1
|
Yadollahvandmiandoab R, Jalalizadeh M, Buosi K, Garcia-Perdomo HA, Reis LO. Immunogenic Cell Death Role in Urothelial Cancer Therapy. Curr Oncol 2022; 29:6700-6713. [PMID: 36135095 PMCID: PMC9498148 DOI: 10.3390/curroncol29090526] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/07/2022] [Accepted: 09/16/2022] [Indexed: 12/05/2022] Open
Abstract
PURPOSE Bladder cancer is the 13th most common cause of cancer death with the highest lifetime cost for treatment of all cancers. This scoping review clarifies the available evidence on the role of a novel therapeutic approach called immunogenic cell death (ICD) in urothelial cancer of the bladder. METHODS In accordance with the recommendations of the Joanna Briggs Institute, we searched MEDLINE (Ovid), EMBASE, CENTRAL databases, and supplemented with manual searches through the conferences, Google scholar, and clinicaltrials.gov for published studies up to April 2022. We included literature that studied molecular mechanisms of ICD and the role of certain danger-associated molecular patterns (DAMPs) in generating ICD, safety and efficacy of different ICD inducers, and their contributions in combination with other urothelial cancer treatments. RESULTS Oncolytic viruses, radiotherapy, certain chemo/chemo radiation therapy combinations, photodynamic therapy, and novel agents were studied as ICD-inducing treatment modalities in the included studies. ICD was observed in vitro (murine or human urothelial carcinoma) in ten studies, eight studies were performed on mouse models (orthotopic or subcutaneous), and five clinical trials assessed patient response to ICD inducing agents. The most common studied DAMPs were Calreticulin, HMGB1, ATP, and Heat Shock Proteins (HSP) 70 and 90, which were either expressed on the cancer cells or released. CONCLUSION ICD inducers were able to generate lasting antitumor immune responses with memory formation in animal studies (vaccination effect). In clinical trials these agents generally had low side effects, except for one trial, and could be used alone or in combination with other cancer treatment strategies in urothelial cancer patients.
Collapse
Affiliation(s)
- Reza Yadollahvandmiandoab
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo 13083-970, Brazil
| | - Mehrsa Jalalizadeh
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo 13083-970, Brazil
| | - Keini Buosi
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo 13083-970, Brazil
| | - Herney Andrés Garcia-Perdomo
- Division of Urology/Urooncology, Department of Surgery, School of Medicine, Universidad del Valle, Cali 72824, Colombia
| | - Leonardo Oliveira Reis
- UroScience, School of Medical Sciences, University of Campinas, UNICAMP, Campinas, Sao Paulo 13083-970, Brazil
- Center for Life Sciences, Pontifical Catholic University of Campinas, PUC-Campinas, Sao Paulo 13087-571, Brazil
| |
Collapse
|
2
|
Tripodi L, Vitale M, Cerullo V, Pastore L. Oncolytic Adenoviruses for Cancer Therapy. Int J Mol Sci 2021; 22:2517. [PMID: 33802281 PMCID: PMC7959120 DOI: 10.3390/ijms22052517] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/24/2021] [Accepted: 02/25/2021] [Indexed: 12/11/2022] Open
Abstract
Many immuno-therapeutic strategies are currently being developed to fight cancer. In this scenario, oncolytic adenoviruses (Onc.Ads) have an interesting role for their peculiar tumor selectivity, safety, and transgene-delivery capability. The major strength of the Onc.Ads is the extraordinary immunogenicity that leads to a strong T-cell response, which, together with the possibility of the delivery of a therapeutic transgene, could be more effective than current strategies. In this review, we travel in the adenovirus (Ads) and Onc.Ads world, focusing on a variety of strategies that can enhance Onc.Ads antitumoral efficacy, passing through tumor microenvironment modulation. Onc.Ads-based therapeutic strategies constitute additional weapons in the fight against cancer and appear to potentiate conventional and immune checkpoint inhibitors (ICIs)-based therapies leading to a promising scenario.
Collapse
Affiliation(s)
- Lorella Tripodi
- SEMM European School for Molecular Medicine, 20123 Milano, Italy;
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy;
| | - Maria Vitale
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy;
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Napoli, Italy
| | - Vincenzo Cerullo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Napoli, Italy
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland
| | - Lucio Pastore
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy;
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, 80131 Napoli, Italy
| |
Collapse
|
3
|
Feng Z, Chen Q. Raised CD40L expression attenuates drug resistance in Adriamycin-resistant THP-1 cells. Exp Ther Med 2020; 19:2188-2194. [PMID: 32104283 PMCID: PMC7027340 DOI: 10.3892/etm.2020.8452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 10/04/2019] [Indexed: 12/18/2022] Open
Abstract
Acute myeloid leukemia is a common hematological malignancy that often exhibits strong drug resistance when treated using conventional chemotherapy. Although numerous studies have been carried out to develop methods of overcoming drug resistance, the results have generally been unsatisfactory. CD40 ligand (CD40L) has been shown to improve the sensitivity of cancer cells to drug treatment. In the present study, Adriamycin (ADM)-resistant human monocytic THP-1 cells (THP-1/A cells) were developed by incubating THP-1 cells with increasing concentrations of ADM. Cells were transfected with CD40L vectors to explore the potential involvement of CD40L in regulating multidrug resistance (MDR) in cancer. Cell proliferation and viability were measured using the Cell Counting Kit-8 assay; cell apoptosis was evaluated by flow cytometry, trypan blue staining and caspase-3 activity; and the expression of MDR-associated protein 1 (MRP1) and permeability glycoprotein (P-gp) was analyzed using western blotting. The results revealed that the protein expression levels of MRP1 and P-gp were downregulated by raised CD40L expression and that the combination of raised CD40L expression with daunorubicin (DNR), a drug from which ADM is derived, significantly increased the extent of cell apoptosis, indicating that drug resistance was effectively attenuated by CD40L. Collectively, these results suggested that CD40L may contribute towards reducing DNR resistance in THP-1/A cells.
