1001
|
|
1002
|
Alspach E, Lussier DM, Miceli AP, Kizhvatov I, DuPage M, Luoma AM, Meng W, Lichti CF, Esaulova E, Vomund AN, Runci D, Ward JP, Gubin MM, Medrano RFV, Arthur CD, White JM, Sheehan KCF, Chen A, Wucherpfennig KW, Jacks T, Unanue ER, Artyomov MN, Schreiber RD. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature 2019; 574:696-701. [PMID: 31645760 PMCID: PMC6858572 DOI: 10.1038/s41586-019-1671-8] [Citation(s) in RCA: 587] [Impact Index Per Article: 97.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 09/12/2019] [Indexed: 12/11/2022]
Abstract
The ability of the immune system to eliminate and shape the immunogenicity of tumours defines the process of cancer immunoediting1. Immunotherapies such as those that target immune checkpoint molecules can be used to augment immune-mediated elimination of tumours and have resulted in durable responses in patients with cancer that did not respond to previous treatments. However, only a subset of patients benefit from immunotherapy and more knowledge about what is required for successful treatment is needed2-4. Although the role of tumour neoantigen-specific CD8+ T cells in tumour rejection is well established5-9, the roles of other subsets of T cells have received less attention. Here we show that spontaneous and immunotherapy-induced anti-tumour responses require the activity of both tumour-antigen-specific CD8+ and CD4+ T cells, even in tumours that do not express major histocompatibility complex (MHC) class II molecules. In addition, the expression of MHC class II-restricted antigens by tumour cells is required at the site of successful rejection, indicating that activation of CD4+ T cells must also occur in the tumour microenvironment. These findings suggest that MHC class II-restricted neoantigens have a key function in the anti-tumour response that is nonoverlapping with that of MHC class I-restricted neoantigens and therefore needs to be considered when identifying patients who will most benefit from immunotherapy.
Collapse
Affiliation(s)
- Elise Alspach
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.,The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Danielle M Lussier
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.,The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Alexander P Miceli
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.,The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Ilya Kizhvatov
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Michel DuPage
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, USA
| | - Adrienne M Luoma
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Wei Meng
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.,The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Cheryl F Lichti
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.,The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Ekaterina Esaulova
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Anthony N Vomund
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Daniele Runci
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.,The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Jeffrey P Ward
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.,The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA.,Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Matthew M Gubin
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.,The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Ruan F V Medrano
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.,The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Cora D Arthur
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.,The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - J Michael White
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Kathleen C F Sheehan
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA.,The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA
| | - Alex Chen
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Tyler Jacks
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Emil R Unanue
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Robert D Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA. .,The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, MO, USA. .,The Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
| |
Collapse
|
1003
|
Wang RJ, Liu W, Mu XY, Yao ZX, Wu K, Zheng Z, Jiang JT, Tan MY, Sun F, Fan J, Wang X, Zheng JH, He YY, Liu ZH. Preoperative CD4 +CD25 +/CD4 + and tumor diameter predict prognosis in male patients with bladder cancer. Biomark Med 2019; 13:1387-1397. [PMID: 31631674 DOI: 10.2217/bmm-2018-0481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: The value of the peripheral blood lymphocyte subpopulation ratios and tumor diameter for prognosis in bladder cancer (BC) patients needs to be explored. Materials & methods: A total of 161 male BC patients and 68 male normal controls were retrospectively reviewed. The value of combining predictor consisted of both CD4+CD25+/CD4+ and computed tomography urography tumor diameter (CTU-D) on stage, overall survival (OS) and recurrence probability was analyzed by logistic regression, Kaplan-Meier method and log-rank test. Results: The combining predictor was a statistically independent risk for stage; dramatic differences in OS and recurrence probability were found between the combining predictor-high (cut-off point >0.08) and combining predictor-low groups (cut-off point ≤0.08). Conclusion: The combining predictor could be a significant predictor for advanced stage, OS and recurrence probability in male patients with BC.
Collapse
Affiliation(s)
- Ren-Jie Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Wei Liu
- General Practice Team, Shanghai Weifang Community Health Service Center, Shanghai 200122, PR China
| | - Xing-Yu Mu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Zhi-Xian Yao
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Ke Wu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Zhong Zheng
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Jun-Tao Jiang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Ming-Yue Tan
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Feng Sun
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Jie Fan
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Xiang Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Jun-Hua Zheng
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Yin-Yan He
- Department of Obstetrics & Gynecology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| | - Zhi-Hong Liu
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, PR China
| |
Collapse
|
1004
|
Kristensen LK, Fröhlich C, Christensen C, Melander MC, Poulsen TT, Galler GR, Lantto J, Horak ID, Kragh M, Nielsen CH, Kjaer A. CD4 + and CD8a + PET imaging predicts response to novel PD-1 checkpoint inhibitor: studies of Sym021 in syngeneic mouse cancer models. Theranostics 2019; 9:8221-8238. [PMID: 31754392 PMCID: PMC6857046 DOI: 10.7150/thno.37513] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/09/2019] [Indexed: 12/17/2022] Open
Abstract
Predicting the outcome of immunotherapy is essential for efficient treatment. The recent clinical success of immunotherapy is increasingly changing the paradigm of cancer treatment. Accordingly, the development of immune-based agents is accelerating and the number of agents in the global immuno-oncology pipeline has grown 60-70% over the past year. However, despite remarkable clinical efficacy in some patients, only few achieve a lasting clinical response. Treatment failure can be attributed to poorly immunogenic tumors that do not attract tumor infiltrating lymphocytes (TILs). Therefore, we developed positron emission tomography (PET) radiotracers for non-invasive detection of CD4+ and CD8a+ TILs in syngeneic mouse tumor models for preclinical studies. Methods: Seven syngeneic mouse tumor models (B16F10, P815, CT26, MC38, Renca, 4T1, Sa1N) were quantified for CD4+ and CD8a+ TILs using flow cytometry and immunohistochemistry (IHC), as well as for tumor growth response to Sym021, a humanized PD-1 antibody cross-reactive with mouse PD-1. Radiotracers were generated from F(ab)'2 fragments of rat-anti-mouse CD4 and CD8a antibodies conjugated to the p-SCN-Bn-Desferrioxamine (SCN-Bn-DFO) chelator and radiolabeled with Zirconium-89 (89Zr-DFO-CD4/89Zr-DFO-CD8a). Tracers were optimized for in vivo PET/CT imaging in CT26 tumor-bearing mice and specificity was evaluated by depletion studies and isotype control imaging. 89Zr-DFO-CD4 and 89Zr-DFO-CD8a PET/CT imaging was conducted in the panel of syngeneic mouse models prior to immunotherapy with Sym021. Results: Syngeneic tumor models were characterized as "hot" or "cold" according to number of TILs determined by flow cytometry and IHC. 89Zr-DFO-CD4 and 89Zr-DFO-CD8a were successfully generated with a radiochemical purity >99% and immunoreactivity >85%. The optimal imaging time-point was 24 hours post-injection of ~1 MBq tracer with 30 µg non-labeled co-dose. Reduced tumor and spleen uptake of 89Zr-DFO-CD8a was observed in CD8a+ depleted mice and the uptake was comparable with that of isotype control (89Zr-DFO-IgG2b) confirming specificity. PET imaging in syngeneic tumor models revealed a varying maximum tumor-to-heart ratio of 89Zr-DFO-CD4 and 89Zr-DFO-CD8a across tumor types and in-between subjects that correlated with individual response to Sym021 at day 10 relative to start of therapy (p=0.0002 and p=0.0354, respectively). The maximum 89Zr-DFO-CD4 tumor-to-heart ratio could be used to stratify mice according to Sym021 therapy response and overall survival was improved in mice with a 89Zr-DFO-CD4 ratio >9 (p=0.0018). Conclusion: We developed 89Zr-DFO-CD4 and 89Zr-DFO-CD8a PET radiotracers for specific detection and whole-body assessment of CD4+ and CD8a+ status. These radiotracers can be used to phenotype preclinical syngeneic mouse tumor models and to predict response to an immune checkpoint inhibitor. We foresee development of such non-invasive in vivo biomarkers for prediction and evaluation of clinical efficacy of immunotherapeutic agents, such as Sym021.
Collapse
Affiliation(s)
- Lotte K. Kristensen
- Minerva Imaging, Copenhagen, Denmark
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Dept. of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Denmark
| | | | - Camilla Christensen
- Minerva Imaging, Copenhagen, Denmark
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Dept. of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Denmark
| | | | | | | | | | | | | | - Carsten H. Nielsen
- Minerva Imaging, Copenhagen, Denmark
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Dept. of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Denmark
| | - Andreas Kjaer
- Dept. of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Dept. of Biomedical Sciences, Rigshospitalet and University of Copenhagen, Denmark
| |
Collapse
|
1005
|
Liao HW, Garris C, Pfirschke C, Rickelt S, Arlauckas S, Siwicki M, Kohler RH, Weissleder R, Sundvold-Gjerstad V, Sveinbjørnsson B, Rekdal Ø, Pittet MJ. LTX-315 sequentially promotes lymphocyte-independent and lymphocyte-dependent antitumor effects. Cell Stress 2019; 3:348-360. [PMID: 31799501 PMCID: PMC6859426 DOI: 10.15698/cst2019.11.204] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/25/2019] [Accepted: 09/30/2019] [Indexed: 12/16/2022] Open
Abstract
LTX-315 is an oncolytic peptide that has antitumor efficacy in mice grafted with various tumor cell lines and is currently being tested in phase II clinical trials. Here we aimed to further evaluate LTX-315 in conditional genetic mouse models of cancer that typically resist current treatment options and to better understand the drug's mode of action in vivo. We report LTX-315 mediates profound antitumor effects against Braf- and Pten-driven melanoma and delays the progression of Kras- and P53-driven soft tissue sarcoma in mice. Additionally, we show in melanoma that LTX-315 triggers two sequential phases of antitumor response. The first phase of response, which begins within minutes of drug delivery into tumors, is defined by disrupted tumor vasculature and decreased tumor burden and occurs independently of lymphocytes. The second phase of response, which continues over weeks, is defined by long-term alteration of the tumor microenvironment; the changes induced by LTX-315 are most notably characterized by CD8+ T cell infiltration. We further show that these CD8+ T cells are involved in suppressing melanoma outgrowth in mice and report similar CD8+ T cell infiltration following LTX-315 treatment in melanoma and sarcoma patients. Taken together, these findings reveal LTX-315's multiple antitumor effects, including disrupting the tumor vasculature and promoting the conversion of poorly immunogenic tumors into ones that display antitumor T cell immunity.
Collapse
Affiliation(s)
- Hsin-Wei Liao
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA, USA
- These authors contributed equally
| | - Christopher Garris
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA, USA
- These authors contributed equally
| | - Christina Pfirschke
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA, USA
| | - Steffen Rickelt
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sean Arlauckas
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA, USA
| | - Marie Siwicki
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA, USA
| | - Rainer H. Kohler
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA, USA
| | | | - Baldur Sveinbjørnsson
- Lytix Biopharma, Oslo, Norway
- Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Øystein Rekdal
- Lytix Biopharma, Oslo, Norway
- Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Mikael J. Pittet
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
1006
|
Bianco A, Perrotta F, Barra G, Malapelle U, Rocco D, De Palma R. Prognostic Factors and Biomarkers of Responses to Immune Checkpoint Inhibitors in Lung Cancer. Int J Mol Sci 2019; 20:E4931. [PMID: 31590386 PMCID: PMC6801651 DOI: 10.3390/ijms20194931] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/28/2019] [Accepted: 10/01/2019] [Indexed: 12/21/2022] Open
Abstract
Manipulation of the immune response is a game changer in lung cancer treatment, revolutionizing management. PD1 and CTLA4 are dynamically expressed on different T cell subsets that can either disrupt or sustain tumor growth. Monoclonal antibodies (MoAbs) against PD1/PDL1 and CTLA4 have shown that inhibitory signals can be impaired, blocking T cell activation and function. MoAbs, used as both single-agents or in combination with standard therapy for the treatment of advanced non-small cell lung cancer (NSCLC), have exhibited advantages in terms of overall survival and response rate; nivolumab, pembrolizumab, atezolizumab and more recently, durvalumab, have already been approved for lung cancer treatment and more compounds are in the pipeline. A better understanding of signaling elicited by these antibodies on T cell subsets, as well as identification of biological determinants of sensitivity, resistance and correlates of efficacy, will help to define the mechanisms of antitumor responses. In addition, the relevance of T regulatory cells (Treg) involved in immune responses in cancer is attracting increasing interest. A major challenge for future research is to understand why a durable response to immune checkpoint inhibitors (ICIs) occurs only in subsets of patients and the mechanisms of resistance after an initial response. This review will explore current understanding and future direction of research on ICI treatment in lung cancer and the impact of tumor immune microenvironment n influencing clinical responses.
Collapse
Affiliation(s)
- Andrea Bianco
- Department of Translational Medical Sciences, University of Campania "L Vanvitelli", 80131 Naples, Italy.
