301
|
Wang C, Ye Y, Hu Q, Bellotti A, Gu Z. Tailoring Biomaterials for Cancer Immunotherapy: Emerging Trends and Future Outlook. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29. [PMID: 28556553 DOI: 10.1002/adma.201606036] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/04/2017] [Indexed: 05/05/2023]
Abstract
Cancer immunotherapy, as a paradigm shift in cancer treatment, has recently received tremendous attention. The active cancer vaccination, immune checkpoint blockage (ICB) and chimeric antigen receptor (CAR) for T-cell-based adoptive cell transfer are among these developments that have achieved a significant increase in patient survival in clinical trials. Despite these advancements, emerging research at the interdisciplinary interface of cancer biology, immunology, bioengineering, and materials science is important to further enhance the therapeutic benefits and reduce side effects. Here, an overview of the latest studies on engineering biomaterials for the enhancement of anticancer immunity is given, including the perspectives of delivery of immunomodulatory therapeutics, engineering immune cells, and constructing immune-modulating scaffolds. The opportunities and challenges in this field are also discussed.
Collapse
Affiliation(s)
- Chao Wang
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Yanqi Ye
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Quanyin Hu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Adriano Bellotti
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Department of Medicine University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Zhen Gu
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, NC, 27695, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Medicine University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| |
Collapse
|
302
|
Candidate immune biomarkers for radioimmunotherapy. Biochim Biophys Acta Rev Cancer 2017; 1868:58-68. [DOI: 10.1016/j.bbcan.2017.02.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/21/2017] [Accepted: 02/25/2017] [Indexed: 12/25/2022]
|
303
|
|
304
|
Amirouchene-Angelozzi N, Swanton C, Bardelli A. Tumor Evolution as a Therapeutic Target. Cancer Discov 2017; 7:2159-8290.CD-17-0343. [PMID: 28729406 DOI: 10.1158/2159-8290.cd-17-0343] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/22/2017] [Accepted: 06/14/2017] [Indexed: 11/16/2022]
Abstract
Recent technological advances in the field of molecular diagnostics (including blood-based tumor genotyping) allow the measurement of clonal evolution in patients with cancer, thus adding a new dimension to precision medicine: time. The translation of this new knowledge into clinical benefit implies rethinking therapeutic strategies. In essence, it means considering as a target not only individual oncogenes but also the evolving nature of human tumors. Here, we analyze the limitations of targeted therapies and propose approaches for treatment within an evolutionary framework.Significance: Precision cancer medicine relies on the possibility to match, in daily medical practice, detailed genomic profiles of a patient's disease with a portfolio of drugs targeted against tumor-specific alterations. Clinical blockade of oncogenes is effective but only transiently; an approach to monitor clonal evolution in patients and develop therapies that also evolve over time may result in improved therapeutic control and survival outcomes. Cancer Discov; 7(8); 1-13. ©2017 AACR.
Collapse
Affiliation(s)
| | - Charles Swanton
- University College London Cancer Institute and The Francis Crick Institute, London, United Kingdom
| | - Alberto Bardelli
- Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia (FPO), IRCCS, Candiolo, Torino, Italy.
- Department of Oncology, University of Torino, Torino, Italy
| |
Collapse
|
305
|
Synthetic melanin bound to subunit vaccine antigens significantly enhances CD8+ T-cell responses. PLoS One 2017; 12:e0181403. [PMID: 28715455 PMCID: PMC5513539 DOI: 10.1371/journal.pone.0181403] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 07/02/2017] [Indexed: 02/07/2023] Open
Abstract
Cytotoxic T-lymphocytes (CTLs) play a key role in immunity against cancer; however, the induction of CTL responses with currently available vaccines remains difficult. Because several reports have suggested that pigmentation and immunity might be functionally linked, we investigated whether melanin can act as an adjuvant in vaccines. Short synthetic peptides (8-35 amino acids long) containing T-cell epitopes were mixed with a solution of L-Dopa, a precursor of melanin. The mixture was then oxidized to generate nanoparticles of melanin-bound peptides. Immunization with melanin-bound peptides efficiently triggered CTL responses in mice, even against self-antigens and at a very low dose of peptides (microgram range). Immunization against a tumor antigen inhibited the growth of established tumors in mice, an effect that was abrogated by the depletion of CD8+ lymphocytes. These results demonstrate the efficacy of melanin as a vaccine adjuvant.
Collapse
|
306
|
Zhang Q, Yuan XF, Lu Y, Li ZZ, Bao SQ, Zhang XL, Yang YY, Fan DM, Zhang YZ, Wu CX, Guo HX, Zhang YJ, Ye Z, Xiong DS. Surface expression of anti-CD3scfv stimulates locoregional immunotherapy against hepatocellular carcinoma depending on the E1A-engineered human umbilical cord mesenchymal stem cells. Int J Cancer 2017. [PMID: 28643325 DOI: 10.1002/ijc.30846] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Tumor antigens is at the core of cancer immunotherapy, however, the ideal antigen selection is difficult especially in poorly immunogenic tumors. In this study, we designed a strategy to modify hepatocellular carcinoma (HCC) cells by surface expressing anti-CD3scfv within the tumor site strictly, which depended on the E1A-engineered human umbilical cord mesenchymal stem cells (HUMSC.E1A) delivery system. Subsequently, membrane-bound anti-CD3scfv actived the lymphocytes which lysed HCC cells bypassing the expression of antigens or MHC restriction. First, we constructed the anti-CD3scfv gene driven by human α-fetoprotein (AFP) promoter into an adenoviral vector and the E1A gene into the lentiviral vector. Our results showed that anti-CD3scfv could specifically express on the surface of HCC cells and activate the lymphocytes to kill target cells effectively in vitro. HUMSC infected by AdCD3scfv followed by LentiR.E1A could support the adenoviral replication and packaging in vitro 36 h after LentiR.E1A infection. Using a subcutaneous HepG2 xenograft model, we confirmed that AdCD3scfv and LentiR.E1A co-transfected HUMSC could migrate selectively to the tumor site and produce considerable adenoviruses. The new generated AdCD3scfv infected and modified tumor cells successfully. Mice injected with the MSC.E1A.AdCD3scfv and lymphocytes significantly inhibited the tumor growth compared with control groups. Furthermore, 5-fluorouracil (5-FU) could sensitize adenovirus infection at low MOI resulting in improved lymphocytes cytotoxicity in vitro and in vivo. In summary, this study provides a promising strategy for solid tumor immunotherapy.
Collapse
Affiliation(s)
- Qing Zhang
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Xiang-Fei Yuan
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.,Institute of Integrative Medicine for Acute Abdominal Diseases, Nankai Hospital, Tianjin, 300100, China
| | - Yang Lu
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Zhen-Zhen Li
- National-Local Joint Engineering Research Center of Biodiagnostics & Biotherapy, The Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Shi-Qi Bao
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Xiao-Long Zhang
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Yuan-Yuan Yang
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Dong-Mei Fan
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Yi-Zhi Zhang
- Central Hospital of Karamay, Karamay, Xinjiang, 834000, China
| | - Chen-Xuan Wu
- the Third Central Hospital of Tianjin Medical University, Tianjin, 300170, China
| | - Hong-Xing Guo
- the Third Central Hospital of Tianjin Medical University, Tianjin, 300170, China
| | - Yan-Jun Zhang
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Zhou Ye
- Central Hospital of Karamay, Karamay, Xinjiang, 834000, China
| | - Dong-Sheng Xiong
- State Key Laboratory of Experimental Hematology, Department of Pharmacy, Institute of Hematology & Hospital of Blood Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| |
Collapse
|
307
|
Rationally combining immunotherapies to improve efficacy of immune checkpoint blockade in solid tumors. Cytokine Growth Factor Rev 2017; 36:5-15. [PMID: 28693973 DOI: 10.1016/j.cytogfr.2017.06.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Revised: 06/19/2017] [Accepted: 06/20/2017] [Indexed: 12/13/2022]
Abstract
With the widespread application of immune checkpoint blocking antibodies (ICBs) for the treatment of advanced cancer, immunotherapy has proven to be capable of yielding unparalleled clinical results. However, despite the initial success of ICB-treatment, still a minority of patients experience durable responses to ICB therapy. A plethora of mechanisms underlie ICB resistance ranging from low immunogenicity, inadequate generation or recruitment of tumor-specific T cells or local suppression by stromal cells to acquired genetic alterations leading to immune escape. Increasing the response rates to ICBs requires insight into the mechanisms underlying resistance and the subsequent design of rational therapeutic combinations on a per patient basis. In this review, we aim to establish order into the mechanisms governing primary and secondary ICB resistance, offer therapeutic options to circumvent different modes of resistance and plea for a personalized medicine approach to maximize immunotherapeutic benefit for all cancer patients.
