our results collectively indicate that MPM tumors constitute a diverse heterogeneity in both the genomic landscape and immune microenvironment, and that mutation/neoantigen load may affect the immune microenvironment in MPM tissues.
Collapse
Affiliation(s)
- Kazuma Kiyotani
- Department of Medicine, The University of Chicago , Chicago, IL, USA
| | - Jae-Hyun Park
- Department of Medicine, The University of Chicago , Chicago, IL, USA
| | - Hiroyuki Inoue
- Department of Medicine, The University of Chicago , Chicago, IL, USA
| | - Aliya Husain
- Department of Pathology, The University of Chicago , Chicago, IL, USA
| | - Sope Olugbile
- Department of Medicine, The University of Chicago , Chicago, IL, USA
| | - Makda Zewde
- Department of Medicine, The University of Chicago , Chicago, IL, USA
| | - Yusuke Nakamura
- Department of Medicine, The University of Chicago, Chicago, IL, USA; Department of Surgery, The University of Chicago, Chicago, IL, USA
| | - Wickii T Vigneswaran
- Department of Surgery, The University of Chicago, Chicago, IL, USA; Department of Thoracic and Cardiovascular Surgery, Loyola University Medical Center, Maywood, IL, USA
| |
Collapse
|
30
|
Botticella A, Defraene G, Nackaerts K, Deroose CM, Coolen J, Nafteux P, Peeters S, Ricardi U, De Ruysscher D. Optimal gross tumor volume definition in lung-sparing intensity modulated radiotherapy for pleural mesothelioma: an in silico study. Acta Oncol 2016; 55:1450-1455. [PMID: 27732127 DOI: 10.1080/0284186x.2016.1234066] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The gross tumor volume (GTV) definition for malignant pleural mesothelioma (MPM) is ill-defined. We therefore investigated which imaging modality is optimal: computed tomography (CT) with intravenous contrast (IVC), positron emission tomography-CT (PET/CT) or magnetic resonance imaging (MRI). MATERIAL AND METHODS Sixteen consecutive patients with untreated stage I-IV MPM were included. Patients with prior pleurodesis were excluded. CT with IVC, 18FDG-PET/CT and MRI (T2 and contrast-enhanced T1) were obtained. CT was rigidly co-registered with PET/CT and with MRI. Three sets of pleural GTVs were defined: GTVCT, GTVCT+PET/CT and GTVCT+MRI. Quantitative and qualitative evaluations of the contoured GTVs were performed. RESULTS Compared to CT-based GTV definition, PET/CT identified additional tumor sites (defined as either separate nodules or greater extent of a known tumor) in 12/16 patients. Compared to either CT or PET/CT, MRI identified additional tumor sites in 15/16 patients (p = .7). The mean GTVCT, GTVCT+PET/CT and GTVCT+MRI [±standard deviation (SD)] were 630.1 cm3 (±302.81), 640.23 cm3 (±302.83) and 660.8 cm3 (±290.8), respectively. Differences in mean volumes were not significant. The mean Jaccard Index was significantly lower in MRI-based contours versus all the others. CONCLUSION As MRI identified additional pleural disease sites in the majority of patients, it may play a role in optimal target volume definition.