Collapse
Affiliation(s)
- Zhongxin Feng
- Department of Hematology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Qi Chen
- Department of Hematology, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| |
Collapse
|
4
|
Ylösmäki E, Cerullo V. Design and application of oncolytic viruses for cancer immunotherapy. Curr Opin Biotechnol 2019; 65:25-36. [PMID: 31874424 DOI: 10.1016/j.copbio.2019.11.016] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 12/28/2022]
Abstract
The approval of the first oncolytic virus (OV) for the treatment of metastatic melanoma and the recent discovery that the use of oncolytic viruses may enhance cancer immunotherapies targeted against various immune checkpoint proteins have attracted great interest in the field of cancer virotherapy. OVs are designed to target and kill cancer cells leaving normal cell unharmed. OV infection and concomitant cancer cell killing stimulate anti-tumour immunity and modulates tumour microenvironment towards less immunosuppressive phenotype. The intrinsic capacity of OVs to turn immunologically cold tumours into immunologically hot tumours, and to increase immune cell and cytokine infiltration, can be further enhanced by arming OVs with transgenes that increase their immunostimulatory activities and direct immune responses specifically towards cancer cells. These OVs, specifically engineered to be used as cancer immunotherapeutics, can be synergized with other immune modulators or cytotoxic agents to achieve the most potent immunotherapy for cancer.
Collapse
Affiliation(s)
- Erkko Ylösmäki
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| | - Vincenzo Cerullo
- Laboratory of Immunovirotherapy, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
5
|
Baxevanis CN, Sofopoulos M, Fortis SP, Perez SA. The role of immune infiltrates as prognostic biomarkers in patients with breast cancer. Cancer Immunol Immunother 2019; 68:1671-1680. [PMID: 30905043 PMCID: PMC11028310 DOI: 10.1007/s00262-019-02327-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 03/19/2019] [Indexed: 02/07/2023]
Abstract
The presence of immune infiltrates in the tumor microenvironment has been documented in many types of cancer. Moreover, the preexistent or endogenous immunity which consists of interactions between intratumoral lymphocytes and tumor cells is mostly relevant for the successful application of various anticancer therapies, including standard chemotherapy, immune checkpoint inhibition-based immunotherapy and targeted therapies. The immunoscore defines densities of intratumoral immune infiltrates which determine poor or favorable prognosis depending on their quantity and quality in the tumor compartments. Results from large clinical studies have demonstrated an association between high densities of cytotoxic and memory TILs in the tumor compartments with improved prognosis. Importantly, we have demonstrated that differential combined densities of immune infiltrates jointly analyzed in the tumor center (TC) and the invasive margin (IM) have a significant prognostic value in breast cancer patients with poor clinicopathological parameters.
Collapse
Affiliation(s)
- Constantin N Baxevanis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Ave., 115 22, Athens, Greece.
| | | | - Sotirios P Fortis
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Ave., 115 22, Athens, Greece
| | - Sonia A Perez
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, 171 Alexandras Ave., 115 22, Athens, Greece
| |
Collapse
|
6
|
Puttmann K, Duggan M, Mortazavi A, Diaz DA, Carson III WE, Sundi D. The Role of Myeloid Derived Suppressor Cells in Urothelial Carcinoma Immunotherapy. Bladder Cancer 2019. [DOI: 10.3233/blc-190219] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Kathleen Puttmann
- Department of Urology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Megan Duggan
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Amir Mortazavi
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Dayssy Alexandra Diaz
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - William E. Carson III
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Debasish Sundi
- Department of Urology, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
7
|
Eriksson E, Milenova I, Wenthe J, Moreno R, Alemany R, Loskog A. IL-6 Signaling Blockade during CD40-Mediated Immune Activation Favors Antitumor Factors by Reducing TGF-β, Collagen Type I, and PD-L1/PD-1. THE JOURNAL OF IMMUNOLOGY 2019; 202:787-798. [PMID: 30617223 DOI: 10.4049/jimmunol.1800717] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 11/22/2018] [Indexed: 12/14/2022]
Abstract
IL-6 plays a role in cancer pathogenesis via its connection to proteins involved in the formation of desmoplastic stroma and to immunosuppression by driving differentiation of myeloid suppressor cells together with TGF-β. Inhibition of IL-6 signaling in the tumor microenvironment may, thus, limit desmoplasia and myeloid suppressor cell differentiation. CD40 signaling can further revert myeloid cell differentiation toward antitumor active phenotypes. Hence, the simultaneous use of IL-6 blockade with CD40 stimuli may tilt the tumor microenvironment to promote antitumor immune responses. In this paper, we evaluated the mechanisms of LOAd713, an oncolytic adenovirus designed to block IL-6R signaling and to provide myeloid cell activation via a trimerized membrane-bound isoleucine zipper (TMZ) CD40L. LOAd713-infected pancreatic cancer cells were killed by oncolysis, whereas infection of stellate cells reduced factors involved in stroma formation, including TGF-β-1 and collagen type I. Virus infection prevented IL-6/GM-CSF-mediated differentiation of myeloid suppressors, but not CD163 macrophages, whereas infection of dendritic cells led to upregulation of maturation markers, including CD83, CD86, IL-12p70, and IFN-γ. Further, IL-6R blockade prevented upregulation of programed death ligand 1 (PD-L1) and PD-1 on the stimulated dendritic cells. These results suggest that LOAd713 can kill infected tumor cells and has the capacity to affect the tumor microenvironment by stimulating stellate cells and myeloid suppressors with TMZ-CD40L and IL-6R blockade. Gene transfer of murine TMZ-CD40L prolonged survival in an animal model. LOAd713 may be an interesting therapeutic option for cancers connected to IL-6 signaling, such as pancreatic cancer.
