51
|
Kasakovski D, Xu L, Li Y. T cell senescence and CAR-T cell exhaustion in hematological malignancies. J Hematol Oncol 2018; 11:91. [PMID: 29973238 PMCID: PMC6032767 DOI: 10.1186/s13045-018-0629-x] [Citation(s) in RCA: 193] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/06/2018] [Indexed: 12/16/2022] Open
Abstract
T cell senescence has been recognized to play an immunosuppressive role in the aging population and cancer patients. Strategies dedicated to preventing or reversing replicative and premature T cell senescence are required to increase the lifespan of human beings and to reduce the morbidity from cancer. In addition, overcoming the T cell terminal differentiation or senescence from lymphoma and leukemia patients is a promising approach to enhance the effectiveness of adoptive cellular immunotherapy (ACT). Chimeric antigen receptor T (CAR-T) cell and T cell receptor-engineered T (TCR-T) cell therapy highly rely on functionally active T cells. However, the mechanisms which drive T cell senescence remain unclear and controversial. In this review, we describe recent progress for restoration of T cell homeostasis from age-related senescence as well as recovery of T cell activation in hematological malignancies.
Collapse
Affiliation(s)
- Dimitri Kasakovski
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632 China
- Department of Anatomy and Molecular Embryology, Institute of Anatomy, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Ling Xu
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632 China
- Department of Hematology, First Affiliated Hospital, School of Medicine, Jinan University, No. 601 West of Huangpu Avenue, Guangzhou, 510632 China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, 510632 China
- Department of Hematology, First Affiliated Hospital, School of Medicine, Jinan University, No. 601 West of Huangpu Avenue, Guangzhou, 510632 China
| |
Collapse
|
52
|
Ruan QZ, Fu JQ, Wu XX, Huang LP, Ruan RS. Rational combinations of in vivo cancer antigen priming and adoptive T-cell therapy mobilize immune and clinical responses in terminal cancers. Cancer Immunol Immunother 2018; 67:907-915. [PMID: 29511794 PMCID: PMC11028218 DOI: 10.1007/s00262-018-2142-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 02/24/2018] [Indexed: 10/17/2022]
Abstract
PURPOSE It is now recognized that solid tumors encroach on the host's immune microenvironment to favor its own proliferation. Strategies to enhance the specificity of the endogenous T-cell population against tumors have been met with limited clinical success. We aimed to devise a two-tier protocol coupling in vivo whole antigen priming with ex vivo cellular expansion to clinically evaluate survival in patients following re-infusion of primed, autologous T cells, thereby determining treatment efficacy. EXPERIMENTAL DESIGN Treatment commenced with the acquisition of whole tumor antigens from tumor cell lines corresponding with patients' primary malignancy. Lysate mixture was inoculated intradermally, while peripheral blood mononuclear cells (PBMCs) were periodically extracted via phlebotomy and expanded in culture ex vivo for re-infusion. Post-treatment tumor-specific T-cell response and cytotoxicity was confirmed via Elispot and real-time cell analyzing (RTCA) assay. Serum cytokine levels and cytotoxicity scores were evaluated for associations with survival status and duration. RESULTS There was a significant increase in cytotoxicity exhibited by T cells measured using both Elispot and RTCA following treatment. Correlation analysis determined significant association between higher post-treatment cytotoxicity scores and survival status (R = 0.52, p = 0.0028) as well as longer survival duration in months (R = 0.59, p = 0.005). CONCLUSIONS Our treatment protocol successfully demonstrated significant correlation between tumor-associated antigen-specific immune response and objective prolongation of survival. Whole-cell cancer antigen priming and adoptive T-cell therapy is, therefore, a highly feasible clinical model which can be easily replicated to positively influence outcome in end-stage malignancy.
Collapse
Affiliation(s)
- Qing Zhao Ruan
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jian Qian Fu
- Department of Oncology, Xiamen 5th Hospital, Xiamen, China
| | - Xiao Xuan Wu
- Xiamen Key Laboratory for Translational Medicine of Cancer Theranostics, School of Pharmaceutical Sciences, Xiamen University, #246-248, Xiangan Nanlu, Xiangan District, Xiamen, China
| | - Li Ping Huang
- Xiamen Key Laboratory for Translational Medicine of Cancer Theranostics, School of Pharmaceutical Sciences, Xiamen University, #246-248, Xiangan Nanlu, Xiangan District, Xiamen, China
| | - Run Sheng Ruan
- Xiamen Key Laboratory for Translational Medicine of Cancer Theranostics, School of Pharmaceutical Sciences, Xiamen University, #246-248, Xiangan Nanlu, Xiangan District, Xiamen, China.
- Zhang Zhou Xing Pu Hospital, Zhang Zhou, China.
| |
Collapse
|
53
|
Garg AD, Agostinis P. Cell death and immunity in cancer: From danger signals to mimicry of pathogen defense responses. Immunol Rev 2018; 280:126-148. [PMID: 29027218 DOI: 10.1111/imr.12574] [Citation(s) in RCA: 308] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immunogenicity of cancer cells is an emerging determinant of anti-cancer immunotherapy. Beyond developing immunostimulatory regimens like dendritic cell-based vaccines, immune-checkpoint blockers, and adoptive T-cell transfer, investigators are beginning to focus on the immunobiology of dying cancer cells and its relevance for the success of anticancer immunotherapies. It is currently accepted that cancer cells may die in response to anti-cancer therapies through regulated cell death programs, which may either repress or increase their immunogenic potential. In particular, the induction of immunogenic cancer cell death (ICD), which is hallmarked by the emission of damage-associated molecular patterns (DAMPs); molecules analogous to pathogen-associated molecular patterns (PAMPs) acting as danger signals/alarmins, is of great relevance in cancer therapy. These ICD-associated danger signals favor immunomodulatory responses that lead to tumor-associated antigens (TAAs)-directed T-cell immunity, which paves way for the removal of residual, treatment-resistant cancer cells. It is also emerging that cancer cells succumbing to ICD can orchestrate "altered-self mimicry" i.e. mimicry of pathogen defense responses, on the levels of nucleic acids and/or chemokines (resulting in type I interferon/IFN responses or pathogen response-like neutrophil activity). In this review, we exhaustively describe the main molecular, immunological, preclinical, and clinical aspects of immunosuppressive cell death or ICD (with respect to apoptosis, necrosis and necroptosis). We also provide an extensive historical background of these fields, with special attention to the self/non-self and danger models, which have shaped the field of cell death immunology.
Collapse
Affiliation(s)
- Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Laboratory, Department for Cellular and Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Laboratory, Department for Cellular and Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| |
Collapse
|
54
|
Heim L, Friedrich J, Engelhardt M, Trufa DI, Geppert CI, Rieker RJ, Sirbu H, Finotto S. NFATc1 Promotes Antitumoral Effector Functions and Memory CD8 + T-cell Differentiation during Non-Small Cell Lung Cancer Development. Cancer Res 2018; 78:3619-3633. [PMID: 29691251 DOI: 10.1158/0008-5472.can-17-3297] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 03/12/2018] [Accepted: 04/20/2018] [Indexed: 11/16/2022]
Abstract
Nuclear factor of activated T cells 1 (NFATc1) is a transcription factor activated by T-cell receptor (TCR) and Ca2+ signaling that affects T-cell activation and effector function. Upon tumor antigen challenge, TCR and calcium-release-activated channels are induced, promoting NFAT dephosphorylation and translocation into the nucleus. In this study, we report a progressive decrease of NFATc1 in lung tumor tissue and in tumor-infiltrating lymphocytes (TIL) of patients suffering from advanced-stage non-small cell lung cancer (NSCLC). Mice harboring conditionally inactivated NFATc1 in T cells (NFATc1ΔCD4) showed increased lung tumor growth associated with impaired T-cell activation and function. Furthermore, in the absence of NFATc1, reduced IL2 influenced the development of memory CD8+ T cells. We found a reduction of effector memory and CD103+ tissue-resident memory (TRM) T cells in the lung of tumor-bearing NFATc1ΔCD4 mice, underlining an impaired cytotoxic T-cell response and a reduced TRM tissue-homing capacity. In CD4+ICOS+ T cells, programmed cell death 1 (PD-1) was induced in the draining lymph nodes of these mice and associated with lung tumor cell growth. Targeting PD-1 resulted in NFATc1 induction in CD4+ and CD8+ T cells in tumor-bearing mice and was associated with increased antitumor cytotoxic functions. This study reveals a role of NFATc1 in the activation and cytotoxic functions of T cells, in the development of memory CD8+ T-cell subsets, and in the regulation of T-cell exhaustion. These data underline the indispensability of NFATc1 for successful antitumor immune responses in patients with NSCLC.Significance: The multifaceted role of NFATc1 in the activation and function of T cells during lung cancer development makes it a critical participant in antitumor immune responses in patients with NSCLC. Cancer Res; 78(13); 3619-33. ©2018 AACR.
Collapse
Affiliation(s)
- Lisanne Heim
- Department of Molecular Pneumology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Juliane Friedrich
- Department of Molecular Pneumology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marina Engelhardt
- Department of Molecular Pneumology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Denis I Trufa
- Department of Thoracic Surgery, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Carol I Geppert
- Institute of Pathology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Ralf J Rieker
- Institute of Pathology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Horia Sirbu
- Department of Thoracic Surgery, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Susetta Finotto
- Department of Molecular Pneumology, University Hospital, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
55
|
Romano E, Rufo N, Korf H, Mathieu C, Garg AD, Agostinis P. BNIP3 modulates the interface between B16-F10 melanoma cells and immune cells. Oncotarget 2018; 9:17631-17644. [PMID: 29707136 PMCID: PMC5915144 DOI: 10.18632/oncotarget.24815] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 02/27/2018] [Indexed: 01/10/2023] Open
Abstract
The hypoxia responsive protein BNIP3, plays an important role in promoting cell death and/or autophagy, ultimately resulting in a cancer type-dependent, tumour-enhancer or tumour-suppressor activity. We previously reported that in melanoma cells, BNIP3 regulates cellular morphology, mitochondrial clearance, cellular viability and maintains protein expression of CD47, a pro-cancerous, immunosuppressive 'don't eat me' signal. Surface exposed CD47 is often up-regulated by cancer cells to avoid clearance by phagocytes and to suppress immunogenic cell death (ICD) elicited by anticancer therapies. However, whether melanoma-associated BNIP3 modulates CD47-associated immunological effects or ICD has not been explored properly. To this end, we evaluated the impact of the genetic ablation of BNIP3 (i.e. BNIP3KD) in melanoma cells, on macrophage-based phagocytosis, polarization and chemotaxis. Additionally, we tested its effects on crucial determinants of chemotherapy-induced ICD (i.e. danger signals), as well as in vivo anticancer vaccination effect. Interestingly, loss of BNIP3 reduced the expression of CD47 both in normoxic and hypoxic conditions while macrophage phagocytosis and chemotaxis were accentuated only when BNIP3KD melanoma cells were exposed to hypoxia. Moreover, when exposed to the ICD inducer mitoxantrone, the loss of melanoma cell-associated BNIP3 did not alter apoptosis induction, but significantly prevented ATP secretion and reduced phagocytic clearance of dying cells. In line with this, prophylactic vaccination experiments showed that the loss of BNIP3 tends to increase the intrinsic resistance of B16-F10 melanoma cells to ICD-associated anticancer vaccination effect in vivo. Thus, normoxic vs. hypoxic and live vs. dying cell contexts influence the ultimate immunomodulatory roles of melanoma cell-associated BNIP3.
Collapse
Affiliation(s)
- Erminia Romano
- Laboratory for Cell Death Research and Therapy (CDRT), Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Nicole Rufo
- Laboratory for Cell Death Research and Therapy (CDRT), Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Hannelie Korf
- Laboratory of Hepatology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
- Laboratory of Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Chantal Mathieu
- Laboratory of Clinical and Experimental Endocrinology (CEE), Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven, Leuven, Belgium
| | - Abhishek D Garg
- Laboratory for Cell Death Research and Therapy (CDRT), Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Laboratory for Cell Death Research and Therapy (CDRT), Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
56
|
Ness N, Andersen S, Khanehkenari MR, Nordbakken CV, Valkov A, Paulsen EE, Nordby Y, Bremnes RM, Donnem T, Busund LT, Richardsen E. The prognostic role of immune checkpoint markers programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1) in a large, multicenter prostate cancer cohort. Oncotarget 2018; 8:26789-26801. [PMID: 28460462 PMCID: PMC5432297 DOI: 10.18632/oncotarget.15817] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 02/20/2017] [Indexed: 12/20/2022] Open
Abstract
Programmed cell death protein 1 (PD-1) and its ligand Programmed death ligand 1 (PD-L1) have gained massive attention in cancer research due to recent availability and their targeted antitumor effects. Their role in prostate cancer is still undetermined. We constructed tissue microarrays from prostatectomy specimens from 535 prostate cancer patients. Following validation of antibodies, immunohistochemistry was used to evaluate the expression of PD-1 in lymphocytes and PD-L1 in epithelial and stromal cells of primary tumors. PD-L1 expression was commonly seen in tumor epithelial cells (92% of cases). Univariate survival analysis revealed a positive association between a high density of PD-1+ lymphocytes and worse clinical failure-free survival, limited to a trend (p = 0.084). In subgroups known to indicate unfavorable prostate cancer prognosis (Gleason grade 9, age < 65, preoperative PSA > 10, pT3) patients with high density of PD-1+ lymphocytes had a significantly higher risk of clinical failure (p = < 0.001, p = 0.025, p = 0.039 and p = 0.011, respectively). In the multivariate analysis, high density of PD-1+ lymphocytes was a significant negative independent prognostic factor for clinical failure-free survival (HR = 2.48, CI 95% 1.12-5.48, p = 0.025).
