1151
|
Chaudhary B, Elkord E. Novel expression of Neuropilin 1 on human tumor-infiltrating lymphocytes in colorectal cancer liver metastases. Expert Opin Ther Targets 2014; 19:147-61. [PMID: 25351619 DOI: 10.1517/14728222.2014.977784] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVES Neuropilin 1 (NRP1) is a transmembrane protein with diverse roles in physiological and pathological settings. NRP1 expression has been reported on T cells in inflammatory microenvironments and in secondary lymphoid tissue. Tumor-infiltrating lymphocytes (TILs) play an important role in cancer prognosis. In this study, we investigated NRP1 expression on TILs and peripheral blood mononuclear cells (PBMCs) from colorectal cancer liver metastases (LI/CRC). METHODS TILs from LI/CRC and PBMCs from healthy donors and patients were analyzed for expression of NRP1, in addition to other Treg-related markers. PBMCs were co-cultured in vitro with tumor tissue and analyzed for NRP1 expression. RESULTS We report for the first time that NRP1 is highly expressed on CD3(+)CD4(+) TILs compared to PBMCs. NRP1 expression correlated closely with CD25 expression in TILs. NRP1 was expressed on both Helios(+) and Helios(-) FoxP3-expressing Tregs and on a FoxP3(-)Helios(-) T cell subset. It was also induced on PBMCs following in vitro co-culture with tumor tissue. CONCLUSIONS NRP1 is upregulated on TILs and can be induced on PBMCs by tumor tissue. Further studies are warranted to define the function of NRP1 on human TILs. As a therapeutic target, NRP1 may allow selective targeting of TIL subsets including suppressive Tregs.
Collapse
Affiliation(s)
- Belal Chaudhary
- United Arab Emirates University, College of Medicine and Health Sciences , PO Box 17666, Al Ain , UAE +971 37137527 ; +971 37671966 ; ;
| | | |
Collapse
|
1152
|
Gibbons RM, Liu X, Pulko V, Harrington SM, Krco CJ, Kwon ED, Dong H. B7-H1 limits the entry of effector CD8(+) T cells to the memory pool by upregulating Bim. Oncoimmunology 2014; 1:1061-1073. [PMID: 23170254 PMCID: PMC3494620 DOI: 10.4161/onci.20850] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Protective T‑cell immunity against cancer and infections is dependent on the generation of a durable effector and memory T‑cell pool. Studies from cancer and chronic infections reveal that B7-H1 (PD-L1) engagement with its receptor PD-1 promotes apoptosis of effector T cells. It is not clear how B7-H1 regulates T‑cell apoptosis and the subsequent impact of B7-H1 on the generation of memory T cells. In immunized B7-H1-deficient mice, we detected an increased expansion of effector CD8+ T cells and a delayed T‑cell contraction followed by the emergence of a protective CD8+ T‑cell memory capable of completely rejecting tumor metastases in the lung. Intracellular staining revealed that antigen-primed CD8+ T cells in B7-H1-deficient mice express lower levels of the pro-apoptotic molecule Bim. The engagement of activated CD8+ T cells by a plate-bound B7-H1 fusion protein led to the upregulation of Bim and increased cell death. Assays based on blocking antibodies determined that both PD-1 and CD80 are involved in the B7-H1-mediated regulation of Bim in activated CD8+ T cells. Our results suggest that B7-H1 may negatively regulate CD8+ T‑cell memory by enhancing the depletion of effector CD8+ T cells through the upregulation of Bim. Our findings may provide a new strategy for targeting B7-H1 signaling in effector CD8+ T cells to achieve protective antitumor memory responses.
Collapse
Affiliation(s)
- Rachel M Gibbons
- Department of Immunology; College of Medicine; Mayo Clinic; Rochester, MN USA
| | | | | | | | | | | | | |
Collapse
|
1153
|
Henick BS, Herbst RS, Goldberg SB. The PD-1 pathway as a therapeutic target to overcome immune escape mechanisms in cancer. Expert Opin Ther Targets 2014; 18:1407-20. [PMID: 25331677 DOI: 10.1517/14728222.2014.955794] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Immunotherapy is emerging as a powerful approach in cancer treatment. Preclinical data predicted the antineoplastic effects seen in clinical trials of programmed death-1 (PD-1) pathway inhibitors, as well as their observed toxicities. The results of early clinical trials are extraordinarily promising in several cancer types and have shaped the direction of ongoing and future studies. AREAS COVERED This review describes the biological rationale for targeting the PD-1 pathway with monoclonal antibodies for the treatment of cancer as a context for examining the results of early clinical trials. It also surveys the landscape of ongoing clinical trials and discusses their anticipated strengths and limitations. EXPERT OPINION PD-1 pathway inhibition represents a new frontier in cancer immunotherapy, which shows clear evidence of activity in various tumor types including NSCLC and melanoma. Ongoing and upcoming trials will examine optimal combinations of these agents, which should further define their role across tumor types. Current limitations include the absence of a reliable companion diagnostic to predict likely responders, as well as lack of data in early-stage cancer when treatment has the potential to increase cure rates.