Collapse
Affiliation(s)
- Emma Eriksson
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Ioanna Milenova
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Jessica Wenthe
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden
| | - Rafael Moreno
- L'Institut d'Investigació Biomèdica de Bellvitge-Institut Català d'Oncologia, L'Hospitalet de Llobregat, 08908 Barcelona, Spain; and
| | - Ramon Alemany
- L'Institut d'Investigació Biomèdica de Bellvitge-Institut Català d'Oncologia, L'Hospitalet de Llobregat, 08908 Barcelona, Spain; and
| | - Angelica Loskog
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 751 85 Uppsala, Sweden; .,Lokon Pharma AB, 751 83 Uppsala, Sweden
| |
Collapse
|
8
|
Zhang W, Zhong Y, Cui H, Wang L, Yang R, Su Z, Xiang B, Wei Q. Combination of calcineurin B subunit (CnB) and 5-fluorouracil reverses 5-fluorouracil-induced immunosuppressive effect and enhances the antitumor activity in hepatocellular carcinoma. Oncol Lett 2017; 14:6135-6142. [PMID: 29113258 DOI: 10.3892/ol.2017.6958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 08/03/2017] [Indexed: 02/05/2023] Open
Abstract
Five-fluorouracil (5-FU) is a widely used chemotherapeutic agent for digestive system tumors; however, continuous use of 5-FU may cause severe side effects, including myelosuppression and immunosuppression. Our previous study revealed that calcineurin B subunit (CnB), an innovative genetic engineering antitumor protein, possesses tumor-suppressive effects with low toxicity. CnB can bind to and activate integrin αM on macrophages, subsequently promoting the expression, and secretion of TNF-related apoptosis-inducing ligand, a specific proapoptotic cytokine. In the present study, whether the combined use of CnB and 5-FU can reverse the myelosuppression, and immunosuppressive effects of 5-FU by reactivating the immune system thus increasing antitumor efficacy, was investigated. It was demonstrated that combined treatment of 5-FU and CnB led to increased tumor-suppressive effects, as indicated by reduced tumor volume and weight when compared with 5-FU or CnB treatment alone in a hepatoma xenograph model. In addition, it was demonstrated that combined treatment inhibited the proliferation of hepatoma cells. Notably, the addition of CnB to 5-FU-based therapy completely reversed the immunosuppressive effect of 5-FU. The spleen index and total number of white blood cells in the combination group were higher compared with that of the 5-FU alone group. Furthermore, pathological examinations indicated that CnB attenuated 5-FU-induced organ damage. Based on these findings, it is proposed that CnB may serve as a novel and promising drug candidate for the improvement of 5-FU-based chemotherapy.