Collapse
Affiliation(s)
- Nora Ness
- Department of Medical Biology, UiT The Arctic University of Norway, N-9037 Tromso, Norway
| | - Sigve Andersen
- Department of Clinical Medicine, UiT The Arctic University of Norway, N-9037 Tromso, Norway.,Department of Oncology, University Hospital of North Norway, N-9038 Tromso, Norway
| | | | - Cecilie V Nordbakken
- Department of Clinical Pathology, University Hospital of North Norway, N-9038 Tromso, Norway
| | - Andrej Valkov
- Department of Clinical Pathology, University Hospital of North Norway, N-9038 Tromso, Norway
| | - Erna-Elise Paulsen
- Department of Clinical Medicine, UiT The Arctic University of Norway, N-9037 Tromso, Norway.,Department of Oncology, University Hospital of North Norway, N-9038 Tromso, Norway
| | - Yngve Nordby
- Department of Clinical Medicine, UiT The Arctic University of Norway, N-9037 Tromso, Norway.,Department of Urology, University Hospital of North Norway, N-9038 Tromso, Norway
| | - Roy M Bremnes
- Department of Clinical Medicine, UiT The Arctic University of Norway, N-9037 Tromso, Norway.,Department of Oncology, University Hospital of North Norway, N-9038 Tromso, Norway
| | - Tom Donnem
- Department of Clinical Medicine, UiT The Arctic University of Norway, N-9037 Tromso, Norway.,Department of Oncology, University Hospital of North Norway, N-9038 Tromso, Norway
| | - Lill-Tove Busund
- Department of Medical Biology, UiT The Arctic University of Norway, N-9037 Tromso, Norway.,Department of Clinical Pathology, University Hospital of North Norway, N-9038 Tromso, Norway
| | - Elin Richardsen
- Department of Medical Biology, UiT The Arctic University of Norway, N-9037 Tromso, Norway.,Department of Clinical Pathology, University Hospital of North Norway, N-9038 Tromso, Norway
| |
Collapse
|
57
|
Zhao K, Yu L, Wang X, He Y, Lu B. Clostridium butyricum regulates visceral hypersensitivity of irritable bowel syndrome by inhibiting colonic mucous low grade inflammation through its action on NLRP6. Acta Biochim Biophys Sin (Shanghai) 2018; 50:216-223. [PMID: 29329362 DOI: 10.1093/abbs/gmx138] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 11/30/2017] [Indexed: 01/30/2023] Open
Abstract
Visceral hypersensitivity induced by stress is quite common in irritable bowel syndrome (IBS) patients. Probiotics play an important role in reducing visceral hypersensitivity in IBS patients. However, the mechanism has not been clearly elucidated. In this study, we investigated the role of nod-like receptor pyrin domain-containing protein 6 (NLRP6) in Clostridium butyricum-regulated IBS induced by stress. Our results showed that NLRP6 was down-regulated in IBS group colon tissues. In addition, IL-18, IL-1β, myeloperoxidase (MPO), d-lactic acid (D-LA), and CD172a were up-regulated in the IBS group of colonic mucous. IL-18 and IL-1β were also increased after the NLRP6 gene was silenced. Pathological score suggested low inflammation of colonic mucous rather than terminal ileum. Water-avoidance stress (WAS) showed visceral hypersensitivity to colonic distension. However, treatment with Clostridium butyricum reversed these results, exerting a beneficial effect. In conclusion, Clostridium butyricum may exert a beneficial action on visceral hypersensitivity of IBS by inhibiting low grade inflammation of colonic mucous through its action on NLRP6.
Collapse
Affiliation(s)
- Kejia Zhao
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Leimin Yu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Xi Wang
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Yibo He
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Bin Lu
- Department of Gastroenterology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| |
Collapse
|
58
|
Karpanen T, Olweus J. The Potential of Donor T-Cell Repertoires in Neoantigen-Targeted Cancer Immunotherapy. Front Immunol 2017; 8:1718. [PMID: 29321773 PMCID: PMC5732232 DOI: 10.3389/fimmu.2017.01718] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/21/2017] [Indexed: 12/30/2022] Open
Abstract
T cells can recognize peptides encoded by mutated genes, but analysis of tumor-infiltrating lymphocytes suggests that very few neoantigens spontaneously elicit T-cell responses. This may be an important reason why immune checkpoint inhibitors are mainly effective in tumors with a high mutational burden. Reasons for clinically insufficient responses to neoantigens might be inefficient priming, inhibition, or deletion of the cognate T cells. Responses can be dramatically improved by cancer immunotherapy such as checkpoint inhibition, but often with temporary effects. By contrast, T cells from human leukocyte antigen (HLA)-matched donors can cure diseases such as chronic myeloid leukemia. The therapeutic effect is mediated by donor T cells recognizing polymorphic peptides for which the donor and patient are disparate, presented on self-HLA. Donor T-cell repertoires are unbiased by the immunosuppressive environment of the tumor. A recent study demonstrated that T cells from healthy individuals are able to respond to neoantigens that are ignored by tumor-infiltrating T cells of melanoma patients. In this review, we discuss possible reasons why neoantigens escape host T cells and how these limitations may be overcome by utilization of donor-derived T-cell repertoires to facilitate rational design of neoantigen-targeted immunotherapy.
Collapse
Affiliation(s)
- Terhi Karpanen
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, and K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| | - Johanna Olweus
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital Radiumhospitalet, and K.G. Jebsen Center for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| |
Collapse
|
59
|
Kumar S, Saini RV, Mahindroo N. Recent advances in cancer immunology and immunology-based anticancer therapies. Biomed Pharmacother 2017; 96:1491-1500. [PMID: 29198747 DOI: 10.1016/j.biopha.2017.11.126] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 11/12/2017] [Accepted: 11/27/2017] [Indexed: 12/14/2022] Open
Abstract
Cancer immunotherapies offer promise for cure of cancer with specificity and minimal toxicity. Recent developments in cancer immunology have led to the better understanding of role of immune regulatory mechanisms in cancer. There is rapid progress in this field in the last few years. Several clinical studies report the efficacy of immunotherapies for treating cancer. The immunology-based anticancer therapies have shown better safety profiles in clinic as compared to other chemotherapeutic agents, thus increasing interest in this area. This review summarizes recent advances in cancer immunology and discusses tumor microenvironment and immunology-based anticancer therapies, including vaccines and therapies targeting immune checkpoints.
Collapse
Affiliation(s)
- Sunil Kumar
- School of Pharmaceutical Sciences, Shoolini University, Post Box 9, Solan, 173212, Himachal Pradesh, India
| | - Reena Vohra Saini
- School of Biotechnology, Shoolini University, Post Box 9, Solan, 173212, Himachal Pradesh, India; Centre of Research on Himalayan Sustainability and Development, Shoolini University, Post Box 9, Solan, 173212, Himachal Pradesh, India
| | - Neeraj Mahindroo
- School of Pharmaceutical Sciences, Shoolini University, Post Box 9, Solan, 173212, Himachal Pradesh, India; Centre of Research on Himalayan Sustainability and Development, Shoolini University, Post Box 9, Solan, 173212, Himachal Pradesh, India.
| |
Collapse
|
60
|
Analysis of non-small cell lung cancer microenvironment indicates preponderance of T cell exhaustion marker expression. Exp Cell Res 2017; 360:205-209. [DOI: 10.1016/j.yexcr.2017.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 09/04/2017] [Accepted: 09/06/2017] [Indexed: 12/22/2022]
|
61
|
Davoodzadeh Gholami M, Kardar GA, Saeedi Y, Heydari S, Garssen J, Falak R. Exhaustion of T lymphocytes in the tumor microenvironment: Significance and effective mechanisms. Cell Immunol 2017; 322:1-14. [PMID: 29079339 DOI: 10.1016/j.cellimm.2017.10.002] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Revised: 10/08/2017] [Accepted: 10/09/2017] [Indexed: 12/23/2022]
Abstract
T lymphocytes play crucial roles in adaptive immune responses to tumors. However, due to different tolerance mechanisms and inhibitory effects of the tumor microenvironment (TME) on T cells, responses to tumors are insufficient. In fact, cellular and molecular suppressive mechanisms repress T cell responses in the TME, resulting in senescent, anergic and exhausted lymphocytes. Exhaustion is a poor responsive status of T cells, with up-regulated expression of inhibitory receptors, decreased production of effective cytokines, and reduced cytotoxic activity. Low immunogenicity of tumor antigens and inadequate presentation of tumor-specific antigens results in inappropriate activation of naive T lymphocytes against tumor antigens. Moreover, when effector cytotoxic T cells enter TME, they encounter a complicated network of cells and cytokines that suppress their effectiveness and turn them into exhausted T cells. Thus, the mechanism of T cell exhaustion in cancer is different from that in chronic infections. In this review we will discuss the main components such as inhibitory receptors, inflammatory cells, stromal cells, cytokine milieu as well as environmental and metabolic conditions in TME which play role in development of exhaustion. Furthermore, recent therapeutic methods available to overcome exhaustion will be discussed.
Collapse
Affiliation(s)
- Mohammad Davoodzadeh Gholami
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Gholam Ali Kardar
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Yousef Saeedi
- Department of Pharmaceutical Sciences, Utrecht University, Netherlands.
| | - Sahel Heydari
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Johan Garssen
- Department of Pharmaceutical Sciences, Utrecht University, Netherlands.
| | - Reza Falak
- Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
62
|
Haymaker C, Yang Y, Wang J, Zou Q, Sahoo A, Alekseev A, Singh D, Ritthipichai K, Hailemichael Y, Hoang ON, Qin H, Schluns KS, Wang T, Overwijk WW, Sun SC, Bernatchez C, Kwak LW, Neelapu SS, Nurieva R. Absence of Grail promotes CD8 + T cell anti-tumour activity. Nat Commun 2017; 8:239. [PMID: 28798332 PMCID: PMC5552797 DOI: 10.1038/s41467-017-00252-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/14/2017] [Indexed: 12/15/2022] Open
Abstract
T-cell tolerance is a major obstacle to successful cancer immunotherapy; thus, developing strategies to break immune tolerance is a high priority. Here we show that expression of the E3 ubiquitin ligase Grail is upregulated in CD8+ T cells that have infiltrated into transplanted lymphoma tumours, and Grail deficiency confers long-term tumour control. Importantly, therapeutic transfer of Grail-deficient CD8+ T cells is sufficient to repress established tumours. Mechanistically, loss of Grail enhances anti-tumour reactivity and functionality of CD8+ T cells. In addition, Grail-deficient CD8+ T cells have increased IL-21 receptor (IL-21R) expression and hyperresponsiveness to IL-21 signalling as Grail promotes IL-21R ubiquitination and degradation. Moreover, CD8+ T cells isolated from lymphoma patients express higher levels of Grail and lower levels of IL-21R, compared with CD8+ T cells from normal donors. Our data demonstrate that Grail is a crucial factor controlling CD8+ T-cell function and is a potential target to improve cytotoxic T-cell activity.Grail is an E3 ubiquitin ligase that inhibits T-cell receptor signalling in CD4+ T cells. Here the authors show Grail also limits IL-21 receptor expression and function in CD8+ T cells, is overactive in these cells in patients with lymphoma, and promotes tumour development in a lymphoma transplant mouse model.
Collapse
Affiliation(s)
- Cara Haymaker
- Department of Melanoma Medical Oncology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Yi Yang
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
- Department of Radiation Oncology, The Second Hospital of Jilin University, No. 218 Ziqiang St., Changchun City, Jilin Province, 130041, China
| | - Junmei Wang
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Qiang Zou
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Anupama Sahoo
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Andrei Alekseev
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Divyendu Singh
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Krit Ritthipichai
- Department of Melanoma Medical Oncology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Yared Hailemichael
- Department of Melanoma Medical Oncology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Oanh N Hoang
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Hong Qin
- Department of Lymphoma/Myeloma, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
- Toni Stephenson Lymphoma Center, City of Hope, 1500 East Duarte Rd., Duarte, CA, 91010, USA
| | - Kimberly S Schluns
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Tiejun Wang
- Department of Radiation Oncology, The Second Hospital of Jilin University, No. 218 Ziqiang St., Changchun City, Jilin Province, 130041, China
| | - Willem W Overwijk
- Department of Melanoma Medical Oncology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Shao-Cong Sun
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Chantale Bernatchez
- Department of Melanoma Medical Oncology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Larry W Kwak
- Department of Lymphoma/Myeloma, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
- Toni Stephenson Lymphoma Center, City of Hope, 1500 East Duarte Rd., Duarte, CA, 91010, USA
| | - Sattva S Neelapu
- Department of Lymphoma/Myeloma, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Roza Nurieva
- Department of Immunology, M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA.
| |
Collapse
|
63
|
Ho PC, Kaech SM. Reenergizing T cell anti-tumor immunity by harnessing immunometabolic checkpoints and machineries. Curr Opin Immunol 2017; 46:38-44. [PMID: 28458087 DOI: 10.1016/j.coi.2017.04.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/03/2017] [Accepted: 04/07/2017] [Indexed: 12/21/2022]
Abstract
T cells patrol our bodies preventing pathogenic infections and malignant cell outgrowth. However, T cells must be properly controlled because aberrant or persistent T cell responses can damage tissues and contribute to autoimmune diseases and other chronic inflammatory diseases including metabolic syndrome. One regulatory mechanism utilized in immune cells is immunometabolic regulation, which ensures immune cells properly respond to systemic and peripheral metabolic cues. Recent work has suggested that deregulated metabolism in tumor cells creates a microenvironmental barrier for mounting effective anti-tumor immune responses. Here, we discuss how tumor cells evade immunosurveillance by modulating metabolic checkpoints in immune cells and discuss how memory T cells could provide effective anti-tumor responses by sustaining metabolic fitness and longevity.
