1
|
Belderbos RA, Vroman H, Aerts JGJV. Cellular Immunotherapy and Locoregional Administration of CAR T-Cells in Malignant Pleural Mesothelioma. Front Oncol 2020; 10:777. [PMID: 32582537 PMCID: PMC7283907 DOI: 10.3389/fonc.2020.00777] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
Malignant pleural mesothelioma (MPM) is a treatment recalcitrant tumor with a poor overall survival (OS). Current approved treatment consists of first line chemotherapy that only modestly increases OS, illustrating the desperate need for other treatment options in MPM. Unfortunately, clinical studies that investigate the effectivity of checkpoint inhibitor (CI) treatment failed to improve clinical outcome over current applied therapies. In general, MPM is characterized as an immunological cold tumor with low T-cell infiltration, which could explain the disappointing results of clinical trials investigating CI treatment in MPM. Currently, many other therapeutic approaches, such as cellular therapies and cancer vaccines are investigated that could induce a tumor-specific immune response and increase of the number of tumor-infiltrating lymphocytes. In this review we will discuss these novel treatment approaches for MPM.
Collapse
Affiliation(s)
- Robert A Belderbos
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, Netherlands.,Erasmus MC Cancer Institute, Erasmus MC Rotterdam, Rotterdam, Netherlands
| | - Heleen Vroman
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, Netherlands.,Erasmus MC Cancer Institute, Erasmus MC Rotterdam, Rotterdam, Netherlands
| | - Joachim G J V Aerts
- Department of Pulmonary Medicine, Erasmus MC Rotterdam, Rotterdam, Netherlands.,Erasmus MC Cancer Institute, Erasmus MC Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
2
|
Zhang M, Yang J, Hua W, Li Z, Xu Z, Qian Q. Monitoring checkpoint inhibitors: predictive biomarkers in immunotherapy. Front Med 2019; 13:32-44. [PMID: 30680606 DOI: 10.1007/s11684-018-0678-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/16/2018] [Indexed: 12/19/2022]
Abstract
Immunotherapy has become the fourth cancer therapy after surgery, chemotherapy, and radiotherapy. In particular, immune checkpoint inhibitors are proved to be unprecedentedly in increasing the overall survival rates of patients with refractory cancers, such as advanced melanoma, non-small cell lung cancer, and renal cell carcinoma. However, inhibitor therapies are only effective in a small proportion of patients with problems, such as side effects and high costs. Therefore, doctors urgently need reliable predictive biomarkers for checkpoint inhibitor therapies to choose the optimal therapies. Here, we review the biomarkers that can serve as potential predictors of the outcomes of immune checkpoint inhibitor treatment, including tumor-specific profiles and tumor microenvironment evaluation and other factors.
Collapse
Affiliation(s)
- Min Zhang
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China
| | - Jingwen Yang
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China
| | - Wenjing Hua
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China
| | - Zhong Li
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China
| | - Zenghui Xu
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China.
| | - Qijun Qian
- ShangHai Cell Therapy Group Co., Ltd., Shanghai, 201805, China.
| |
Collapse
|
3
|
Gilani SNS, Mehta A, Garcia-Fadrique A, Rowaiye B, Jenei V, Dayal S, Chandrakumaran K, Carr N, Mohamed F, Cecil T, Moran B. Outcomes of cytoreductive surgery with hyperthermic intraperitoneal chemotherapy for peritoneal mesothelioma and predictors of survival. Int J Hyperthermia 2018; 34:578-584. [PMID: 29431036 DOI: 10.1080/02656736.2018.1434902] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) benefits selected patients with peritoneal mesothelioma. We present the outcomes of this treatment strategy in a UK peritoneal malignancy national referral centre. METHODS Observational retrospective analysis of data prospectively collected in a dedicated peritoneal malignancy database between March 1998 and January 2016. RESULTS Of 1586 patients treated for peritoneal malignancy, 76 (4.8%) underwent surgery for peritoneal mesothelioma. Median age was 49 years (range 21-73 years). 34 patients (45%) were female. Of the 76 patients, 39 (51%) had low grade histological subtypes (mostly multicystic mesothelioma), and 37 (49%) had diffuse malignant peritoneal mesothelioma (DMPM; mostly epithelioid mesothelioma). Complete cytoreduction was achieved in 52 patients (68%) and maximal tumour debulking (MTD) was performed in 20 patients (26%); the remaining 4 patients (5%) underwent a laparotomy with biopsy only. HIPEC was administered in 67 patients (88%). Median overall (OS) and disease-free survival (DFS) after CRS was 97.8 (80.2-115.4) and 58.8 (47.4-70.3) months, respectively. After complete cytoreduction, 100% overall survival was observed amongst patients with low-grade disease. Ki-67 proliferation index was significantly associated with survival outcomes after complete cytoreduction for DMPM and was an independent predictor of decreased survival. CONCLUSION With adequate patient selection (guided by histological classification and Ki-67 proliferation index) and complete cytoreduction with HIPEC, satisfactory outcomes can be achieved in selected patients with peritoneal mesothelioma.