Collapse
Affiliation(s)
- Wenlong Zhang
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing 100875, P.R. China
| | - Youxiu Zhong
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing 100875, P.R. China
| | - Hongfei Cui
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing 100875, P.R. China
| | - Liya Wang
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing 100875, P.R. China
| | - Rui Yang
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing 100875, P.R. China
| | - Zhenyi Su
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing 100875, P.R. China.,Department of Biochemistry, School of Medicine, Southeast University, Nanjing, Jiangsu, P.R. China
| | - Benqiong Xiang
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing 100875, P.R. China
| | - Qun Wei
- Department of Biochemistry and Molecular Biology, Gene Engineering and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing 100875, P.R. China
| |
Collapse
|
9
|
Liang M, Zhan F, Zhao J, Li Q, Wuyang J, Mu G, Li D, Zhang Y, Huang X. CPA-7 influences immune profile and elicits anti-prostate cancer effects by inhibiting activated STAT3. BMC Cancer 2016; 16:504. [PMID: 27435207 PMCID: PMC4952363 DOI: 10.1186/s12885-016-2488-6] [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: 02/24/2016] [Accepted: 06/30/2016] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Platinum-based chemotherapy is emerging as the first line of treatment for castration resistant prostate cancer. Among the family of platinum (IV)-based compounds, a member known as CPA-7 inhibits the growth of multiple cancer cell lines. However, how and to what extent CPA-7 elicits its anti-prostate cancer effects in vivo is largely unknown. METHODS In this study, we firstly assessed the potential toxicity of the synthesized CPA-7 in a prostate cancer model as well as in normal mice. Next, we evaluated the in vitro effects of CPA-7 on the growth of prostate cancer cells using cell counting assay, and calculated the tumor sizes and cumulative survival rate of the tumor bearing mice by Kaplan-Meier method during CPA-7 treatment. Then we measured the expression level of the activated form of STAT3 (one targets of CPA-7) and its transcriptive activity post CPA-7 treatment by synergistically using western blot, IHC, and firefly luciferase reporter assays. Finally, effects of CPA-7 on immune cell trafficking in the tumor draining lymph nodes and in the spleens are evaluated with flow cytometry. RESULTS Treatment with CPA-7 significantly inhibited growth of prostate cancer cells in vitro, and also in mice resulting in a prolonged survival and a decreased recurrence rate. These therapeutic effects are due, at least in part, to functional depletion of STAT3 in prostate tumor tissue as well as in the surrounding areas of tumor cell invasion. CPA-7 treatment also resulted in a reduced level of regulatory T cells and increased levels of cytotoxic T and T helper cells in the spleen and in tumor infiltrating lymph nodes. This favorable effect on immune cell trafficking may account for the amnestic immune response against recurrent prostate cancer. CONCLUSIONS CPA-7 is a promising new therapeutic agent for prostate cancer that both inhibits tumor cell proliferation and stimulates anti-tumor immunity. It has potential as first line treatment and/or as an adjuvant for refractory prostate cancer.
Collapse
Affiliation(s)
- Meihua Liang
- Department of Endocrinology, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Fei Zhan
- Department of Gastrointestinal Medical Oncology, The Affiliated Tumor Hospital of Harbin Medical University, Harbin, 150081, China
| | - Juan Zhao
- Biotherapy Center, Tumor Hospital of Harbin Medical University, Harbin, 150081, China
| | - Qi Li
- Biotherapy Center, Tumor Hospital of Harbin Medical University, Harbin, 150081, China
| | - Jiazi Wuyang
- Biotherapy Center, Tumor Hospital of Harbin Medical University, Harbin, 150081, China
| | - Guannan Mu
- Biotherapy Center, Tumor Hospital of Harbin Medical University, Harbin, 150081, China
| | - Dianjun Li
- Biotherapy Center, Tumor Hospital of Harbin Medical University, Harbin, 150081, China
| | - Yanqiao Zhang
- Department of Gastrointestinal Medical Oncology, The Affiliated Tumor Hospital of Harbin Medical University, Harbin, 150081, China.
| | - Xiaoyi Huang
- Biotherapy Center, Tumor Hospital of Harbin Medical University, Harbin, 150081, China. .,Center of Translational Medicine, Harbin Medical University, Harbin, 150086, China.
| |
Collapse
|
10
|
Cook AM, Lesterhuis WJ, Nowak AK, Lake RA. Chemotherapy and immunotherapy: mapping the road ahead. Curr Opin Immunol 2015; 39:23-9. [PMID: 26724433 DOI: 10.1016/j.coi.2015.12.003] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 12/07/2015] [Accepted: 12/08/2015] [Indexed: 12/17/2022]
Abstract
Cancer immunotherapy, and in particular checkpoint blockade, is now standard clinical care for a growing number of cancers. Cytotoxic drugs have been the primary weapon against cancer for a long time and have typically been understood because of their capacity to directly kill tumour cells. It is now clear that these drugs are potential partners for checkpoint blockade and different drugs can influence the immune response to cancer through a wide variety of mechanisms. Some of these relate to immunogenic cell death, whilst others relate to changes in antigen-presentation, tumour cell targeting, or depletion of immunosuppressive cells. Here, we review some recent advances in our understanding of the immunological changes associated with chemotherapy, discuss progress in combining chemotherapy with checkpoint blockade, and comment on the difficulties encountered in translating promising preclinical data into successful treatments for cancer patients.
Collapse
Affiliation(s)
- Alistair M Cook
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Australia; School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | - W Joost Lesterhuis
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Australia; School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | - Anna K Nowak
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Australia; School of Medicine and Pharmacology, University of Western Australia, Perth, Australia; Department of Medical Oncology, Sir Charles Gairdner Hospital, Perth, Australia
| | - Richard A Lake
- National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Australia; School of Medicine and Pharmacology, University of Western Australia, Perth, Australia.
| |
Collapse
|
11
|
Evidence for Oncolytic Virotherapy: Where Have We Got to and Where Are We Going? Viruses 2015; 7:6291-312. [PMID: 26633468 PMCID: PMC4690862 DOI: 10.3390/v7122938] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/20/2015] [Accepted: 11/25/2015] [Indexed: 12/13/2022] Open
Abstract
The last few years have seen an increased interest in immunotherapy in the treatment of malignant disease. In particular, there has been significant enthusiasm for oncolytic virotherapy, with a large amount of pre-clinical data showing promise in animal models in a wide range of tumour types. How do we move forward into the clinical setting and translate something which has such potential into meaningful clinical outcomes? Here, we review how the field of oncolytic virotherapy has developed thus far and what the future may hold.