Collapse
Affiliation(s)
- Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Vaud 1066, Switzerland; Ludwig Lausanne Branch, University of Lausanne, Lausanne, Vaud 1066, Switzerland.
| | - Susan M Kaech
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06519, USA.
| |
Collapse
|
64
|
Granier C, Dariane C, Combe P, Verkarre V, Urien S, Badoual C, Roussel H, Mandavit M, Ravel P, Sibony M, Biard L, Radulescu C, Vinatier E, Benhamouda N, Peyromaure M, Oudard S, Méjean A, Timsit MO, Gey A, Tartour E. Tim-3 Expression on Tumor-Infiltrating PD-1 +CD8 + T Cells Correlates with Poor Clinical Outcome in Renal Cell Carcinoma. Cancer Res 2017; 77:1075-1082. [PMID: 27872087 DOI: 10.1158/0008-5472.can-16-0274] [Citation(s) in RCA: 168] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 10/08/2016] [Accepted: 10/29/2016] [Indexed: 11/16/2022]
Abstract
Inhibitory receptors expressed by T cells mediate tolerance to tumor antigens, with coexpression of these receptors exacerbating this dysfunctional state. Using the VectraR automated multiparametric immunofluorescence technique, we quantified intratumoral CD8+ T cells coexpressing the inhibitory receptors PD-1 and Tim-3 from patients with renal cell carcinoma (RCC). A second validation cohort measured the same parameters by cytometry. The percentage of tumor-infiltrating CD8+ T cells coexpressing PD-1 and Tim-3 correlated with an aggressive phenotype and a larger tumor size at diagnosis. Coexpression of PD-1 and Tim-3 above the median conferred a higher risk of relapse and a poorer 36-month overall survival. Notably, other CD8+T-cell subsets did not exert a similar effect on overall survival. Moreover, only the PD-1+Tim-3+ subset of CD8+ T cells exhibited impaired function after stimulation. Our findings establish intratumoral Tim-3+PD1+CD8+ T cells as critical mediators of an aggressive phenotype in RCC. Use of the Vectra tool may be useful to identify similarly critical prognostic and predictive biomarkers in other tumor types and their response to immunotherapy. Cancer Res; 77(5); 1075-82. ©2016 AACR.
Collapse
Affiliation(s)
- Clémence Granier
- INSERM U970, Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
| | - Charles Dariane
- INSERM U970, Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
- Department of Urology, Hôpital Européen Georges Pompidou, Paris, France
| | - Pierre Combe
- INSERM U970, Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
- Service d'Oncologie Médicale, Hôpital Européen Georges Pompidou, Paris, France
| | - Virginie Verkarre
- Department of Pathology, Hôpital Européen Georges Pompidou, Paris, France
| | - Saïk Urien
- EA08: Evaluation des Thérapeutiques et Pharmacologie Périnatale et Pédiatrique, Unité de Recherche Clinique Paris Centre, Paris, France
- CIC 1419 INSERM Cochin-Necker, Paris, France
| | - Cécile Badoual
- INSERM U970, Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
- Department of Pathology, Hôpital Européen Georges Pompidou, Paris, France
| | - Hélène Roussel
- INSERM U970, Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
- Department of Pathology, Hôpital Européen Georges Pompidou, Paris, France
| | - Marion Mandavit
- INSERM U970, Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
| | - Patrice Ravel
- CNRS UMR5048 Centre de Biochimie Structurale, INSERM U554, Université de Montpellier 1 et 2, Montpellier, France
| | | | - Lucie Biard
- Université Paris Diderot Paris 7, Paris, France
| | | | - Emeline Vinatier
- INSERM U970, Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
- Service d'Immunologie Biologique, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Nadine Benhamouda
- INSERM U970, Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
- Service d'Immunologie Biologique, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | | | - Stéphane Oudard
- INSERM U970, Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
- Service d'Oncologie Médicale, Hôpital Européen Georges Pompidou, Paris, France
| | - Arnaud Méjean
- Department of Urology, Hôpital Européen Georges Pompidou, Paris, France
| | | | - Alain Gey
- INSERM U970, Université Paris Descartes, Sorbonne Paris-Cité, Paris, France
- Service d'Immunologie Biologique, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| | - Eric Tartour
- INSERM U970, Université Paris Descartes, Sorbonne Paris-Cité, Paris, France.
- Service d'Immunologie Biologique, AP-HP, Hôpital Européen Georges Pompidou, Paris, France
| |
Collapse
|
65
|
Antonia SJ, Vansteenkiste JF, Moon E. Immunotherapy: Beyond Anti-PD-1 and Anti-PD-L1 Therapies. Am Soc Clin Oncol Educ Book 2017; 35:e450-8. [PMID: 27249753 DOI: 10.1200/edbk_158712] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Advanced-stage non-small cell lung cancer (NSCLC) and small cell lung cancer are cancers in which chemotherapy produces a survival benefit, although it is small. We now know that anti-PD-1/PD-L1 has substantial clinical activity in both of these diseases, with an overall response rate (ORR) of 15%-20%. These responses are frequently rapid and durable, increase median overall survival (OS) compared with chemotherapy, and produce long-term survivors. Despite these very significant results, many patients do not benefit from anti-PD-1/PD-L1. This is because of the potential for malignancies to co-opt myriad immunosuppressive mechanisms other than aberrant expression of PD-L1. Conceptually, these can be divided into three categories. First, for some patients there is likely a failure to generate sufficient functional tumor antigen-specific T cells. Second, for others, tumor antigen-specific T cells may be generated but fail to enter into the tumor parenchyma. Finally, there are a large number of immunosuppressive mechanisms that have the potential to be operational within the tumor microenvironment: surface membrane immune checkpoint proteins PD-1, CTLA-4, LAG3, TIM3, BTLA, and adenosine A2AR; soluble factors and metabolic alterations interleukin (IL)-10, transforming growth factor (TGF)-β, adenosine, IDO, and arginase; and inhibitory cells, cancer-associated fibroblasts (CAFs), regulatory T cells, myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages. In this article, we discuss three strategies to generate more tumor-reactive T cells for patients: anti-CTLA-4, therapeutic tumor vaccination, and adoptive cellular therapy, with T cells redirected to tumor antigens using T-cell receptor (TCR) or chimeric antigen receptor (CAR) gene modification. We also review some of the various strategies in development to thwart tumor microenvironment immunosuppressive mechanisms. Strategies to drive more T cells into tumors remain a significant challenge.
Collapse
Affiliation(s)
- Scott J Antonia
- From the Department of Thoracic Oncology Moffitt Cancer Center, Tampa, FL; Respiratory Oncology Unit, University Hospital KU Leuven, Leuven, Belgium; Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Johan F Vansteenkiste
- From the Department of Thoracic Oncology Moffitt Cancer Center, Tampa, FL; Respiratory Oncology Unit, University Hospital KU Leuven, Leuven, Belgium; Hospital of the University of Pennsylvania, Philadelphia, PA
| | - Edmund Moon
- From the Department of Thoracic Oncology Moffitt Cancer Center, Tampa, FL; Respiratory Oncology Unit, University Hospital KU Leuven, Leuven, Belgium; Hospital of the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
66
|
Yip J, Geng X, Shen J, Ding Y. Cerebral Gluconeogenesis and Diseases. Front Pharmacol 2017; 7:521. [PMID: 28101056 PMCID: PMC5209353 DOI: 10.3389/fphar.2016.00521] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 12/15/2016] [Indexed: 01/08/2023] Open
Abstract
The gluconeogenesis pathway, which has been known to normally present in the liver, kidney, intestine, or muscle, has four irreversible steps catalyzed by the enzymes: pyruvate carboxylase, phosphoenolpyruvate carboxykinase, fructose 1,6-bisphosphatase, and glucose 6-phosphatase. Studies have also demonstrated evidence that gluconeogenesis exists in brain astrocytes but no convincing data have yet been found in neurons. Astrocytes exhibit significant 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 activity, a key mechanism for regulating glycolysis and gluconeogenesis. Astrocytes are unique in that they use glycolysis to produce lactate, which is then shuttled into neurons and used as gluconeogenic precursors for reduction. This gluconeogenesis pathway found in astrocytes is becoming more recognized as an important alternative glucose source for neurons, specifically in ischemic stroke and brain tumor. Further studies are needed to discover how the gluconeogenesis pathway is controlled in the brain, which may lead to the development of therapeutic targets to control energy levels and cellular survival in ischemic stroke patients, or inhibit gluconeogenesis in brain tumors to promote malignant cell death and tumor regression. While there are extensive studies on the mechanisms of cerebral glycolysis in ischemic stroke and brain tumors, studies on cerebral gluconeogenesis are limited. Here, we review studies done to date regarding gluconeogenesis to evaluate whether this metabolic pathway is beneficial or detrimental to the brain under these pathological conditions.
Collapse
Affiliation(s)
- James Yip
- Department of Neurosurgery, Wayne State University School of Medicine Detroit, MI, USA
| | - Xiaokun Geng
- Department of Neurosurgery, Wayne State University School of MedicineDetroit, MI, USA; China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China; Department of Neurology, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China
| | - Jiamei Shen
- Department of Neurosurgery, Wayne State University School of MedicineDetroit, MI, USA; China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of MedicineDetroit, MI, USA; China-America Institute of Neuroscience, Beijing Luhe Hospital, Capital Medical UniversityBeijing, China
| |
Collapse
|
67
|
Bu X, Yao Y, Li X. Immune Checkpoint Blockade in Breast Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:383-402. [PMID: 29282694 DOI: 10.1007/978-981-10-6020-5_18] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer immunotherapy is emerging as the most promising novel strategy for cancer treatment. Cancer immunotherapy is broadly categorized into three forms: immune checkpoint modulation, adoptive cell transfer, and cancer vaccine. Immune checkpoint blockade is demonstrated as the most clinically effective treatment with low immune-related adverse events (irAE). Blockade of PD-1/PD-L1 and CTLA-4 has achieved remarkable success in treating various types of tumors, which sparks great interests in this therapeutic strategy and expands the role of immune checkpoint blockade in treating tumors including breast cancer. Based on the notable results obtained from clinical trials, the United States' Food and Drug Administration (FDA) has approved multiple CTLA-4 monoclonal antibodies as well as the PD-1/PD-L1 monoclonal antibodies for treatment of different types of tumors. The theories of immunoediting, T-cell exhaustions, and co-stimulatory/co-inhibitory pathways are immunological foundations for immune checkpoint blockade therapy. Breast cancers such as triple negative breast cancer and HER-2 negative breast cancer respond to immune checkpoint blockade therapy due to their high immunogenicity. PD-1/PD-L1 blockade has just received FDA approval as a standard cancer therapy for solid tumors such as breast cancer. Development of immune checkpoint blockade focuses on two directions: one is to identify proper biomarkers of immune checkpoint blockade in breast cancer, and the other is to combine therapies with PD-1/PD-L1 blockade antibodies to achieve optimal clinical outcomes.
Collapse
Affiliation(s)
- Xia Bu
- Department of Medical Oncology, The First Affiliated Hospital, Henan University Cancer Center, School of Medicine, Henan University, Kaifeng, People's Republic of China.
| | - Yihui Yao
- Department of Medical Oncology, The First Affiliated Hospital, Henan University Cancer Center, School of Medicine, Henan University, Kaifeng, People's Republic of China
| | - Xiaoyu Li
- Department of Hematology, The First Affiliated Hospital, Henan University Cancer Center, School of Medicine, Henan University, Kaifeng, People's Republic of China
| |
Collapse
|
68
|
Singer M, Wang C, Cong L, Marjanovic ND, Kowalczyk MS, Zhang H, Nyman J, Sakuishi K, Kurtulus S, Gennert D, Xia J, Kwon JYH, Nevin J, Herbst RH, Yanai I, Rozenblatt-Rosen O, Kuchroo VK, Regev A, Anderson AC. A Distinct Gene Module for Dysfunction Uncoupled from Activation in Tumor-Infiltrating T Cells. Cell 2016; 166:1500-1511.e9. [PMID: 27610572 DOI: 10.1016/j.cell.2016.08.052] [Citation(s) in RCA: 271] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/14/2016] [Accepted: 08/23/2016] [Indexed: 02/01/2023]
Abstract
Reversing the dysfunctional T cell state that arises in cancer and chronic viral infections is the focus of therapeutic interventions; however, current therapies are effective in only some patients and some tumor types. To gain a deeper molecular understanding of the dysfunctional T cell state, we analyzed population and single-cell RNA profiles of CD8(+) tumor-infiltrating lymphocytes (TILs) and used genetic perturbations to identify a distinct gene module for T cell dysfunction that can be uncoupled from T cell activation. This distinct dysfunction module is downstream of intracellular metallothioneins that regulate zinc metabolism and can be identified at single-cell resolution. We further identify Gata-3, a zinc-finger transcription factor in the dysfunctional module, as a regulator of dysfunction, and we use CRISPR-Cas9 genome editing to show that it drives a dysfunctional phenotype in CD8(+) TILs. Our results open novel avenues for targeting dysfunctional T cell states while leaving activation programs intact.