- Department of Pneumology and Oncology, A.O. dei Colli, Hosp. V Monaldi, 80131 Naples, Italy.
| | - Fabio Perrotta
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, 86100 Campobasso, Italy.
| | - Giusi Barra
- Department of Precision Medicine, University of Campania "L Vanvitelli", 80131 Naples, Italy.
| | - Umberto Malapelle
- Department of Public Health, University of Naples "Federico II", 80131 Naples, Italy.
| | - Danilo Rocco
- Department of Pneumology and Oncology, A.O. dei Colli, Hosp. V Monaldi, 80131 Naples, Italy.
| | - Raffaele De Palma
- Department of Precision Medicine, University of Campania "L Vanvitelli", 80131 Naples, Italy.
| |
Collapse
|
1007
|
Koutsoumpli G, Ip PP, Schepel I, Hoogeboom BN, Boerma A, Daemen T. Alphavirus-based hepatitis C virus therapeutic vaccines: can universal helper epitopes enhance HCV-specific cytotoxic T lymphocyte responses? Ther Adv Vaccines Immunother 2019; 7:2515135519874677. [PMID: 31620673 PMCID: PMC6777054 DOI: 10.1177/2515135519874677] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 07/31/2019] [Indexed: 12/28/2022] Open
Abstract
Background: Antigen-specific T cell immune responses play a pivotal role in resolving
acute and chronic hepatitis C virus (HCV) infections. Currently, no
prophylactic or therapeutic vaccines against HCV are available. We
previously demonstrated the preclinical potency of therapeutic HCV vaccines
based on recombinant Semliki Forest virus (SFV) replicon particles. However,
clinical trials do not always meet the high expectations of preclinical
studies, thus, optimization of vaccine strategies is crucial. In efforts to
further increase the frequency of HCV-specific immune responses in the
candidate SFV-based vaccines, the authors assessed whether inclusion of
three strong, so-called universal helper T cell epitopes, and an endoplasmic
reticulum localization, and retention signal (collectively termed
sigHELP-KDEL cassette) could enhance HCV-specific immune responses. Methods: We included the sigHELP-KDEL cassette in two of the candidate SFV-based HCV
vaccines, targeting NS3/4A and NS5A/B proteins. We characterized the new
constructs in vitro for the expression and stability of the
transgene-encoded proteins. Their immune efficacy with respect to
HCV-specific immune responses in vivo was compared with the
parental SFV vaccine expressing the corresponding HCV antigen. Further
characterization of the functionality of the HCV-specific CD8+ T
cells was assessed by surface and intracellular cytokine staining and flow
cytometry analysis. Results: Moderate, but significantly, enhanced frequencies of antigen-specific immune
responses were achieved upon lower/suboptimal dosage immunization. In
optimal dosage immunization, the inclusion of the cassette did not further
increase the frequencies of HCV-specific CD8+ T cells when
compared with the parental vaccines and the frequencies of effector and
memory populations were identical. Conclusion: We hypothesize that the additional effect of the sigHELP-KDEL cassette in
SFV-based vaccines depends on the immunogenicity, nature, and stability of
the target antigen expressed by the vaccine.
Collapse
Affiliation(s)
- Georgia Koutsoumpli
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Peng Peng Ip
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Ilona Schepel
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Baukje Nynke Hoogeboom
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Annemarie Boerma
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Toos Daemen
- Department of Medical Microbiology, Tumor Virology and Cancer Immunotherapy, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, Groningen, 9713 AV, the Netherlands
| |
Collapse
|
1008
|
Proliferation-competent Tcf1+ CD8 T cells in dysfunctional populations are CD4 T cell help independent. Proc Natl Acad Sci U S A 2019; 116:20070-20076. [PMID: 31530725 PMCID: PMC6778176 DOI: 10.1073/pnas.1902701116] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
T cell maintenance in chronic infection and cancer follows a hierarchical order. Short-lived effector CD8 T cells are constitutively replaced from a proliferation-competent Tcf1-expressing progenitor population. This occurs spontaneously at low levels and increases in magnitude upon blocking PD-1 signaling. We explore how CD4 T cell help controls transition and survival of the progenitors and their progeny by utilizing single-cell RNA sequencing. Unexpectedly, absence of CD4 help caused reductions in cell numbers only among terminally differentiated cells while proliferation-competent progenitor cells remained unaffected with regard to their numbers and their overall phenotype. In fact, upon restoration of a functional CD4 compartment, the progenitors began to regenerate the effector CD8 T cells. Thus, unlike memory T cells for which secondary expansion requires CD4 T cell help, this is not a necessity for proliferation-competent progenitor cells in dysfunctional populations. Our data therefore reveals that proliferation-competent cells in dysfunctional populations show a previously unrecognized uncoupling of CD4 T cell help that is otherwise required by conventional memory T cells.
Collapse
|
1009
|
Wu M, Li X, Zhang T, Liu Z, Zhao Y. Identification of a Nine-Gene Signature and Establishment of a Prognostic Nomogram Predicting Overall Survival of Pancreatic Cancer. Front Oncol 2019; 9:996. [PMID: 31612115 PMCID: PMC6776930 DOI: 10.3389/fonc.2019.00996] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Accepted: 09/17/2019] [Indexed: 12/29/2022] Open
Abstract
Background: Pancreatic cancer is highly lethal and aggressive with increasing trend of mortality in both genders. An effective prediction model is needed to assess prognosis of patients for optimization of treatment. Materials and Methods: Seven datasets of mRNA expression and clinical data were obtained from gene expression omnibus (GEO) database. Level 3 mRNA expression and clinicopathological data were obtained from The Cancer Genome Atlas pancreatic ductal adenocarcinoma (TCGA-PAAD) dataset. Differentially expressed genes (DEGs) between pancreatic tumor and normal tissue were identified by integrated analysis of multiple GEO datasets. Univariate and Lasso Cox regression analyses were applied to identify overall survival-related DEGs and establish a prognostic gene signature whose performance was evaluated by Kaplan-Meier curve, receiver operating characteristic (ROC), Harrell's concordance index (C-index) and calibration curve. GSE62452 and GSE57495 were used for external validation. Gene set enrichment analysis (GSEA) and tumor immunity analysis were applied to elucidate the molecular mechanisms and immune relevance. Multivariate Cox regression analysis was used to identify independent prognostic factors in pancreatic cancer. Finally, a prognostic nomogram was established based on the TCGA PAAD dataset. Results: A nine-gene signature comprising MET, KLK10, COL17A1, CEP55, ANKRD22, ITGB6, ARNTL2, MCOLN3, and SLC25A45 was established to predict overall survival of pancreatic cancer. The ROC curve and C-index indicated good performance of the nine-gene signature at predicting overall survival in the TCGA dataset and external validation datasets relative to classic AJCC staging. The nine-gene signature could classify patients into high- and low-risk groups with distinct overall survival and differentiate tumor from normal tissue. Univariate Cox regression revealed that the nine-gene signature was an independent prognostic factor in pancreatic cancer. The nomogram incorporating the gene signature and clinical prognostic factors was superior to AJCC staging in predicting overall survival. The high-risk group was enriched with multiple oncological signatures and aggressiveness-related pathways and associated with significantly lower levels of CD4+ T cell infiltration. Conclusion: Our study identified a nine-gene signature and established a prognostic nomogram that reliably predict overall survival in pancreatic cancer. The findings may be beneficial to therapeutic customization and medical decision-making.
Collapse
Affiliation(s)
- Mengwei Wu
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobin Li
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Taiping Zhang
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Ziwen Liu
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yupei Zhao
- Department of General Surgery, Peking Union Medical College, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
1010
|
Wolf Y, Bartok O, Patkar S, Eli GB, Cohen S, Litchfield K, Levy R, Jiménez-Sánchez A, Trabish S, Lee JS, Karathia H, Barnea E, Day CP, Cinnamon E, Stein I, Solomon A, Bitton L, Pérez-Guijarro E, Dubovik T, Shen-Orr SS, Miller ML, Merlino G, Levin Y, Pikarsky E, Eisenbach L, Admon A, Swanton C, Ruppin E, Samuels Y. UVB-Induced Tumor Heterogeneity Diminishes Immune Response in Melanoma. Cell 2019; 179:219-235.e21. [PMID: 31522890 PMCID: PMC6863386 DOI: 10.1016/j.cell.2019.08.032] [Citation(s) in RCA: 258] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 05/30/2019] [Accepted: 08/15/2019] [Indexed: 12/12/2022]
Abstract
Although clonal neo-antigen burden is associated with improved response to immune therapy, the functional basis for this remains unclear. Here we study this question in a novel controlled mouse melanoma model that enables us to explore the effects of intra-tumor heterogeneity (ITH) on tumor aggressiveness and immunity independent of tumor mutational burden. Induction of UVB-derived mutations yields highly aggressive tumors with decreased anti-tumor activity. However, single-cell-derived tumors with reduced ITH are swiftly rejected. Their rejection is accompanied by increased T cell reactivity and a less suppressive microenvironment. Using phylogenetic analyses and mixing experiments of single-cell clones, we dissect two characteristics of ITH: the number of clones forming the tumor and their clonal diversity. Our analysis of melanoma patient tumor data recapitulates our results in terms of overall survival and response to immune checkpoint therapy. These findings highlight the importance of clonal mutations in robust immune surveillance and the need to quantify patient ITH to determine the response to checkpoint blockade.
Collapse
Affiliation(s)
- Yochai Wolf
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Osnat Bartok
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sushant Patkar
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Gitit Bar Eli
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Sapir Cohen
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Kevin Litchfield
- Cancer Evolution and Genome Instability Laboratory, Francis Crick Institute, London NW1 1AT, UK; Cancer Research UK Lung Cancer Centre of Excellence London, University College London Cancer Institute, London WC1E 6DD, UK
| | - Ronen Levy
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Alejandro Jiménez-Sánchez
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Sophie Trabish
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Joo Sang Lee
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Hiren Karathia
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Eilon Barnea
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Einat Cinnamon
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Ilan Stein
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Adam Solomon
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Lital Bitton
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Eva Pérez-Guijarro
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Tania Dubovik
- Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Shai S Shen-Orr
- Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | - Martin L Miller
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge CB2 0RE, UK
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Yishai Levin
- The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Eli Pikarsky
- The Lautenberg Center for Immunology and Cancer Research, IMRIC, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Lea Eisenbach
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Arie Admon
- Faculty of Biology, Technion - Israel Institute of Technology, Haifa, Israel
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, Francis Crick Institute, London NW1 1AT, UK; Cancer Research UK Lung Cancer Centre of Excellence London, University College London Cancer Institute, London WC1E 6DD, UK; Department of Medical Oncology, University College London Hospitals, London NW1 2BU, UK
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
1011
|
Follicular CD4 T Cells Tutor CD8 Early Memory Precursors: An Initiatory Journey to the Frontier of B Cell Territory. iScience 2019; 20:100-109. [PMID: 31563850 PMCID: PMC6817688 DOI: 10.1016/j.isci.2019.09.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 08/02/2019] [Accepted: 09/11/2019] [Indexed: 01/12/2023] Open
Abstract
The early events of CD8 memory generation remain largely unknown. Here we report that as early as 2 days after antigen priming, very early memory precursors can be identified by their expression of the chemokine receptor CXCR5. These early precursors, which have an effector phenotype, expand and temporarily migrate to the T-B cell zone junction where they interact with follicular CD4+ T cells (Tfh). Remarkably, this interaction with Tfh, hitherto considered as exclusive B cell helpers, is required for CD8 memory precursors to become highly competent memory cells. CD40 and interleukin-21 signaling are involved in the help provided to CXCR5+CD8 memory precursors. This study thus unveils critical early steps in the generation of CD8 memory, identifies CXCR5 as the earliest known marker of CD8 memory precursors, suggests a major helper role for Tfh, and points to possible coordination between the pathways of CD8 and B cell memory generation at the T-B-cell zone junction.
Collapse
|
1012
|
The impact of endoplasmic reticulum stress responses in dendritic cell immunobiology. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:153-176. [PMID: 31759430 DOI: 10.1016/bs.ircmb.2019.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) are critical for bridging innate and adaptive immunity. They do so by presenting antigens to T cells, and by expressing diverse molecules that further promote T cell activation, differentiation and memory formation. During this process, intracellular and extracellular factors can perturb the protein-folding capacity of endoplasmic reticulum (ER) and induce a cellular state of "ER stress," which is controlled and resolved by the unfolded protein response (UPR). Interestingly, various studies have shown that DCs can activate UPR-related pathways even in the absence of global ER stress, and that this process can modulate their normal activity. In other settings, such as cancer, adverse microenvironmental conditions have been demonstrated to evoke severe ER stress and persistent activation of the UPR in tumor-infiltrating DCs. This process disrupts their metabolism and local antigen-presenting capacity, hence impeding the initiation and maintenance of anti-cancer immunity. Here, we review recent findings on how canonical and non-canonical UPR activation impacts DC immunobiology at the steady-state, upon activation via pattern recognition receptors, and under diverse pathological conditions. We also discuss the potential therapeutic implications that targeting the UPR in DCs may have in the context of cancer and in other pathologies such as graft-versus-host disease.