Collapse
|
308
|
Donia M, Harbst K, van Buuren M, Kvistborg P, Lindberg MF, Andersen R, Idorn M, Munir Ahmad S, Ellebæk E, Mueller A, Fagone P, Nicoletti F, Libra M, Lauss M, Hadrup SR, Schmidt H, Andersen MH, Thor Straten P, Nilsson JA, Schumacher TN, Seliger B, Jönsson G, Svane IM. Acquired Immune Resistance Follows Complete Tumor Regression without Loss of Target Antigens or IFNγ Signaling. Cancer Res 2017; 77:4562-4566. [PMID: 28655789 DOI: 10.1158/0008-5472.can-16-3172] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 04/08/2017] [Accepted: 06/22/2017] [Indexed: 11/16/2022]
Abstract
Cancer immunotherapy can result in durable tumor regressions in some patients. However, patients who initially respond often experience tumor progression. Here, we report mechanistic evidence of tumoral immune escape in an exemplary clinical case: a patient with metastatic melanoma who developed disease recurrence following an initial, unequivocal radiologic complete regression after T-cell-based immunotherapy. Functional cytotoxic T-cell responses, including responses to one mutant neoantigen, were amplified effectively with therapy and generated durable immunologic memory. However, these immune responses, including apparently effective surveillance of the tumor mutanome, did not prevent recurrence. Alterations of the MHC class I antigen-processing and presentation machinery (APM) in resistant cancer cells, but not antigen loss or impaired IFNγ signaling, led to impaired recognition by tumor-specific CD8+ T cells. Our results suggest that future immunotherapy combinations should take into account targeting cancer cells with intact and impaired MHC class I-related APM. Loss of target antigens or impaired IFNγ signaling does not appear to be mandatory for tumor relapse after a complete radiologic regression. Personalized studies to uncover mechanisms leading to disease recurrence within each individual patient are warranted. Cancer Res; 77(17); 4562-6. ©2017 AACR.
Collapse
Affiliation(s)
- Marco Donia
- Department of Hematology, Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark. .,Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Katja Harbst
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Sweden
| | - Marit van Buuren
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Pia Kvistborg
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Rikke Andersen
- Department of Hematology, Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark.,Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Manja Idorn
- Department of Hematology, Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark
| | - Shamaila Munir Ahmad
- Department of Hematology, Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark
| | - Eva Ellebæk
- Department of Hematology, Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark.,Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Anja Mueller
- Institute for Medical Immunology, Martin Luther University, Halle-Wittenberg, Germany
| | - Paolo Fagone
- Department of Bio-medical Sciences, University of Catania, Catania, Italy
| | | | - Massimo Libra
- Department of Bio-medical Sciences, University of Catania, Catania, Italy
| | - Martin Lauss
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Sweden
| | - Sine Reker Hadrup
- Department of Hematology, Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark
| | - Henrik Schmidt
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Mads Hald Andersen
- Department of Hematology, Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark
| | - Per Thor Straten
- Department of Hematology, Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark
| | - Jonas A Nilsson
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Ton N Schumacher
- Sahlgrenska Cancer Center, University of Gothenburg, Gothenburg, Sweden
| | - Barbara Seliger
- Institute for Medical Immunology, Martin Luther University, Halle-Wittenberg, Germany
| | - Göran Jönsson
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Faculty of Medicine, Lund University, Sweden
| | - Inge Marie Svane
- Department of Hematology, Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark. .,Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
309
|
Kunert A, Obenaus M, Lamers CHJ, Blankenstein T, Debets R. T-cell Receptors for Clinical Therapy: In Vitro Assessment of Toxicity Risk. Clin Cancer Res 2017. [PMID: 28645940 DOI: 10.1158/1078-0432.ccr-17-1012] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Adoptive therapy with T-cell receptor (TCR)-engineered T cells has shown promising results in the treatment of patients with tumors, and the number of TCRs amenable for clinical testing is expanding rapidly. Notably, adoptive therapy with T cells is challenged by treatment-related side effects, which calls for cautious selection of target antigens and TCRs that goes beyond their mere ability to induce high T-cell reactivity. Here, we propose a sequence of in vitro assays to improve selection of TCRs and exemplify risk assessments of on-target as well as off-target toxicities using TCRs directed against cancer germline antigens. The proposed panel of assays covers parameters considered key to safety, such as expression of target antigen in healthy tissues, determination of a TCR's recognition motif toward its cognate peptide, and a TCR's cross-reactivity toward noncognate peptides. Clin Cancer Res; 23(20); 6012-20. ©2017 AACR.
Collapse
Affiliation(s)
- Andre Kunert
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | - Matthias Obenaus
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Institute of Immunology, Charité Campus Buch, Berlin, Germany
| | - Cor H J Lamers
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Thomas Blankenstein
- Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.,Institute of Immunology, Charité Campus Buch, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Reno Debets
- Laboratory of Tumor Immunology, Department of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| |
Collapse
|
310
|
Doorduijn EM, Sluijter M, Salvatori DC, Silvestri S, Maas S, Arens R, Ossendorp F, van der Burg SH, van Hall T. CD4 + T Cell and NK Cell Interplay Key to Regression of MHC Class I low Tumors upon TLR7/8 Agonist Therapy. Cancer Immunol Res 2017. [PMID: 28637878 DOI: 10.1158/2326-6066.cir-16-0334] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
One of the next challenges in cancer immunotherapy is the resistance of tumors to T-cell-based treatments through loss of MHC class I. Here, we show that under these circumstances, the Toll-like receptor (TLR)-7/8 ligand imiquimod, but not the TLR3 ligand poly I:C or TLR9 ligand CpG, mediated an effective antitumor response. The rejection of these immune-escaped cancers was mediated by NK cells and CD4+ T cells, whereas activated CD8+ T cells were dispensable. Application of the innate immune stimulator at a distant site activated NK cells and thereby elicited tumor-specific T-cell responses in tumor-bearing mice. Mechanistically, imiquimod activated NK cells to kill tumor cells, resulting in release of tumor antigens and induction of tumor-specific CD4+ T cells. These T helper cells provoked a strong induction of CXCL9 and CXCL10 in the tumor environment. Simultaneously, imiquimod induced the expression of the cognate chemokine receptor CXCR3 on peripheral lymphocytes. This ignited intratumoral CD4+ T-cell infiltration and accumulation, which was critical for tumor rejection; CXCR3 blocking antibodies mitigated the clinical response. In the effector phase, NK cell recruitment to tumors and their activation depended on CD4+ T cells. Together, we have uncovered a potent immune axis of tumor-specific CD4+ T cells and NK cells that eliminates escaped MHC-Ilow tumors. Cancer Immunol Res; 5(8); 642-53. ©2017 AACR.
Collapse
Affiliation(s)
- Elien M Doorduijn
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Marjolein Sluijter
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Daniela C Salvatori
- Central Laboratory Animal Facility, Leiden University Medical Center (LUMC), Leiden, the Netherlands.,Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Serenella Silvestri
- Central Laboratory Animal Facility, Leiden University Medical Center (LUMC), Leiden, the Netherlands.,Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Saskia Maas
- Central Laboratory Animal Facility, Leiden University Medical Center (LUMC), Leiden, the Netherlands.,Department of Anatomy and Embryology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Ramon Arens
- Immunohematology and Blood Transfusion, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Ferry Ossendorp
- Immunohematology and Blood Transfusion, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands.
| |
Collapse
|
311
|
Matsushita H, Hasegawa K, Oda K, Yamamoto S, Nishijima A, Imai Y, Asada K, Ikeda Y, Karasaki T, Fujiwara K, Aburatani H, Kakimi K. The frequency of neoantigens per somatic mutation rather than overall mutational load or number of predicted neoantigens per se is a prognostic factor in ovarian clear cell carcinoma. Oncoimmunology 2017; 6:e1338996. [PMID: 28920005 DOI: 10.1080/2162402x.2017.1338996] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 06/02/2017] [Indexed: 01/21/2023] Open
Abstract
Neoantigens derived from tumor-specific somatic mutations are excellent targets for anti-tumor immune responses. In ovarian clear cell carcinoma (OCCC), checkpoint blockade yields durable responses in a subset of patients. To approach the question of why only some patients respond, we first investigated neoantigen loads and immune signatures using exome sequencing and expression array data for 74 OCCC patients treated conventionally. Neither the number of missense mutations nor total predicted neoantigens assessed in the tumor correlated with clinical outcomes. However, the number of neoantigens per missense mutation ("neoAg frequency") did correlate with clinical outcomes. Cox multivariate regression analysis demonstrated that low neoAg frequencies correlated with increased progression-free survival (PFS) and was an independent predictive factor for PFS in OCCC (p = 0.032), especially at stage I-II (p = 0.0045). Immunity-associated genes including those related to effector memory CD8 T cells were dominantly expressed in tumors with low neoAg frequencies in stage I-II patients, suggesting CD8 T cell-mediated elimination of immunogenic sub-clones expressing neoantigens (immunoediting) had occurred. In contrast, we observed decreased HLA-A, -B, and -C expression (p = 0.036, p = 0.026, and p = 0.030, respectively) as well as increased ratios of CTLA-4, PD-1, Tim-3, and LAG3 to CD8A expression (p = 0.0064, p = 0.017, p = 0.033 and p = 0.0136, respectively) in stage I-II tumors with high neoAg frequencies. Constrained anti-tumor immunity may thus result in limited immunoediting, and poor prognosis. Our results show that neoAg frequency in OCCC is an independent prognostic factor for clinical outcome and may become a potential candidate biomarker for immunomodulatory agent-based treatments.