Collapse
Affiliation(s)
- Meromit Singer
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Chao Wang
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Le Cong
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nemanja D Marjanovic
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biology, Koch Institute and Ludwig Center, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | | | - Huiyuan Zhang
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jackson Nyman
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kaori Sakuishi
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Sema Kurtulus
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - David Gennert
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Junrong Xia
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | - John Y H Kwon
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - James Nevin
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | - Itai Yanai
- Institute for Computational Medicine and Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | | | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| | - Aviv Regev
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biology, Koch Institute and Ludwig Center, Massachusetts Institute of Technology, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - Ana C Anderson
- Evergrande Center for Immunologic Diseases and Ann Romney Center for Neurologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
69
|
Magnetic nanoparticle-induced hyperthermia with appropriate payloads: Paul Ehrlich’s “magic (nano)bullet” for cancer theranostics? Cancer Treat Rev 2016; 50:217-227. [DOI: 10.1016/j.ctrv.2016.09.016] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 09/16/2016] [Accepted: 09/22/2016] [Indexed: 01/11/2023]
|
70
|
Perspectives in immunotherapy: meeting report from the “Immunotherapy Bridge”, Napoli, December 5th 2015. J Immunother Cancer 2016. [PMCID: PMC5067891 DOI: 10.1186/s40425-016-0168-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Harnessing the immune system and preventing immune escape, the immunotherapy of cancer provides great potential for clinical application, in broad patient populations, achieving both conventional and unconventional clinical responses. After the substantial advances in melanoma, the focus of cancer immunotherapy has expanded to include many other cancers. Targeting immune checkpoints and further mechanisms used by tumors to avoid anticancer immunity, different approaches are under evaluation, including combination therapies. The first Immunotherapy Bridge meeting focused on various cancer types including melanoma, non-small cell lung cancer, renal cell, breast and ovarian carcinoma, and discussed mechanisms of action of single agents and combination strategies, and the prediction of clinical responses.
Collapse
|
71
|
Abstract
Recent progress in cancer immunotherapy emphasizes the importance of understanding immune-regulatory pathways in tumours. Dysfunction of antitumour T cells may be due to mechanisms that are evolutionarily conserved or acquired by somatic mutations. The dysfunctional state of T cells has been termed 'exhaustion', on the basis of similarities to dysfunctional T cells in chronic infections. However, despite shared properties, recent studies have identified marked differences between T cell dysfunction in cancer and chronic infection. In this Review, we discuss T cell-intrinsic molecular alterations and metabolic communication in the tumour microenvironment. Identification of the underlying molecular drivers of T cell dysfunction is essential for the continued progress of cancer research and therapy.
Collapse
Affiliation(s)
- Daniel E Speiser
- Department of Oncology, Ludwig Cancer Research, University of Lausanne, Biopole 3 - 02DB92, Chemin des Boveresses 155, CH-1066 Epalinges, Switzerland.,Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, Ontario Cancer Institute, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada
| | - Ping-Chih Ho
- Department of Oncology, Ludwig Cancer Research, University of Lausanne, Biopole 3 - 02DB92, Chemin des Boveresses 155, CH-1066 Epalinges, Switzerland
| | - Grégory Verdeil
- Department of Oncology, Ludwig Cancer Research, University of Lausanne, Biopole 3 - 02DB92, Chemin des Boveresses 155, CH-1066 Epalinges, Switzerland
| |
Collapse
|
72
|
Waugh KA, Leach SM, Moore BL, Bruno TC, Buhrman JD, Slansky JE. Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model. THE JOURNAL OF IMMUNOLOGY 2016; 197:1477-88. [PMID: 27371726 DOI: 10.4049/jimmunol.1600589] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/09/2016] [Indexed: 12/21/2022]
Abstract
Mechanisms of self-tolerance often result in CD8(+) tumor-infiltrating lymphocytes (TIL) with a hypofunctional phenotype incapable of tumor clearance. Using a transplantable colon carcinoma model, we found that CD8(+) T cells became tolerized in <24 h in an established tumor environment. To define the collective impact of pathways suppressing TIL function, we compared genome-wide mRNA expression of tumor-specific CD8(+) T cells from the tumor and periphery. Notably, gene expression induced during TIL hypofunction more closely resembled self-tolerance than viral exhaustion. Differential gene expression was refined to identify a core set of genes that defined hypofunctional TIL; these data comprise the first molecular profile of tumor-specific TIL that are naturally responding and represent a polyclonal repertoire. The molecular profile of TIL was further dissected to determine the extent of overlap and distinction between pathways that collectively restrict T cell functions. As suggested by the molecular profile of TIL, protein expression of inhibitory receptor LAG-3 was differentially regulated throughout prolonged late-G1/early-S phase of the cell cycle. Our data may accelerate efficient identification of combination therapies to boost anti-tumor function of TIL specifically against tumor cells.
Collapse
Affiliation(s)
| | - Sonia M Leach
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206
| | - Brandon L Moore
- University of Colorado School of Medicine, Aurora, CO 80045; and
| | - Tullia C Bruno
- University of Colorado School of Medicine, Aurora, CO 80045; and
| | | | - Jill E Slansky
- University of Colorado School of Medicine, Aurora, CO 80045; and
| |
Collapse
|
73
|
Nguyen HH, Kim T, Song SY, Park S, Cho HH, Jung SH, Ahn JS, Kim HJ, Lee JJ, Kim HO, Cho JH, Yang DH. Naïve CD8(+) T cell derived tumor-specific cytotoxic effectors as a potential remedy for overcoming TGF-β immunosuppression in the tumor microenvironment. Sci Rep 2016; 6:28208. [PMID: 27306834 PMCID: PMC4910083 DOI: 10.1038/srep28208] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/23/2016] [Indexed: 01/04/2023] Open
Abstract
Despite of the potential implications for cancer immunotherapy, conventional approaches using in vitro expanded CD8+ T cells have suboptimal outcomes, mostly due to loss of functionality from cellular exhaustion. We therefore investigated the phenotypic and functional differences among in vitro activated CD8+ T cells of three different sources, namely naïve (NTeff), memory (MTeff) and tumor-infiltrating lymphocytes (TILeff) from human and mice, to better understand mechanisms behind potent effector functions and potential for overcoming current limitations. In line with the greater proliferation activity and longer telomere lengths of NTeff populations, cells of naïve origin exhibited significantly less amounts of T cell exhaustion markers than those of MTeff and TILeff, and moreover, acquired distinct expression patterns of memory-promoting transcription factors, T-bet and Eomes, induced in a rapid and sustainable manner. NTeff cells appeared to have lower expression of Foxp1 and were refractory to apoptosis upon TGF-β conditioning, implying better survival potential and resistance to tumor-induced immune suppression. Of CD8+ T cell pools activated to tumor-specific CTLs, naïve cell generated effectors possessed the most potent cytotoxic activity, validating implications for use in rational design of adoptive immunotherapy.
Collapse
Affiliation(s)
- Hong Hanh Nguyen
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Therasa Kim
- Department of Medical Sciences, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Sang Yun Song
- Department of Thoracic and Cardiovascular Surgery, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Somang Park
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Hyang Hee Cho
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Sung-Hoon Jung
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Jae-Sook Ahn
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Hyeoung-Joon Kim
- Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Je-Jung Lee
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| | - Hee-Ok Kim
- Academy of Immunology &Microbiology (AIM), Institute for Basic Science (IBS), Pohang, Republic of Korea
| | - Jae-Ho Cho
- Academy of Immunology &Microbiology (AIM), Institute for Basic Science (IBS), Pohang, Republic of Korea
| | - Deok-Hwan Yang
- Research Center for Cancer Immunotherapy, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea.,Department of Hematology-Oncology, Chonnam National University Hwasun Hospital, Hwasun, Jeollanam-do, Republic of Korea
| |
Collapse
|
74
|
Effector, Memory, and Dysfunctional CD8(+) T Cell Fates in the Antitumor Immune Response. J Immunol Res 2016; 2016:8941260. [PMID: 27314056 PMCID: PMC4893440 DOI: 10.1155/2016/8941260] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/28/2016] [Indexed: 12/31/2022] Open
Abstract
The adaptive immune system plays a pivotal role in the host's ability to mount an effective, antigen-specific immune response against tumors. CD8(+) tumor-infiltrating lymphocytes (TILs) mediate tumor rejection through recognition of tumor antigens and direct killing of transformed cells. In growing tumors, TILs are often functionally impaired as a result of interaction with, or signals from, transformed cells and the tumor microenvironment. These interactions and signals can lead to transcriptional, functional, and phenotypic changes in TILs that diminish the host's ability to eradicate the tumor. In addition to effector and memory CD8(+) T cells, populations described as exhausted, anergic, senescent, and regulatory CD8(+) T cells have been observed in clinical and basic studies of antitumor immune responses. In the context of antitumor immunity, these CD8(+) T cell subsets remain poorly characterized in terms of fate-specific biomarkers and transcription factor profiles. Here we discuss the current characterization of CD8(+) T cell fates in antitumor immune responses and discuss recent insights into how signals in the tumor microenvironment influence TIL transcriptional networks to promote CD8(+) T cell dysfunction.
Collapse
|
75
|
Affiliation(s)
- Katherine A Waugh
- University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA
| | - Sonia M Leach
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA
| | - Jill E Slansky
- University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA
| |
Collapse
|
76
|
Newick K, O'Brien S, Sun J, Kapoor V, Maceyko S, Lo A, Puré E, Moon E, Albelda SM. Augmentation of CAR T-cell Trafficking and Antitumor Efficacy by Blocking Protein Kinase A Localization. Cancer Immunol Res 2016; 4:541-51. [PMID: 27045023 DOI: 10.1158/2326-6066.cir-15-0263] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 03/04/2016] [Indexed: 11/16/2022]
Abstract
Antitumor treatments based on the infusion of T cells expressing chimeric antigen receptors (CAR T cells) are still relatively ineffective for solid tumors, due to the presence of immunosuppressive mediators [such as prostaglandin E2 (PGE2) and adenosine] and poor T-cell trafficking. PGE2 and adenosine activate protein kinase A (PKA), which then inhibits T-cell receptor (TCR) activation. This inhibition process requires PKA to localize to the immune synapse via binding to the membrane protein ezrin. We generated CAR T cells that expressed a small peptide called the "regulatory subunit I anchoring disruptor" (RIAD) that inhibits the association of PKA with ezrin, thus blunting the negative effects of PKA on TCR activation. After exposure to PGE2 or adenosine in vitro, CAR-RIAD T cells showed increased TCR signaling, released more cytokines, and showed enhanced killing of tumor cells compared with CAR T cells. When injected into tumor-bearing mice, the antitumor efficacy of murine and human CAR-RIAD T cells was enhanced compared with that of CAR T cells, due to resistance to tumor-induced hypofunction and increased T-cell infiltration of established tumors. Subsequent in vitro assays showed that both mouse and human CAR-RIAD cells migrated more efficiently than CAR cells did in response to the chemokine CXCL10 and also had better adhesion to various matrices. Thus, the intracellular addition of the RIAD peptide to adoptively transferred CAR T cells augments their efficacy by increasing their effector function and by improving trafficking into tumor sites. This treatment strategy, therefore, shows potential clinical application for treating solid tumors. Cancer Immunol Res; 4(6); 541-51. ©2016 AACR.
Collapse
Affiliation(s)
- Kheng Newick
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shaun O'Brien
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jing Sun
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Veena Kapoor
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven Maceyko
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Albert Lo
- Department of Biomedical Sciences, School of Veterinary Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ellen Puré
- Department of Biomedical Sciences, School of Veterinary Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Edmund Moon
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Steven M Albelda
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
77
|
Ho PC, Liu PS. Metabolic communication in tumors: a new layer of immunoregulation for immune evasion. J Immunother Cancer 2016; 4:4. [PMID: 26885366 PMCID: PMC4754991 DOI: 10.1186/s40425-016-0109-1] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/13/2016] [Indexed: 01/04/2023] Open
Abstract
The success of cancer immunotherapy reveals the power of host immunity on killing cancer cells and the feasibility to unleash restraints of anti-tumor immunity. However, the immunosuppressive tumor microenvironment and low immunogenicity of cancer cells restrict the therapeutic efficacy of cancer immunotherapies in a small fraction of patients. Therefore deciphering the underlying mechanisms promoting the generation of an immunosuppressive tumor microenvironment is direly needed to better harness host anti-tumor immunity. Early works revealed that deregulated metabolic activities in cancer cells support unrestricted proliferation and survival by producing macromolecules. Intriguingly, recent studies uncovered that metabolic switch in immune and endothelial cells modulate cellular activities and contribute to the progression of several diseases, including cancers. Herein, we review the progress on immunometabolic regulations on fine-tuning activities of immune cells and discuss how metabolic communication between cancer and infiltrating immune cells contributes to cancer immune evasion. Moreover, we would like to discuss how we might exploit this knowledge to improve current immunotherapies and the unresolved issues in this field.