Collapse
Affiliation(s)
- Brian S Henick
- Yale School of Medicine, Department of Internal Medicine, Resident in Internal Medicine , 333 Cedar Street, FMP 125, New Haven, CT 06520-8028 , USA
| | | | | |
Collapse
|
1154
|
Abstract
Despite extensive investigation over the past three decades, cancer immunotherapy has produced limited success, with few agents achieving approval by the Food and Drug Administration and even the most effective helping only a minority of patients, primarily with melanoma or renal cancer. In recent years, immune checkpoints that maintain physiologic self-tolerance have been implicated in the down-regulation of anti-tumor immunity. Efforts to restore latent anti-tumor immunity have focused on antibody-based interventions targeting CTL antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) on T lymphocytes and its principal ligand (PD-L1) on tumor cells. Ipilimumab, an antibody targeting CTLA-4, appears to restore tumor immunity at the priming phase, whereas anti-PD-1/PD-L1 antibodies restore immune function in the tumor microenvironment. Although ipilimumab can produce durable long-term responses in patients with advanced melanoma, it is associated with significant immune-related toxicities. By contrast, antibodies targeting either PD-1 or PD-L1 have produced significant anti-tumor activity with considerably less toxicity. Activity was seen in patients with melanoma and renal cancer, as well as those with non-small-cell lung, bladder and head and neck cancers, tumors not previously felt to be sensitive to immunotherapy. The tolerability of PD-1-pathway blockers and their unique mechanism of action have made them ideal backbones for combination regimen development. Combination approaches involving cytotoxic chemotherapy, anti-angiogenic agents, alternative immune-checkpoint inhibitors, immunostimulatory cytokines and cancer vaccines are currently under clinical investigation. Current efforts focus on registration trials of single agents and combinations in various diseases and disease settings and identifying predictive biomarkers of response.
Collapse
Affiliation(s)
- George K Philips
- Department of Medicine, Georgetown University Hospital, 3800 Reservoir Road NW, Washington DC 20007, USA
| | - Michael Atkins
- Department of Oncology, Georgetown-Lombardi Comprehensive Cancer Center, Research Building-E501, 3970 Reservoir Road NW, Washington DC 20057, USA
| |
Collapse
|
1155
|
Altvater B, Kailayangiri S, Theimann N, Ahlmann M, Farwick N, Chen C, Pscherer S, Neumann I, Mrachatz G, Hansmeier A, Hardes J, Gosheger G, Juergens H, Rossig C. Common Ewing sarcoma-associated antigens fail to induce natural T cell responses in both patients and healthy individuals. Cancer Immunol Immunother 2014; 63:1047-60. [PMID: 24973179 PMCID: PMC11028878 DOI: 10.1007/s00262-014-1574-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2014] [Accepted: 06/16/2014] [Indexed: 11/24/2022]
Abstract
Disseminated or relapsed Ewing sarcoma (EwS) has remained fatal in the majority of patients. A promising approach to preventing relapse after conventional therapy is to establish tumor antigen-specific immune control. Efficient and specific T cell memory against the tumor depends on the expansion of rare T cells with native specificity against target antigens overexpressed by the tumor. Candidate antigens in EwS include six-transmembrane epithelial antigen of the prostate-1 (STEAP1), and the human cancer/testis antigens X-antigen family member 1 (XAGE1) and preferentially expressed antigen in melanoma (PRAME). Here, we screened normal donors and EwS patients for the presence of circulating T cells reactive with overlapping peptide libraries of these antigens by IFN-γ Elispot analysis. The majority of 22 healthy donors lacked detectable memory T cell responses against STEAP1, XAGE1 and PRAME. Moreover, ex vivo detection of T cells specific for these antigens in both blood and bone marrow were limited to a minority of EwS patients and required nonspecific T cell prestimulation. Cytotoxic T cells specific for the tumor-associated antigens were efficiently and reliably generated by in vitro priming using professional antigen-presenting cells and optimized cytokine stimulation; however, these T cells failed to interact with native antigen processed by target cells and with EwS cells expressing the antigen. We conclude that EwS-associated antigens fail to induce efficient T cell receptor (TCR)-mediated antitumor immune responses even under optimized conditions. Strategies based on TCR engineering could provide a more effective means to manipulating T cell immunity toward targeted elimination of tumor cells.