Collapse
Affiliation(s)
- Hirokazu Matsushita
- Department of Immunotherapeutics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan.,Gynecologic Oncology Translational Research Unit, Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, Japan
| | - Katsutoshi Oda
- Department of Obstetrics and Gynecology, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Shogo Yamamoto
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo, Japan
| | - Akira Nishijima
- Department of Obstetrics and Gynecology, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan.,Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo, Japan
| | - Yuichi Imai
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan.,Gynecologic Oncology Translational Research Unit, Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, Japan
| | - Kayo Asada
- Department of Obstetrics and Gynecology, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan.,Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo, Japan
| | - Yuji Ikeda
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan.,Gynecologic Oncology Translational Research Unit, Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, Japan.,Department of Obstetrics and Gynecology, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Takahiro Karasaki
- Department of Immunotherapeutics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Saitama, Japan.,Gynecologic Oncology Translational Research Unit, Project Research Division, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
312
|
Zhang X, Kim S, Hundal J, Herndon JM, Li S, Petti AA, Soysal SD, Li L, McLellan MD, Hoog J, Primeau T, Myers N, Vickery TL, Sturmoski M, Hagemann IS, Miller CA, Ellis MJ, Mardis ER, Hansen T, Fleming TP, Goedegebuure SP, Gillanders WE. Breast Cancer Neoantigens Can Induce CD8 + T-Cell Responses and Antitumor Immunity. Cancer Immunol Res 2017; 5:516-523. [PMID: 28619968 DOI: 10.1158/2326-6066.cir-16-0264] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Revised: 04/06/2017] [Accepted: 05/31/2017] [Indexed: 11/16/2022]
Abstract
Next-generation sequencing technologies have provided insights into the biology and mutational landscape of cancer. Here, we evaluate the relevance of cancer neoantigens in human breast cancers. Using patient-derived xenografts from three patients with advanced breast cancer (xenografts were designated as WHIM30, WHIM35, and WHIM37), we sequenced exomes of tumor and patient-matched normal cells. We identified 2,091 (WHIM30), 354 (WHIM35), and 235 (WHIM37) nonsynonymous somatic mutations. A computational analysis identified and prioritized HLA class I-restricted candidate neoantigens expressed in the dominant tumor clone. Each candidate neoantigen was evaluated using peptide-binding assays, T-cell cultures that measure the ability of CD8+ T cells to recognize candidate neoantigens, and preclinical models in which we measured antitumor immunity. Our results demonstrate that breast cancer neoantigens can be recognized by the immune system, and that human CD8+ T cells enriched for prioritized breast cancer neoantigens were able to protect mice from tumor challenge with autologous patient-derived xenografts. We conclude that next-generation sequencing and epitope-prediction strategies can identify and prioritize candidate neoantigens for immune targeting in breast cancer. Cancer Immunol Res; 5(7); 516-23. ©2017 AACR.
Collapse
Affiliation(s)
- Xiuli Zhang
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Samuel Kim
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Jasreet Hundal
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - John M Herndon
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Shunqiang Li
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Allegra A Petti
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Savas D Soysal
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri.,Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Lijin Li
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Mike D McLellan
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | - Jeremy Hoog
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Tina Primeau
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Nancy Myers
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Tammi L Vickery
- Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri
| | - Mark Sturmoski
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - Ian S Hagemann
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Chris A Miller
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri.,Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Matthew J Ellis
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri.,Lester and Sue Smith Breast Care Center, Oncology/Medicine and MCB, Baylor College of Medicine, Houston, Texas
| | - Elaine R Mardis
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri.,Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Ted Hansen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri
| | - Timothy P Fleming
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - S Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri
| | - William E Gillanders
- Department of Surgery, Washington University School of Medicine, St. Louis, Missouri. .,The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
313
|
Balan S, Finnigan J, Bhardwaj N. Dendritic Cell Strategies for Eliciting Mutation-Derived Tumor Antigen Responses in Patients. Cancer J 2017; 23:131-137. [PMID: 28410301 PMCID: PMC5520811 DOI: 10.1097/ppo.0000000000000251] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Dendritic cells (DCs) are equipped for sensing danger signals and capturing, processing, and presenting antigens to naive or effector cells and are critical in inducing humoral and adaptive immunity. Successful vaccinations are those that activate DCs to elicit both cellular and humoral responses, as well as long-lasting memory response against the target of interest. Recently, it has become apparent that tumor cells can provide new sources of antigens through nonsynonymous mutations or frame-shift mutations, leading to potentially hundreds of mutation-derived tumor antigens (MTAs) or neoantigens. T cells recognizing MTA have been detected in cancer patients and can even lead to tumor regression. Designing MTA-specific vaccination strategies will have to take into account the adjuvant activity of DC subsets and the best formulation to elicit an effective immune response. We discuss the potential of human DCs to prime MTA-specific responses.
Collapse
Affiliation(s)
- Sreekumar Balan
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY NY
| | - John Finnigan
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY NY
| | - Nina Bhardwaj
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, NY NY
| |
Collapse
|
314
|
Abstract
Development of "immune-based targeted therapy" in oncology has limited experience with signal pathway modulation. However, as we have become better versed in understanding immune function related to anticancer response, "hints" of specific targets associated with sensitivity and resistance have been identified with targeted immune therapy. This brief review summarizes the relationship of several targeted immune therapeutics and activity associated clinical responsiveness.
Collapse
Affiliation(s)
| | - John Nemunaitis
- Gradalis, Inc., Dallas, TX, USA.,Mary Crowley Cancer Research Center, Dallas, TX, USA
| |
Collapse
|
315
|
Kanuma T, Yamamoto T, Kobiyama K, Moriishi E, Masuta Y, Kusakabe T, Ozasa K, Kuroda E, Jounai N, Ishii KJ. CD63-Mediated Antigen Delivery into Extracellular Vesicles via DNA Vaccination Results in Robust CD8+ T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2017; 198:4707-4715. [DOI: 10.4049/jimmunol.1600731] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 04/11/2017] [Indexed: 12/16/2022]
|
316
|
Lu Z, Zhang C, Sheng J, Shen J, Liu B. T cell receptor β-chain repertoire analysis reveals the association between neoantigens and tumour-infiltrating lymphocytes in multifocal papillary thyroid carcinoma. Int J Cancer 2017; 141:377-382. [PMID: 28431188 DOI: 10.1002/ijc.30743] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/12/2017] [Accepted: 03/14/2017] [Indexed: 12/30/2022]
Affiliation(s)
- Zheming Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing); Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute; No. 52 Fucheng Road, Haidian District Beijing 100142 China
| | - Chaoting Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing); Department of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute; No. 52 Fucheng Road, Haidian District Beijing 100142 China
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing); Laboratory of Genetics, Peking University Cancer Hospital & Institute; No. 52 Fucheng Road, Haidian District Beijing 100142 China
| | - Jindong Sheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing); Department of Head & Neck Surgery, Peking University Cancer Hospital & Institute; No. 52 Fucheng Road, Haidian District Beijing 100142 China
| | - Jing Shen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing); Core laboratory, Peking University Cancer Hospital & Institute; No. 52 Fucheng Road, Haidian District Beijing 100142 China
| | - Baoguo Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing); Department of Head & Neck Surgery, Peking University Cancer Hospital & Institute; No. 52 Fucheng Road, Haidian District Beijing 100142 China
| |
Collapse
|
317
|
Munn DH, Sharma MD, Johnson TS, Rodriguez P. IDO, PTEN-expressing Tregs and control of antigen-presentation in the murine tumor microenvironment. Cancer Immunol Immunother 2017; 66:1049-1058. [PMID: 28488123 DOI: 10.1007/s00262-017-2010-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 05/02/2017] [Indexed: 01/16/2023]
Abstract
The tumor microenvironment is profoundly immunosuppressive. This creates a major barrier for attempts to combine immunotherapy with conventional chemotherapy or radiation, because the tumor antigens released by these cytotoxic agents are not cross-presented in an immunogenic fashion. In this Focused Research Review, we focus on mouse preclinical studies exploring the role of immunosuppressive Tregs expressing the PTEN lipid phosphatase, and the links between PTEN+ Tregs and the immunoregulatory enzyme indoleamine 2,3-dioxygenase (IDO). IDO has received attention because it can be expressed by a variety of human tumor types in vivo, but IDO can also be induced in host immune cells of both humans and mice in response to inflammation, infection or dying (apoptotic) cells. Mechanistically, IDO and PTEN+ Tregs are closely connected, with IDO causing activation of the PTEN pathway in Tregs. Genetic ablation or pharmacologic inhibition of PTEN in mouse Tregs destabilizes their suppressive phenotype, and this prevents transplantable and autochthonous tumors from creating their normal immunosuppressive microenvironment. Genetic ablation of either IDO or PTEN+ Tregs in mice results in a fundamental defect in the ability to maintain tolerance to antigens associated with apoptotic cells, including dying tumor cells. Consistent with this, pharmacologic inhibitors of either pathway show synergy when combined with cytotoxic agents such as chemotherapy or radiation. Thus, we propose that IDO and PTEN+ Tregs represent closely linked checkpoints that can influence the choice between immune activation versus tolerance to dying tumor cells.