Collapse
Affiliation(s)
- Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland ; Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| | - Pu-Ste Liu
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland ; Ludwig Center for Cancer Research, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
78
|
Robert-Tissot C, Speiser DE. Anticancer Teamwork: Cross-Presenting Dendritic Cells Collaborate with Therapeutic Monoclonal Antibodies. Cancer Discov 2016; 6:17-9. [DOI: 10.1158/2159-8290.cd-15-1366] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
79
|
Abstract
The microenvironment is increasingly recognized to have key roles in cancer, and biomaterials provide a means to engineer microenvironments both in vitro and in vivo to study and manipulate cancer. In vitro cancer models using 3D matrices recapitulate key elements of the tumour microenvironment and have revealed new aspects of cancer biology. Cancer vaccines based on some of the same biomaterials have, in parallel, allowed for the engineering of durable prophylactic and therapeutic anticancer activity in preclinical studies, and some of these vaccines have moved to clinical trials. The impact of biomaterials engineering on cancer treatment is expected to further increase in importance in the years to come.
Collapse
Affiliation(s)
- Luo Gu
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
| | - David J Mooney
- Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138, USA
| |
Collapse
|
80
|
Garg AD, Galluzzi L, Apetoh L, Baert T, Birge RB, Bravo-San Pedro JM, Breckpot K, Brough D, Chaurio R, Cirone M, Coosemans A, Coulie PG, De Ruysscher D, Dini L, de Witte P, Dudek-Peric AM, Faggioni A, Fucikova J, Gaipl US, Golab J, Gougeon ML, Hamblin MR, Hemminki A, Herrmann M, Hodge JW, Kepp O, Kroemer G, Krysko DV, Land WG, Madeo F, Manfredi AA, Mattarollo SR, Maueroder C, Merendino N, Multhoff G, Pabst T, Ricci JE, Riganti C, Romano E, Rufo N, Smyth MJ, Sonnemann J, Spisek R, Stagg J, Vacchelli E, Vandenabeele P, Vandenberk L, Van den Eynde BJ, Van Gool S, Velotti F, Zitvogel L, Agostinis P. Molecular and Translational Classifications of DAMPs in Immunogenic Cell Death. Front Immunol 2015; 6:588. [PMID: 26635802 PMCID: PMC4653610 DOI: 10.3389/fimmu.2015.00588] [Citation(s) in RCA: 304] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 11/02/2015] [Indexed: 12/22/2022] Open
Abstract
The immunogenicity of malignant cells has recently been acknowledged as a critical determinant of efficacy in cancer therapy. Thus, besides developing direct immunostimulatory regimens, including dendritic cell-based vaccines, checkpoint-blocking therapies, and adoptive T-cell transfer, researchers have started to focus on the overall immunobiology of neoplastic cells. It is now clear that cancer cells can succumb to some anticancer therapies by undergoing a peculiar form of cell death that is characterized by an increased immunogenic potential, owing to the emission of the so-called “damage-associated molecular patterns” (DAMPs). The emission of DAMPs and other immunostimulatory factors by cells succumbing to immunogenic cell death (ICD) favors the establishment of a productive interface with the immune system. This results in the elicitation of tumor-targeting immune responses associated with the elimination of residual, treatment-resistant cancer cells, as well as with the establishment of immunological memory. Although ICD has been characterized with increased precision since its discovery, several questions remain to be addressed. Here, we summarize and tabulate the main molecular, immunological, preclinical, and clinical aspects of ICD, in an attempt to capture the essence of this phenomenon, and identify future challenges for this rapidly expanding field of investigation.
Collapse
Affiliation(s)
- Abhishek D Garg
- Cell Death Research and Therapy Laboratory, Department of Cellular Molecular Medicine, KU Leuven - University of Leuven , Leuven , Belgium
| | - Lorenzo Galluzzi
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; Université Pierre et Marie Curie , Paris , France ; Gustave Roussy Comprehensive Cancer Institute , Villejuif , France
| | - Lionel Apetoh
- U866, INSERM , Dijon , France ; Faculté de Médecine, Université de Bourgogne , Dijon , France ; Centre Georges François Leclerc , Dijon , France
| | - Thais Baert
- Department of Gynaecology and Obstetrics, UZ Leuven , Leuven , Belgium ; Laboratory of Gynaecologic Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven , Leuven , Belgium
| | - Raymond B Birge
- Department of Microbiology, Biochemistry, and Molecular Genetics, University Hospital Cancer Center, Rutgers Cancer Institute of New Jersey, New Jersey Medical School , Newark, NJ , USA
| | - José Manuel Bravo-San Pedro
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; Université Pierre et Marie Curie , Paris , France ; Gustave Roussy Comprehensive Cancer Institute , Villejuif , France
| | - Karine Breckpot
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel , Jette , Belgium
| | - David Brough
- Faculty of Life Sciences, University of Manchester , Manchester , UK
| | - Ricardo Chaurio
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nurnberg , Erlangen , Germany
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome , Rome , Italy
| | - An Coosemans
- Department of Gynaecology and Obstetrics, UZ Leuven , Leuven , Belgium ; Laboratory of Gynaecologic Oncology, Department of Oncology, Leuven Cancer Institute, KU Leuven , Leuven , Belgium
| | - Pierre G Coulie
- de Duve Institute, Université Catholique de Louvain , Brussels , Belgium
| | - Dirk De Ruysscher
- Department of Radiation Oncology, University Hospitals Leuven, KU Leuven - University of Leuven , Leuven , Belgium
| | - Luciana Dini
- Department of Biological and Environmental Science and Technology, University of Salento , Salento , Italy
| | - Peter de Witte
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven , Leuven , Belgium
| | - Aleksandra M Dudek-Peric
- Cell Death Research and Therapy Laboratory, Department of Cellular Molecular Medicine, KU Leuven - University of Leuven , Leuven , Belgium
| | | | - Jitka Fucikova
- SOTIO , Prague , Czech Republic ; Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic
| | - Udo S Gaipl
- Department of Radiation Oncology, Universitätsklinikum Erlangen , Erlangen , Germany
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw , Warsaw , Poland
| | | | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital , Boston, MA , USA
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Transplantation Laboratory, Haartman Institute, University of Helsinki , Helsinki , Finland ; Helsinki University Hospital Comprehensive Cancer Center , Helsinki , Finland ; TILT Biotherapeutics Ltd. , Helsinki , Finland
| | - Martin Herrmann
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nurnberg , Erlangen , Germany
| | - James W Hodge
- Recombinant Vaccine Group, Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health , Bethesda, MD , USA
| | - Oliver Kepp
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; Université Pierre et Marie Curie , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute , Villejuif , France
| | - Guido Kroemer
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; Université Pierre et Marie Curie , Paris , France ; Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute , Villejuif , France ; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP , Paris , France ; Department of Women's and Children's Health, Karolinska University Hospital , Stockholm , Sweden
| | - Dmitri V Krysko
- Molecular Signaling and Cell Death Unit, Inflammation Research Center, VIB , Ghent , Belgium ; Department of Biomedical Molecular Biology, Ghent University , Ghent , Belgium
| | - Walter G Land
- Molecular ImmunoRheumatology, INSERM UMRS1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France
| | - Frank Madeo
- Institute of Molecular Biosciences, NAWI Graz, University of Graz , Graz , Austria ; BioTechMed Graz , Graz , Austria
| | - Angelo A Manfredi
- IRRCS Istituto Scientifico San Raffaele, Università Vita-Salute San Raffaele , Milan , Italy
| | - Stephen R Mattarollo
- Translational Research Institute, University of Queensland Diamantina Institute, University of Queensland , Wooloongabba, QLD , Australia
| | - Christian Maueroder
- Department of Internal Medicine 3 - Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nurnberg , Erlangen , Germany
| | - Nicolò Merendino
- Laboratory of Cellular and Molecular Nutrition, Department of Ecological and Biological Sciences, Tuscia University , Viterbo , Italy
| | - Gabriele Multhoff
- Department of Radiation Oncology, Klinikum rechts der Isar, Technische Universität München , Munich , Germany
| | - Thomas Pabst
- Department of Medical Oncology, University Hospital , Bern , Switzerland
| | - Jean-Ehrland Ricci
- INSERM, U1065, Université de Nice-Sophia-Antipolis, Centre Méditerranéen de Médecine Moléculaire (C3M), Équipe "Contrôle Métabolique des Morts Cellulaires" , Nice , France
| | - Chiara Riganti
- Department of Oncology, University of Turin , Turin , Italy
| | - Erminia Romano
- Cell Death Research and Therapy Laboratory, Department of Cellular Molecular Medicine, KU Leuven - University of Leuven , Leuven , Belgium
| | - Nicole Rufo
- Cell Death Research and Therapy Laboratory, Department of Cellular Molecular Medicine, KU Leuven - University of Leuven , Leuven , Belgium
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Insitute , Herston, QLD , Australia ; School of Medicine, University of Queensland , Herston, QLD , Australia
| | - Jürgen Sonnemann
- Department of Paediatric Haematology and Oncology, Children's Clinic, Jena University Hospital , Jena , Germany
| | - Radek Spisek
- SOTIO , Prague , Czech Republic ; Department of Immunology, 2nd Faculty of Medicine, University Hospital Motol, Charles University , Prague , Czech Republic
| | - John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Institut du Cancer de Montréal, Faculté de Pharmacie, Université de Montréal , Montreal, QC , Canada
| | - Erika Vacchelli
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers , Paris , France ; U1138, INSERM , Paris , France ; Université Paris Descartes, Sorbonne Paris Cité , Paris , France ; Université Pierre et Marie Curie , Paris , France ; Gustave Roussy Comprehensive Cancer Institute , Villejuif , France
| | - Peter Vandenabeele
- Molecular Signaling and Cell Death Unit, Inflammation Research Center, VIB , Ghent , Belgium ; Department of Biomedical Molecular Biology, Ghent University , Ghent , Belgium
| | - Lien Vandenberk
- Laboratory of Pediatric Immunology, Department of Microbiology and Immunology, KU Leuven - University of Leuven , Leuven , Belgium
| | - Benoit J Van den Eynde
- Ludwig Institute for Cancer Research, de Duve Institute, Université Catholique de Louvain , Brussels , Belgium
| | - Stefaan Van Gool
- Laboratory of Pediatric Immunology, Department of Microbiology and Immunology, KU Leuven - University of Leuven , Leuven , Belgium
| | - Francesca Velotti
- Department of Ecological and Biological Sciences, Tuscia University , Viterbo , Italy
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute , Villejuif , France ; University of Paris Sud , Le Kremlin-Bicêtre , France ; U1015, INSERM , Villejuif , France ; Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 507 , Villejuif , France
| | - Patrizia Agostinis
- Cell Death Research and Therapy Laboratory, Department of Cellular Molecular Medicine, KU Leuven - University of Leuven , Leuven , Belgium
| |
Collapse
|
81
|
Waugh KA, Leach SM, Slansky JE. Targeting Transcriptional Regulators of CD8+ T Cell Dysfunction to Boost Anti-Tumor Immunity. Vaccines (Basel) 2015; 3:771-802. [PMID: 26393659 PMCID: PMC4586477 DOI: 10.3390/vaccines3030771] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/09/2015] [Accepted: 09/10/2015] [Indexed: 02/07/2023] Open
Abstract
Transcription is a dynamic process influenced by the cellular environment: healthy, transformed, and otherwise. Genome-wide mRNA expression profiles reflect the collective impact of pathways modulating cell function under different conditions. In this review we focus on the transcriptional pathways that control tumor infiltrating CD8+ T cell (TIL) function. Simultaneous restraint of overlapping inhibitory pathways may confer TIL resistance to multiple mechanisms of suppression traditionally referred to as exhaustion, tolerance, or anergy. Although decades of work have laid a solid foundation of altered transcriptional networks underlying various subsets of hypofunctional or “dysfunctional” CD8+ T cells, an understanding of the relevance in TIL has just begun. With recent technological advances, it is now feasible to further elucidate and utilize these pathways in immunotherapy platforms that seek to increase TIL function.
Collapse
Affiliation(s)
- Katherine A Waugh
- University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA.
| | - Sonia M Leach
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206, USA.
| | - Jill E Slansky
- University of Colorado School of Medicine, 12800 East 19th Avenue, Mail Stop 8333, Aurora, CO 80045, USA.
| |
Collapse
|
82
|
Phosphoenolpyruvate Is a Metabolic Checkpoint of Anti-tumor T Cell Responses. Cell 2015; 162:1217-28. [PMID: 26321681 DOI: 10.1016/j.cell.2015.08.012] [Citation(s) in RCA: 1083] [Impact Index Per Article: 108.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 06/03/2015] [Accepted: 07/21/2015] [Indexed: 02/06/2023]
Abstract
Activated T cells engage aerobic glycolysis and anabolic metabolism for growth, proliferation, and effector functions. We propose that a glucose-poor tumor microenvironment limits aerobic glycolysis in tumor-infiltrating T cells, which suppresses tumoricidal effector functions. We discovered a new role for the glycolytic metabolite phosphoenolpyruvate (PEP) in sustaining T cell receptor-mediated Ca(2+)-NFAT signaling and effector functions by repressing sarco/ER Ca(2+)-ATPase (SERCA) activity. Tumor-specific CD4 and CD8 T cells could be metabolically reprogrammed by increasing PEP production through overexpression of phosphoenolpyruvate carboxykinase 1 (PCK1), which bolstered effector functions. Moreover, PCK1-overexpressing T cells restricted tumor growth and prolonged the survival of melanoma-bearing mice. This study uncovers new metabolic checkpoints for T cell activity and demonstrates that metabolic reprogramming of tumor-reactive T cells can enhance anti-tumor T cell responses, illuminating new forms of immunotherapy.