Collapse
Affiliation(s)
- Syeda Nadia Shah Gilani
- a Peritoneal Malignancy Institute, Basingstoke and North Hampshire Hospital , Basingstoke , UK
| | - Akash Mehta
- a Peritoneal Malignancy Institute, Basingstoke and North Hampshire Hospital , Basingstoke , UK
| | - Alfonso Garcia-Fadrique
- b Department of General and Digestive Surgery , Valencia Oncology Institute Foundation , Valencia , Spain
| | - Babatunde Rowaiye
- a Peritoneal Malignancy Institute, Basingstoke and North Hampshire Hospital , Basingstoke , UK
| | - Veronika Jenei
- c Cancer Sciences Unit , University of Southampton School of Medicine , Southampton , UK
| | - Sanjeev Dayal
- a Peritoneal Malignancy Institute, Basingstoke and North Hampshire Hospital , Basingstoke , UK
| | - Kandiah Chandrakumaran
- a Peritoneal Malignancy Institute, Basingstoke and North Hampshire Hospital , Basingstoke , UK
| | - Norman Carr
- a Peritoneal Malignancy Institute, Basingstoke and North Hampshire Hospital , Basingstoke , UK
| | - Faheez Mohamed
- a Peritoneal Malignancy Institute, Basingstoke and North Hampshire Hospital , Basingstoke , UK
| | - Tom Cecil
- a Peritoneal Malignancy Institute, Basingstoke and North Hampshire Hospital , Basingstoke , UK
| | - Brendan Moran
- a Peritoneal Malignancy Institute, Basingstoke and North Hampshire Hospital , Basingstoke , UK
| |
Collapse
|
4
|
Tremelimumab as second-line or third-line treatment in relapsed malignant mesothelioma (DETERMINE): a multicentre, international, randomised, double-blind, placebo-controlled phase 2b trial. Lancet Oncol 2017; 18:1261-1273. [PMID: 28729154 DOI: 10.1016/s1470-2045(17)30446-1] [Citation(s) in RCA: 303] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 05/05/2017] [Accepted: 05/11/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND New therapeutic strategies for malignant mesothelioma are urgently needed. In the DETERMINE study, we investigated the effects of the cytotoxic-T-lymphocyte-associated antigen 4 (CTLA-4) monoclonal antibody tremelimumab in patients with previously treated advanced malignant mesothelioma. METHODS DETERMINE was a double-blind, placebo-controlled, phase 2b trial done at 105 study centres across 19 countries in patients with unresectable pleural or peritoneal malignant mesothelioma who had progressed after one or two previous systemic treatments for advanced disease. Eligible patients were aged 18 years or older with Eastern Cooperative Oncology Group performance status of 0 or 1 and measurable disease as defined in the modified Response Evaluation Criteria In Solid Tumors (RECIST) version 1.0 for pleural mesothelioma or RECIST version 1.1 for peritoneal mesothelioma. Patients were randomly assigned (2:1) in blocks of three, stratified by European Organisation for Research and Treatment of Cancer status (low risk vs high risk), line of therapy (second line vs third line), and anatomic site (pleural vs peritoneal), by use of an interactive voice or web system, to receive intravenous tremelimumab (10 mg/kg) or placebo every 4 weeks for 7 doses and every 12 weeks thereafter until a treatment discontinuation criterion was met. The primary endpoint was overall survival in the intention-to-treat population. Safety was assessed in all patients who received at least one dose of study drug. The trial is ongoing but no longer recruiting participants, and is registered with ClinicalTrials.gov, number NCT01843374. FINDINGS Between May 17, 2013, and Dec 4, 2014, 571 patients were randomly assigned to receive tremelimumab (n=382) or placebo (n=189), of whom 569 patients received treatment (two patients in the tremelimumab group were excluded from the safety population because they did not receive treatment). At the data cutoff date (Jan 24, 2016), 307 (80%) of 382 patients had died in the tremelimumab group and 154 (81%) of 189 patients had died in the placebo group. Median overall survival in the intention-to-treat population did not differ between the treatment groups: 