Collapse
Affiliation(s)
- David H Munn
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Room CN4141, Augusta, GA, 30912, USA. .,Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA.
| | - Madhav D Sharma
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Room CN4141, Augusta, GA, 30912, USA.,Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Theodore S Johnson
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Room CN4141, Augusta, GA, 30912, USA.,Department of Pediatrics, Medical College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Paulo Rodriguez
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Room CN4141, Augusta, GA, 30912, USA
| |
Collapse
|
318
|
Case Report: Immune-mediated Complete Response in a Patient With Recurrent Advanced Ewing Sarcoma (EWS) After Vigil Immunotherapy. J Pediatr Hematol Oncol 2017; 39:e183-e186. [PMID: 28338569 DOI: 10.1097/mph.0000000000000822] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ewing sarcoma is a highly resistant disease with a <10% chance of survival at 5 years after failure of frontline chemotherapy. This is a case report of an Ewing sarcoma patient with metastatic disease recurrence <2 years after standard chemotherapy/radiation who achieved a durable and sustained complete response after 2 series of treatments with Vigil (GMCSF/bi-shRNA furin DNA autologous tumor immunotherapy) serially manufactured from first and second recurrences with ELISPOT assay correlation. Results support justification of further testing of Vigil with ELISPOT assay as a biomarker to assess level of immune response and correlation with disease control.
Collapse
|
319
|
Wang Q, Wu X. Primary and acquired resistance to PD-1/PD-L1 blockade in cancer treatment. Int Immunopharmacol 2017; 46:210-219. [DOI: 10.1016/j.intimp.2017.03.015] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 03/12/2017] [Accepted: 03/14/2017] [Indexed: 01/08/2023]
|
320
|
NY-ESO-1 Protein Cancer Vaccine With Poly-ICLC and OK-432: Rapid and Strong Induction of NY-ESO-1-specific Immune Responses by Poly-ICLC. J Immunother 2017; 40:140-147. [PMID: 28338507 DOI: 10.1097/cji.0000000000000162] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We conducted a clinical trial of a cancer vaccine using NY-ESO-1 protein with polyinosinic-polycytidylic acid-poly-L-lysine carboxymethylcellulose (poly-ICLC) and/or OK-432 against solid tumors. A total of 15 patients were sequentially enrolled in 4 cohorts. Patients in cohort 1 received NY-ESO-1 protein; cohort 2a received NY-ESO-1 protein+OK-432; cohort 2b received NY-ESO-1 protein+poly-ICLC; cohort 3 received NY-ESO-1 protein+OK-432+poly-ICLC with Montanide ISA-51. The endpoints of this trial were safety, NY-ESO-1 immune responses, and clinical response. Vaccine-related adverse events observed were fever and injection-site reaction (grade 1). Two patients showed stable disease after vaccination. NY-ESO-1 antibodies were observed in 4 patients at the baseline (sero-positive) and augmented in all patients after vaccination. Eleven patients showed a conversion of negative antibody responses at baseline to positive after vaccination (seroconversion). The seroconversions were observed in all 11 sero-negative patients by the fourth immunization; in particular, it was observed by the second immunization in patients with poly-ICLC, and these induced antibody responses were stronger than those in patients immunized without poly-ICLC. The number of NY-ESO-1-specific interferon (IFN)γ-producing T cells was increased in patients immunized with poly-ICLC and/or OK-432, and furthermore, the increase of IFNγ-producing CD8 T cells in patients immunized with poly-ICLC was significantly higher than that in patients without poly-ICLC. Nonspecific activations of T-cell or antigen presenting cells were not observed. Our present study showed that poly-ICLC is a promising adjuvant for cancer vaccines.
Collapse
|
321
|
Pfeifer CR, Alvey CM, Irianto J, Discher DE. Genome variation across cancers scales with tissue stiffness - an invasion-mutation mechanism and implications for immune cell infiltration. ACTA ACUST UNITED AC 2017; 2:103-114. [PMID: 29082336 DOI: 10.1016/j.coisb.2017.04.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Many different types of soft and solid tumors have now been sequenced, and meta-analyses suggest that genomic variation across tumors scales with the stiffness of the tumors' tissues of origin. The opinion expressed here is based on a review of current genomics data, and it considers multiple 'mechanogenomics' mechanisms to potentially explain this scaling of mutation rate with tissue stiffness. Since stiff solid tissues have higher density of fibrous collagen matrix, which should decrease tissue porosity, cancer cell proliferation could be affected and so could invasion into stiff tissues as the nucleus is squeezed sufficiently to enhance DNA damage. Diversification of a cancer genome after constricted migration is now clear. Understanding genome changes that give rise to neo-antigens is important to selection as well as to the development of immunotherapies, and we discuss engineered monocytes/macrophages as particularly relevant to understanding infiltration into solid tumors.
Collapse
Affiliation(s)
- Charlotte R Pfeifer
- Physical Sciences Oncology Center at Penn (PSOC@Penn), University of Pennsylvania, Philadelphia, PA 19104.,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104.,Graduate Group / Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA 19104
| | - Cory M Alvey
- Physical Sciences Oncology Center at Penn (PSOC@Penn), University of Pennsylvania, Philadelphia, PA 19104.,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104.,Graduate Group / Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104
| | - Jerome Irianto
- Physical Sciences Oncology Center at Penn (PSOC@Penn), University of Pennsylvania, Philadelphia, PA 19104.,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104
| | - Dennis E Discher
- Physical Sciences Oncology Center at Penn (PSOC@Penn), University of Pennsylvania, Philadelphia, PA 19104.,Molecular & Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA 19104.,Graduate Group / Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, PA 19104.,Graduate Group / Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
322
|
Elements of cancer immunity and the cancer-immune set point. Nature 2017; 541:321-330. [PMID: 28102259 DOI: 10.1038/nature21349] [Citation(s) in RCA: 3331] [Impact Index Per Article: 475.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/11/2016] [Indexed: 02/06/2023]
Abstract
Immunotherapy is proving to be an effective therapeutic approach in a variety of cancers. But despite the clinical success of antibodies against the immune regulators CTLA4 and PD-L1/PD-1, only a subset of people exhibit durable responses, suggesting that a broader view of cancer immunity is required. Immunity is influenced by a complex set of tumour, host and environmental factors that govern the strength and timing of the anticancer response. Clinical studies are beginning to define these factors as immune profiles that can predict responses to immunotherapy. In the context of the cancer-immunity cycle, such factors combine to represent the inherent immunological status - or 'cancer-immune set point' - of an individual.