Collapse
|
12
|
Rushworth D, Alpert A, Santana-Carrero R, Olivares S, Spencer D, Cooper LJN. Antithymidylate resistance enables transgene selection and cell survival for T cells in the presence of 5-fluorouracil and antifolates. Gene Ther 2015; 23:119-28. [PMID: 26273805 DOI: 10.1038/gt.2015.88] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Revised: 07/11/2015] [Accepted: 08/05/2015] [Indexed: 12/31/2022]
Abstract
Antithymidylates (AThy) constitute a class of drugs used in the treatment of cancers such as lung, colon, breast and pancreas. These drugs inhibit DNA synthesis by targeting the enzymes dihydrofolate reductase (DHFR) and/or thymidylate synthase (TYMS). AThys effectively inhibit cancer cells, and also inhibit T cells, preventing anticancer immunity, which might otherwise develop from AThy-induced cancer destruction. We establish that T cells expressing mutant DHFR--DHFR L22F, F31S (DHFR(FS))--and/or mutant TYMS--TYMS T51S, G52S (TYMS(SS))-effectively survive in toxic concentrations of AThys methotrexate, pemetrexed and 5-fluorouracil. Furthermore, we show that DHFR(FS) permitted rapid selection of an inducible suicide transgene in T cells. These findings demonstrate that AThy resistances prevent AThy cytotoxicity to T cells while permitting selection of important transgenes. This technological development could enhance in vitro and in vivo survival and selection of T-cell therapeutics being designed for a broad range of cancers.
Collapse
Affiliation(s)
- D Rushworth
- Division of Pediatrics, Children's Cancer Hospital, The University of Texas MD Anderson Cancer Center, Unit 907, Houston, TX, USA.,The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - A Alpert
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - R Santana-Carrero
- The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA.,University of Puerto Rico School of Medicine, San Juan, Puerto Rico, United States Minor Outlying Islands
| | - S Olivares
- Division of Pediatrics, Children's Cancer Hospital, The University of Texas MD Anderson Cancer Center, Unit 907, Houston, TX, USA
| | - D Spencer
- Bellicum Pharmaceuticals, Houston, TX, USA
| | - L J N Cooper
- Division of Pediatrics, Children's Cancer Hospital, The University of Texas MD Anderson Cancer Center, Unit 907, Houston, TX, USA.,The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| |
Collapse
|
13
|
Hassan GS, Stagg J, Mourad W. Role of CD154 in cancer pathogenesis and immunotherapy. Cancer Treat Rev 2015; 41:431-40. [PMID: 25843228 DOI: 10.1016/j.ctrv.2015.03.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 03/19/2015] [Accepted: 03/20/2015] [Indexed: 01/11/2023]
Abstract
Many factors and molecules have been investigated as potential players in the pathogenesis or immunosurveillance of cancer. Among these, CD154 has been recognized as a co-stimulatory molecule with high potential for treating cancer, in addition to its contribution in the development of the disease. CD154 was initially described for its pivotal role in T cell-dependent humoral responses via an interaction with its classical receptor, CD40. Subsequent studies showed that CD154 is also implicated in cell-mediated immunity and inflammation via an interaction with CD40 alone or in combination with newly identified receptors, members of the integrin family, leading to the development of chronic inflammatory and autoimmune diseases. In the current article, we present an overview of the role of CD154 as a potential etiological factor in tumors inducing proliferation of malignant cells, their rescue from apoptosis and their invasiveness. In addition, this review describes the immuno-regulatory functions of CD154 against cancer reflected by its stimulation of antigen-presenting cells and the subsequent activation of effector cells, its enhancement of malignant cells' immunogenicity, its modulation of immune settings around tumors, and its initiation of proliferation inhibiting effects in malignant cells. In vitro as well as in vivo studies are outlined and a particular attention is given to clinical studies and progress reached at this point. Findings reviewed herein will improve our knowledge of the role of the CD154 system in cancers from causative to immunotherapeutic functions, paving the way for the identification of new targets for prevention and/or treatment of malignant disorders.
Collapse
Affiliation(s)
- Ghada S Hassan
- Centre de Recherche-Centre Hospitalier de l'Université de Montréal (CR-CHUM), Montréal, Quebec, Canada
| | - John Stagg
- Centre de Recherche-Centre Hospitalier de l'Université de Montréal (CR-CHUM), Montréal, Quebec, Canada
| | - Walid Mourad
- Centre de Recherche-Centre Hospitalier de l'Université de Montréal (CR-CHUM), Montréal, Quebec, Canada.