Collapse
|
1013
|
Tondini E, Arakelian T, Oosterhuis K, Camps M, van Duikeren S, Han W, Arens R, Zondag G, van Bergen J, Ossendorp F. A poly-neoantigen DNA vaccine synergizes with PD-1 blockade to induce T cell-mediated tumor control. Oncoimmunology 2019; 8:1652539. [PMID: 31646082 PMCID: PMC6791440 DOI: 10.1080/2162402x.2019.1652539] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 12/20/2022] Open
Abstract
The combination of immune-stimulating strategies has the potency to improve immunotherapy of cancer. Vaccination against neoepitopes derived from patient tumor material can generate tumor-specific T cell immunity, which could reinforce the efficacy of checkpoint inhibitor therapies such as anti-PD-1 treatment. DNA vaccination is a versatile platform that allows the inclusion of multiple neoantigen-coding sequences in a single formulation and therefore represents an ideal platform for neoantigen vaccination. We developed an anti-tumor vaccine based on a synthetic DNA vector designed to contain multiple cancer-specific epitopes in tandem. The DNA vector encoded a fusion gene consisting of three neoepitopes derived from the mouse colorectal tumor MC38 and their natural flanking sequences as 40 amino acid stretches. In addition, we incorporated as reporter epitopes the helper and CTL epitope sequences of ovalbumin. The poly-neoantigen DNA vaccine elicited T cell responses to all three neoantigens and induced functional CD8 and CD4 T cell responses to the reporter antigen ovalbumin after intradermal injection in mice. The DNA vaccine was effective in preventing outgrowth of B16 melanoma expressing ovalbumin in a prophylactic setting. Moreover, the combination of therapeutic DNA vaccination and anti-PD-1 treatment was synergistic in controlling MC38 tumor growth whereas individual treatments did not succeed. These data demonstrate the potential of DNA vaccination to target multiple neoepitopes in a single formulation and highlight the cooperation between vaccine-based and checkpoint blockade immunotherapies for the successful eradication of established tumors.
Collapse
Affiliation(s)
- Elena Tondini
- Dept. Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Tsolere Arakelian
- Dept. Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Marcel Camps
- Dept. Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne van Duikeren
- Dept. Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Wanda Han
- Immunetune BV, Leiden, The Netherlands
| | - Ramon Arens
- Dept. Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Ferry Ossendorp
- Dept. Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
1014
|
Fultang L, Panetti S, Ng M, Collins P, Graef S, Rizkalla N, Booth S, Lenton R, Noyvert B, Shannon-Lowe C, Middleton G, Mussai F, De Santo C. MDSC targeting with Gemtuzumab ozogamicin restores T cell immunity and immunotherapy against cancers. EBioMedicine 2019; 47:235-246. [PMID: 31462392 PMCID: PMC6796554 DOI: 10.1016/j.ebiom.2019.08.025] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 08/08/2019] [Accepted: 08/12/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Targeting of MDSCs is a major clinical challenge in the era of immunotherapy. Antibodies which deplete MDSCs in murine models can reactivate T cell responses. In humans such approaches have not developed due to difficulties in identifying targets amenable to clinical translation. METHODS RNA-sequencing of M-MDSCs and G-MDSCs from cancer patients was undertaken. Flow cytometry and immunohistochemistry of blood and tumours determined MDSC CD33 expression. MDSCs were treated with Gemtuzumab ozogamicin and internalisation kinetics, and cell death mechanisms determined by flow cytometry, confocal microscopy and electron microscopy. Effects on T cell proliferation and CAR-T cell anti-tumour cytotoxicity were identified in the presence of Gemtuzumab ozogamicin. FINDINGS RNA-sequencing of human M-MDSCs and G-MDSCs identified transcriptomic differences, but that CD33 is a common surface marker. Flow cytometry indicated CD33 expression is higher on M-MDSCs, and CD33+ MDSCs are found in the blood and tumours regardless of cancer subtype. Treatment of human MDSCs leads to Gemtuzumab ozogamicin internalisation, increased p-ATM, and cell death; restoring T cell proliferation. Anti-GD2-/mesothelin-/EGFRvIII-CAR-T cell activity is enhanced in combination with the anti-MDSC effects of Gemtuzumab ozogamicin. INTERPRETATION The study identifies that M-MDSCs and G-MDSCs are transcriptomically different but CD33 is a therapeutic target on peripheral and infiltrating MDSCs across cancer subtypes. The immunotoxin Gemtuzumab ozogamicin can deplete MDSCs providing a translational approach to reactivate T cell and CAR-T cell responses against multiple cancers. In the rare conditions of HLH/MAS gemtuzumab ozogamicin provides a novel anti-myeloid strategy. FUND: This work was supported by Cancer Research UK, CCLG, Treating Children with Cancer, and the alumni and donors to the University of Birmingham.
Collapse
Affiliation(s)
- Livingstone Fultang
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Silvia Panetti
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Margaret Ng
- Department of Anatomic Pathology, The Chinese University of Hong Kong, Hong Kong
| | - Paul Collins
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Suzanne Graef
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Nagy Rizkalla
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Sarah Booth
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Richard Lenton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Boris Noyvert
- CRUK Birmingham Centre and Centre for Computational Biology, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Claire Shannon-Lowe
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Gary Middleton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Francis Mussai
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| | - Carmela De Santo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| |
Collapse
|
1015
|
Galaine J, Turco C, Vauchy C, Royer B, Mercier-Letondal P, Queiroz L, Loyon R, Mouget V, Boidot R, Laheurte C, Lakkis Z, Jary M, Adotévi O, Borg C, Godet Y. CD4 T cells target colorectal cancer antigens upregulated by oxaliplatin. Int J Cancer 2019; 145:3112-3125. [PMID: 31396953 DOI: 10.1002/ijc.32620] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 06/07/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022]
Abstract
Immune checkpoint blockade has proven its efficacy in hypermutated subtypes of metastatic colorectal cancers (mCRC). Immunogenic potential can also be observed with conventional chemotherapies, but this property has never been explored thoroughly in CRC patients. The CRC therapeutic arsenal includes oxaliplatin, a well-characterized platinum drug already described as immunogenic. Here, we investigated the impact of the oxaliplatin-based treatment on mCRC immunopeptidome. We demonstrated that oxaliplatin-resistant CRC cell lines overexpressed telomerase reverse transcriptase (TERT), colorectal-associated-tumor antigen-1 (COA-1) and mesothelin tumor-associated antigens. We identified new HLA class-II-restricted and promiscuous peptides derived from COA-1 and mesothelin. The two naturally processed peptides COA-1331-345 and Meso366-380 appear to be the most immunogenic in mCRC patients. A prospective cohort of 162 mCRC patients enabled us to explore the impact of oxaliplatin exposure on the antitumor-specific immune response. Interestingly, chemotherapy-naive mCRC patients present high immune CD4 T-cell responses directed against TERT, COA-1 and mesothelin-derived peptides. These antitumor T-cell responses were maintained after 3 months of oxaliplatin-based treatment. Altogether, these findings highlight the interest of immunostimulatory agents to improve the management of chemoresistant mCRC patients. Finally, the high frequency of immune responses targeting the new immunogenic peptides derived from COA-1 and mesothelin support their use in immunomonitoring strategies.
Collapse
Affiliation(s)
- Jeanne Galaine
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Célia Turco
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Gastrointestinal Surgery, Besançon, France
| | - Charline Vauchy
- INSERM CIC-1431, University Hospital of Besançon, Clinical Investigation Center un Biotherapy, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France
| | - Bernard Royer
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of pharmacotoxicology, Besançon, France
| | - Patricia Mercier-Letondal
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Lise Queiroz
- INSERM CIC-1431, University Hospital of Besançon, Clinical Investigation Center un Biotherapy, Fédération Hospitalo-Universitaire INCREASE, LabEx LipSTIC, Besançon, France
| | - Romain Loyon
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Virginie Mouget
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Romain Boidot
- Centre Georges-François Leclerc, Platform for Transfer to Cancer Biology, Dijon, France
| | - Caroline Laheurte
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,EFS Bourgogne Franche-Comté, INSERM CIC-1431, CHRU Besançon, Plateforme de BioMonitoring, Besançon, France
| | - Zaher Lakkis
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Gastrointestinal Surgery, Besançon, France
| | - Marine Jary
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, Besançon, France
| | - Olivier Adotévi
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, Besançon, France
| | - Christophe Borg
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France.,University Hospital of Besançon, Department of Medical Oncology, Besançon, France
| | - Yann Godet
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| |
Collapse
|
1016
|
Bogen B, Fauskanger M, Haabeth OA, Tveita A. CD4 + T cells indirectly kill tumor cells via induction of cytotoxic macrophages in mouse models. Cancer Immunol Immunother 2019; 68:1865-1873. [PMID: 31448380 DOI: 10.1007/s00262-019-02374-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 08/01/2019] [Indexed: 02/04/2023]
Abstract
It is well recognized that CD4+ T cells may play an important role in immunosurveillance and immunotherapy against cancer. However, the details of how these cells recognize and eliminate the tumor cells remain incompletely understood. For the past 25 years, we have focused on how CD4+ T cells reject multiple myeloma cells in a murine model (MOPC315). In our experimental system, the secreted tumor-specific antigen is taken up by tumor-infiltrating macrophages that process it and present a neoepitope [a V region-derived idiotypic (Id) peptide] on MHC class II molecules to Th1 cells. Stimulated Th1 cells produce IFNγ, which activates macrophages in a manner that elicits an M1-like, tumoricidal phenotype. Through an inducible nitric oxide synthetase (iNOS)-dependent mechanism, the M1 macrophages secrete nitric oxide (NO) that diffuses into neighboring tumor cells. Inside the tumor cells, NO-derived reactive nitrogen species, including peroxynitrite, causes nitrosylation of proteins and triggers apoptosis by the intrinsic apoptotic pathway. This mode of indirect tumor recognition by CD4+ T cells operates independently of MHC class II expression on cancer cells. However, secretion of the tumor-specific antigen, and uptake and MHCII presentation on macrophages, is required for rejection. Similar mechanisms can also be observed in a B-lymphoma model and in the unrelated B16 melanoma model. Our findings reveal a novel mechanism by which CD4+ T cells kill tumor cells indirectly via induction of intratumoral cytotoxic macrophages. The data suggest that induction of M1 polarization of tumor-infiltrating macrophages, by CD4+ T cells or through other means, could serve as an immunotherapeutic strategy.
Collapse
Affiliation(s)
- Bjarne Bogen
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
- Department of Immunology, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424, Oslo, Norway.
- KG Jebsen Centre for Influenza Vaccine Research, Oslo, Norway.
| | - Marte Fauskanger
- Department of Immunology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ole Audun Haabeth
- Department of Immunology, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424, Oslo, Norway
| | - Anders Tveita
- Department of Immunology, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424, Oslo, Norway
| |
Collapse
|
1017
|
Bai Y, Wang Y, Zhang X, Fu J, Xing X, Wang C, Gao L, Liu Y, Shi L. Potential applications of nanoparticles for tumor microenvironment remodeling to ameliorate cancer immunotherapy. Int J Pharm 2019; 570:118636. [PMID: 31446027 DOI: 10.1016/j.ijpharm.2019.118636] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 08/17/2019] [Accepted: 08/19/2019] [Indexed: 02/07/2023]
Abstract
In recent years, researchers have made significant innovations in the field of tumor immunotherapy based on the knowledge of biology, oncology, and immunology. Tumor immunotherapy involves the use of immune checkpoint inhibitors and CAR (chimeric antigen receptor)-T cell therapy. As compared with conventional chemotherapy, immunotherapy is a potential approach to induce a more powerful immune response against tumor in the patient suffering from the advanced stage malignancy. Regardless of the developments made, a large number of clinical studies have confirmed that a substantial number of cancer patients still demonstrate non-responsiveness to immunotherapy, mainly due to the immunomodulating interactions of tumor cells with the immunosuppressive tumor microenvironment (iTME). It leads to immune tolerance of tumors and influences the efficacy of immunotherapy. This immune failure could be attributed to a complex immunosuppressive network comprising stromal and inflammatory cells, vessel system, ECM (extracellular matrix) and the cytokines released in tumor microenvironment (TME). The antitumor immune activity can be enhanced at different stages of tumor development by selective suppression of inhibitory pathways in the TME. This specific task can be achieved by using nano-sized drug delivery tools which are specific in their action and biocompatible in nature. Several recent studies have described the use of nanoparticles for iTME remodeling through the specific elimination of immunosuppressive cells, obstructing immune checkpoints, promotion of inflammatory cytokines, and amending the regulatory cells of the immune system. The efficacy of current immunotherapy can be improved by nanoparticle-mediated remodeling of iTME.