Collapse
|
83
|
Leidinger P, Backes C, Dahmke IN, Galata V, Huwer H, Stehle I, Bals R, Keller A, Meese E. What makes a blood cell based miRNA expression pattern disease specific?--a miRNome analysis of blood cell subsets in lung cancer patients and healthy controls. Oncotarget 2015; 5:9484-97. [PMID: 25344866 PMCID: PMC4253448 DOI: 10.18632/oncotarget.2419] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
There is evidence of blood-borne miRNA signatures for various human diseases. To dissect the origin of disease-specific miRNA expression in human blood, we separately analyzed the miRNome of different immune cell subtypes, each in lung cancer patients and healthy individuals. Each immune cell type revealed a specific miRNA expression pattern also dependinging on the cell origin, line of defense, and function. The overall expression pattern of each leukocyte subtype showed great similarities between patients and controls. However, for each cell subtype we identified miRNAs that were deregulated in lung cancer patients including hsa-miR-21, a well-known oncomiR associated with poor lung cancer prognosis that was up-regulated in all leukocyte subtype comparisons of cancer versus controls. While the miRNome of cells of the adaptive immune system allowed only a weak separation between patients and controls, cells of the innate immune system allowed perfect or nearly perfect classification. Leukocytes of lung cancer patients show a cancer-specific miRNA expression profile. Our data also show that cancer specific miRNA expression pattern of whole blood samples are not determined by a single cell type. The data indicate that additional blood components, like erythrocytes, platelets, or exosomes might contribute to the disease specificity of a miRNA signature.
Collapse
Affiliation(s)
- Petra Leidinger
- Institute of Human Genetics, Medical School, Saarland University, Building 60, 66421 Homburg/Saar, Germany
| | - Christina Backes
- Department of Clinical Bioinformatics, Saarland University, Building E2.1, 66123 Saarbrücken, Germany
| | - Indra N Dahmke
- Institute for Clinical and Experimental Surgery, Saarland University, Building 65, 66421 Homburg/Saar, Germany
| | - Valentina Galata
- Department of Clinical Bioinformatics, Saarland University, Building E2.1, 66123 Saarbrücken, Germany
| | - Hanno Huwer
- Department of Thoracic Surgery, Voelklingen Heart Center, 66333 Voelklingen Germany
| | - Ingo Stehle
- Department of Pneumology, Medical School, Saarland University, Building 91, 66421 Homburg/Saar, Germany
| | - Robert Bals
- Department of Pneumology, Medical School, Saarland University, Building 91, 66421 Homburg/Saar, Germany
| | - Andreas Keller
- Department of Clinical Bioinformatics, Saarland University, Building E2.1, 66123 Saarbrücken, Germany
| | - Eckart Meese
- Institute of Human Genetics, Medical School, Saarland University, Building 60, 66421 Homburg/Saar, Germany
| |
Collapse
|
84
|
Gassner FJ, Zaborsky N, Catakovic K, Rebhandl S, Huemer M, Egle A, Hartmann TN, Greil R, Geisberger R. Chronic lymphocytic leukaemia induces an exhausted T cell phenotype in the TCL1 transgenic mouse model. Br J Haematol 2015; 170:515-22. [PMID: 25940792 PMCID: PMC4687418 DOI: 10.1111/bjh.13467] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 03/15/2015] [Indexed: 12/31/2022]
Abstract
Although chronic lymphocytic leukaemia (CLL) is a B cell malignancy, earlier studies have indicated a role of T cells in tumour growth and disease progression. In particular, the functional silencing of antigen-experienced T cells, called T cell exhaustion, has become implicated in immune evasion in CLL. In this study, we tested whether T cell exhaustion is recapitulated in the TCL1(tg) mouse model for CLL. We show that T cells express high levels of the inhibitory exhaustion markers programmed cell death 1 (PDCD1, also termed PD-1) and lymphocyte-activation gene 3 (LAG3), whereas CLL cells express high levels of CD274 (also termed PD-ligand 1). In addition, the fraction of exhausted T cells increases with CLL progression. Finally, we demonstrate that exhausted T cells are reinvigorated towards CLL cytotoxicity by inhibition of PDCD1/CD274 interaction in vivo. These results suggest that T cell exhaustion contributes to CLL pathogenesis and that interference with PDCD1/CD274 signalling holds high potential for therapeutic approaches.
Collapse
MESH Headings
- Animals
- Gene Expression Regulation, Leukemic/genetics
- Gene Expression Regulation, Leukemic/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Mice
- Mice, Transgenic
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
Collapse
Affiliation(s)
- Franz J Gassner
- Laboratory for Immunological and Molecular Cancer Research, 3 Medical Department with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Centre, Paracelsus Medical UniversitySalzburg, Austria
- Salzburg Cancer Research InstituteSalzburg, Austria
| | - Nadja Zaborsky
- Laboratory for Immunological and Molecular Cancer Research, 3 Medical Department with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Centre, Paracelsus Medical UniversitySalzburg, Austria
- Salzburg Cancer Research InstituteSalzburg, Austria
| | - Kemal Catakovic
- Laboratory for Immunological and Molecular Cancer Research, 3 Medical Department with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Centre, Paracelsus Medical UniversitySalzburg, Austria
- Salzburg Cancer Research InstituteSalzburg, Austria
| | - Stefan Rebhandl
- Laboratory for Immunological and Molecular Cancer Research, 3 Medical Department with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Centre, Paracelsus Medical UniversitySalzburg, Austria
- Salzburg Cancer Research InstituteSalzburg, Austria
| | - Michael Huemer
- Laboratory for Immunological and Molecular Cancer Research, 3 Medical Department with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Centre, Paracelsus Medical UniversitySalzburg, Austria
- Salzburg Cancer Research InstituteSalzburg, Austria
| | - Alexander Egle
- Laboratory for Immunological and Molecular Cancer Research, 3 Medical Department with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Centre, Paracelsus Medical UniversitySalzburg, Austria
- Salzburg Cancer Research InstituteSalzburg, Austria
| | - Tanja N Hartmann
- Laboratory for Immunological and Molecular Cancer Research, 3 Medical Department with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Centre, Paracelsus Medical UniversitySalzburg, Austria
- Salzburg Cancer Research InstituteSalzburg, Austria
| | - Richard Greil
- Laboratory for Immunological and Molecular Cancer Research, 3 Medical Department with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Centre, Paracelsus Medical UniversitySalzburg, Austria
- Salzburg Cancer Research InstituteSalzburg, Austria
| | - Roland Geisberger
- Laboratory for Immunological and Molecular Cancer Research, 3 Medical Department with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Centre, Paracelsus Medical UniversitySalzburg, Austria
- Salzburg Cancer Research InstituteSalzburg, Austria
| |
Collapse
|
85
|
|
86
|
Verdeil G, Fuertes Marraco SA, Murray T, Speiser DE. From T cell "exhaustion" to anti-cancer immunity. Biochim Biophys Acta Rev Cancer 2015; 1865:49-57. [PMID: 26123831 DOI: 10.1016/j.bbcan.2015.06.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 06/18/2015] [Accepted: 06/23/2015] [Indexed: 12/14/2022]
Abstract
The immune system has the potential to protect from malignant diseases for extended periods of time. Unfortunately, spontaneous immune responses are often inefficient. Significant effort is required to develop reliable, broadly applicable immunotherapies for cancer patients. A major innovation was transplantation with hematopoietic stem cells from genetically distinct donors for patients with hematologic malignancies. In this setting, donor T cells induce long-term remission by keeping cancer cells in check through powerful allogeneic graft-versus-leukemia effects. More recently, a long awaited breakthrough for patients with solid tissue cancers was achieved, by means of therapeutic blockade of T cell inhibitory receptors. In untreated cancer patients, T cells are dysfunctional and remain in a state of T cell "exhaustion". Nonetheless, they often retain a high potential for successful defense against cancer, indicating that many T cells are not entirely and irreversibly exhausted but can be mobilized to become highly functional. Novel antibody therapies that block inhibitory receptors can lead to strong activation of anti-tumor T cells, mediating clinically significant anti-cancer immunity for many years. Here we review these new treatments and the current knowledge on tumor antigen-specific T cells.
Collapse
Affiliation(s)
- Grégory Verdeil
- Ludwig Cancer Research Center and Department of Oncology, Clinical Tumor Biology & Immunotherapy Group, Lausanne University Hospital Center (CHUV) and University of Lausanne, Route de la Corniche 9A, CH-1066 Epalinges, Switzerland
| | - Silvia A Fuertes Marraco
- Ludwig Cancer Research Center and Department of Oncology, Clinical Tumor Biology & Immunotherapy Group, Lausanne University Hospital Center (CHUV) and University of Lausanne, Route de la Corniche 9A, CH-1066 Epalinges, Switzerland
| | - Timothy Murray
- Ludwig Cancer Research Center and Department of Oncology, Clinical Tumor Biology & Immunotherapy Group, Lausanne University Hospital Center (CHUV) and University of Lausanne, Route de la Corniche 9A, CH-1066 Epalinges, Switzerland
| | - Daniel E Speiser
- Ludwig Cancer Research Center and Department of Oncology, Clinical Tumor Biology & Immunotherapy Group, Lausanne University Hospital Center (CHUV) and University of Lausanne, Route de la Corniche 9A, CH-1066 Epalinges, Switzerland.
| |
Collapse
|
87
|
Fuertes Marraco SA, Neubert NJ, Verdeil G, Speiser DE. Inhibitory Receptors Beyond T Cell Exhaustion. Front Immunol 2015; 6:310. [PMID: 26167163 PMCID: PMC4481276 DOI: 10.3389/fimmu.2015.00310] [Citation(s) in RCA: 175] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 05/30/2015] [Indexed: 12/15/2022] Open
Abstract
Inhibitory receptors (iRs) are frequently associated with "T cell exhaustion". However, the expression of iRs is also dependent on T cell differentiation and activation. Therapeutic blockade of various iRs, also referred to as "checkpoint blockade", is showing -unprecedented results in the treatment of cancer patients. Consequently, the clinical potential in this field is broad, calling for increased research efforts and rapid refinements in the understanding of iR function. In this review, we provide an overview on the significance of iR expression for the interpretation of T cell functionality. We summarize how iRs have been strongly associated with "T cell exhaustion" and illustrate the parallel evidence on the importance of T cell differentiation and activation for the expression of iRs. The differentiation subsets of CD8 T cells (naïve, effector, and memory cells) show broad and inherent differences in iR expression, while activation leads to strong upregulation of iRs. Therefore, changes in iR expression during an immune response are often concomitant with T cell differentiation and activation. Sustained expression of iRs in chronic infection and in the tumor microenvironment likely reflects a specialized T cell differentiation. In these situations of prolonged antigen exposure and chronic inflammation, T cells are "downtuned" in order to limit tissue damage. Furthermore, we review the novel "checkpoint blockade" treatments and the potential of iRs as biomarkers. Finally, we provide recommendations for the immune monitoring of patients to interpret iR expression data combined with parameters of activation and differentiation of T cells.
Collapse
Affiliation(s)
- Silvia A. Fuertes Marraco
- Ludwig Cancer Research Center, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Natalie J. Neubert
- Ludwig Cancer Research Center, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Grégory Verdeil
- Ludwig Cancer Research Center, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Daniel E. Speiser
- Ludwig Cancer Research Center, University of Lausanne, Lausanne, Switzerland
- Department of Oncology, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| |
Collapse
|
88
|
Schreiner J, Thommen DS, Herzig P, Bacac M, Klein C, Roller A, Belousov A, Levitsky V, Savic S, Moersig W, Uhlenbrock F, Heinzelmann-Schwarz VA, Umana P, Pisa P, von Bergwelt-Baildon M, Lardinois D, Müller P, Karanikas V, Zippelius A. Expression of inhibitory receptors on intratumoral T cells modulates the activity of a T cell-bispecific antibody targeting folate receptor. Oncoimmunology 2015; 5:e1062969. [PMID: 27057429 DOI: 10.1080/2162402x.2015.1062969] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 06/08/2015] [Accepted: 06/10/2015] [Indexed: 12/18/2022] Open
Abstract
T-cell bispecific antibodies (TCBs) are a novel therapeutic tool designed to selectively recruit T-cells to tumor cells and simultaneously activate them. However, it is currently unknown whether the dysfunctional state of T-cells, embedded into the tumor microenvironment, imprints on the therapeutic activity of TCBs. We performed a comprehensive analysis of activation and effector functions of tumor-infiltrating T-cells (TILs) in different tumor types, upon stimulation by a TCB targeting folate receptor 1 and CD3 (FolR1-TCB). We observed a considerable heterogeneity in T-cell activation, cytokine production and tumor cell killing upon exposure to FolR1-TCB among different FolR1-expressing tumors. Of note, tumors presenting with a high frequency of PD-1hi TILs displayed significantly impaired tumor cell killing and T-cell function. Further characterization of additional T-cell inhibitory receptors revealed that PD-1hi TILs defined a T-cell subset with particularly high levels of multiple inhibitory receptors compared with PD-1int and PD-1neg T-cells. PD-1 blockade could restore cytokine secretion but not cytotoxicity of TILs in a subset of patients with scarce PD-1hi expressing cells; in contrast, patients with abundance of PD-1hi expressing T-cells did not benefit from PD-1 blockade. Our data highlight that FolR1-TCB is a promising novel immunotherapeutic treatment option which is capable of activating intratumoral T-cells in different carcinomas. However, its therapeutic efficacy may be substantially hampered by a pre-existing dysfunctional state of T-cells, reflected by abundance of intratumoral PD-1hi T-cells. These findings present a rationale for combinatorial approaches of TCBs with other therapeutic strategies targeting T-cell dysfunction.