7·7 months (95% CI 6·8-8·9) in the tremelimumab group and 7·3 months (5·9-8·7) in the placebo group (hazard ratio 0·92 [95% CI 0·76-1·12], p=0·41). Treatment-emergent adverse events of grade 3 or worse occurred in 246 (65%) of 380 patients in the tremelimumab group and 91 (48%) of 189 patients in the placebo group; the most common were dyspnoea (34 [9%] patients in the tremelimumab group vs 27 [14%] patients in the placebo group), diarrhoea (58 [15%] vs one [<1%]), and colitis (26 [7%] vs none). The most common serious adverse events were diarrhoea (69 [18%] patients in the tremelimumab group vs one [<1%] patient in the placebo group), dyspnoea (29 [8%] vs 24 [13%]), and colitis (24 [6%] vs none). Treatment-emergent events leading to death occurred in 36 (9%) of 380 patients in the tremelimumab group and 12 (6%) of 189 in the placebo group; those leading to the death of more than one patient were mesothelioma (three [1%] patients in the tremelimumab group vs two [1%] in the placebo group), dyspnoea (three [1%] vs two [1%]); respiratory failure (one [<1%] vs three [2%]), myocardial infarction (three [1%] vs none), lung infection (three [1%] patients vs none), cardiac failure (one [<1%] vs one [<1%]), and colitis (two [<1%] vs none). Treatment-related adverse events leading to death occurred in five (1%) patients in the tremelimumab group and none in the placebo group. The causes of death were lung infection in one patient, intestinal perforation and small intestinal obstruction in one patient; colitis in two patients, and neuritis and skin ulcer in one patient. INTERPRETATION Tremelimumab did not significantly prolong overall survival compared with placebo in patients with previously treated malignant mesothelioma. The safety profile of tremelimumab was consistent with the known safety profile of CTLA-4 inhibitors. Investigations into whether immunotherapy combination regimens can provide greater efficacy than monotherapies in malignant mesothelioma are ongoing. FUNDING AstraZeneca.
Collapse
|
5
|
Spencer KR, Wang J, Silk AW, Ganesan S, Kaufman HL, Mehnert JM. Biomarkers for Immunotherapy: Current Developments and Challenges. Am Soc Clin Oncol Educ Book 2017; 35:e493-503. [PMID: 27249758 DOI: 10.1200/edbk_160766] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Immunotherapy has revolutionized cancer therapy and has been named the cancer advance of the year for 2016. Checkpoint inhibitors have demonstrated unprecedented rates of durable responses in some of the most difficult-to-treat cancers; however, many treated patients do not respond, and the potential for serious side effects exists. There is a growing need to identify biomarkers that will improve the selection of patients who will best respond to therapy, further elucidate drug mechanisms of action, and help tailor therapy regimens. Biomarkers are being explored at the soluble, cellular, and genomic levels, and examples in immunotherapy include serum proteins, tumor-specific receptor expression patterns, factors in the tumor microenvironment, circulating immune and tumor cells, and host genomic factors. The search for reliable biomarkers is limited by our incomplete understanding of how immunotherapies modify the already complex immune response to cancer, as well as the contribution of immuno-editing to a dynamic and inducible tumor microenvironment and immune milieu. Furthermore, there has been little extension of any candidate assay into large, prospective studies, and the lack of standardization in measurement and interpretation restricts their validity. Both tumor-infiltrating lymphocytes and PD-L1 expression within the tumor microenvironment have been recognized as having both prognostic and predictive value for patients treated with immunotherapy. Alternately, a larger panel of gene signatures, chemokines, and other factors that correlate with response has been proposed. In this article, we will explore the status of current biomarker candidates.