Collapse
Affiliation(s)
- Yuzhuo Bai
- Extrathoracic and Thyroid Mammary Surgery, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yun Wang
- Extrathoracic and Thyroid Mammary Surgery, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Xudong Zhang
- Encephalopathy Center, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Jianhua Fu
- Reproductive Center, Jilin Provincial People's Hospital, Changchun 130021, China
| | - Xiuli Xing
- Physical Examination Center, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Chunlan Wang
- Respiratory Department, First Clinical College, Academy of Traditional Chinese Medicine, Changchun 130021, China
| | - Longlan Gao
- Brain Surgery, Liaoyuan Hospital of Traditional Chinese Medicine, Liaoyuan 136000, China
| | - Yu Liu
- Brain Surgery, Liaoyuan Hospital of Traditional Chinese Medicine, Liaoyuan 136000, China
| | - Li Shi
- Department of Oncology and Hematology, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China.
| |
Collapse
|
1018
|
Gold Nanoparticle-Mediated Photoporation Enables Delivery of Macromolecules over a Wide Range of Molecular Weights in Human CD4+ T Cells. CRYSTALS 2019. [DOI: 10.3390/cryst9080411] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The modification of CD4+ T cells with exogenous nucleic acids or proteins is a critical step in several research and therapeutic applications, such as HIV studies and cancer immunotherapies. However, efficient cell transfections are not always easily achieved when working with these primary hard-to-transfect cells. While the modification of T cells is typically performed by viral transduction or electroporation, their use is associated with safety issues or cytotoxicity. Vapor nanobubble (VNB) photoporation with sensitizing gold nanoparticles (AuNPs) has recently emerged as a new technology for safe and flexible cell transfections. In this work, we evaluated the potential of VNB photoporation as a novel technique for the intracellular delivery of macromolecules in primary human CD4+ T cells using fluorescent dextrans as model molecules. Our results show that VNB photoporation enables efficient delivery of fluorescent dextrans of 10 kDa in Jurkat (>60% FD10+ cells) as well as in primary human CD4+ T cells (±40% FD10+ cells), with limited cell toxicity (>70% cell viability). We also demonstrated that the technique allows the delivery of dextrans that are up to 500 kDa in Jurkat cells, suggesting its applicability for the delivery of biological macromolecules with a wide range of molecular weights. Altogether, VNB photoporation represents a promising technique for the universal delivery of macromolecules in view of engineering CD4+ T cells for use in a wide variety of research and therapeutic applications.
Collapse
|
1019
|
Distinct prognostic value of circulating anti-telomerase CD4 + Th1 immunity and exhausted PD-1 +/TIM-3 + T cells in lung cancer. Br J Cancer 2019; 121:405-416. [PMID: 31358938 PMCID: PMC6738094 DOI: 10.1038/s41416-019-0531-5] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 06/25/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Despite the critical roles of Th1-polarised CD4+ T cells in cancer immunosurveillance, the translation of their potential to clinical use remains challenging. Here, we investigate the clinical relevance of circulating antitumor Th1 immunity in non-small cell lung cancer (NSCLC). METHODS The circulating antitumor Th1 response was assessed by the ELISpot assay in 170 NSCLC patients using a mixture of HLA class II-restricted peptides from telomerase (TERT). Phenotyping of blood immune cells was performed by flow cytometry. RESULTS TERT-reactive CD4 T-cell response was detected in 35% of NSCLC patients before any treatment. Functional analysis showed that these cells were effector memory and Th1 polarised capable to produce effector cytokines, such as IFN-γ, TNF-α and IL-2. The presence of anti-TERT Th1 response was inversely correlated with the level of exhausted PD-1+/TIM-3+CD4 T cells. The level of these two immune parameters differentially affected the survival, so that increased level of anti-TERT Th1 response and low rate of exhausted PD-1+TIM-3+CD4+ T cells were associated with a better prognosis. CONCLUSIONS Systemic anti-TERT Th1 response plays a strong antitumor protective role in NSCLC. This study underlines the potential interest of monitoring circulating antitumor Th1 response for patients' stratification and therapy decision.
Collapse
|
1020
|
Tumor Vasculatures: A New Target for Cancer Immunotherapy. Trends Pharmacol Sci 2019; 40:613-623. [PMID: 31331639 DOI: 10.1016/j.tips.2019.07.001] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 06/30/2019] [Accepted: 07/01/2019] [Indexed: 12/24/2022]
Abstract
Immune cells rely on a functional vascular network to enter tissues. In solid tumors, blood vessels are abnormal and dysfunctional and, thus, immune effector cell infiltration is impaired. Although normalizing the tumor vasculature has been shown to improve the efficacy of cancer immunotherapies, recent studies suggest that enhanced immune stimulation also, in turn, improves tumor vascular normalization. Thus, this new paradigm of immune system-tumor vasculature mutual reprogramming opens the possibility of identifying new cancer treatment strategies that combine vascular targeting and immunotherapies. Here, we highlight current evidence supporting immune system-tumor vasculature crosstalk and outline how this relationship can provide new rationales for developing more effective combination immunotherapy strategies for treating human cancers.
Collapse
|
1021
|
Sprooten J, Ceusters J, Coosemans A, Agostinis P, De Vleeschouwer S, Zitvogel L, Kroemer G, Galluzzi L, Garg AD. Trial watch: dendritic cell vaccination for cancer immunotherapy. Oncoimmunology 2019; 8:e1638212. [PMID: 31646087 PMCID: PMC6791419 DOI: 10.1080/2162402x.2019.1638212] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022] Open
Abstract
Dendritic- cells (DCs) have received considerable attention as potential targets for the development of anticancer vaccines. DC-based anticancer vaccination relies on patient-derived DCs pulsed with a source of tumor-associated antigens (TAAs) in the context of standardized maturation-cocktails, followed by their reinfusion. Extensive evidence has confirmed that DC-based vaccines can generate TAA-specific, cytotoxic T cells. Nonetheless, clinical efficacy of DC-based vaccines remains suboptimal, reflecting the widespread immunosuppression within tumors. Thus, clinical interest is being refocused on DC-based vaccines as combinatorial partners for T cell-targeting immunotherapies. Here, we summarize the most recent preclinical/clinical development of anticancer DC vaccination and discuss future perspectives for DC-based vaccines in immuno-oncology.
Collapse
Affiliation(s)
- Jenny Sprooten
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jolien Ceusters
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and Immunotherapy, ImmunOvar Research Group, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
- Department of Gynecology and Obstetrics, UZ Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
- Center for Cancer Biology (CCB), VIB, Leuven, Belgium
| | - Steven De Vleeschouwer
- Research Group Experimental Neurosurgery and Neuroanatomy, KU Leuven, Leuven, Belgium
- Department of Neurosurgery, UZ Leuven, Leuven, Belgium
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, 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
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China
- Department of Women’s and Children’s Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA
- Université de Paris Descartes, Paris, France
| | - Abhishek D. Garg
- Cell Death Research & Therapy (CDRT) unit, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| |
Collapse
|
1022
|
Zhang MY, Guo J, Hu XM, Zhao SQ, Li SL, Wang J. An in vivo anti-tumor effect of eckol from marine brown algae by improving the immune response. Food Funct 2019; 10:4361-4371. [PMID: 31276149 DOI: 10.1039/c9fo00865a] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The anti-cancer activities of brown algae and some active extracts or components from brown algae have been demonstrated. But the anti-tumor activities of eckol, a new natural phlorotannin derived from marine brown algae, are poorly understood. In order to investigate the in vivo anti-tumor effect and its potential mechanisms of eckol in a sarcoma 180 (S180) xenograft-bearing animal model, S180 xenograft-bearing mice were randomly divided into 4 groups: model control, and eckol low-dose (0.25 mg kg-1), middle-dose (0.5 mg kg-1) and high-dose (1.0 mg kg-1) groups. After eckol administration, the tumor inhibition, tumor tissue histology, thymus index and spleen index were measured. The apoptotic tumor cells were detected using the terminal deoxynucleotidyl transferase mediated dUTP nick end labelling (TUNEL) assay. The protein expression levels of cleaved Caspase-3 and Caspase-9 (two key apoptotic proteins), Bcl-2 and Bax (two key anti-apoptosis-related genes), as well as epidermal growth factor receptor (EGFR, a well-known cell proliferation-stimulating molecule in tumorigenesis) and p-EGFR in tumor tissues were determined by western blot. A carbon particle clearance test, measurement of serum cytokine levels, a splenic T lymphocyte proliferation test, and T lymphocyte subpopulation analysis were used to evaluate the effect of eckol on the immune function of tumor-bearing mice. Moreover, CD11c+-dendritic cell (DC) infiltration in tumor tissues was detected by immunohistochemistry, and the surface molecules on bone marrow-derived DCs were analyzed using flow cytometry. The pro-apoptosis and anti-proliferation activities of eckol were manifested by the increased TUNEL-positive apoptotic cells, the upregulated Caspase-3 and Caspase-9 expression, and the downregulated expression of Bcl-2, Bax, EGFR and p-EGFR in eckol-treated transplanted S180 tumors. Most importantly, eckol stimulated the mononuclear phagocytic system, recruited and activated DCs, promoted the tumor-specific Th1 responses, increased the CD4+/CD8+ T lymphocyte ratio, and enhanced cytotoxic T lymphocyte responses in the eckol-treated animals, suggesting its potent stimulatory property on innate and adaptive immune responses. This study suggested that eckol might act as a functional food constituent derived from marine brown algae with a potential in vivo anti-tumor effect achieved by improving the immune response.
Collapse
Affiliation(s)
- Meng-Ya Zhang
- Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Wuhan University of Science and Technology, Wuhan, China.
| | | | | | | | | | | |
Collapse
|
1023
|
Rodríguez-Rodríguez N, Madera-Salcedo IK, Bugarin-Estrada E, Sánchez-Miranda E, Torres-García D, Cervantes-Torres J, Fragoso G, Rosetti F, Crispín JC, Sciutto E. The helminth-derived peptide GK-1 induces an anti-tumoral CD8 T cell response associated with downregulation of the PD-1/PD-L1 pathway. Clin Immunol 2019; 212:108240. [PMID: 31299381 DOI: 10.1016/j.clim.2019.07.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 07/04/2019] [Accepted: 07/08/2019] [Indexed: 12/17/2022]
Abstract
CD8 T cells can kill malignant cells in an antigen-specific manner. However, anti-tumoral responses are usually limited by suppressive factors that curb the effector responses of tumor-infiltrating CD8 T cells. Therapeutic strategies to overcome intra-tumoral T cell suppression, for example immune checkpoint inhibition, have been clinically effective in patients with cancer. Here, we provide data that demonstrates that GK-1, a peptide derived from the parasite Taenia crassiceps, promotes an anti-melanoma CD8 T cell response with heightened effector characteristics that leads to an increased amount of tumor-infiltrating CD44+ IFN-γ-producing CD8 T cells. The response induced by GK-1 was associated with a reduction in the expression of PD-1 and PD-L1 on tumor-infiltrating CD8 and dendritic cells, respectively, effects that led to a dramatic decrease in tumor burden. Our results suggest that the immunomodulatory properties of GK-1 may promote a CD8 T cell response that may be therapeutically useful in the setting of cancer.
Collapse
Affiliation(s)
- Noé Rodríguez-Rodríguez
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Iris K Madera-Salcedo
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Emmanuel Bugarin-Estrada
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Elizabeth Sánchez-Miranda
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Diana Torres-García
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jacquelynne Cervantes-Torres
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gladis Fragoso
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Florencia Rosetti
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - José C Crispín
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| | - Edda Sciutto
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
1024
|
Chen YQ, Li PC, Pan N, Gao R, Wen ZF, Zhang TY, Huang F, Wu FY, Ou XL, Zhang JP, Zhu XJ, Hu HM, Chen K, Cai YL, Wang LX. Tumor-released autophagosomes induces CD4 + T cell-mediated immunosuppression via a TLR2-IL-6 cascade. J Immunother Cancer 2019; 7:178. [PMID: 31300052 PMCID: PMC6625067 DOI: 10.1186/s40425-019-0646-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 06/19/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND CD4+ T cells are critical effectors of anti-tumor immunity, but how tumor cells influence CD4+ T cell effector function is not fully understood. Tumor cell-released autophagosomes (TRAPs) are being recognized as critical modulators of host anti-tumor immunity during tumor progression. Here, we explored the mechanistic aspects of TRAPs in the modulation of CD4+ T cells in the tumor microenvironment. METHODS TRAPs isolated from tumor cell lines and pleural effusions or ascites of cancer patients were incubated with CD4+ T cells to examine the function and mechanism of TRAPs in CD4+ T cell differentiation and function. TRAPs-elicited CD4+ T cells were tested for their suppression of effector T cell function, induction of regulatory B cells, and promotion of tumorigenesis and metastasis in a mouse model. RESULTS Heat shock protein 90α (HSP90α) on the surface of TRAPs from malignant effusions of cancer patients and tumor cell lines stimulated CD4+ T cell production of IL-6 via a TLR2-MyD88-NF-κB signal cascade. TRAPs-induced autocrine IL-6 further promoted CD4+ T cells secretion of IL-10 and IL-21 via STAT3. Notably, TRAPs-elicited CD4+ T cells inhibited CD4+ and CD8+ effector T cell function in an IL-6- and IL-10-dependent manner and induced IL-10-producing regulatory B cells (Bregs) via IL-6, IL-10 and IL-21, thereby promoting tumor growth and metastasis. Consistently, inhibition of tumor autophagosome formation or IL-6 secretion by CD4+ T cells markedly retarded tumor growth. Furthermore, B cell or CD4+ T cell depletion impeded tumor growth by increasing effector T cell function. CONCLUSIONS HSP90α on the surface of TRAPs programs the immunosuppressive functions of CD4+ T cells to promote tumor growth and metastasis. TRAPs or their membrane-bound HSP90α represent important therapeutic targets to reverse cancer-associated immunosuppression and improve immunotherapy.