Collapse
MESH Headings
- Adolescent
- Adult
- Antigen-Presenting Cells/drug effects
- Antigen-Presenting Cells/immunology
- Antigens, Neoplasm/biosynthesis
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/pharmacology
- Bone Marrow Cells/drug effects
- Bone Marrow Cells/immunology
- Case-Control Studies
- Cell Line, Tumor
- Child
- Child, Preschool
- Epitopes, T-Lymphocyte/immunology
- Female
- Humans
- K562 Cells
- Male
- Oxidoreductases/biosynthesis
- Oxidoreductases/immunology
- Oxidoreductases/pharmacology
- Sarcoma, Ewing/blood
- Sarcoma, Ewing/immunology
- Sarcoma, Ewing/pathology
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Young Adult
Collapse
Affiliation(s)
- Bianca Altvater
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Sareetha Kailayangiri
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Nadine Theimann
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Martina Ahlmann
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Nicole Farwick
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Christiane Chen
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Sibylle Pscherer
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Ilka Neumann
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Gabriele Mrachatz
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Anna Hansmeier
- Department of Internal Medicine A, Hematology and Oncology, University Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Jendrik Hardes
- Department of Orthopedic Surgery, University Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Georg Gosheger
- Department of Orthopedic Surgery, University Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Heribert Juergens
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
| | - Claudia Rossig
- Department of Pediatric Hematology and Oncology, University Children’s Hospital Muenster, Albert-Schweitzer Campus 1, 48149 Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003 – CiM), University of Muenster, Münster, Germany
| |
Collapse
|
1156
|
The perspective of immunotherapy: new molecules and new mechanisms of action in immune modulation. Curr Opin Oncol 2014; 26:204-14. [PMID: 24424272 DOI: 10.1097/cco.0000000000000054] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Targeting CTLA-4, the patriarch of immune checkpoint modulators, is currently the only immunotherapeutic approach that has achieved significant clinical benefit in melanoma phase III trials. In this review, recent new ideas about the mechanism of action of anti-CTLA antibodies, other new molecules to target, and rationales for combination therapies will be discussed. RECENT FINDINGS Although the clinical efficacy of the anti-CTLA-4 monoclonal antibody (mAb) ipilimumab is meanwhile without doubt, its mechanism of action is still not fully understood. Recent data indicate that, besides modulation of the TCR signal, CTLA-4 mAbs can mediate regulatory T-cell depletion in an Fc gamma receptor dependent manner.Blockade of the molecules PD-1 and PD-L1 has given promising clinical responses (and this beyond melanoma), whereas their complex expression and interaction pattern makes a clear statement about the mechanism of action challenging.Additional other co-inhibitory and co-stimulatory molecules have been identified recently. Combinations of immune checkpoint modulators themselves or with other therapies, such as adoptive cell therapy, targeted therapy or radiotherapy, will improve the outcomes further. SUMMARY Immune checkpoint blockade is currently the most promising systemic therapeutic approach to achieve long-lasting responses or even cure in melanoma and other malignancies.
Collapse
|
1157
|
T cell differentiation in chronic infection and cancer: functional adaptation or exhaustion? Nat Rev Immunol 2014; 14:768-74. [PMID: 25257362 DOI: 10.1038/nri3740] [Citation(s) in RCA: 223] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chronic viral infections and malignant tumours induce T cells that have a reduced ability to secrete effector cytokines and have upregulated expression of the inhibitory receptor PD1 (programmed cell death protein 1). These features have so far been considered to mark terminally differentiated 'exhausted' T cells. However, several recent clinical and experimental observations indicate that phenotypically exhausted T cells can still mediate a crucial level of pathogen or tumour control. In this Opinion article, we propose that the exhausted phenotype results from a differentiation process in which T cells stably adjust their effector capacity to the needs of chronic infection. We argue that this phenotype is optimized to cause minimal tissue damage while still mediating a critical level of pathogen control. In contrast to the presently held view of functional exhaustion, this new concept better reflects the pathophysiology and clinical manifestations of persisting infections, and it provides a rationale for emerging therapies that enhance T cell activity in chronic infection and cancer by blocking inhibitory receptors.