| |
Collapse
|
14
|
Kepp O, Senovilla L, Vitale I, Vacchelli E, Adjemian S, Agostinis P, Apetoh L, Aranda F, Barnaba V, Bloy N, Bracci L, Breckpot K, Brough D, Buqué A, Castro MG, Cirone M, Colombo MI, Cremer I, Demaria S, Dini L, Eliopoulos AG, Faggioni A, Formenti SC, Fučíková J, Gabriele L, Gaipl US, Galon J, Garg A, Ghiringhelli F, Giese NA, Guo ZS, Hemminki A, Herrmann M, Hodge JW, Holdenrieder S, Honeychurch J, Hu HM, Huang X, Illidge TM, Kono K, Korbelik M, Krysko DV, Loi S, Lowenstein PR, Lugli E, Ma Y, Madeo F, Manfredi AA, Martins I, Mavilio D, Menger L, Merendino N, Michaud M, Mignot G, Mossman KL, Multhoff G, Oehler R, Palombo F, Panaretakis T, Pol J, Proietti E, Ricci JE, Riganti C, Rovere-Querini P, Rubartelli A, Sistigu A, Smyth MJ, Sonnemann J, Spisek R, Stagg J, Sukkurwala AQ, Tartour E, Thorburn A, Thorne SH, Vandenabeele P, Velotti F, Workenhe ST, Yang H, Zong WX, Zitvogel L, Kroemer G, Galluzzi L. Consensus guidelines for the detection of immunogenic cell death. Oncoimmunology 2014; 3:e955691. [PMID: 25941621 PMCID: PMC4292729 DOI: 10.4161/21624011.2014.955691] [Citation(s) in RCA: 640] [Impact Index Per Article: 58.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/04/2014] [Indexed: 02/07/2023] Open
Abstract
Apoptotic cells have long been considered as intrinsically tolerogenic or unable to elicit immune responses specific for dead cell-associated antigens. However, multiple stimuli can trigger a functionally peculiar type of apoptotic demise that does not go unnoticed by the adaptive arm of the immune system, which we named "immunogenic cell death" (ICD). ICD is preceded or accompanied by the emission of a series of immunostimulatory damage-associated molecular patterns (DAMPs) in a precise spatiotemporal configuration. Several anticancer agents that have been successfully employed in the clinic for decades, including various chemotherapeutics and radiotherapy, can elicit ICD. Moreover, defects in the components that underlie the capacity of the immune system to perceive cell death as immunogenic negatively influence disease outcome among cancer patients treated with ICD inducers. Thus, ICD has profound clinical and therapeutic implications. Unfortunately, the gold-standard approach to detect ICD relies on vaccination experiments involving immunocompetent murine models and syngeneic cancer cells, an approach that is incompatible with large screening campaigns. Here, we outline strategies conceived to detect surrogate markers of ICD in vitro and to screen large chemical libraries for putative ICD inducers, based on a high-content, high-throughput platform that we recently developed. Such a platform allows for the detection of multiple DAMPs, like cell surface-exposed calreticulin, extracellular ATP and high mobility group box 1 (HMGB1), and/or the processes that underlie their emission, such as endoplasmic reticulum stress, autophagy and necrotic plasma membrane permeabilization. We surmise that this technology will facilitate the development of next-generation anticancer regimens, which kill malignant cells and simultaneously convert them into a cancer-specific therapeutic vaccine.
Collapse
Key Words
- APC, antigen-presenting cell
- ATF6, activating transcription factor 6
- ATP release
- BAK1, BCL2-antagonist/killer 1
- BAX, BCL2-associated X protein
- BCL2, B-cell CLL/lymphoma 2 protein
- CALR, calreticulin
- CTL, cytotoxic T lymphocyte
- DAMP, damage-associated molecular pattern
- DAPI, 4′,6-diamidino-2-phenylindole
- DiOC6(3), 3,3′-dihexyloxacarbocyanine iodide
- EIF2A, eukaryotic translation initiation factor 2A
- ER, endoplasmic reticulum
- FLT3LG, fms-related tyrosine kinase 3 ligand
- G3BP1, GTPase activating protein (SH3 domain) binding protein 1
- GFP, green fluorescent protein
- H2B, histone 2B
- HMGB1
- HMGB1, high mobility group box 1
- HSP, heat shock protein
- HSV-1, herpes simplex virus type I
- ICD, immunogenic cell death
- IFN, interferon
- IL, interleukin
- MOMP, mitochondrial outer membrane permeabilization
- PDIA3, protein disulfide isomerase family A
- PI, propidium iodide
- RFP, red fluorescent protein
- TLR, Toll-like receptor
- XBP1, X-box binding protein 1
- autophagy
- calreticulin
- endoplasmic reticulum stress
- immunotherapy
- member 3
- Δψm, mitochondrial transmembrane potential
Collapse
Affiliation(s)
- Oliver Kepp
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Laura Senovilla
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
- INSERM; U1015; Villejuif, France
| | - Ilio Vitale
- Regina Elena National Cancer Institute; Rome, Italy
| | - Erika Vacchelli
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Sandy Adjemian
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Molecular Cell Biology Laboratory; Department of Immunology; Institute of Biomedical Sciences; University of São Paulo; São Paulo, Brazil
| | - Patrizia Agostinis
- Cell Death Research and Therapy (CDRT) Laboratory; Department of Cellular and Molecular Medicine; University of Leuven; Leuven, Belgium
| | - Lionel Apetoh
- INSERM; UMR866; Dijon, France
- Centre Georges François Leclerc; Dijon, France
- Université de Bourgogne; Dijon, France
| | - Fernando Aranda
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Vincenzo Barnaba