Collapse
Affiliation(s)
- Yong-Qiang Chen
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Peng-Cheng Li
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Ning Pan
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Rong Gao
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Zhi-Fa Wen
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Tian-Yu Zhang
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Fang Huang
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Fang-Yuan Wu
- Department of Obstetrics and Gynecology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Xi-Long Ou
- Department of Gastroenterology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Jin-Ping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Xue-Jun Zhu
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.,Jiangsu Province Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Hong-Ming Hu
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.,Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Kang Chen
- Department of Obstetrics and Gynecology and Barbara Ann Karmanos Cancer Institute, Wayne State University, Mucosal Immunology Studies Team, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Detroit, MI, 48201, USA.,Jiangsu Province Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yun-Lang Cai
- Department of Obstetrics and Gynecology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.
| | - Li-Xin Wang
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.
| |
Collapse
|
1025
|
Chen YQ, Li PC, Pan N, Gao R, Wen ZF, Zhang TY, Huang F, Wu FY, Ou XL, Zhang JP, Zhu XJ, Hu HM, Chen K, Cai YL, Wang LX. Tumor-released autophagosomes induces CD4 + T cell-mediated immunosuppression via a TLR2-IL-6 cascade. J Immunother Cancer 2019. [PMID: 31300052 DOI: 10.1186/s40425-019-0646-5.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND CD4+ T cells are critical effectors of anti-tumor immunity, but how tumor cells influence CD4+ T cell effector function is not fully understood. Tumor cell-released autophagosomes (TRAPs) are being recognized as critical modulators of host anti-tumor immunity during tumor progression. Here, we explored the mechanistic aspects of TRAPs in the modulation of CD4+ T cells in the tumor microenvironment. METHODS TRAPs isolated from tumor cell lines and pleural effusions or ascites of cancer patients were incubated with CD4+ T cells to examine the function and mechanism of TRAPs in CD4+ T cell differentiation and function. TRAPs-elicited CD4+ T cells were tested for their suppression of effector T cell function, induction of regulatory B cells, and promotion of tumorigenesis and metastasis in a mouse model. RESULTS Heat shock protein 90α (HSP90α) on the surface of TRAPs from malignant effusions of cancer patients and tumor cell lines stimulated CD4+ T cell production of IL-6 via a TLR2-MyD88-NF-κB signal cascade. TRAPs-induced autocrine IL-6 further promoted CD4+ T cells secretion of IL-10 and IL-21 via STAT3. Notably, TRAPs-elicited CD4+ T cells inhibited CD4+ and CD8+ effector T cell function in an IL-6- and IL-10-dependent manner and induced IL-10-producing regulatory B cells (Bregs) via IL-6, IL-10 and IL-21, thereby promoting tumor growth and metastasis. Consistently, inhibition of tumor autophagosome formation or IL-6 secretion by CD4+ T cells markedly retarded tumor growth. Furthermore, B cell or CD4+ T cell depletion impeded tumor growth by increasing effector T cell function. CONCLUSIONS HSP90α on the surface of TRAPs programs the immunosuppressive functions of CD4+ T cells to promote tumor growth and metastasis. TRAPs or their membrane-bound HSP90α represent important therapeutic targets to reverse cancer-associated immunosuppression and improve immunotherapy.
Collapse
Affiliation(s)
- Yong-Qiang Chen
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Peng-Cheng Li
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Ning Pan
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Rong Gao
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Zhi-Fa Wen
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Tian-Yu Zhang
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Fang Huang
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Fang-Yuan Wu
- Department of Obstetrics and Gynecology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China
| | - Xi-Long Ou
- Department of Gastroenterology, Zhongda Hospital, Medical School of Southeast University, Nanjing, 210009, China
| | - Jin-Ping Zhang
- Institutes of Biology and Medical Sciences, Soochow University, Suzhou, 215123, China
| | - Xue-Jun Zhu
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.,Jiangsu Province Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Hong-Ming Hu
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.,Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, 97213, USA
| | - Kang Chen
- Department of Obstetrics and Gynecology and Barbara Ann Karmanos Cancer Institute, Wayne State University, Mucosal Immunology Studies Team, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Detroit, MI, 48201, USA.,Jiangsu Province Hospital of Traditional Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China
| | - Yun-Lang Cai
- Department of Obstetrics and Gynecology, Zhongda Hospital, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.
| | - Li-Xin Wang
- Department of Microbiology and Immunology, Medical School of Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China.
| |
Collapse
|
1026
|
Liu M, Wen M, Shen S, Zhang Z, Chen G, Zhang W. One‐Pot, Multicomponent Strategy for Designing Lymphoseek‐Inspired Hetero‐Glycoadjuvant@AuNPs. Macromol Rapid Commun 2019; 40:e1900215. [DOI: 10.1002/marc.201900215] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/31/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Mengjie Liu
- M. Liu, M. Wen, S. Shen, Prof. G. Chen, Prof. W. ZhangCenter for Soft Condensed Matter Physicsand Interdisciplinary Research & School of Physical Scienceand TechnologyJiangsu Key Laboratory of Thin FilmsSoochow University Suzhou 215006 P. R. China
| | - Ming Wen
- M. Liu, M. Wen, S. Shen, Prof. G. Chen, Prof. W. ZhangCenter for Soft Condensed Matter Physicsand Interdisciplinary Research & School of Physical Scienceand TechnologyJiangsu Key Laboratory of Thin FilmsSoochow University Suzhou 215006 P. R. China
| | - Shuyi Shen
- M. Liu, M. Wen, S. Shen, Prof. G. Chen, Prof. W. ZhangCenter for Soft Condensed Matter Physicsand Interdisciplinary Research & School of Physical Scienceand TechnologyJiangsu Key Laboratory of Thin FilmsSoochow University Suzhou 215006 P. R. China
| | - Zhengbiao Zhang
- State and Local Joint Engineering Laboratoryfor Novel Functional Polymeric MaterialsSoochow University Suzhou 215123 China
| | - Gaojian Chen
- M. Liu, M. Wen, S. Shen, Prof. G. Chen, Prof. W. ZhangCenter for Soft Condensed Matter Physicsand Interdisciplinary Research & School of Physical Scienceand TechnologyJiangsu Key Laboratory of Thin FilmsSoochow University Suzhou 215006 P. R. China
| | - Weidong Zhang
- M. Liu, M. Wen, S. Shen, Prof. G. Chen, Prof. W. ZhangCenter for Soft Condensed Matter Physicsand Interdisciplinary Research & School of Physical Scienceand TechnologyJiangsu Key Laboratory of Thin FilmsSoochow University Suzhou 215006 P. R. China
- State and Local Joint Engineering Laboratoryfor Novel Functional Polymeric MaterialsSoochow University Suzhou 215123 China
| |
Collapse
|
1027
|
Shergold AL, Millar R, Nibbs RJ. Understanding and overcoming the resistance of cancer to PD-1/PD-L1 blockade. Pharmacol Res 2019; 145:104258. [DOI: 10.1016/j.phrs.2019.104258] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/01/2019] [Accepted: 05/01/2019] [Indexed: 12/22/2022]
|
1028
|
Wang HF, Wang SS, Huang MC, Liang XH, Tang YJ, Tang YL. Targeting Immune-Mediated Dormancy: A Promising Treatment of Cancer. Front Oncol 2019; 9:498. [PMID: 31297335 PMCID: PMC6607988 DOI: 10.3389/fonc.2019.00498] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 05/24/2019] [Indexed: 02/05/2023] Open
Abstract
Immune-mediated dormancy is when the immune system keeps proliferating tumor cells unchanged, mostly via cytotoxic activity of immune cells. Cancer dormancy, especially immune-mediated dormancy, may be the explanation for tumor refractory and may be responsible for resistance to conventional chemo- and radiotherapies. Here, we will describe different scenarios as to how the immune cells and cytokines involved in cancer progression are connected with the initiation of dormancy and cancer treatment. Two distinct treatment methods, such as maintaining metastatic tumor cells dormant and awakening them, are also discussed. A better understanding of immune-mediated dormancy will help to design novel and effective immunotherapies and will likely increase the efficiency of tumor treatment inhibiting metastasis.
Collapse
Affiliation(s)
- Hao-Fan Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Sha-Sha Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Mei-Chang Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China.,Hubei Key Laboratory of Industrial Microbiology, Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei University of Technology, Wuhan, China
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
1029
|
Huff WX, Kwon JH, Henriquez M, Fetcko K, Dey M. The Evolving Role of CD8 +CD28 - Immunosenescent T Cells in Cancer Immunology. Int J Mol Sci 2019; 20:ijms20112810. [PMID: 31181772 PMCID: PMC6600236 DOI: 10.3390/ijms20112810] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/05/2019] [Accepted: 06/06/2019] [Indexed: 12/18/2022] Open
Abstract
Functional, tumor-specific CD8+ cytotoxic T lymphocytes drive the adaptive immune response to cancer. Thus, induction of their activity is the ultimate aim of all immunotherapies. Success of anti-tumor immunotherapy is precluded by marked immunosuppression in the tumor microenvironment (TME) leading to CD8+ effector T cell dysfunction. Among the many facets of CD8+ T cell dysfunction that have been recognized—tolerance, anergy, exhaustion, and senescence—CD8+ T cell senescence is incompletely understood. Naïve CD8+ T cells require three essential signals for activation, differentiation, and survival through T-cell receptor, costimulatory receptors, and cytokine receptors. Downregulation of costimulatory molecule CD28 is a hallmark of senescent T cells and increased CD8+CD28− senescent populations with heterogeneous roles have been observed in multiple solid and hematogenous tumors. T cell senescence can be induced by several factors including aging, telomere damage, tumor-associated stress, and regulatory T (Treg) cells. Tumor-induced T cell senescence is yet another mechanism that enables tumor cell resistance to immunotherapy. In this paper, we provide a comprehensive overview of CD8+CD28− senescent T cell population, their origin, their function in immunology and pathologic conditions, including TME and their implication for immunotherapy. Further characterization and investigation into this subset of CD8+ T cells could improve the efficacy of future anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Wei X Huff
- Department of Neurosurgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Jae Hyun Kwon
- Department of Neurosurgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Mario Henriquez
- Department of Neurosurgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Kaleigh Fetcko
- Department of Neurology, University of Illinois at Chicago School of Medicine, Chicago, IL 60612, USA.
| | - Mahua Dey
- Department of Neurosurgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
1030
|
Zhao Y, Ge X, He J, Cheng Y, Wang Z, Wang J, Sun L. The prognostic value of tumor-infiltrating lymphocytes in colorectal cancer differs by anatomical subsite: a systematic review and meta-analysis. World J Surg Oncol 2019; 17:85. [PMID: 31118034 PMCID: PMC6532263 DOI: 10.1186/s12957-019-1621-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 04/23/2019] [Indexed: 01/15/2023] Open
Abstract
PURPOSE In colorectal cancer (CRC), whether the immune score can be used to predict the clinical prognosis of the patient has not been completely established. Besides, the prognostic values of tumor-infiltrating lymphocytes (TILs) in different anatomical locations, counting sites, and subtypes have been controversial. The purpose of this meta-analysis is to analyze and determine the prognostic value of TILs indices including TIL subsets, infiltrating sites, and anatomical sites. METHODS Relevant literature was obtained by searching PubMed and Google Scholar. The pooled hazard ratio (HR) of the overall survival (OS), disease-free survival (DFS), and cancer-specific survival (CSS) was computed to investigate the prognostic significance of CD3+, CD8+, CD45RO+, and FOXP3+ T cells. RESULTS A total of 22 studies involving 5108 patients were included in the meta-analysis. In CC, based on T cell subtypes analysis, the final results indicated that CD8+ and FOXP3+ infiltrating cells, but not CD3+ T cells were prognostic markers for DFS and OS. In addition, with regard to the counting location of TILs, subgroup analysis revealed that only high FOXP3+ infiltrates in the tumor stroma (ST) were significantly associated with OS (HR = 0.38, 95% confidence interval (CI) = 0.22-0.67, P = 0.0007), whereas in invasive margin (IM), high density of CD3+ infiltrating cells indicated increased DFS (HR = 0.76, 95% CI = 0.62-0.93, P = 0.008). At the tumor center (TC), high CD8+ T cells infiltration was associated with improved DFS (HR = 0.50, 95% CI = 0.38-0.65, P < 0.00001). In RC, whether CSS or OS, high-density TIL was associated with improved prognosis. CONCLUSION In a single counting site, high-density TILs reflect favorable prognostic value in CC or RC. For CC, more prospective studies are needed to verify whether different anatomical sites affect the distribution of TILs and thus the prognosis of patients. For RC, further studies should analyze the prognostic value of the immune score.