Collapse
Affiliation(s)
- Angela Botticella
- Department of Oncology, Experimental Radiation Oncology, KU Leuven–University of Leuven, Leuven, Belgium
| | - Gilles Defraene
- Department of Oncology, Experimental Radiation Oncology, KU Leuven–University of Leuven, Leuven, Belgium
| | - Kristiaan Nackaerts
- Respiratory Diseases/Respiratory Oncology Unit, KU Leuven–University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Christophe M. Deroose
- Department Imaging and Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven–University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Johan Coolen
- Radiology Department, University Hospitals Leuven, Leuven, Belgium
| | - Philippe Nafteux
- Department of Thoracic Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Stephanie Peeters
- Department of Radiation Oncology, KU Leuven–University of Leuven, University Hospitals Leuven, Leuven, Belgium
| | - Umberto Ricardi
- Department of Oncology, Radiation Oncology, University of Turin, Turin, Italy
| | - Dirk De Ruysscher
- Department of Radiation Oncology, KU Leuven–University of Leuven, University Hospitals Leuven, Leuven, Belgium
- GROW–School for Oncology and Developmental Biology, Department of Radiation Oncology (MAASTRO clinic), Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
31
|
Marcq E, Siozopoulou V, De Waele J, van Audenaerde J, Zwaenepoel K, Santermans E, Hens N, Pauwels P, van Meerbeeck JP, Smits ELJ. Prognostic and predictive aspects of the tumor immune microenvironment and immune checkpoints in malignant pleural mesothelioma. Oncoimmunology 2016; 6:e1261241. [PMID: 28197385 DOI: 10.1080/2162402x.2016.1261241] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/27/2016] [Accepted: 11/10/2016] [Indexed: 12/29/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is an aggressive cancer with a poor prognosis and an increasing incidence, for which novel therapeutic strategies are urgently required. Since the immune system has been described to play a presumed role in the protection against MPM, characterization of its tumor immune microenvironment (TME) and immune checkpoints can identify new immunotherapeutic targets and their predictive and/or prognostic value. To characterize the TME and the immune checkpoint expression profile, we performed immunohistochemistry (IHC) on formalin-fixed paraffin embedded (FFPE) tissue sections from 54 MPM patients (40 at time of diagnosis; 14 treated with chemotherapy). We stained for PD-1, PD-L1, TIM-3, LAG-3, CD4, CD8, CD45RO, granzyme B, FoxP3 and CD68. Furthermore, we analyzed the relationship between the immunological parameters and survival, as well as response to chemotherapy. We found that TIM-3, PD-1 and PD-L1 were expressed on both immune and tumor cells. Strikingly, PD-1 and PD-L1 expression on tumor cells was only seen in unpretreated samples. No LAG-3 expression was observed. CD45RO expression in the stroma was an independent negative predictive factor for response on chemotherapy, while CD4 and TIM-3 expression in lymphoid aggregates were independent prognostic factors for better outcome. Our data propose TIM-3 as a promising new target in mesothelioma. Chemotherapy influences the expression of immune checkpoints and therefore further research on the best combination treatment schedule is required.
Collapse
Affiliation(s)
- Elly Marcq
- Center for Oncological Research, University of Antwerp , Antwerp, Belgium
| | - Vasiliki Siozopoulou
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
| | - Jorrit De Waele
- Center for Oncological Research, University of Antwerp , Antwerp, Belgium
| | | | - Karen Zwaenepoel
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
| | - Eva Santermans
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University , Diepenbeek, Belgium
| | - Niel Hens
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Diepenbeek, Belgium; Centre for Health Economics Research and Modeling Infectious Diseases, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | - Patrick Pauwels
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Department of Pathology, Antwerp University Hospital, Antwerp, Belgium
| | - Jan P van Meerbeeck
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Thoracic Oncology/MOCA, Antwerp University Hospital, Antwerp, Belgium
| | - Evelien L J Smits
- Center for Oncological Research, University of Antwerp, Antwerp, Belgium; Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
32
|
Thapa B, Watkins DN, John T. Immunotherapy for malignant mesothelioma: reality check. Expert Rev Anticancer Ther 2016; 16:1167-1176. [DOI: 10.1080/14737140.2016.1241149] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
33
|
Management of malignant pleural mesothelioma-part 2: therapeutic approaches : Consensus of the Austrian Mesothelioma Interest Group (AMIG). Wien Klin Wochenschr 2016; 128:618-26. [PMID: 27457872 PMCID: PMC5033993 DOI: 10.1007/s00508-016-1036-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 06/09/2016] [Indexed: 12/29/2022]
Abstract
Treatment of malignant pleural mesothelioma (MPM) depends on performance status of the patient, tumor stage, and histological differentiation. Chemotherapy (CHT) can be administered as first- and second-line treatment in unresectable MPM or as neoadjuvant or adjuvant treatment before or after surgery. A combination of an antifolate and platinum-based CHT is the only approved standard of care. Several targeted and immunotherapies are in evaluation and further studies are warranted to determine the therapeutic value of these new treatment options. Radiotherapy (RT) can be considered either as adjuvant treatment after surgery or for palliation of pain-related tumor growth. Recent data support the use of RT in a neoadjuvant setting. Macroscopic complete resection by pleurectomy/decortication (P/D) or extrapleural pneumonectomy (EPP) is indicated in selected patients with good performance status. Surgery should only be applied as part of a multimodality treatment (MMT) in combination with chemo- and/or radiotherapy. In a large number of cases, palliative attempts are needed to improve quality of life and to achieve symptom control.