- Departement of Internal Medicine and Medical Sciences; University of Rome La Sapienza; Rome, Italy
- Istituto Pasteur; Fondazione Cenci Bolognetti; Rome, Italy
| | - Norma Bloy
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Laura Bracci
- Department of Hematology; Oncology and Molecular Medicine; Istituto Superiore di Sanità (ISS); Rome, Italy
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy (LMCT); Department of Biomedical Sciences Medical School of the Free University of Brussels (VUB); Jette, Belgium
| | - David Brough
- Faculty of Life Sciences; University of Manchester; Manchester, UK
| | - Aitziber Buqué
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Maria G. Castro
- Department of Neurosurgery and Cell and Developmental Biology; University of Michigan School of Medicine; Ann Arbor, MI USA
| | - Mara Cirone
- Department of Experimental Medicine; University of Rome La Sapienza; Rome, Italy
| | - Maria I. Colombo
- Laboratorio de Biología Celular y Molecular; Instituto de Histología y Embriología (IHEM); Facultad de Ciencias Médicas; Universidad Nacional de Cuyo; CONICET; Mendoza, Argentina
| | - Isabelle Cremer
- INSERM; U1138; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Equipe 13; Center de Recherche des Cordeliers; Paris, France
| | - Sandra Demaria
- Department of Pathology; New York University School of Medicine; New York, NY USA
| | - Luciana Dini
- Department of Biological and Environmental Science and Technology (DiSTeBA); University of Salento; Lecce, Italy
| | - Aristides G. Eliopoulos
- Molecular and Cellular Biology Laboratory; Division of Basic Sciences; University of Crete Medical School; Heraklion, Greece
- Institute of Molecular Biology and Biotechnology; Foundation of Research and Technology - Hellas; Heraklion, Greece
| | - Alberto Faggioni
- Department of Experimental Medicine; University of Rome La Sapienza; Rome, Italy
| | - Silvia C. Formenti
- Department of Radiation Oncology; NewYork University School of Medicine and Langone Medical Center; New York, NY USA
| | - Jitka Fučíková
- Department of Immunology; 2 Faculty of Medicine and University Hospital Motol, Charles University; Prague, Czech Republic
- Sotio; Prague, Czech Republic
| | - Lucia Gabriele
- Department of Hematology; Oncology and Molecular Medicine; Istituto Superiore di Sanità (ISS); Rome, Italy
| | - Udo S. Gaipl
- Department of Radiation Oncology; University Hospital Erlangen; University of Erlangen-Nürnberg; Erlangen, Germany
| | - Jérôme Galon
- INSERM; U1138; Paris, France
- Université Pierre et Marie Curie/Paris VI; Paris, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Laboratory of Integrative Cancer Immunology; Center de Recherche des Cordeliers; Paris, France
| | - Abhishek Garg
- Cell Death Research and Therapy (CDRT) Laboratory; Department of Cellular and Molecular Medicine; University of Leuven; Leuven, Belgium
| | - François Ghiringhelli
- INSERM; UMR866; Dijon, France
- Centre Georges François Leclerc; Dijon, France
- Université de Bourgogne; Dijon, France
| | - Nathalia A. Giese
- European Pancreas Center; Department of Surgery; University Hospital Heidelberg; Heidelberg, Germany
| | - Zong Sheng Guo
- Department of Surgery; University of Pittsburgh; Pittsburgh, PA USA
| | - Akseli Hemminki
- Cancer Gene Therapy Group; Transplantation laboratory; Haartman Institute; University of Helsinki; Helsinki, Finland
| | - Martin Herrmann
- Department of Internal Medicine 3; University of Erlangen-Nuremberg; Erlangen, Germany
| | - James W. Hodge
- Laboratory of Tumor Immunology and Biology; Center for Cancer Research; National Cancer Institute (NCI), National Institutes of Health (NIH); Bethesda, MD USA
| | - Stefan Holdenrieder
- Institute of Clinical Chemistry and Clinical Pharmacology; University Hospital Bonn; Bonn, Germany
| | - Jamie Honeychurch
- Faculty of Medical and Human Sciences, Institute of Cancer Studies; Manchester Academic Health Sciences Center; University of Manchester; Manchester, UK
| | - Hong-Min Hu
- Cancer Research and Biotherapy Center; Second Affiliated Hospital of Southeast University; Nanjing, China
- Laboratory of Cancer Immunobiology; Earle A. Chiles Research Institute; Providence Portland Medical Center; Portland, OR USA
| | - Xing Huang
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
| | - Tim M. Illidge
- Faculty of Medical and Human Sciences, Institute of Cancer Studies; Manchester Academic Health Sciences Center; University of Manchester; Manchester, UK
| | - Koji Kono
- Department of Surgery; National University of Singapore; Singapore, Singapore
- Cancer Science Institute of Singapore; National University of Singapore; Singapore, Singapore
| | | | - Dmitri V. Krysko
- VIB Inflammation Research Center; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
| | - Sherene Loi
- Division of Cancer Medicine and Division of Research; Peter MacCallum Cancer Center; East Melbourne; Victoria, Australia
| | - Pedro R. Lowenstein
- Department of Neurosurgery and Cell and Developmental Biology; University of Michigan School of Medicine; Ann Arbor, MI USA
| | - Enrico Lugli
- Unit of Clinical and Experimental Immunology; Humanitas Clinical and Research Center; Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan; Rozzano, Italy
| | - Yuting Ma