Collapse
Affiliation(s)
- Yamei Zhao
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Xiaoxu Ge
- Department of Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Jiawei He
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Yi Cheng
- Departments of Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Zhanhuai Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China
| | - Jian Wang
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China.
| | - Lifeng Sun
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, People's Republic of China.
| |
Collapse
|
1031
|
Chen H, Chong W, Wu Q, Yao Y, Mao M, Wang X. Association of LRP1B Mutation With Tumor Mutation Burden and Outcomes in Melanoma and Non-small Cell Lung Cancer Patients Treated With Immune Check-Point Blockades. Front Immunol 2019; 10:1113. [PMID: 31164891 PMCID: PMC6536574 DOI: 10.3389/fimmu.2019.01113] [Citation(s) in RCA: 127] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 05/01/2019] [Indexed: 01/04/2023] Open
Abstract
Background: Tumor mutation burden (TMB) have been served as the most prevalent biomarkers to predict immunotherapy response. LRP1B (low-density lipoprotein receptor-related protein 1B) is frequently mutated in melanoma, non-small cell lung cancer (NSCLC) and other tumors; however, its association with TMB and survival in patients with immunotherapy remains unknown. Methods: We curated somatic mutation data and clinicopathologic information from 332 melanoma immunotherapy samples for discovery and 113 NSCLC samples for further corroboration. Bayesian variants non-negative matrix factorization was used to extract tumor mutational signatures. Multivariate Cox and logistic regression models were applied to adjust confounding factors. The CIBERSORT and GSEA algorithm were separately used to infer leukocyte relative abundance and significantly enriched pathways. Results: Patients with LRP1B mutation were identified to be associated with prolonged survival in both immunotherapy cohort. Higher tumor mutation burden was found in LRP1B mutated patients, and the association remained significant after controlling for age, gender, stage, mutations in TP53 and ATR, and mutational signatures. Immune response and cell cycle regulation circuits were among the top enriched pathways in samples with LRP1B mutations. Conclusion: Our studies suggested sequencing even a single, frequently mutated gene may provide insight into genome-wide mutational burden, and may serve as a biomarker to predict immune response.
Collapse
Affiliation(s)
- Hao Chen
- Clinical Epidemiology Unit, Qilu Hospital of Shandong University, Jinan, China.,Key Laboratory of Cancer Prevention and Therapy of Tianjin, Department of Epidemiology and Biostatistics, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wei Chong
- Key Laboratory of Cancer Prevention and Therapy, Department of Breast Cancer Pathology and Research Laboratory, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Qian Wu
- Department of Respiratory Medicine, Central Hospital of Zibo, Zibo, China
| | - Yueliang Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Mao
- Key Laboratory of Tumor Immunopathology, Ministry of Education of China, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xin Wang
- Department of Epidemiology and Biostatistics, First Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
1032
|
Couture A, Garnier A, Docagne F, Boyer O, Vivien D, Le-Mauff B, Latouche JB, Toutirais O. HLA-Class II Artificial Antigen Presenting Cells in CD4 + T Cell-Based Immunotherapy. Front Immunol 2019; 10:1081. [PMID: 31156634 PMCID: PMC6533590 DOI: 10.3389/fimmu.2019.01081] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 04/29/2019] [Indexed: 12/11/2022] Open
Abstract
CD4+ T cells differentiate into various T helper subsets characterized by distinct cytokine secreting profiles that confer them effector functions adapted to a variety of infectious or endogenous threats. Regulatory CD4+ T cells are another specialized subset that plays a fundamental role in the maintenance of immune tolerance to self-antigens. Manipulating effector or regulatory CD4+ T cells responses is a promising immunotherapy strategy for, respectively, chronical viral infections and cancer, or severe autoimmune diseases and transplantation. Adoptive cell therapy (ACT) is an emerging approach that necessitates defining robust and efficient methods for the in vitro expansion of antigen-specific T cells then infused into patients. To address this challenge, artificial antigen presenting cells (AAPCs) have been developed. They constitute a reliable and easily usable platform to stimulate and amplify antigen-specific CD4+ T cells. Here, we review the recent advances in understanding the functions of CD4+ T cells in immunity and in immune tolerance, and their use for ACT. We also describe the characteristics of different AAPC models and the way to improve their stimulating functions. Finally, we discuss the potential interest of these AAPCs, both as fundamental tools to decipher CD4+ T cell responses and as reagents to generate clinical grade antigen-specific CD4+ T cells for immunotherapy.
Collapse
Affiliation(s)
- Alexandre Couture
- UNIROUEN, Inserm U1245, Institute for Research and Innovation in Biomedicine, Normandie University, Rouen, France
| | - Anthony Garnier
- Inserm U1237, Physiopathology and Imaging of Neurological Disorders, Caen University Hospital, Caen, France
| | - Fabian Docagne
- Inserm U1237, Physiopathology and Imaging of Neurological Disorders, Caen University Hospital, Caen, France
| | - Olivier Boyer
- Department of Immunology and Biotherapy, Inserm U1234, Institute for Research and Innovation in Biomedicine, UNIROUEN, Rouen University Hospital, Normandie University, Rouen, France
| | - Denis Vivien
- Inserm U1237, Physiopathology and Imaging of Neurological Disorders, Caen University Hospital, Caen, France
- Department of Clinical Research, Caen University Hospital, Caen, France
| | - Brigitte Le-Mauff
- Inserm U1237, Physiopathology and Imaging of Neurological Disorders, Caen University Hospital, Caen, France
- Department of Immunology and Immunopathology, Caen University Hospital, Caen, France
| | - Jean-Baptiste Latouche
- UNIROUEN, Inserm U1245, Institute for Research and Innovation in Biomedicine, Normandie University, Rouen, France
- Department of Genetics, Rouen University Hospital, Rouen, France
| | - Olivier Toutirais
- Inserm U1237, Physiopathology and Imaging of Neurological Disorders, Caen University Hospital, Caen, France
- Department of Immunology and Immunopathology, Caen University Hospital, Caen, France
- French Blood Service (Etablissement Français du Sang), Caen, France
| |
Collapse
|
1033
|
Noubade R, Majri-Morrison S, Tarbell KV. Beyond cDC1: Emerging Roles of DC Crosstalk in Cancer Immunity. Front Immunol 2019; 10:1014. [PMID: 31143179 PMCID: PMC6521804 DOI: 10.3389/fimmu.2019.01014] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 04/23/2019] [Indexed: 01/03/2023] Open
Abstract
Dendritic cells (DCs) efficiently process and present antigens to T cells, and by integrating environmental signals, link innate and adaptive immunity. DCs also control the balance between tolerance and immunity, and are required for T-cell mediated anti-tumor immunity. One subset of classical DCs, cDC1, are particularly important for eliciting CD8 T cells that can kill tumor cells. cDC1s are superior in antigen cross-presentation, a process of presenting exogenous antigens on MHC class I to activate CD8+ T cells. Tumor-associated cDC1s can transport tumor antigen to the draining lymph node and cross-present tumor antigens, resulting in priming and activation of cytotoxic T cells. Although cross-presenting cDC1s are critical for eliciting anti-tumor T cell responses, the role and importance of other DC subsets in anti-tumor immunity is not as well-characterized. Recent literature in other contexts suggests that critical crosstalk between DC subsets can significantly alter biological outcomes, and these DC interactions likely also contribute significantly to tumor-specific immune responses. Therefore, antigen presentation by cDC1s may be necessary but not sufficient for maximal immune responses against cancer. Here, we discuss recent advances in the understanding of DC subset interactions to maximize anti-tumor immunity, and propose that such interactions should be considered for the development of better DC-targeted immunotherapies.
Collapse
Affiliation(s)
- Rajkumar Noubade
- Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Sonia Majri-Morrison
- Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| | - Kristin V Tarbell
- Department of Inflammation and Oncology, Amgen Research, Amgen Inc., South San Francisco, CA, United States
| |
Collapse
|
1034
|
Guo Q, Huang F, Goncalves C, Del Rincón SV, Miller WH. Translation of cancer immunotherapy from the bench to the bedside. Adv Cancer Res 2019; 143:1-62. [PMID: 31202357 DOI: 10.1016/bs.acr.2019.03.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The tremendous success of immune checkpoint blockades has revolutionized cancer management. Our increased understanding of the cell types that compose the tumor microenvironment (TME), including those of the innate and adaptive immune system, has helped to shape additional immune modulatory strategies in cancer care. Pre-clinical and clinical investigations targeting novel checkpoint interactions and key pathways that regulate cancer immunity continue to increase rapidly. Various combinatorial drug regimens are being tested in attempt to achieve durable response and survival rates of patients with cancer. This review provides an overview of specific components of the TME, an introduction to novel immune checkpoints, followed by a survey of present day and future combination immune modulatory therapies. The idea that the immune system can recognize and destroy tumor cells was first described in the cancer immunosurveillance hypothesis of Burnet and Thomas. However, early experimental evidence failed to support the concept. It was not until the late 1990s when seminal papers clearly showed the existence of cancer immunosurveillance, leading to the cancer immunoediting hypothesis. In this century, progress in the understanding of negative regulators of the immune response led to the discovery that inhibition of these regulators in patients with cancer could lead to dramatic and durable remissions. Drs. Tasuku Honjo and James P. Allison were awarded the Nobel Prize in 2018 for their pioneering work in this field. We now see rapid advances in cancer immunology and emerging effective therapies revolutionizing cancer care across tumor types in the clinic, while pre-clinical research is moving from a focus on the malignant cells themselves to dissect the highly heterogenic and complex multi-cellular tumor microenvironment (TME).
Collapse
Affiliation(s)
- Qianyu Guo
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Fan Huang
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Christophe Goncalves
- Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Sonia V Del Rincón
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada
| | - Wilson H Miller
- Division of Experimental Medicine, Faculty of Medicine, McGill University, Montreal, QC, Canada; Jewish General Hospital, Segal Cancer Centre, Department of Oncology, Montreal, QC, Canada; Rossy Cancer Network, Montreal, QC, Canada.
| |
Collapse
|
1035
|
Qin Y, Lee Y, Seo J, Kim T, Shin JH, Park SH. NIH3T3 Directs Memory-Fated CTL Programming and Represses High Expression of PD-1 on Antitumor CTLs. Front Immunol 2019; 10:761. [PMID: 31031760 PMCID: PMC6470252 DOI: 10.3389/fimmu.2019.00761] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 03/21/2019] [Indexed: 11/15/2022] Open
Abstract
Memory CD8+ T cells have long been considered a promising population for adoptive cell therapy (ACT) due to their long-term persistence and robust re-stimulatory response. NIH3T3 is an immortalized mouse embryonic fibroblast cell line. We report that NIH3T3-conditioned medium (CM) can augment effector functions of CTLs following antigen priming and confer phenotypic and transcriptional properties of central memory cells. After NIH3T3-CM-educated CTLs were infused into naïve mice, they predominantly developed to central memory cells. A large number of NIH3T3-CM-educated CTLs with high functionality persisted and infiltrated to tumor mass. In addition, NIH3T3-CM inhibited CTLs expression of PD-1 in vitro and repressed their high expression of PD-1 in tumor microenvironment after adoptive transfer. Consequently, established tumor models showed that infusion of NIH3T3-CM-educated CTLs dramatically improved CTL mediated-antitumor immunity. Furthermore, NIH3T3-CM also promoted human CD8+ T cells differentiation into memory cells. These results suggest that NIH3T3-CM-programmed CTLs are good candidates for adoptive transfer in tumor therapy.
Collapse
Affiliation(s)
- Yingyu Qin
- Department of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| | - Yuna Lee
- ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Jaeho Seo
- ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Taehyun Kim
- ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Jung Hoon Shin
- ImmunoMax Co., Ltd, Korea University, Seoul, South Korea
| | - Se-Ho Park
- Department of Life Sciences and Biotechnology, Korea University, Seoul, South Korea
| |
Collapse
|
1036
|
Belnoue E, Mayol JF, Carboni S, Di Berardino Besson W, Dupuychaffray E, Nelde A, Stevanovic S, Santiago-Raber ML, Walker PR, Derouazi M. Targeting self and neo-epitopes with a modular self-adjuvanting cancer vaccine. JCI Insight 2019; 5:127305. [PMID: 31013258 DOI: 10.1172/jci.insight.127305] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Induction of a potent CD4 and CD8 T-cell response against tumor-specific and tumor-associated antigen is critical for eliminating tumor cells. Recent vaccination strategies have been hampered by an inefficacious and low amplitude immune response. Here we describe a self-adjuvanted chimeric protein vaccine platform to address these challenges, characterized by a multidomain construction incorporating (i) a cell penetrating peptide (CPP) allowing internalization of several multiantigenic Major Histocompatibility Complex (MHC)-restricted peptides within (ii) the multiantigenic domain (Mad) and (iii) a TLR2/4 agonist domain (TLRag). Functionality of the resulting chimeric protein is based on the combined effect of the above-mentioned three different domains for simultaneous activation of antigen presenting cells and antigen cross-presentation, leading to an efficacious multiantigenic and multiallelic cellular immune response. Helper and cytotoxic T-cell responses were observed against model-, neo- and self-antigens, and were highly potent in several murine tumor models. The safety and the immunogenicity of a human vaccine candidate designed for colorectal cancer treatment was demonstrated in a non-human primate model. This newly engineered therapeutic vaccine approach is promising for the treatment of poorly infiltrated tumors that do not respond to currently marketed immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | | | - Annika Nelde
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Tubingen, Germany
| | - Stefan Stevanovic
- University of Tübingen, Interfaculty Institute for Cell Biology, Department of Immunology, Tubingen, Germany
| | | | - Paul R Walker
- Centre for Translational Research in Onco-Hematology, Department of Internal Medicine Specialties, University of Geneva and Division of Oncology, Geneva University Hospitals, Geneva, Switzerland
| | | |
Collapse
|
1037
|
Fennemann FL, de Vries IJM, Figdor CG, Verdoes M. Attacking Tumors From All Sides: Personalized Multiplex Vaccines to Tackle Intratumor Heterogeneity. Front Immunol 2019; 10:824. [PMID: 31040852 PMCID: PMC6476980 DOI: 10.3389/fimmu.2019.00824] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/28/2019] [Indexed: 12/23/2022] Open
Abstract
Tumor vaccines are an important asset in the field of cancer immunotherapy. Whether prophylactic or therapeutic, these vaccines aim to enhance the T cell-mediated anti-tumor immune response that is orchestrated by dendritic cells. Although promising preclinical and early-stage clinical results have been obtained, large-scale clinical implementation of cancer vaccination is stagnating due to poor clinical response. The challenges of clinical efficacy of tumor vaccines can be mainly attributed to tumor induced immunosuppression and poor immunogenicity of the chosen tumor antigens. Recently, intratumor heterogeneity and the relation with tumor-specific neoantigen clonality were put in the equation.In this perspective we provide an overview of recent studies showing how personalized tumor vaccines containing multiple neoantigens can broaden and enhance the anti-tumor immune response. Furthermore, we summarize advances in the understanding of the intratumor mutational landscape containing different tumor cell subclones and the temporal and spatial diversity of neoantigen presentation and burden, and the relation between these factors with respect to tumor immunogenicity. Together, the presented knowledge calls for the investment in the characterization of neoantigens in the context of intratumor heterogeneity to improve clinical efficacy of personalized tumor vaccines.