Collapse
|
323
|
Milano G. Resistance to immunotherapy: clouds in a bright sky. Invest New Drugs 2017; 35:649-654. [PMID: 28401366 DOI: 10.1007/s10637-017-0456-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 03/15/2017] [Indexed: 01/05/2023]
Abstract
Two major challenges persist for an optimal management of immunotherapy: i) identifying those patients who will benefit from this type of therapy, and ii) determining the biological, cellular and molecular mechanisms that trigger disease progression while on therapy. There is a consensual view in favor of standardizing practices currently used to measure programmed death ligand 1 (PD-L1) expression that relates to innate resistance. The tumor mutation landscape has been widely explored as a potential predictor of treatment efficacy. In contrast, our knowledge is rather limited as concerns the mechanisms sustaining acquired resistance to checkpoint blockade immunotherapy in patients under treatment. Upregulation of T cell immunoglobulin mucin domain 3 (TIM-3) in CD8+ T-cells has been reported in patients developing acquired resistance to anti-PD-1 treatment. Resistance mechanisms are even more complex for combinatorial strategies linking immunotherapeutic agents and conventional therapies, an area that is expanding rapidly. However, with the arrival of advanced analytical methods such as mass cytometry, there is reason for optimism. These methods can identify cellular mechanisms governing response to therapy and resistance. The clinical use of inhibitors of tumor-microenvironment-modulated pathways, such as those targeting indoleamine 2, 3-dioxygenase (IDO), hold promise for resistance management. Graphical abstract Clouds in a bright sky - Joseph Mallord William Turner.
Collapse
Affiliation(s)
- Gérard Milano
- Oncopharmacology Unit, Centre Antoine-Lacassagne, 33 avenue de Valombrose, 06189, Nice Cedex 2, France.
| |
Collapse
|
324
|
IDO and galectin-3 hamper the ex vivo generation of clinical grade tumor-specific T cells for adoptive cell therapy in metastatic melanoma. Cancer Immunol Immunother 2017; 66:913-926. [PMID: 28401257 PMCID: PMC5489610 DOI: 10.1007/s00262-017-1995-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 03/27/2017] [Indexed: 12/23/2022]
Abstract
Adoptive T cell transfer (ACT) with ex vivo-expanded tumor-reactive T cells proved to be successful for the treatment of metastatic melanoma patients. Mixed lymphocyte tumor cell cultures (MLTC) can be used to generate tumor-specific T cells for ACT; however, in a number of cases tumor-reactive T cell, expansion is far from optimal. We hypothesized that this is due to tumor intrinsic and extrinsic factors and aimed to identify and manipulate these factors so to optimize our clinical, GMP-compliant MLTC protocol. We found that the tumor cell produced IDO and/or galectin-3, and the accumulation of CD4+CD25hiFoxP3+ T cells suppressed the expansion of tumor-specific T cells in the MLTC. Strategies to eliminate CD4+CD25hiFoxP3+ T cells during culture required the depletion of the whole CD4+ T cell population and were found to be undesirable. Blocking of IDO and galectin-3 was feasible and resulted in improved efficiency of the MLTC. Implementation of these findings in clinical protocols for ex vivo expansion of tumor-reactive T cells holds promise for an increased therapeutic potential of adoptive cell transfer treatments with tumor-specific T cells.
Collapse
|
325
|
MHC class II restricted neoantigen: A promising target in tumor immunotherapy. Cancer Lett 2017; 392:17-25. [DOI: 10.1016/j.canlet.2016.12.039] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/22/2016] [Accepted: 12/24/2016] [Indexed: 01/06/2023]
|
326
|
Bhinder B, Elemento O. Towards a better cancer precision medicine: systems biology meets immunotherapy. CURRENT OPINION IN SYSTEMS BIOLOGY 2017; 2:67-73. [PMID: 28989987 PMCID: PMC5628760 DOI: 10.1016/j.coisb.2017.01.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Systems biology approaches that embrace the complexity of cancer are starting to gain traction in the development of new anticancer therapeutic strategies. In this review we describe how genomic analyses are helping improve our understanding of response to immunotherapy, a front-runner in cancer treatment. We argue that systems-level approaches are needed to help understand the concerted impact of tumor-specific and immune-specific molecular features on clinical outcomes, predict responders and unravel the complexity of tumor ecosystems. This integrated approach will propel immunotherapy into the exciting world of precision medicine.
Collapse
Affiliation(s)
- Bhavneet Bhinder
- Department of Physiology and Biophysics, Institute for Computational Biomedicine and Institute for Precision Medicine, Weill Cornell Medical College, 1305 York Avenue, New York, New York 10021, USA
| | - Olivier Elemento
- Department of Physiology and Biophysics, Institute for Computational Biomedicine and Institute for Precision Medicine, Weill Cornell Medical College, 1305 York Avenue, New York, New York 10021, USA
| |
Collapse
|
327
|
Kuai R, Ochyl LJ, Bahjat KS, Schwendeman A, Moon JJ. Designer vaccine nanodiscs for personalized cancer immunotherapy. NATURE MATERIALS 2017; 16:489-496. [PMID: 28024156 PMCID: PMC5374005 DOI: 10.1038/nmat4822] [Citation(s) in RCA: 742] [Impact Index Per Article: 106.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 11/08/2016] [Indexed: 05/04/2023]
Abstract
Despite the tremendous potential of peptide-based cancer vaccines, their efficacy has been limited in humans. Recent innovations in tumour exome sequencing have signalled the new era of personalized immunotherapy with patient-specific neoantigens, but a general methodology for stimulating strong CD8α+ cytotoxic T-lymphocyte (CTL) responses remains lacking. Here we demonstrate that high-density lipoprotein-mimicking nanodiscs coupled with antigen (Ag) peptides and adjuvants can markedly improve Ag/adjuvant co-delivery to lymphoid organs and sustain Ag presentation on dendritic cells. Strikingly, nanodiscs elicited up to 47-fold greater frequencies of neoantigen-specific CTLs than soluble vaccines and even 31-fold greater than perhaps the strongest adjuvant in clinical trials (that is, CpG in Montanide). Moreover, multi-epitope vaccination generated broad-spectrum T-cell responses that potently inhibited tumour growth. Nanodiscs eliminated established MC-38 and B16F10 tumours when combined with anti-PD-1 and anti-CTLA-4 therapy. These findings represent a new powerful approach for cancer immunotherapy and suggest a general strategy for personalized nanomedicine.
Collapse
Affiliation(s)
- Rui Kuai
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Lukasz J. Ochyl
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Keith S. Bahjat
- Discovery Research, Bristol-Myers Squibb Biologics Discovery California, Redwood City, CA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Co-corresponding authors. J.J.M. () or A.S. ()
| | - James J. Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
- Co-corresponding authors. J.J.M. () or A.S. ()
| |
Collapse
|
328
|
Openshaw MR, Richards CJ, Guttery DS, Shaw JA, Thomas AL. The genetics of gastroesophageal adenocarcinoma and the use of circulating cell free DNA for disease detection and monitoring. Expert Rev Mol Diagn 2017; 17:459-470. [DOI: 10.1080/14737159.2017.1308824] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
329
|
Jiang N. Immune engineering: from systems immunology to engineering immunity. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2017; 1:54-62. [PMID: 29038795 DOI: 10.1016/j.cobme.2017.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The smallpox vaccine represents the earliest attempt in engineering immunity. The recent success of chimeric antigen receptor T cells (CAR-T cells) in cancer once again demonstrates the clinical potential of immune engineering. Inspired by this success, diverse approaches have been used to boost various aspects of immunity: engineering dendritic cells (DCs), natural killer (NK) cells, T cells, antibodies, cytokines, small peptides, and others. With recent development of various high-throughput technologies (of which engineers, especially biomedical engineers/bioengineers contributed significantly), such as immune repertoire sequencing, and analytical methods, a systems level of understanding immunity (or the lack of it) beyond model animals has provided critical insights into the human immune system. This review focuses on recent progressed made in systems biology and the engineering of adaptive immunity.
Collapse
Affiliation(s)
- Ning Jiang
- Department of Biomedical engineering, Cockrell School of Engineering, University of Texas at Austin, Austin, TX 78712, USA.,Institute for Cellular and Molecular Biology, College of Natural Sciences, University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
330
|
Zajac P, Schultz-Thater E, Tornillo L, Sadowski C, Trella E, Mengus C, Iezzi G, Spagnoli GC. MAGE-A Antigens and Cancer Immunotherapy. Front Med (Lausanne) 2017; 4:18. [PMID: 28337438 PMCID: PMC5340762 DOI: 10.3389/fmed.2017.00018] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 02/09/2017] [Indexed: 12/30/2022] Open
Abstract
MAGE-A antigens are expressed in a variety of cancers of diverse histological origin and germinal cells. Due to their relatively high tumor specificity, they represent attractive targets for active specific and adoptive cancer immunotherapies. Here, we (i) review past and ongoing clinical studies targeting these antigens, (ii) analyze advantages and disadvantages of different therapeutic approaches, and (iii) discuss possible improvements in MAGE-A-specific immunotherapies.