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Frank Madeo
- Institute of Molecular Biosciences; University of Graz; Graz, Austria
| | - Angelo A. Manfredi
- University Vita-Salute San Raffaele; Milano, Italy
- San Raffaele Scientific Institute; Milano, Italy
| | - Isabelle Martins
- Gustave Roussy Cancer Campus; Villejuif, France
- INSERM, U1030; Villejuif, France
- Faculté de Médecine; Université Paris-Sud/Paris XI; Kremlin-Bicêtre, France
| | - Domenico Mavilio
- Unit of Clinical and Experimental Immunology; Humanitas Clinical and Research Center; Milan, Italy
- Department of Medical Biotechnologies and Translational Medicine, University of Milan; Rozzano, Italy
| | - Laurie Menger
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Cancer Immunology Unit, Research Department of Haematology; University College London (UCL) Cancer Institute; London, UK
| | - Nicolò Merendino
- Department of Ecological and Biological Sciences (DEB), Tuscia University; Viterbo, Italy
| | - Michael Michaud
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Gregoire Mignot
- Cellular and Molecular Immunology and Endocrinology, Oniris; Nantes, France
| | - Karen L. Mossman
- Department of Pathology and Molecular Medicine; McMaster Immunology Research Center; Hamilton, Canada
- Institute for Infectious Disease Research; McMaster University; Hamilton, Canada
| | - Gabriele Multhoff
- Department of Radiation Oncology; Klinikum rechts der Isar; Technical University of Munich; Munich, Germany
| | - Rudolf Oehler
- Comprehensive Cancer Center; Medical University of Vienna; Vienna, Austria
| | - Fabio Palombo
- Departement of Internal Medicine and Medical Sciences; University of Rome La Sapienza; Rome, Italy
- Istituto Pasteur; Fondazione Cenci Bolognetti; Rome, Italy
| | | | - Jonathan Pol
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
| | - Enrico Proietti
- Department of Hematology; Oncology and Molecular Medicine; Istituto Superiore di Sanità (ISS); Rome, Italy
| | - Jean-Ehrland Ricci
- INSERM; U1065; Nice, France
- Equipe “Contrôle Métabolique des Morts Cellulaires,” Center Méditerranéen de Médecine Moléculaire (C3M); Nice, France
- Faculté de Médecine; Université de Nice Sophia Antipolis; Nice, France
- Centre Hospitalier Universitaire de Nice; Nice, France
| | - Chiara Riganti
- Department of Oncology and Subalpine Center for Research and Experimental Medicine (CeRMS); University of Turin; Turin, Italy
| | - Patrizia Rovere-Querini
- University Vita-Salute San Raffaele; Milano, Italy
- San Raffaele Scientific Institute; Milano, Italy
| | - Anna Rubartelli
- Cell Biology Unit; Azienda Ospedaliera Universitaria San Martino; Istituto Nazionale per la Ricerca sul Cancro; Genova, Italy
| | | | - Mark J. Smyth
- Immunology in Cancer and Infection Laboratory; QIMR Berghofer Medical Research Institute; Herston, Australia
- School of Medicine, University of Queensland; Herston, Australia
| | - Juergen Sonnemann
- Department of Pediatric Haematology and Oncology; Jena University Hospital, Children's Clinic; Jena, Germany
| | - Radek Spisek
- Department of Immunology; 2 Faculty of Medicine and University Hospital Motol, Charles University; Prague, Czech Republic
- Sotio; Prague, Czech Republic
| | - John Stagg
- Centre de Recherche du Center Hospitalier de l’Université de Montréal; Faculté de Pharmacie, Université de Montréal; Montréal, Canada
| | - Abdul Qader Sukkurwala
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Department of Pathology, Dow International Medical College; Dow University of Health Sciences; Karachi, Pakistan
| | - Eric Tartour
- INSERM; U970; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
| | - Andrew Thorburn
- Department of Pharmacology; University of Colorado School of Medicine; Aurora, CO USA
| | | | - Peter Vandenabeele
- VIB Inflammation Research Center; Ghent, Belgium
- Department of Biomedical Molecular Biology; Ghent University; Ghent, Belgium
- Methusalem Program; Ghent University; Ghent, Belgium
| | - Francesca Velotti
- Department of Ecological and Biological Sciences (DEB), Tuscia University; Viterbo, Italy
| | - Samuel T. Workenhe
- Department of Pathology and Molecular Medicine; McMaster Immunology Research Center; Hamilton, Canada
- Institute for Infectious Disease Research; McMaster University; Hamilton, Canada
| | - Haining Yang
- University of Hawaii Cancer Center; Honolulu, HI USA
| | - Wei-Xing Zong
- Department of Molecular Genetics and Microbiology; Stony Brook University; Stony Brook, NY USA
| | - Laurence Zitvogel
- INSERM; U1015; Villejuif, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Centre d’Investigation Clinique Biothérapie 507 (CICBT507); Gustave Roussy Cancer Campus; Villejuif, France
| | - Guido Kroemer
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Metabolomics and Cell Biology Platforms; Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
- Pôle de Biologie; Hôpital Européen Georges Pompidou; AP-HP; Paris, France
| | - Lorenzo Galluzzi
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer; Center de Recherche des Cordeliers; Paris, France
- INSERM; U1138; Paris, France
- Gustave Roussy Cancer Campus; Villejuif, France
- Université Paris Descartes/Paris V; Sorbonne Paris Cité; Paris, France
| |
Collapse
|