Collapse
Affiliation(s)
- Felix L Fennemann
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - I Jolanda M de Vries
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Medical Oncology, Radboud University Medical Center, Nijmegen, Netherlands
| | - Carl G Figdor
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Division of Immunotherapy, Oncode Institute, Radboud University Medical Center, Nijmegen, Netherlands.,Institute for Chemical Immunology, Nijmegen, Netherlands
| | - Martijn Verdoes
- Department of Tumor Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Institute for Chemical Immunology, Nijmegen, Netherlands
| |
Collapse
|
1038
|
Axelrod ML, Cook RS, Johnson DB, Balko JM. Biological Consequences of MHC-II Expression by Tumor Cells in Cancer. Clin Cancer Res 2019; 25:2392-2402. [PMID: 30463850 PMCID: PMC6467754 DOI: 10.1158/1078-0432.ccr-18-3200] [Citation(s) in RCA: 316] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 10/02/2018] [Accepted: 11/16/2018] [Indexed: 12/20/2022]
Abstract
Immunotherapy has emerged as a key pillar of cancer treatment. To build upon the recent successes of immunotherapy, intense research efforts are aimed at a molecular understanding of antitumor immune responses, identification of biomarkers of immunotherapy response and resistance, and novel strategies to circumvent resistance. These studies are revealing new insight into the intricacies of tumor cell recognition by the immune system, in large part through MHCs. Although tumor cells widely express MHC-I, a subset of tumors originating from a variety of tissues also express MHC-II, an antigen-presenting complex traditionally associated with professional antigen-presenting cells. MHC-II is critical for antigen presentation to CD4+ T lymphocytes, whose role in antitumor immunity is becoming increasingly appreciated. Accumulating evidence demonstrates that tumor-specific MHC-II associates with favorable outcomes in patients with cancer, including those treated with immunotherapies, and with tumor rejection in murine models. Herein, we will review current research regarding tumor-enriched MHC-II expression and regulation in a range of human tumors and murine models, and the possible therapeutic applications of tumor-specific MHC-II.
Collapse
Affiliation(s)
- Margaret L Axelrod
- Department of Medicine, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee
- Cancer Biology Graduate Program, Vanderbilt University, Nashville, Tennessee
| | - Rebecca S Cook
- Cancer Biology Graduate Program, Vanderbilt University, Nashville, Tennessee
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Vanderbilt University, Nashville, Tennessee.
- Cancer Biology Graduate Program, Vanderbilt University, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| |
Collapse
|
1039
|
Shi MJ, Meng XY, Wu QJ, Zhou XH. High CD3D/CD4 ratio predicts better survival in muscle-invasive bladder cancer. Cancer Manag Res 2019; 11:2987-2995. [PMID: 31114346 PMCID: PMC6489580 DOI: 10.2147/cmar.s191105] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 02/07/2019] [Indexed: 01/03/2023] Open
Abstract
Background: Bladder cancer is a common malignancy that affects the human urinary tract. Muscle-invasive bladder cancer (MIBC) is aggressive and has poor prognosis. Previous studies have reported that the tumor-infiltrating lymphocytes (TILs) were associated with MIBC outcome; however, inconsistency remains and mRNA level TIL markers’ prognostic significance in MIBC is unclear. Materials and methods: In the present study, we reanalyzed data from four public datasets (the Cancer Genome Atlas for investigation; and CIT, GSE5287, and GSE31684 for validation) to examine the prognostic significance of CD3D, CD4, CD8A, CD3D/CD4 and CD3D/CD8A in MIBC. Results: We found that the CD3D/CD4 ratio was a stable independent prognostic factor in MIBC (beta = −0.87, P = 0.025); high CD3D/CD4 ratio predicted better survival in MIBC, and the power of this association was much stronger in basal-squamous tumors (beta = −4.73, P = 2.67E-06). We also noted that the CD4 expression was significantly higher than CD3D (P < 0.05), indicating the presence of CD3−CD4+ cells which could be immune-suppressing. Conclusion: The CD3D/CD4 ratio can be viewed as a prognostic marker and a rough measurement for the interaction between immune-effecting CD3+ TILs and immune-suppressing CD3−CD4+ cells in MIBC, and this interaction may play a particularly important role in anti-cancer immunity in basal-squamous tumors as it has a very strong association with survival in this subtype, and may be used to select potential responders to immunotherapy.
Collapse
Affiliation(s)
- Ming-Jun Shi
- Institut Curie, PSL Research University, CNRS, UMR 144, Paris F-75005, France.,Department of Urology, Beijing Friendship Hospital, Capital Medical Univeristy , Beijing, 430071, People's Republic of China
| | - Xiang-Yu Meng
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, People's Republic of China.,Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, People's Republic of China
| | - Qiu-Ji Wu
- Department of Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, People's Republic of China
| | - Xiong-Hui Zhou
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, People's Republic of China
| |
Collapse
|
1040
|
Immunotherapeutics in Multiple Myeloma: How Can Translational Mouse Models Help? JOURNAL OF ONCOLOGY 2019; 2019:2186494. [PMID: 31093282 PMCID: PMC6481018 DOI: 10.1155/2019/2186494] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 03/04/2019] [Indexed: 12/30/2022]
Abstract
Multiple myeloma (MM) is usually diagnosed in older adults at the time of immunosenescence, a collection of age-related changes in the immune system that contribute to increased susceptibility to infection and cancer. The MM tumor microenvironment and cumulative chemotherapies also add to defects in immunity over the course of disease. In this review we discuss how mouse models have furthered our understanding of the immune defects caused by MM and enabled immunotherapeutics to progress to clinical trials, but also question the validity of using immunodeficient models for these purposes. Immunocompetent models, in particular the 5T series and Vk⁎MYC models, are increasingly being utilized in preclinical studies and are adding to our knowledge of not only the adaptive immune system but also how the innate system might be enhanced in anti-MM activity. Finally we discuss the concept of immune profiling to target patients who might benefit the most from immunotherapeutics, and the use of humanized mice and 3D culture systems for personalized medicine.
Collapse
|
1041
|
Duong HTT, Thambi T, Yin Y, Lee JE, Seo YK, Jeong JH, Lee DS. Smart pH-Responsive Nanocube-Controlled Delivery of DNA Vaccine and Chemotherapeutic Drugs for Chemoimmunotherapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:13058-13068. [PMID: 30888149 DOI: 10.1021/acsami.8b21185] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The combination of chemotherapeutic agents with immune stimulating agents for treating degenerative diseases, called chemoimmunotherapy, has emerged as a promising cancer treatment modality. Despite the tremendous potential, chemoimmunotherapy by the combination of drugs and immune stimulators often suffers because of the lack of controlled delivery nanostructures in the microenvironment. To this end, we show that by using pH-responsive smart nanocubes (NCs), cancer cells and tumor-associated immune cells can be precisely targeted with a chemotherapeutic agent (doxorubicin, DOX) and immune stimulating agent (plasmid ovalbumin, pOVA) for enhanced chemoimmunotherapy. The pH-responsive smart NCs protect payloads from nuclease degradation and avoid renal clearance and undergo supersensitive structural change at the extracellular tumor regions that mediate efficient release. Concurrent release of pOVA vaccines encoding tumor-specific antigen laden with polyplexes were loaded on tumor-associated immune cells and produce antigen-specific humoral immune response, whereas DOX enables effective infiltration into the cancer cells and is involved in the eradication of tumor tissues. The amount of anti-OVA IgG1 antibody produced by the intravenous administration of NC formulation was similar to that of free OVA formulation. Importantly, the combined delivery of pDNA and DOX using NCs showed significantly enhanced antitumor efficacy in B16/OVA melanoma tumor xenografts, which remarkably outperforms the monotherapy counterparts. These results suggest that pH-responsive smart NCs laden with pDNA and DOX provide a promising nanostructure for chemoimmunotherapy that simultaneously involves cancer cell killing and stimulates antigen-specific immune response to prevent cancer recurrence.
Collapse
|
1042
|
Tang C, Makusheva Y, Sun H, Han W, Iwakura Y. Myeloid C-type lectin receptors in skin/mucoepithelial diseases and tumors. J Leukoc Biol 2019; 106:903-917. [PMID: 30964564 PMCID: PMC6850291 DOI: 10.1002/jlb.2ri0119-031r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/11/2019] [Accepted: 03/20/2019] [Indexed: 12/30/2022] Open
Abstract
Myeloid C‐type lectin receptors (CLRs), which consist of an extracellular carbohydrate recognition domain and intracellular signal transducing motif such as the immunoreceptor tyrosine‐based activation motif (ITAM) or immunoreceptor tyrosine‐based inhibitory motif (ITIM), are innate immune receptors primarily expressed on myeloid lineage cells such as dendritic cells (DCs) and Mϕs. CLRs play important roles in host defense against infection by fungi and bacteria by recognizing specific carbohydrate components of these pathogens. However, these immune receptors also make important contributions to immune homeostasis of mucosa and skin in mammals by recognizing components of microbiota, as well as by recognizing self‐components such as alarmins from dead cells and noncanonical non‐carbohydrate ligands. CLR deficiency not only induces hypersensitivity to infection, but also causes dysregulation of muco‐cutaneous immune homeostasis, resulting in the development of allergy, inflammation, autoimmunity, and tumors. In this review, we introduce recent discoveries regarding the roles of myeloid CLRs in the immune system exposed to the environment, and discuss the roles of these lectin receptors in the development of colitis, asthma, psoriasis, atopic dermatitis, and cancer. Although some CLRs are suggested to be involved in the development of these diseases, the function of CLRs and their ligands still largely remain to be elucidated.
Collapse
Affiliation(s)
- Ce Tang
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yulia Makusheva
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Haiyang Sun
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Wei Han
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| | - Yoichiro Iwakura
- Center for Animal Disease Models, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Japan
| |
Collapse
|
1043
|
Rezalotfi A, Ahmadian E, Aazami H, Solgi G, Ebrahimi M. Gastric Cancer Stem Cells Effect on Th17/Treg Balance; A Bench to Beside Perspective. Front Oncol 2019; 9:226. [PMID: 31024835 PMCID: PMC6464032 DOI: 10.3389/fonc.2019.00226] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 03/13/2019] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer stem cells (GCSCs), a small population among tumor cells, are responsible for tumor initiation, development, metastasis, and recurrence. They play a crucial role in immune evasion, immunomodulation, and impairment of effector immunity and believed to be emerged to change the balance of the immune system, importantly CD4+ T cells in the chronic inflamed tumor site. However, different subtypes of innate and adaptive immune cells are involved in the formation of the immune system in the tumor microenvironment, we would look at T cells in this study. Tumor microenvironment induces differentiation of CD4+ T cells into different subsets of T cells, mainly suppressive regulatory T cells (Treg), and T helper 17 (Th17) cells, although their exact role in tumor immunity is still under debate depending on tumor types and stages. Counterbalance between Th17 and Treg cells in the gastrointestinal system result in the homeostasis and normal function of the immune system, particularly mucosal immunity. Recent data demonstrated a high infiltration of Th17 and Treg cells into the gastric tumor site and proved that tumor microenvironment might disturb the balance between Th17 and Treg. It is possible to assume an association between activation of CSCs which contribute to metastasis in late stages, and the imbalanced Th17/Treg cells observed in advanced gastric cancer patients. This review intends to clarify the importance of gastric tumor microenvironment specifically CSCs in relation to Th17/Tregs balance firstly and to highlight the relevance of imbalanced Th17/Treg subsets in determining the stages and behavior of the tumor secondly. Finally, the present study suggests a clinical approach looking at the plasticity of T cells with a focus on Th17 as a promising dedicated arm in cancer immunotherapy.