Collapse
|
34
|
Bonelli MA, Fumarola C, La Monica S, Alfieri R. New therapeutic strategies for malignant pleural mesothelioma. Biochem Pharmacol 2016; 123:8-18. [PMID: 27431778 DOI: 10.1016/j.bcp.2016.07.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 07/14/2016] [Indexed: 12/31/2022]
Abstract
Malignant pleural mesothelioma (MPM) is a rare and aggressive malignant disease affecting the mesothelium, commonly associated to asbestos exposure. Therapeutic actions are limited due to the late stage at which most patients are diagnosed and the intrinsic chemo-resistance of the tumor. The recommended systemic therapy for MPM is cisplatin/pemetrexed regimen with a mean overall survival of about 12months and a median progression free survival of less than 6months. Considering that the incidence of this tumor is expected to increase in the next decade and that its prognosis is poor, novel therapeutic approaches are urgently needed. For some tumors, such as lung cancer and breast cancer, druggable oncogenic alterations have been identified and targeted therapy is an important option for these patients. For MPM, clinical guidelines do not recommend biological targeted therapy, mainly because of poor target definition or inappropriate trial design. Further studies are required for a full comprehension of the molecular pathogenesis of MPM and for the development of new target agents. This review updates pre-clinical and clinical data on the efficacy of targeted therapy and immune checkpoint inhibition in the treatment of mesothelioma. Finally, future perspectives in this deadly disease are also discussed.
Collapse
Affiliation(s)
- Mara A Bonelli
- Unit of Experimental Oncology, Department of Clinical and Experimental Medicine, University of Parma, Via Volturno 39, 43126 Parma, Italy.
| | - Claudia Fumarola
- Unit of Experimental Oncology, Department of Clinical and Experimental Medicine, University of Parma, Via Volturno 39, 43126 Parma, Italy.
| | - Silvia La Monica
- Unit of Experimental Oncology, Department of Clinical and Experimental Medicine, University of Parma, Via Volturno 39, 43126 Parma, Italy.
| | - Roberta Alfieri
- Unit of Experimental Oncology, Department of Clinical and Experimental Medicine, University of Parma, Via Volturno 39, 43126 Parma, Italy.
| |
Collapse
|
35
|
Ceresoli GL, Bonomi M, Sauta MG. Immune checkpoint inhibitors in malignant pleural mesothelioma: promises and challenges. Expert Rev Anticancer Ther 2016; 16:673-5. [DOI: 10.1080/14737140.2016.1191951] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Giovanni Luca Ceresoli
- Thoracic and GU Oncology Unit, Department of Medical Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Maria Bonomi
- Thoracic and GU Oncology Unit, Department of Medical Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| | - Maria Grazia Sauta
- Thoracic and GU Oncology Unit, Department of Medical Oncology, Cliniche Humanitas Gavazzeni, Bergamo, Italy
| |
Collapse
|