Collapse
|
1158
|
Kedmi M, Avigdor A, Nagler A. Anti-PD-1-targeted therapies focusing on lymphatic malignancies: biological rationale, clinical challenges and opportunities. Acta Haematol 2014; 133:129-35. [PMID: 25247668 DOI: 10.1159/000362151] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 03/09/2014] [Indexed: 01/01/2023]
Abstract
Cancer immunotherapy with tumor-directed antibodies has generally been very successful, while T-cell immunotherapy has been less effective. Some lymphoid malignancies can be cured with immunochemotherapy but nevertheless many patients relapse or progress in spite of maximal therapy. Both solid tumors and lymphoid malignancies develop mechanisms in order to escape destruction by the intact immune system. One such mechanism is mediated through immune checkpoints. PD-1 (programmed cell death protein-1, which is expressed on activated T and B cells, natural killer cells and myeloid cells, is one of those checkpoints. This review focuses on the effect of PD-1 activation on lymphoid malignancies and its role as a therapeutic target.
Collapse
Affiliation(s)
- Meirav Kedmi
- Hematology Division, Chaim Sheba Medical Center, Tel Hashomer, and Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | |
Collapse
|
1159
|
Kvistborg P, Philips D, Kelderman S, Hageman L, Ottensmeier C, Joseph-Pietras D, Welters MJP, van der Burg S, Kapiteijn E, Michielin O, Romano E, Linnemann C, Speiser D, Blank C, Haanen JB, Schumacher TN. Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci Transl Med 2014; 6:254ra128. [PMID: 25232180 DOI: 10.1126/scitranslmed.3008918] [Citation(s) in RCA: 306] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Anti-CTLA-4 treatment improves the survival of patients with advanced-stage melanoma. However, although the anti-CTLA-4 antibody ipilimumab is now an approved treatment for patients with metastatic disease, it remains unknown by which mechanism it boosts tumor-specific T cell activity. In particular, it is unclear whether treatment amplifies previously induced T cell responses or whether it induces new tumor-specific T cell reactivities. Using a combination ultraviolet (UV)-induced peptide exchange and peptide-major histocompatibility complex (pMHC) combinatorial coding, we monitored immune reactivity against a panel of 145 melanoma-associated epitopes in a cohort of patients receiving anti-CTLA-4 treatment. Comparison of pre- and posttreatment T cell reactivities in peripheral blood mononuclear cell samples of 40 melanoma patients demonstrated that anti-CTLA-4 treatment induces a significant increase in the number of detectable melanoma-specific CD8 T cell responses (P = 0.0009). In striking contrast, the magnitude of both virus-specific and melanoma-specific T cell responses that were already detected before start of therapy remained unaltered by treatment (P = 0.74). The observation that anti-CTLA-4 treatment induces a significant number of newly detected T cell responses-but only infrequently boosts preexisting immune responses-provides strong evidence for anti-CTLA-4 therapy-enhanced T cell priming as a component of the clinical mode of action.
Collapse
Affiliation(s)
- Pia Kvistborg
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands.
| | - Daisy Philips
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands
| | - Sander Kelderman
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands
| | - Lois Hageman
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands
| | - Christian Ottensmeier
- National Institute for Health Research Southampton Experimental Cancer Medicine Centre and Southampton University Hospitals, Tremona Road, Southampton, Hampshire SO16 6YD, UK
| | - Deborah Joseph-Pietras
- National Institute for Health Research Southampton Experimental Cancer Medicine Centre and Southampton University Hospitals, Tremona Road, Southampton, Hampshire SO16 6YD, UK
| | - Marij J P Welters
- Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Sjoerd van der Burg
- Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Ellen Kapiteijn
- Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, Netherlands
| | - Olivier Michielin
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Rue Pierre-Decker 4, 1011 Lausanne, Switzerland
| | - Emanuela Romano
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Rue Pierre-Decker 4, 1011 Lausanne, Switzerland
| | - Carsten Linnemann
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands
| | - Daniel Speiser
- Ludwig Center for Cancer Research, Department of Oncology, University of Lausanne, Rue Pierre-Decker 4, 1011 Lausanne, Switzerland
| | - Christian Blank
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands
| | - John B Haanen
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands
| | - Ton N Schumacher
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, Netherlands.