Collapse
Affiliation(s)
- Paul Zajac
- Oncology Surgery, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| | - Elke Schultz-Thater
- Oncology Surgery, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| | - Luigi Tornillo
- Department of Pathology, University Hospital of Basel , Basel , Switzerland
| | - Charlotte Sadowski
- Oncology Surgery, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| | - Emanuele Trella
- Oncology Surgery, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| | - Chantal Mengus
- Oncology Surgery, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| | - Giandomenica Iezzi
- Cancer Immunotherapy, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| | - Giulio C Spagnoli
- Oncology Surgery, Department of Biomedicine, University Hospital of Basel , Basel , Switzerland
| |
Collapse
|
331
|
Rodríguez-Otero P, Paiva B, Engelhardt M, Prósper F, San Miguel JF. Is immunotherapy here to stay in multiple myeloma? Haematologica 2017; 102:423-432. [PMID: 28082344 PMCID: PMC5394971 DOI: 10.3324/haematol.2016.152504] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/17/2016] [Indexed: 12/28/2022] Open
Abstract
Immune escape and impaired immune surveillance have been identified as emerging hallmarks of cancer.1 Multiple myeloma represents a genuine example of disrupted immune surveillance characterized by: impaired antibody production, deregulation of the T and natural killer cell compartment, disruption of antigen presentation machinery, upregulation of inhibitory surface ligands, and recruitment of immunosuppressive cells. Although the potential value of immunotherapeutic interventions had a clear antecedent in the graft-versus-myeloma effect induced by allogeneic stem cell transplant and donor lymphocyte infusions, it is only recently that this field has faced a real revolution. In this review we discuss the current results obtained with immune approaches in patients with multiple myeloma that have placed this disease under the scope of immuno-oncology, bringing new therapeutic opportunities for the treatment of multiple myeloma patients.
Collapse
Affiliation(s)
- Paula Rodríguez-Otero
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Bruno Paiva
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Monika Engelhardt
- Department of Medicine I, Hematology, Oncology and Stem Cell Transplantation, Medical Center - University of Freiburg, Faculty of Medicine, Germany
| | - Felipe Prósper
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Jesús F San Miguel
- Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| |
Collapse
|
332
|
Anagnostou V, Smith KN, Forde PM, Niknafs N, Bhattacharya R, White J, Zhang T, Adleff V, Phallen J, Wali N, Hruban C, Guthrie VB, Rodgers K, Naidoo J, Kang H, Sharfman W, Georgiades C, Verde F, Illei P, Li QK, Gabrielson E, Brock MV, Zahnow CA, Baylin SB, Scharpf RB, Brahmer JR, Karchin R, Pardoll DM, Velculescu VE. Evolution of Neoantigen Landscape during Immune Checkpoint Blockade in Non-Small Cell Lung Cancer. Cancer Discov 2017; 7:264-276. [PMID: 28031159 PMCID: PMC5733805 DOI: 10.1158/2159-8290.cd-16-0828] [Citation(s) in RCA: 679] [Impact Index Per Article: 97.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 12/22/2016] [Accepted: 12/27/2016] [Indexed: 02/06/2023]
Abstract
Immune checkpoint inhibitors have shown significant therapeutic responses against tumors containing increased mutation-associated neoantigen load. We have examined the evolving landscape of tumor neoantigens during the emergence of acquired resistance in patients with non-small cell lung cancer after initial response to immune checkpoint blockade with anti-PD-1 or anti-PD-1/anti-CTLA-4 antibodies. Analyses of matched pretreatment and resistant tumors identified genomic changes resulting in loss of 7 to 18 putative mutation-associated neoantigens in resistant clones. Peptides generated from the eliminated neoantigens elicited clonal T-cell expansion in autologous T-cell cultures, suggesting that they generated functional immune responses. Neoantigen loss occurred through elimination of tumor subclones or through deletion of chromosomal regions containing truncal alterations, and was associated with changes in T-cell receptor clonality. These analyses provide insight into the dynamics of mutational landscapes during immune checkpoint blockade and have implications for the development of immune therapies that target tumor neoantigens.Significance: Acquired resistance to immune checkpoint therapy is being recognized more commonly. This work demonstrates for the first time that acquired resistance to immune checkpoint blockade can arise in association with the evolving landscape of mutations, some of which encode tumor neoantigens recognizable by T cells. These observations imply that widening the breadth of neoantigen reactivity may mitigate the development of acquired resistance. Cancer Discov; 7(3); 264-76. ©2017 AACR.See related commentary by Yang, p. 250This article is highlighted in the In This Issue feature, p. 235.
Collapse
MESH Headings
- Adult
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/therapeutic use
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- CTLA-4 Antigen/genetics
- CTLA-4 Antigen/immunology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Cell Cycle Checkpoints/drug effects
- Cell Cycle Checkpoints/immunology
- Cohort Studies
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/immunology
- Female
- Humans
- Immunotherapy
- Ipilimumab/pharmacology
- Ipilimumab/therapeutic use
- Janus Kinase 1/genetics
- Janus Kinase 2/genetics
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Male
- Middle Aged
- Mutation
- Nivolumab
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
Collapse
Affiliation(s)
- Valsamo Anagnostou
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kellie N Smith
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Patrick M Forde
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Noushin Niknafs
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Rohit Bhattacharya
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
| | - James White
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Vilmos Adleff
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Neha Wali
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Carolyn Hruban
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Violeta B Guthrie
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Kristen Rodgers
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jarushka Naidoo
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hyunseok Kang
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William Sharfman
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christos Georgiades
- Department of Radiology and Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Franco Verde
- Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Peter Illei
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qing Kay Li
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Edward Gabrielson
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Malcolm V Brock
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cynthia A Zahnow
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Stephen B Baylin
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert B Scharpf
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Julie R Brahmer
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rachel Karchin
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Drew M Pardoll
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Victor E Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland.
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Institute for Computational Medicine, Johns Hopkins University, Baltimore, Maryland
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
333
|
McGranahan N, Swanton C. Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future. Cell 2017; 168:613-628. [PMID: 28187284 DOI: 10.1016/j.cell.2017.01.018] [Citation(s) in RCA: 1710] [Impact Index Per Article: 244.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 01/03/2017] [Accepted: 01/18/2017] [Indexed: 12/12/2022]
Abstract
Intratumor heterogeneity, which fosters tumor evolution, is a key challenge in cancer medicine. Here, we review data and technologies that have revealed intra-tumor heterogeneity across cancer types and the dynamics, constraints, and contingencies inherent to tumor evolution. We emphasize the importance of macro-evolutionary leaps, often involving large-scale chromosomal alterations, in driving tumor evolution and metastasis and consider the role of the tumor microenvironment in engendering heterogeneity and drug resistance. We suggest that bold approaches to drug development, harnessing the adaptive properties of the immune-microenvironment while limiting those of the tumor, combined with advances in clinical trial-design, will improve patient outcome.
Collapse
Affiliation(s)
- Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK; Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK
| | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, Paul O'Gorman Building, 72 Huntley Street, London WC1E 6BT, UK; Translational Cancer Therapeutics Laboratory, The Francis Crick Institute, 1 Midland Rd, London NW1 1AT, UK; Department of Medical Oncology, University College London Hospitals, 235 Euston Rd, Fitzrovia, London NW1 2BU, UK.
| |
Collapse
|
334
|
Affiliation(s)
- Vessela N Kristensen
- From the Department of Clinical Molecular Biology (EpiGen), Medical Division, Akershus University Hospital, Lørenskog, and the Department of Genetics, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo - both in Norway
| |
Collapse
|
335
|
Ping Y, Liu C, Zhang Y. T-cell receptor-engineered T cells for cancer treatment: current status and future directions. Protein Cell 2017; 9:254-266. [PMID: 28108950 PMCID: PMC5829268 DOI: 10.1007/s13238-016-0367-1] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/16/2016] [Indexed: 12/20/2022] Open
Abstract
T-cell receptor (TCR)-engineered T cells are a novel option for adoptive cell therapy used for the treatment of several advanced forms of cancer. Work using TCR-engineered T cells began more than two decades ago, with numerous preclinical studies showing that such cells could mediate tumor lysis and eradication. The success of these trials provided the foundation for clinical trials, including recent clinical successes using TCR-engineered T cells to target New York esophageal squamous cell carcinoma (NY-ESO-1). These successes demonstrate the potential of this approach to treat cancer. In this review, we provide a perspective on the current and future applications of TCR-engineered T cells for the treatment of cancer. Our summary focuses on TCR activation and both pre-clinical and clinical applications of TCR-engineered T cells. We also discuss how to enhance the function of TCR-engineered T cells and prolong their longevity in the tumor microenvironment.