Collapse
Affiliation(s)
- Jens Schreiner
- Laboratory of Cancer Immunology, Department of Biomedicine , Basel, Switzerland
| | - Daniela S Thommen
- Laboratory of Cancer Immunology, Department of Biomedicine, Basel, Switzerland; Department of Medical Oncology, University Hospital Basel, Basel, Switzerland
| | - Petra Herzig
- Laboratory of Cancer Immunology, Department of Biomedicine , Basel, Switzerland
| | - Marina Bacac
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Christian Klein
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Andreas Roller
- Roche Pharma Research and Early Development, Roche Innovation Center Penzberg, Roche Innovation Center Penzberg , Penzberg, Germany
| | - Anton Belousov
- Roche Pharma Research and Early Development, Roche Innovation Center Penzberg, Roche Innovation Center Penzberg , Penzberg, Germany
| | - Victor Levitsky
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Spasenija Savic
- Institute of Pathology, University Hospital Basel , Basel, Switzerland
| | - Wolfgang Moersig
- Department of Surgery, University Hospital Basel , Basel, Switzerland
| | | | | | - Pablo Umana
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Pavel Pisa
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | | | - Didier Lardinois
- Department of Surgery, University Hospital Basel , Basel, Switzerland
| | - Philipp Müller
- Laboratory of Cancer Immunology, Department of Biomedicine , Basel, Switzerland
| | - Vaios Karanikas
- Roche Pharma Research and Early Development, Roche Innovation Center Zurich , Schlieren, Switzerland
| | - Alfred Zippelius
- Laboratory of Cancer Immunology, Department of Biomedicine, Basel, Switzerland; Department of Medical Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
89
|
Jiang Y, Li Y, Zhu B. T-cell exhaustion in the tumor microenvironment. Cell Death Dis 2015; 6:e1792. [PMID: 26086965 PMCID: PMC4669840 DOI: 10.1038/cddis.2015.162] [Citation(s) in RCA: 751] [Impact Index Per Article: 75.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/27/2015] [Accepted: 04/30/2015] [Indexed: 12/24/2022]
Abstract
T-cell exhaustion was originally identified during chronic infection in mice, and was subsequently observed in humans with cancer. The exhausted T cells in the tumor microenvironment show overexpressed inhibitory receptors, decreased effector cytokine production and cytolytic activity, leading to the failure of cancer elimination. Restoring exhausted T cells represents an inspiring strategy for cancer treatment, which has yielded promising results and become a significant breakthrough in the cancer immunotherapy. In this review, we overview the updated understanding on the exhausted T cells in cancer and their potential regulatory mechanisms and discuss current therapeutic interventions targeting exhausted T cells in clinical trials.
Collapse
Affiliation(s)
- Y Jiang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Y Li
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Center for Experimental Therapeutics and Reperfusion Injury, Perioperative and Pain Medicine, Harvard Institutes of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - B Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| |
Collapse
|
90
|
Mac Keon S, Ruiz MS, Gazzaniga S, Wainstok R. Dendritic cell-based vaccination in cancer: therapeutic implications emerging from murine models. Front Immunol 2015; 6:243. [PMID: 26042126 PMCID: PMC4438595 DOI: 10.3389/fimmu.2015.00243] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/06/2015] [Indexed: 01/29/2023] Open
Abstract
Dendritic cells (DCs) play a pivotal role in the orchestration of immune responses, and are thus key targets in cancer vaccine design. Since the 2010 FDA approval of the first cancer DC-based vaccine (Sipuleucel-T), there has been a surge of interest in exploiting these cells as a therapeutic option for the treatment of tumors of diverse origin. In spite of the encouraging results obtained in the clinic, many elements of DC-based vaccination strategies need to be optimized. In this context, the use of experimental cancer models can help direct efforts toward an effective vaccine design. This paper reviews recent findings in murine models regarding the antitumoral mechanisms of DC-based vaccination, covering issues related to antigen sources, the use of adjuvants and maturing agents, and the role of DC subsets and their interaction in the initiation of antitumoral immune responses. The summary of such diverse aspects will highlight advantages and drawbacks in the use of murine models, and contribute to the design of successful DC-based translational approaches for cancer treatment.
Collapse
Affiliation(s)
- Soledad Mac Keon
- Laboratorio de Cancerología, Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires IIBBA-CONICET , Buenos Aires , Argentina
| | - María Sol Ruiz
- Centro de Investigaciones Oncológicas, Fundación para la Investigación, Docencia y Prevención del Cáncer (FUCA) , Buenos Aires , Argentina
| | - Silvina Gazzaniga
- Laboratorio de Biología Tumoral, Departamento de Química Biológica IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| | - Rosa Wainstok
- Laboratorio de Cancerología, Fundación Instituto Leloir, Instituto de Investigaciones Bioquímicas de Buenos Aires IIBBA-CONICET , Buenos Aires , Argentina ; Laboratorio de Biología Tumoral, Departamento de Química Biológica IQUIBICEN-CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires , Buenos Aires , Argentina
| |
Collapse
|
91
|
Veliça P, Zech M, Henson S, Holler A, Manzo T, Pike R, Santos E Sousa P, Zhang L, Heinz N, Schiedlmeier B, Pule M, Stauss H, Chakraverty R. Genetic Regulation of Fate Decisions in Therapeutic T Cells to Enhance Tumor Protection and Memory Formation. Cancer Res 2015; 75:2641-52. [PMID: 25904681 DOI: 10.1158/0008-5472.can-14-3283] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 03/26/2015] [Indexed: 11/16/2022]
Abstract
A key challenge in the field of T-cell immunotherapy for cancer is creating a suitable platform for promoting differentiation of effector cells while at the same time enabling self-renewal needed for long-term memory. Although transfer of less differentiated memory T cells increases efficacy through greater expansion and persistence in vivo, the capacity of such cells to sustain effector functions within immunosuppressive tumor microenvironments may still be limiting. We have therefore directly compared the impact of effector versus memory differentiation of therapeutic T cells in tumor-bearing mice by introducing molecular switches that regulate cell fate decisions via mTOR. Ectopic expression of RAS homolog enriched in brain (RHEB) increased mTORC1 signaling, promoted a switch to aerobic glycolysis, and increased expansion of effector T cells. By rapidly infiltrating tumors, RHEB-transduced T cells significantly reduced the emergence of immunoedited escape variants. In contrast, expression of proline-rich Akt substrate of 40 kDa (PRAS40) inhibited mTORC1, promoted quiescence, and blocked tumor infiltration. Fate mapping studies following transient expression of PRAS40 demonstrated that mTORC1(low) T cells made no contribution to initial tumor control but instead survived to become memory cells proficient in generating recall immunity. Our data support the design of translational strategies for generating heterogeneous T-cell immunity against cancer, with the appropriate balance between promoting effector differentiation and self-renewal. Unlike pharmacologic inhibitors, the genetic approach described here allows for upregulation as well as inhibition of the mTORC1 pathway and is highly selective for the therapeutic T cells without affecting systemic mTORC1 functions.
Collapse
Affiliation(s)
- Pedro Veliça
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom. Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Mathias Zech
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Sian Henson
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Angelika Holler
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Teresa Manzo
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom. Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Rebecca Pike
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Pedro Santos E Sousa
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom. Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Lei Zhang
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom. Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | | | | | - Martin Pule
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Hans Stauss
- Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Ronjon Chakraverty
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom. Institute of Immunity and Transplantation, University College London, London, United Kingdom.
| |
Collapse
|
92
|
Wald N, Legat A, Meyer C, Speiser DE, Goormaghtigh E. An infrared spectral signature of human lymphocyte subpopulations from peripheral blood. Analyst 2015; 140:2257-65. [PMID: 25553786 DOI: 10.1039/c4an02247e] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Metastatic melanomas are frequently refractory to most adjuvant therapies such as chemotherapies and radiotherapies. Recently, immunotherapies have shown good results in the treatment of some metastatic melanomas. Immune cell infiltration in the tumor has been associated with successful immunotherapy. More generally, tumor infiltrating lymphocytes (TILs) in the primary tumor and in metastases of melanoma patients have been demonstrated to correlate positively with favorable clinical outcomes. Altogether, these findings suggest the importance of being able to identify, quantify and characterize immune infiltration at the tumor site for a better diagnostic and treatment choice. In this paper, we used Fourier Transform Infrared (FTIR) imaging to identify and quantify different subpopulations of T cells: the cytotoxic T cells (CD8+), the helper T cells (CD4+) and the regulatory T cells (T reg). As a proof of concept, we investigated pure populations isolated from human peripheral blood from 6 healthy donors. These subpopulations were isolated from blood samples by magnetic labeling and purities were assessed by Fluorescence Activated Cell Sorting (FACS). The results presented here show that Fourier Transform Infrared (FTIR) imaging followed by supervised Partial Least Square Discriminant Analysis (PLS-DA) allows an accurate identification of CD4+ T cells and CD8+ T cells (>86%). We then developed a PLS regression allowing the quantification of T reg in a different mix of immune cells (e.g. Peripheral Blood Mononuclear Cells (PBMCs)). Altogether, these results demonstrate the sensitivity of infrared imaging to detect the low biological variability observed in T cell subpopulations.
Collapse
Affiliation(s)
- N Wald
- Center for Structural Biology and Bioinformatics, Laboratory for the Structure and Function of Biological Membranes; Université Libre de Bruxelles, Campus Plaine, Bld du Triomphe 2, CP206/2, B1050 Brussels, Belgium.
| | | | | | | | | |
Collapse
|
93
|
Schmittnaegel M, Levitsky V, Hoffmann E, Georges G, Mundigl O, Klein C, Knoetgen H. Committing Cytomegalovirus-Specific CD8 T Cells to Eliminate Tumor Cells by Bifunctional Major Histocompatibility Class I Antibody Fusion Molecules. Cancer Immunol Res 2015; 3:764-76. [PMID: 25691327 DOI: 10.1158/2326-6066.cir-15-0037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 02/07/2015] [Indexed: 11/16/2022]
Abstract
Tumor cells escape immune eradication through multiple mechanisms, including loss of antigenicity and local suppression of effector lymphocytes. To counteract these obstacles, we aimed to direct the unique cytomegalovirus (CMV)-specific immune surveillance against tumor cells. We developed a novel generation of fusion proteins composed of a tumor antigen-specific full immunoglobulin connected to a single major histocompatibility class I complex bearing a covalently linked virus-derived peptide (pMHCI-IgG). Here, we show that tumor antigen-expressing cancer cells, which are decorated with pMHCI-IgGs containing a HLA-A*0201 molecule associated with a CMV-derived peptide, are specifically eliminated through engagement of antigen-specific CD8(+) T cells isolated from peripheral blood mononuclear cell preparations of CMV-infected humans. These CD8(+) T cells act without additional expansion, preactivation, or provision of costimulatory signals. Elimination of tumor cells is induced at similar concentrations and with similar time kinetics as those seen with bispecific T-cell engagers (BiTE). However, while BiTE-like reagents indiscriminately activate T cells through binding to the T-cell receptor complex, pMHCI-IgGs selectively engage antigen-specific, constantly renewable, differentiated effector cytotoxic T lymphocytes to tumor cells, thereby representing a novel class of anticancer immunotherapeutics with potentially improved safety and efficacy profiles.
Collapse
Affiliation(s)
- Martina Schmittnaegel
- Large Molecule Research, Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Penzberg, Germany
| | - Victor Levitsky
- Discovery Oncology, Roche Innovation Center Zurich, Roche Pharma Research and Early Development, Zurich, Switzerland
| | - Eike Hoffmann
- Large Molecule Research, Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Penzberg, Germany
| | - Guy Georges
- Large Molecule Research, Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Penzberg, Germany
| | - Olaf Mundigl
- Large Molecule Research, Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Penzberg, Germany
| | - Christian Klein
- Discovery Oncology, Roche Innovation Center Zurich, Roche Pharma Research and Early Development, Zurich, Switzerland
| | - Hendrik Knoetgen
- Large Molecule Research, Roche Innovation Center Penzberg, Roche Pharma Research and Early Development, Penzberg, Germany.
| |
Collapse
|
94
|
Jing W, Gershan JA, Weber J, Tlomak D, McOlash L, Sabatos-Peyton C, Johnson BD. Combined immune checkpoint protein blockade and low dose whole body irradiation as immunotherapy for myeloma. J Immunother Cancer 2015; 3:2. [PMID: 25614821 PMCID: PMC4302511 DOI: 10.1186/s40425-014-0043-z] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 12/01/2014] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Multiple myeloma is characterized by the presence of transformed neoplastic plasma cells in the bone marrow and is generally considered to be an incurable disease. Successful treatments will likely require multi-faceted approaches incorporating conventional drug therapies, immunotherapy and other novel treatments. Our lab previously showed that a combination of transient lymphodepletion (sublethal whole body irradiation) and PD-1/PD-L1 blockade generated anti-myeloma T cell reactivity capable of eliminating established disease. We hypothesized that blocking a combination of checkpoint receptors in the context of low-dose, lymphodepleting whole body radiation would boost anti-tumor immunity. METHODS To test our central hypothesis, we utilized a 5T33 murine multiple myeloma model. Myeloma-bearing mice were treated with a low dose of whole body irradiation and combinations of blocking antibodies to PD-L1, LAG-3, TIM-3, CD48 (the ligand for 2B4) and CTLA4. RESULTS Temporal phenotypic analysis of bone marrow from myeloma-bearing mice demonstrated that elevated percentages of PD-1, 2B4, LAG-3 and TIM-3 proteins were expressed on T cells. When PD-L1 blockade was combined with blocking antibodies to LAG-3, TIM-3 or CTLA4, synergistic or additive increases in survival were observed (survival rates improved from ~30% to >80%). The increased survival rates correlated with increased frequencies of tumor-reactive CD8 and CD4 T cells. When stimulated in vitro with myeloma cells, CD8 T cells from treated mice produced elevated levels proinflammatory cytokines. Cytokines were spontaneously released from CD4 T cells isolated from mice treated with PD-L1 plus CTLA4 blocking antibodies. CONCLUSIONS These data indicate that blocking PD-1/PD-L1 interactions in conjunction with other immune checkpoint proteins provides synergistic anti-tumor efficacy following lymphodepletive doses of whole body irradiation. This strategy is a promising combination strategy for myeloma and other hematologic malignancies.