Collapse
Affiliation(s)
- Alaleh Rezalotfi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Elmira Ahmadian
- Faculty of Biological Sciences and Technology, Department of Animal Sciences, Shahid Beheshti University, Tehran, Iran
| | - Hossein Aazami
- Metabolic Disorders Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Students Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasem Solgi
- Department of Immunology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
1044
|
Scheller L, Fussenegger M. From synthetic biology to human therapy: engineered mammalian cells. Curr Opin Biotechnol 2019; 58:108-116. [PMID: 30933864 DOI: 10.1016/j.copbio.2019.02.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/07/2019] [Accepted: 02/28/2019] [Indexed: 01/05/2023]
Abstract
Mammalian synthetic biology has evolved to become a key driver of biomedical innovation in the area of cell therapy. Advances in receptor engineering, immunotherapy and cell implants promise new treatment options for complex diseases. Synthetic receptors have already found applications in cellular immunotherapy for cancer treatment, and are being introduced into the field of cell implants. Here, we discuss prospects for the next generation of engineered mammalian cells for human therapy, highlighting selected recent studies.
Collapse
Affiliation(s)
- Leo Scheller
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058, Basel, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, CH-4058, Basel, Switzerland; University of Basel, Faculty of Science, Mattenstrasse 26, CH-4058, Basel, Switzerland.
| |
Collapse
|
1045
|
Abstract
Toll-like receptors (TLRs) are associated with tumor growth and immunosuppression, as well as apoptosis and immune system activation. TLRs can activate apoptosis and innate and adaptive immunity pathways, which can be pharmacologically targeted for the development of anticancer oncotherapies. Several studies and clinical trials indicate that TLR agonists are promising adjuvants or elements of novel therapies, particularly when used in conjunction with chemotherapy or radiotherapy. An increasing number of studies suggest that the activation of TLRs in various cancer types is related to oncotherapy; however, before this finding can be applied to clinical practice, additional studies are required. Research suggests that TLR agonists may have potential applications in cancer therapy; nevertheless, because TLR signaling can also promote tumorigenesis, a critical and comprehensive evaluation of TLR action is warranted. This review focuses on recent studies that have assessed the strengths and weaknesses of utilizing TLR agonists as potential anticancer agents.
Collapse
Affiliation(s)
- Caiqi Liu
- Department of Gastroenterology, Second Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| | - Ci Han
- Department of Critical Care Medicine, Third Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| | - Jinfeng Liu
- Department of Pain, Second Affiliated Hospital of Harbin Medical University, Harbin, P.R. China
| |
Collapse
|
1046
|
Chemin K, Gerstner C, Malmström V. Effector Functions of CD4+ T Cells at the Site of Local Autoimmune Inflammation-Lessons From Rheumatoid Arthritis. Front Immunol 2019; 10:353. [PMID: 30915067 PMCID: PMC6422991 DOI: 10.3389/fimmu.2019.00353] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 02/11/2019] [Indexed: 12/17/2022] Open
Abstract
Infiltration of memory CD4+ T cells in synovial joints of Rheumatoid Arthritis (RA) patients has been reported since decades. Moreover, several genome wide association studies (GWAS) pinpointing a key genetic association between the HLA-DR locus and RA have led to the generally agreed hypothesis that CD4+ T cells are directly implicated in the disease. Still, RA is a heterogeneous disease and much effort has been made to understand its different facets. T cell differentiation is driven by mechanisms including antigen stimulation, co-stimulatory signals and cytokine milieu, all of which are abundant in the rheumatic joint, implying that any T cells migrating into the joint may be further affected locally. In parallel to the characterization and classification of T-cell subsets, the contribution of different effector T cells to RA has been investigated in numerous studies though sometimes with contradictory results. In particular, the frequency of Th1 and Th17 cells has been assessed in the synovial joints with various results that could, at least partly, be explained by the stage of the disease. For regulatory T cells, it is largely accepted that they accumulate in RA synovial fluid and that the equilibrium between regulatory T cells and effector cells is a key factor in controlling inflammation processes involved in RA. Recent phenotypic studies describe the possible implication of a novel subset of peripheral T helper cells (Tph) important for T-B cell cross talk and plasma cell differentiation in the RA joint of ACPA+ (autoantibodies against citrullinated proteins) RA patients. Finally, cytotoxic CD4+ T cells, historically described as increased in the peripheral blood of RA patients have attracted new attention in the last years. In view of the recently identified peripheral T-cell subsets, we will integrate immunological data as well as information on genetic variants and therapeutic strategy outcomes into our current understanding of the width of effector T cells. We will also integrate tissue-resident memory T cell aspects, and discuss similarities and differences with inflammatory conditions in skin (psoriasis) and mucosal organs (Crohn's disease).
Collapse
Affiliation(s)
- Karine Chemin
- Division of Rheumatology, Department of Medicine, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Christina Gerstner
- Division of Rheumatology, Department of Medicine, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Vivianne Malmström
- Division of Rheumatology, Department of Medicine, Karolinska Institute, Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
1047
|
Keenan TE, Burke KP, Van Allen EM. Genomic correlates of response to immune checkpoint blockade. Nat Med 2019; 25:389-402. [PMID: 30842677 PMCID: PMC6599710 DOI: 10.1038/s41591-019-0382-x] [Citation(s) in RCA: 340] [Impact Index Per Article: 56.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 01/29/2019] [Accepted: 02/01/2019] [Indexed: 12/12/2022]
Abstract
Despite impressive durable responses, immune checkpoint inhibitors do not provide a long-term benefit to the majority of patients with cancer. Understanding genomic correlates of response and resistance to checkpoint blockade may enhance benefits for patients with cancer by elucidating biomarkers for patient stratification and resistance mechanisms for therapeutic targeting. Here we review emerging genomic markers of checkpoint blockade response, including those related to neoantigens, antigen presentation, DNA repair, and oncogenic pathways. Compelling evidence also points to a role for T cell functionality, checkpoint regulators, chromatin modifiers, and copy-number alterations in mediating selective response to immune checkpoint blockade. Ultimately, efforts to contextualize genomic correlates of response into the larger understanding of tumor immune biology will build a foundation for the development of novel biomarkers and therapies to overcome resistance to checkpoint blockade.
Collapse
Affiliation(s)
- Tanya E Keenan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kelly P Burke
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
1048
|
Iiizumi S, Ohtake J, Murakami N, Kouro T, Kawahara M, Isoda F, Hamana H, Kishi H, Nakamura N, Sasada T. Identification of Novel HLA Class II-Restricted Neoantigens Derived from Driver Mutations. Cancers (Basel) 2019; 11:cancers11020266. [PMID: 30813491 PMCID: PMC6406322 DOI: 10.3390/cancers11020266] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/30/2022] Open
Abstract
Neoantigens derived from tumor-specific genetic mutations might be suitable targets for cancer immunotherapy because of their high immunogenicity. In the current study, we evaluated the immunogenicity of 10 driver mutations that are frequently expressed in various cancers using peripheral blood mononuclear cells from healthy donors (n = 25). Of the 10 synthetic peptides (27-mer) derived from these mutations, the six peptides from KRAS-G12D, KRAS-G12R, KRAS-G13D, NRAS-Q61R, PIK3CA-H1047R, and C-Kit-D816V induced T cell responses, suggesting that frequent driver mutations are not always less immunogenic. In particular, immune responses to PIK3CA-H1047R, C-Kit-D816V, KRAS-G13D, and NRAS-Q61R were observed in more than 10% of the donors. All six peptides induced human leukocyte antigen (HLA) class II-restricted CD4+ T cell responses; notably, PIK3CA-H1047R contained at least two different CD4+ T cell epitopes restricted to different HLA class II alleles. In addition, PIK3CA-H1047R and C-Kit-D816V induced antigen-specific CD8+ T cells as well, indicating that they might contain both HLA class I- and class II-restricted epitopes. Since the identified neoantigens might be shared by patients with various types of cancers and are not easily lost due to immune escape, they have the potential to be promising off-the-shelf cancer immunotherapy targets in patients with the corresponding mutations.
Collapse
Affiliation(s)
- Susumu Iiizumi
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
- Research & Early Development Division, BrightPath Biotherapeutics Co., Ltd., 3-25-22 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan.
| | - Junya Ohtake
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
| | - Naoko Murakami
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
- Research & Early Development Division, BrightPath Biotherapeutics Co., Ltd., 3-25-22 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan.
| | - Taku Kouro
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
| | - Mamoru Kawahara
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
| | - Fumiko Isoda
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
- Research & Early Development Division, BrightPath Biotherapeutics Co., Ltd., 3-25-22 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan.
| | - Hiroshi Hamana
- Department of Innovative Cancer Immunotherapy, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Hiroyuki Kishi
- Department of Immunology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Norihiro Nakamura
- Research & Early Development Division, BrightPath Biotherapeutics Co., Ltd., 3-25-22 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan.
| | - Tetsuro Sasada
- Department of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa 241-8515, Japan.
| |
Collapse
|
1049
|
Brentville VA, Symonds P, Cook KW, Daniels I, Pitt T, Gijon M, Vaghela P, Xue W, Shah S, Metheringham RL, Durrant LG. T cell repertoire to citrullinated self-peptides in healthy humans is not confined to the HLA-DR SE alleles; Targeting of citrullinated self-peptides presented by HLA-DP4 for tumour therapy. Oncoimmunology 2019; 8:e1576490. [PMID: 31069134 PMCID: PMC6492960 DOI: 10.1080/2162402x.2019.1576490] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/06/2018] [Accepted: 12/11/2018] [Indexed: 11/26/2022] Open
Abstract
Post-translational modifications are induced in stressed cells which cause them to be recognised by the system. One such modification is citrullination where the positive charged arginine is modified to a neutral citrulline. We demonstrate most healthy donors show an oligoclonal CD4 response in vitro to at least one citrullinated vimentin or enolase peptide. Unlike rheumatoid arthritis patients, these T cell responses were not restricted by HLA-DRB1 shared epitope (SE) alleles, suggesting they could be presented by other MHC class II alleles. As HLA-DP is less polymorphic than HLA-DR, we investigated whether the common allele, HLA-DP4 could present citrullinated epitopes. The modification of arginine to citrulline enhanced binding of the peptides to HLA-DP4 and induced high-frequency CD4 responses in HLA-DP4 transgenic mouse models. Our previous studies have shown that tumours present citrullinated peptides restricted through HLA-DR4 which are good targets for anti-tumour immunity. In this study, we show that citrullinated vimentin and enolase peptides also induced strong anti-tumour immunity (100% survival, p < 0.0001) against established B16 tumours and against the LLC/2 lung cancer model (p = 0.034) both expressing HLA-DP4. Since most tumours do not constitutively express MHC class II molecules, models were engineered that expressed MHC class II under the control of an IFNγ inducible promoter. Immunisation with citrullinated peptides resulted in 90% survival (p < 0.001) against established B16 HHD tumour expressing IFNγ inducible DP4. These studies show that citrullinated peptides can be presented by a range of MHC class II molecules, including for the first time HLA-DP4, and are strong targets for anti-tumour immunity.
Collapse
Affiliation(s)
- Victoria A Brentville
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham, UK
| | - Peter Symonds
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham, UK
| | - Katherine W Cook
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham, UK
| | - Ian Daniels
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham, UK
| | - Tracy Pitt
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham, UK
| | - Mohamed Gijon
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham, UK
| | - Poonam Vaghela
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham, UK
| | - Wei Xue
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham, UK
| | - Sabaria Shah
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham, UK
| | - Rachael L Metheringham
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham, UK
| | - Lindy G Durrant
- Scancell Limited, Academic Department of Clinical Oncology, University of Nottingham, City Hospital Campus, Nottingham, UK.,Academic Department of Clinical Oncology, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham, City Hospital Campus, Nottingham, UK
| |
Collapse
|
1050
|
Yamaguchi K. Tacrolimus treatment for infertility related to maternal-fetal immune interactions. Am J Reprod Immunol 2019; 81:e13097. [PMID: 30689243 DOI: 10.1111/aji.13097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 12/29/2022] Open
Abstract
Many approaches have been used to achieve successful pregnancies in patients with infertility, though existing treatments remain unsatisfactory in patients with infertility caused by abnormal maternal-fetal immunity. However, our understanding of the immunological aspects of infertility has steadily progressed, aided by recent research into organ transplantation and cancer. The results of these recent analyses have led to the development and evaluation of several candidate immunological treatments, but the use of immunological treatments remains a novel approach. The current paper presents the hypothesis that tacrolimus may have potential as a candidate agent for the treatment of maternal-fetal immunity-related infertility.
Collapse
Affiliation(s)
- Koushi Yamaguchi
- Center of Maternal-Fetal, Neonatal and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|