Collapse
Affiliation(s)
- Yu Ping
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chaojun Liu
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China. .,Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China. .,Engineering Key Laboratory for Cell Therapy of Henan Province, Zhengzhou, 450052, China.
| |
Collapse
|
336
|
Noguchi T, Ward JP, Gubin MM, Arthur CD, Lee SH, Hundal J, Selby MJ, Graziano RF, Mardis ER, Korman AJ, Schreiber RD. Temporally Distinct PD-L1 Expression by Tumor and Host Cells Contributes to Immune Escape. Cancer Immunol Res 2017; 5:106-117. [PMID: 28073774 DOI: 10.1158/2326-6066.cir-16-0391] [Citation(s) in RCA: 226] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2016] [Accepted: 12/29/2016] [Indexed: 12/31/2022]
Abstract
Antibody blockade of programmed death-1 (PD-1) or its ligand, PD-L1, has led to unprecedented therapeutic responses in certain tumor-bearing individuals, but PD-L1 expression's prognostic value in stratifying cancer patients for such treatment remains unclear. Reports conflict on the significance of correlations between PD-L1 on tumor cells and positive clinical outcomes to PD-1/PD-L1 blockade. We investigated this issue using genomically related, clonal subsets from the same methylcholanthrene-induced sarcoma: a highly immunogenic subset that is spontaneously eliminated in vivo by adaptive immunity and a less immunogenic subset that forms tumors in immunocompetent mice, but is sensitive to PD-1/PD-L1 blockade therapy. Using CRISPR/Cas9-induced loss-of-function approaches and overexpression gain-of-function techniques, we confirmed that PD-L1 on tumor cells is key to promoting tumor escape. In addition, the capacity of PD-L1 to suppress antitumor responses was inversely proportional to tumor cell antigenicity. PD-L1 expression on host cells, particularly tumor-associated macrophages (TAM), was also important for tumor immune escape. We demonstrated that induction of PD-L1 on tumor cells was IFNγ-dependent and transient, but PD-L1 induction on TAMs was of greater magnitude, only partially IFNγ dependent, and was stable over time. Thus, PD-L1 expression on either tumor cells or host immune cells could lead to tumor escape from immune control, indicating that total PD-L1 expression in the immediate tumor microenvironment may represent a more accurate biomarker for predicting response to PD-1/PD-L1 blockade therapy, compared with monitoring PD-L1 expression on tumor cells alone. Cancer Immunol Res; 5(2); 106-17. ©2017 AACR.
Collapse
Affiliation(s)
- Takuro Noguchi
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - Jeffrey P Ward
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri.,Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Matthew M Gubin
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - Cora D Arthur
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - Sang Hun Lee
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri
| | - Jasreet Hundal
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri
| | | | | | - Elaine R Mardis
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri.,Department of Genetics, Washington University School of Medicine, St. Louis, Missouri
| | | | - Robert D Schreiber
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, Missouri. .,Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St Louis, Missouri
| |
Collapse
|
337
|
Genetically Modified T-Cell-Based Adoptive Immunotherapy in Hematological Malignancies. J Immunol Res 2017; 2017:5210459. [PMID: 28116322 PMCID: PMC5237740 DOI: 10.1155/2017/5210459] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 12/05/2016] [Indexed: 12/24/2022] Open
Abstract
A significant proportion of hematological malignancies remain limited in treatment options. Immune system modulation serves as a promising therapeutic approach to eliminate malignant cells. Cytotoxic T lymphocytes (CTLs) play a central role in antitumor immunity; unfortunately, nonspecific approaches for targeted recognition of tumor cells by CTLs to mediate tumor immune evasion in hematological malignancies imply multiple mechanisms, which may or may not be clinically relevant. Recently, genetically modified T-cell-based adoptive immunotherapy approaches, including chimeric antigen receptor (CAR) T-cell therapy and engineered T-cell receptor (TCR) T-cell therapy, promise to overcome immune evasion by redirecting the specificity of CTLs to tumor cells. In clinic trials, CAR-T-cell- and TCR-T-cell-based adoptive immunotherapy have produced encouraging clinical outcomes, thereby demonstrating their therapeutic potential in mitigating tumor development. The purpose of the present review is to (1) provide a detailed overview of the multiple mechanisms for immune evasion related with T-cell-based therapies; (2) provide a current summary of the applications of CAR-T-cell- as well as neoantigen-specific TCR-T-cell-based adoptive immunotherapy and routes taken to overcome immune evasion; and (3) evaluate alternative approaches targeting immune evasion via optimization of CAR-T and TCR-T-cell immunotherapies.
Collapse
|
338
|
Ritz D, Gloger A, Neri D, Fugmann T. Purification of soluble HLA class I complexes from human serum or plasma deliver high quality immuno peptidomes required for biomarker discovery. Proteomics 2016; 17. [PMID: 27862975 DOI: 10.1002/pmic.201600364] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Revised: 10/24/2016] [Accepted: 11/10/2016] [Indexed: 12/29/2022]
Abstract
Soluble human leukocyte antigen class I (sHLA)-peptide complexes have been suggested to play a role in the modulation of immune responses and in immune evasion of cancer cells. The set of peptides eluted from sHLA molecules could serve as biomarker for the monitoring of patients with cancer or other conditions. Here, we describe an improved sHLA peptidomics methodology resulting in the identification of 1816 to 2761 unique peptide sequences from triplicate analyses of serum or plasma taken from three healthy donors. More than 90% of the identified peptides were 8-11mers and 74% of these sequences were predicted to bind to cognate HLA alleles, confirming the quality of the resulting immunopeptidomes. In comparison to the HLA peptidome of cultured cells, the plasma-derived peptides were predicted to have a higher stability in complex with the cognate HLA molecules and mainly derived from proteins of the plasma membrane or from the extracellular space. The sHLA peptidomes can efficiently be characterized by using the new methodology, thus serving as potential source of biomarkers in various pathological conditions.
Collapse
Affiliation(s)
| | - Andreas Gloger
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Dario Neri
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | | |
Collapse
|
339
|
Bassani-Sternberg M, Bräunlein E, Klar R, Engleitner T, Sinitcyn P, Audehm S, Straub M, Weber J, Slotta-Huspenina J, Specht K, Martignoni ME, Werner A, Hein R, H. Busch D, Peschel C, Rad R, Cox J, Mann M, Krackhardt AM. Direct identification of clinically relevant neoepitopes presented on native human melanoma tissue by mass spectrometry. Nat Commun 2016; 7:13404. [PMID: 27869121 PMCID: PMC5121339 DOI: 10.1038/ncomms13404] [Citation(s) in RCA: 486] [Impact Index Per Article: 60.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 09/30/2016] [Indexed: 02/07/2023] Open
Abstract
Although mutations may represent attractive targets for immunotherapy, direct identification of mutated peptide ligands isolated from human leucocyte antigens (HLA) on the surface of native tumour tissue has so far not been successful. Using advanced mass spectrometry (MS) analysis, we survey the melanoma-associated immunopeptidome to a depth of 95,500 patient-presented peptides. We thereby discover a large spectrum of attractive target antigen candidates including cancer testis antigens and phosphopeptides. Most importantly, we identify peptide ligands presented on native tumour tissue samples harbouring somatic mutations. Four of eleven mutated ligands prove to be immunogenic by neoantigen-specific T-cell responses. Moreover, tumour-reactive T cells with specificity for selected neoantigens identified by MS are detected in the patient's tumour and peripheral blood. We conclude that direct identification of mutated peptide ligands from primary tumour material by MS is possible and yields true neoepitopes with high relevance for immunotherapeutic strategies in cancer.