Collapse
Affiliation(s)
- Weiqing Jing
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Jill A Gershan
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - James Weber
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Dominique Tlomak
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Laura McOlash
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | | | - Bryon D Johnson
- Division of Hematology/Oncology/Transplant, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| |
Collapse
|
95
|
Rotte A, Bhandaru M, Zhou Y, McElwee KJ. Immunotherapy of melanoma: Present options and future promises. Cancer Metastasis Rev 2015; 34:115-28. [DOI: 10.1007/s10555-014-9542-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
96
|
Stephen TL, Rutkowski MR, Allegrezza MJ, Perales-Puchalt A, Tesone AJ, Svoronos N, Nguyen JM, Sarmin F, Borowsky ME, Tchou J, Conejo-Garcia JR. Transforming growth factor β-mediated suppression of antitumor T cells requires FoxP1 transcription factor expression. Immunity 2014; 41:427-439. [PMID: 25238097 DOI: 10.1016/j.immuni.2014.08.012] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 08/27/2014] [Indexed: 12/15/2022]
Abstract
Tumor-reactive T cells become unresponsive in advanced tumors. Here we have characterized a common mechanism of T cell unresponsiveness in cancer driven by the upregulation of the transcription factor Forkhead box protein P1 (Foxp1), which prevents CD8⁺ T cells from proliferating and upregulating Granzyme-B and interferon-γ in response to tumor antigens. Accordingly, Foxp1-deficient lymphocytes induced rejection of incurable tumors and promoted protection against tumor rechallenge. Mechanistically, Foxp1 interacted with the transcription factors Smad2 and Smad3 in preactivated CD8⁺ T cells in response to microenvironmental transforming growth factor-β (TGF-β), and was essential for its suppressive activity. Therefore, Smad2 and Smad3-mediated c-Myc repression requires Foxp1 expression in T cells. Furthermore, Foxp1 directly mediated TGF-β-induced c-Jun transcriptional repression, which abrogated T cell activity. Our results unveil a fundamental mechanism of T cell unresponsiveness different from anergy or exhaustion, driven by TGF-β signaling on tumor-associated lymphocytes undergoing Foxp1-dependent transcriptional regulation.
Collapse
Affiliation(s)
- Tom L Stephen
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Melanie R Rutkowski
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Michael J Allegrezza
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Alfredo Perales-Puchalt
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Amelia J Tesone
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Nikolaos Svoronos
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Jenny M Nguyen
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Fahmida Sarmin
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Mark E Borowsky
- Helen F. Graham Cancer Center, Christiana Care Health System, 4701 Ogletown-Stanton Road, Newark, DE 19713, USA
| | - Julia Tchou
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania, Philadelphia, PA 19104-1693, USA; Rena Rowan Breast Center, University of Pennsylvania, Philadelphia, PA 19104-1693, USA; Abramson Cancer Center Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-1693, USA
| | - Jose R Conejo-Garcia
- Tumor Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
97
|
Garber HR, Mirza A, Mittendorf EA, Alatrash G. Adoptive T-cell therapy for Leukemia. MOLECULAR AND CELLULAR THERAPIES 2014; 2:25. [PMID: 26056592 PMCID: PMC4452065 DOI: 10.1186/2052-8426-2-25] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/02/2014] [Indexed: 01/15/2023]
Abstract
Allogeneic stem cell transplantation (alloSCT) is the most robust form of adoptive cellular therapy (ACT) and has been tremendously effective in the treatment of leukemia. It is one of the original forms of cancer immunotherapy and illustrates that lymphocytes can specifically recognize and eliminate aberrant, malignant cells. However, because of the high morbidity and mortality that is associated with alloSCT including graft-versus-host disease (GvHD), refining the anti-leukemia immunity of alloSCT to target distinct antigens that mediate the graft-versus-leukemia (GvL) effect could transform our approach to treating leukemia, and possibly other hematologic malignancies. Over the past few decades, many leukemia antigens have been discovered that can separate malignant cells from normal host cells and render them vulnerable targets. In concert, the field of T-cell engineering has matured to enable transfer of ectopic high-affinity antigen receptors into host or donor cells with greater efficiency and potency. Many preclinical studies have demonstrated that engineered and conventional T-cells can mediate lysis and eradication of leukemia via one or more leukemia antigen targets. This evidence now serves as a foundation for clinical trials that aim to cure leukemia using T-cells. The recent clinical success of anti-CD19 chimeric antigen receptor (CAR) cells for treating patients with acute lymphoblastic leukemia and chronic lymphocytic leukemia displays the potential of this new therapeutic modality. In this review, we discuss some of the most promising leukemia antigens and the novel strategies that have been implemented for adoptive cellular immunotherapy of lymphoid and myeloid leukemias. It is important to summarize the data for ACT of leukemia for physicians in-training and in practice and for investigators who work in this and related fields as there are recent discoveries already being translated to the patient setting and numerous accruing clinical trials. We primarily focus on ACT that has been used in the clinical setting or that is currently undergoing preclinical testing with a foreseeable clinical endpoint.
Collapse
Affiliation(s)
- Haven R Garber
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center Houston, Houston, 77030 Texas
| | - Asma Mirza
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center Houston, Houston, 77030 Texas
| | - Elizabeth A Mittendorf
- Department Surgical Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Gheath Alatrash
- Department of Stem Cell Transplantation and Cellular Therapy, University of Texas MD Anderson Cancer Center Houston, Houston, 77030 Texas
| |
Collapse
|
98
|
Feng Y, Zhang N, Jacobs KM, Jiang W, Yang LV, Li Z, Zhang J, Lu JQ, Hu XH. Polarization imaging and classification of Jurkat T and Ramos B cells using a flow cytometer. Cytometry A 2014; 85:817-26. [PMID: 25044756 DOI: 10.1002/cyto.a.22504] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Revised: 03/21/2014] [Accepted: 06/18/2014] [Indexed: 12/23/2022]
Abstract
Label-free and rapid classification of cells can have awide range of applications in biology. We report a robust method of polarization diffraction imaging flow cytometry (p-DIFC) for achieving this goal. Coherently scattered light signals are acquired from single cells excited by a polarized laser beam in the form of two cross-polarized diffraction images. Image texture and intensity parameters are extracted with a gray level co-occurrence matrix (GLCM) algorithm to obtain an optimized set of feature parameters as the morphological "fingerprints" for automated cell classification. We selected the Jurkat T cells and Ramos B cells to test the p-DIFC method's capacity for cell classification. After detailed statistical analysis, we found that the optimized feature vectors yield accuracies of classification between the Jurkat and Ramos ranging from 97.8% to 100% among different cell data sets. Confocal imaging and three-dimensional reconstruction were applied to gain insights on the ability of p-DIFC method for classifying the two cell lines of highly similar morphology. Based on these results we conclude that the p-DIFC method has the capacity to discriminate cells of high similarity in their morphology with "fingerprints" features extracted from the diffraction images, which may be attributed to subtle but statistically significant differences in the nucleus-to-cell volume ratio in the case of Jurkat and Ramos cells.
Collapse
Affiliation(s)
- Yuanming Feng
- Department of Biomedical Engineering, Tianjin University, Tianjin, 300072, China
| | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Comber JD, Philip R. MHC class I antigen presentation and implications for developing a new generation of therapeutic vaccines. THERAPEUTIC ADVANCES IN VACCINES 2014; 2:77-89. [PMID: 24790732 DOI: 10.1177/2051013614525375] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Major histocompatibility complex class I (MHC-I) presented peptide epitopes provide a 'window' into the changes occurring in a cell. Conventionally, these peptides are generated by proteolysis of endogenously synthesized proteins in the cytosol, loaded onto MHC-I molecules, and presented on the cell surface for surveillance by CD8(+) T cells. MHC-I restricted processing and presentation alerts the immune system to any infectious or tumorigenic processes unfolding intracellularly and provides potential targets for a cytotoxic T cell response. Therefore, therapeutic vaccines based on MHC-I presented peptide epitopes could, theoretically, induce CD8(+) T cell responses that have tangible clinical impacts on tumor eradication and patient survival. Three major methods have been used to identify MHC-I restricted epitopes for inclusion in peptide-based vaccines for cancer: genetic, motif prediction and, more recently, immunoproteomic analysis. Although the first two methods are capable of identifying T cell stimulatory epitopes, these have significant disadvantages and may not accurately represent epitopes presented by a tumor cell. In contrast, immunoproteomic methods can overcome these disadvantages and identify naturally processed and presented tumor associated epitopes that induce more clinically relevant tumor specific cytotoxic T cell responses. In this review, we discuss the importance of using the naturally presented MHC-I peptide repertoire in formulating peptide vaccines, the recent application of peptide-based vaccines in a variety of cancers, and highlight the pros and cons of the current state of peptide vaccines.
Collapse
Affiliation(s)
| | - Ramila Philip
- Immunotope, Inc., Pennsylvania Biotechnology Center, 3805 Old Easton Road, Doylestown, PA 18902, USA
| |
Collapse
|
100
|
Moon EK, Wang LC, Dolfi DV, Wilson CB, Ranganathan R, Sun J, Kapoor V, Scholler J, Puré E, Milone MC, June CH, Riley JL, Wherry EJ, Albelda SM. Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumors. Clin Cancer Res 2014; 20:4262-73. [PMID: 24919573 DOI: 10.1158/1078-0432.ccr-13-2627] [Citation(s) in RCA: 336] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE Immunotherapy using vaccines or adoptively transferred tumor-infiltrating lymphocytes (TIL) is limited by T-cell functional inactivation within the solid tumor microenvironment. The purpose of this study was to determine whether a similar tumor-induced inhibition occurred with genetically modified cytotoxic T cells expressing chimeric antigen receptors (CAR) targeting tumor-associated antigens. EXPERIMENTAL DESIGN Human T cells expressing CAR targeting mesothelin or fibroblast activation protein and containing CD3ζ and 4-1BB cytoplasmic domains were intravenously injected into immunodeficient mice bearing large, established human mesothelin-expressing flank tumors. CAR TILs were isolated from tumors at various time points and evaluated for effector functions and status of inhibitory pathways. RESULTS CAR T cells were able to traffic into tumors with varying efficiency and proliferate. They were able to slow tumor growth, but did not cause regressions or cures. The CAR TILs underwent rapid loss of functional activity that limited their therapeutic efficacy. This hypofunction was reversible when the T cells were isolated away from the tumor. The cause of the hypofunction seemed to be multifactorial and was associated with upregulation of intrinsic T-cell inhibitory enzymes (diacylglycerol kinase and SHP-1) and the expression of surface inhibitory receptors (PD1, LAG3, TIM3, and 2B4). CONCLUSIONS Advanced-generation human CAR T cells are reversibly inactivated within the solid tumor microenvironment of some tumors by multiple mechanisms. The model described here will be an important tool for testing T cell-based strategies or systemic approaches to overcome this tumor-induced inhibition. Our results suggest that PD1 pathway antagonism may augment human CAR T-cell function.
Collapse
MESH Headings
- Animals
- BALB 3T3 Cells
- Cytotoxicity, Immunologic/immunology
- Endopeptidases
- Female
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/immunology
- GPI-Linked Proteins/metabolism
- Gelatinases/genetics
- Gelatinases/immunology
- Gelatinases/metabolism
- Humans
- Immunotherapy, Adoptive
- Lymphocytes, Tumor-Infiltrating/immunology
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Mesothelin
- Mesothelioma/immunology
- Mesothelioma/metabolism
- Mesothelioma/therapy
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Serine Endopeptidases/genetics
- Serine Endopeptidases/immunology
- Serine Endopeptidases/metabolism
- Signal Transduction
- T-Lymphocytes/immunology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Edmund K Moon
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine,
| | - Liang-Chuan Wang
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | | | - Caleph B Wilson
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine; and
| | | | - Jing Sun
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - Veena Kapoor
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - John Scholler
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine; and
| | - Ellen Puré
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael C Milone
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine; and
| | - Carl H June
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine; and
| | - James L Riley
- Department of Microbiology and Institute for Immunology
| | - E John Wherry
- Department of Microbiology and Institute for Immunology
| | - Steven M Albelda
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| |
Collapse
|