Collapse
Affiliation(s)
| | - Jianfeng Wang
- From the Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Ann W Silk
- From the Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Shridar Ganesan
- From the Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Howard L Kaufman
- From the Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Janice M Mehnert
- From the Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| |
Collapse
|
6
|
Unique fractal evaluation and therapeutic implications of mitochondrial morphology in malignant mesothelioma. Sci Rep 2016; 6:24578. [PMID: 27080907 PMCID: PMC4832330 DOI: 10.1038/srep24578] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/30/2016] [Indexed: 12/15/2022] Open
Abstract
Malignant mesothelioma (MM), is an intractable disease with limited therapeutic options and grim survival rates. Altered metabolic and mitochondrial functions are hallmarks of MM and most other cancers. Mitochondria exist as a dynamic network, playing a central role in cellular metabolism. MM cell lines display a spectrum of altered mitochondrial morphologies and function compared to control mesothelial cells. Fractal dimension and lacunarity measurements are a sensitive and objective method to quantify mitochondrial morphology and most importantly are a promising predictor of response to mitochondrial inhibition. Control cells have high fractal dimension and low lacunarity and are relatively insensitive to mitochondrial inhibition. MM cells exhibit a spectrum of sensitivities to mitochondrial inhibitors. Low mitochondrial fractal dimension and high lacunarity correlates with increased sensitivity to the mitochondrial inhibitor metformin. Lacunarity also correlates with sensitivity to Mdivi-1, a mitochondrial fission inhibitor. MM and control cells have similar sensitivities to cisplatin, a chemotherapeutic agent used in the treatment of MM. Neither oxidative phosphorylation nor glycolytic activity, correlated with sensitivity to either metformin or mdivi-1. Our results suggest that mitochondrial inhibition may be an effective and selective therapeutic strategy in mesothelioma, and identifies mitochondrial morphology as a possible predictor of response to targeted mitochondrial inhibition.
Collapse
|
7
|
Viti A, Bertolaccini L, Terzi A. Biologic therapy and gene therapy in the multimodality treatment of malignant pleural mesothelioma. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:248. [PMID: 26605294 DOI: 10.3978/j.issn.2305-5839.2015.09.40] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The last years have witnessed an abrupt paradigm shift in cancer treatment owing to the discoveries concerning the relationships between the immune system and neoplastic cells. In the field of malignant mesothelioma, which, despite painstaking efforts, remains an incurable form of cancer, the researchers' attention has been seized by a variety of new biologic approaches, including both viral gene therapy and active immunotherapy. The former is meant to induce programmed cell death by introducing a specific gene in the target cell, this gene encoding a specific protein with anticancer activity. Active immunotherapy, on the other hand, tires to induce an active response of the immune system, whose surveillance may be easily dodged by cancer cells. In fact, this mechanism seems to play an important role in the development, growth and diffusion of malignant mesothelioma which easily manages to hinder the immune response. A thorough understanding of the relationships existing between mesothelioma and immune system is the basis for the success of those immune therapies, which are showing promising results in the preclinical setting, especially when combined with other approaches, such as cytoreductive surgery.
Collapse
Affiliation(s)
- Andrea Viti
- Thoracic Surgery Unit, Sacro Cuore - Don Calabria Research Hospital, Negrar Verona, Italy
| | - Luca Bertolaccini
- Thoracic Surgery Unit, Sacro Cuore - Don Calabria Research Hospital, Negrar Verona, Italy
| | - Alberto Terzi
- Thoracic Surgery Unit, Sacro Cuore - Don Calabria Research Hospital, Negrar Verona, Italy
| |
Collapse
|