Collapse
Affiliation(s)
- Michal Bassani-Sternberg
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried 82152, Germany
| | - Eva Bräunlein
- IIIrd Medical Department, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, Munich 81675, Germany
| | - Richard Klar
- IIIrd Medical Department, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, Munich 81675, Germany
| | - Thomas Engleitner
- IInd Medical Department, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, Munich 81675, Germany
- German Cancer Consortium of Translational Cancer Research (DKTK) and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Pavel Sinitcyn
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried 82152, Germany
| | - Stefan Audehm
- IIIrd Medical Department, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, Munich 81675, Germany
| | - Melanie Straub
- Institute of Pathology, Technische Universität München, Ismaningerstr. 22, Munich 81675, Germany
| | - Julia Weber
- IInd Medical Department, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, Munich 81675, Germany
- German Cancer Consortium of Translational Cancer Research (DKTK) and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Julia Slotta-Huspenina
- Institute of Pathology, Technische Universität München, Ismaningerstr. 22, Munich 81675, Germany
- MRI-TUM-Biobank at the Institute of Pathology, Technische Universität München, Ismaningerstr. 22, Munich 81675, Germany
| | - Katja Specht
- Institute of Pathology, Technische Universität München, Ismaningerstr. 22, Munich 81675, Germany
| | - Marc E. Martignoni
- Surgery Department, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, Munich, 81675, Germany
| | - Angelika Werner
- Surgery Department, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, Munich, 81675, Germany
| | - Rüdiger Hein
- Dermatology Department, Klinikum rechts der Isar, Technische Universität München, Biedersteiner Str 29, Munich 80802, Germany
| | - Dirk H. Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Trogerstr. 30, Munich 81675, Germany
| | - Christian Peschel
- IIIrd Medical Department, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, Munich 81675, Germany
- German Cancer Consortium of Translational Cancer Research (DKTK) and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Roland Rad
- IInd Medical Department, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, Munich 81675, Germany
- German Cancer Consortium of Translational Cancer Research (DKTK) and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Jürgen Cox
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried 82152, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Am Klopferspitz 18, Martinsried 82152, Germany
| | - Angela M. Krackhardt
- IIIrd Medical Department, Klinikum rechts der Isar, Technische Universität München, Ismaningerstr. 22, Munich 81675, Germany
- German Cancer Consortium of Translational Cancer Research (DKTK) and German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| |
Collapse
|
340
|
Collins DC, Sundar R, Lim JSJ, Yap TA. Towards Precision Medicine in the Clinic: From Biomarker Discovery to Novel Therapeutics. Trends Pharmacol Sci 2016; 38:25-40. [PMID: 27871777 DOI: 10.1016/j.tips.2016.10.012] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/18/2016] [Accepted: 10/19/2016] [Indexed: 02/08/2023]
Abstract
Precision medicine continues to be the benchmark to which we strive in cancer research. Seeking out actionable aberrations that can be selectively targeted by drug compounds promises to optimize treatment efficacy and minimize toxicity. Utilizing these different targeted agents in combination or in sequence may further delay resistance to treatments and prolong antitumor responses. Remarkable progress in the field of immunotherapy adds another layer of complexity to the management of cancer patients. Corresponding advances in companion biomarker development, novel methods of serial tumor assessments, and innovative trial designs act synergistically to further precision medicine. Ongoing hurdles such as clonal evolution, intra- and intertumor heterogeneity, and varied mechanisms of drug resistance continue to be challenges to overcome. Large-scale data-sharing and collaborative networks using next-generation sequencing (NGS) platforms promise to take us further into the cancer 'ome' than ever before, with the goal of achieving successful precision medicine.
Collapse
Affiliation(s)
- Dearbhaile C Collins
- The Institute of Cancer Research and Royal Marsden Hospital, Downs Road, London SM2 5PT, UK
| | - Raghav Sundar
- The Institute of Cancer Research and Royal Marsden Hospital, Downs Road, London SM2 5PT, UK
| | - Joline S J Lim
- The Institute of Cancer Research and Royal Marsden Hospital, Downs Road, London SM2 5PT, UK
| | - Timothy A Yap
- The Institute of Cancer Research and Royal Marsden Hospital, Downs Road, London SM2 5PT, UK.
| |
Collapse
|
341
|
Abstract
Immune checkpoint inhibitors (ICI) based cancer immunotherapy has recently attracted considerable interest in the field of cancer therapy. The relevant immunotherapeutic agents do not directly attack the tumor, but boost the body's immune system to recognize and kill cancer cells. In this commentary, recent efforts utilizing immunoengineering for local delivery of these immune checkpoint antibodies are introduced. Future opportunities and challenges in this research theme are also commented.
Collapse
Affiliation(s)
- Chao Wang
- a Joint Department of Biomedical Engineering , University of North Carolina at Chapel Hill and North Carolina State University , Raleigh , NC , USA.,b Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Yanqi Ye
- a Joint Department of Biomedical Engineering , University of North Carolina at Chapel Hill and North Carolina State University , Raleigh , NC , USA.,b Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , NC , USA
| | - Zhen Gu
- a Joint Department of Biomedical Engineering , University of North Carolina at Chapel Hill and North Carolina State University , Raleigh , NC , USA.,b Division of Molecular Pharmaceutics and Center for Nanotechnology in Drug Delivery, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill , Chapel Hill , NC , USA.,c Department of Medicine University of North Carolina School of Medicine Chapel Hill , NC , USA
| |
Collapse
|
342
|
Redeker A, Arens R. Improving Adoptive T Cell Therapy: The Particular Role of T Cell Costimulation, Cytokines, and Post-Transfer Vaccination. Front Immunol 2016; 7:345. [PMID: 27656185 PMCID: PMC5011476 DOI: 10.3389/fimmu.2016.00345] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/24/2016] [Indexed: 12/22/2022] Open
Abstract
Adoptive cellular therapy (ACT) is a form of immunotherapy whereby antigen-specific T cells are isolated or engineered, expanded ex vivo, and transferred back to patients. Clinical benefit after ACT has been obtained in treatment of infection, various hematological malignancies, and some solid tumors; however, due to poor functionality and persistence of the transferred T cells, the efficacy of ACT in the treatment of most solid tumors is often marginal. Hence, much effort is undertaken to improve T cell function and persistence in ACT and significant progress is being made. Herein, we will review strategies to improve ACT success rates in the treatment of cancer and infection. We will deliberate on the most favorable phenotype for the tumor-specific T cells that are infused into patients and on how to obtain T cells bearing this phenotype by applying novel ex vivo culture methods. Moreover, we will discuss T cell function and persistence after transfer into patients and how these factors can be manipulated by means of providing costimulatory signals, cytokines, blocking antibodies to inhibitory molecules, and vaccination. Incorporation of these T cell stimulation strategies and combinations of the different treatment modalities are likely to improve clinical response rates further.
Collapse
Affiliation(s)
- Anke Redeker
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center , Leiden , Netherlands
| | - Ramon Arens
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center , Leiden , Netherlands
| |
Collapse
|
343
|
Santegoets SJAM, Welters MJP, van der Burg SH. Monitoring of the Immune Dysfunction in Cancer Patients. Vaccines (Basel) 2016; 4:vaccines4030029. [PMID: 27598210 PMCID: PMC5041023 DOI: 10.3390/vaccines4030029] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/25/2016] [Accepted: 08/27/2016] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy shows promising clinical results in patients with different types of cancer, but its full potential is not reached due to immune dysfunction as a result of several suppressive mechanisms that play a role in cancer development and progression. Monitoring of immune dysfunction is a prerequisite for the development of strategies aiming to alleviate cancer-induced immune suppression. At this point, the level at which immune dysfunction occurs has to be established, the underlying mechanism(s) need to be known, as well as the techniques to assess this. While it is relatively easy to measure general signs of immune suppression, it turns out that accurate monitoring of the frequency and function of immune-suppressive cells is still difficult. A lack of truly specific markers, the phenotypic complexity among suppressive cells of the same lineage, but potentially with different functions and functional assays that may not cover every mechanistic aspect of immune suppression are among the reasons complicating proper assessments. Technical innovations in flow and mass cytometry will allow for more complete sets of markers to precisely determine phenotype and associated function. There is, however, a clear need for functional assays that recapitulate more of the mechanisms employed to suppress the immune system.
Collapse
Affiliation(s)
- Saskia J A M Santegoets
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Marij J P Welters
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
344
|
Schumacher TN, Hacohen N. Neoantigens encoded in the cancer genome. Curr Opin Immunol 2016; 41:98-103. [PMID: 27518850 DOI: 10.1016/j.coi.2016.07.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 07/27/2016] [Accepted: 07/27/2016] [Indexed: 02/07/2023]
Abstract
Somatic mutations in the genome represent one of the major drivers of malignancy. However, non-synonymous mutations are also a source of mutated peptides that are presented by HLA molecules to induce protective CD4 and CD8 T cell responses. Consistent with this notion, the mutation burden of a tumor is correlated with local immunity as well as outcome of therapy and patient survival. Furthermore, neoantigen-specific T cells appear sufficient to control tumors prophylactically and therapeutically. While the role of neoantigens as a determinant of the foreignness of human cancers is now well established, major questions, including the relative importance of clonal vs subclonal neoantigens, and CD4 vs CD8 T cells, remain unanswered. We expect continued animal studies to address some of the open issues and ongoing clinical trials to establish the utility of therapeutic strategies to enhance neoantigen-specific T cell responses in human cancer.
Collapse
Affiliation(s)
- Ton N Schumacher
- Division of Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | - Nir Hacohen
- Cancer Center and Center for Cancer Immunology, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|