| |
Collapse
|
1160
|
Abstract
Optimal T cell response is dependent not only on T cell receptor activation, but also on additional signaling from coreceptors. The main coreceptors include B7 and tumor necrosis factor family members. They exert costimulatory or coinhibitory effects, and their balance determines the fate of T cell response. In normal conditions, costimulators facilitate the development of protective immune response, whereas coinhibitors dampen inflammation to avoid organ/tissue damage from excessive immune reaction. In the tumor microenvironment, the balance is garbled: inhibitory pathways predominate, and T cell response is impaired. The importance of cosignaling in the tumor immune response has been experimentally and clinically demonstrated. New therapeutic strategies targeting T cell cosignaling, especially coinhibitory molecules, are under active experimental and clinical investigation. This review summarizes the functions of main T cell cosignaling axes and discusses their clinical application.
Collapse
|
1161
|
PD-1(+) CD8(+) T cells are exhausted in tumours and functional in draining lymph nodes of colorectal cancer patients. Br J Cancer 2014; 111:1391-9. [PMID: 25093496 PMCID: PMC4183848 DOI: 10.1038/bjc.2014.416] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/30/2014] [Accepted: 07/02/2014] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The blockade of PD-1-PD-L1 pathway is emerging as an effective therapeutic strategy for several advanced cancers. But the immune regulatory role of PD-1-PD-L1 pathway is not clear in colorectal cancer (CRC) patients. This study aims to evaluate the role of PD-1-PD-L1 pathway in CD8(+) T-cell functions in tumour-draining lymph nodes (TDLNs) and tumours of CRC patients. METHODS PD-1 expression on CD8(+) T cells was examined by flow cytometry, and PD-L1 expression in TDLNs and tumour tissues were examined by immunohistochemistry. Production of IFN-γ, IL-2 and expression of granzyme B, perforin in CD8(+) T cells were detected by intracellular staining. RESULTS PD-1 expression is markedly upregulated on CD8(+) T cells in TDLNs and tumours compared with that in peripheral blood. PD-1-expressing CD8(+) T cells are competent for production of cytokine (IL-2 and IFN-γ) and perforin in the tumour-free lymph nodes (TFLNs), but exhibit exhausted phenotypes in tumours. In addition, PD-L1 is highly expressed in tumours rather than TFLNs, which is closely correlated with the impairment of IFN-γ production of tumour-infiltrating PD-1(+) CD8(+) T cells. CONCLUSIONS Our findings suggest a suppressive effect of PD-1 on CD8(+) T-cell function in tumours, but not in TFLNs.
Collapse
|
1162
|
Sundar R, Soong R, Cho BC, Brahmer JR, Soo RA. Immunotherapy in the treatment of non-small cell lung cancer. Lung Cancer 2014; 85:101-9. [PMID: 24880938 PMCID: PMC4332778 DOI: 10.1016/j.lungcan.2014.05.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 05/07/2014] [Indexed: 12/12/2022]
Abstract
Advances in the understanding of the role of the immune system in tumor immunosurveillance have resulted in the recognition that tumors can evade immune destruction via the dysregulation of co-inhibitory or checkpoint signals. This has led to the development of a generation immunotherapeutic agents targeting the immune checkpoint pathway. Recent early phase studies of immune checkpoint modulators, such as CTLA-4, PD-1 and PD-L1 inhibitors in NSCLC have reported promising results with prolonged clinical responses and tolerable toxicity. This article provides an overview of co-stimulatory and inhibitory molecules that regulate the immune response to tumors, recent therapies that have been developed to exploit these interactions and the role of predictive biomarkers in treatment selection.
Collapse
Affiliation(s)
- Raghav Sundar
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore
| | - Richie Soong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Pathology, National University Health System, Singapore
| | - Byoung-Chul Cho
- Division of Medical Oncology, Yonsei Cancer Center, Seoul, South Korea
| | - Julie R Brahmer
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD, United States
| | - Ross A Soo
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore; Department of Surgery, University of Western Australia, Australia.
| |
Collapse
|
1163
|
Hinrichs CS, Rosenberg SA. Exploiting the curative potential of adoptive T-cell therapy for cancer. Immunol Rev 2014; 257:56-71. [PMID: 24329789 DOI: 10.1111/imr.12132] [Citation(s) in RCA: 358] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Adoptive T-cell therapy (ACT) is a potent and flexible cancer treatment modality that can induce complete, durable regression of certain human malignancies. Long-term follow-up of patients receiving tumor-infiltrating lymphocytes (TILs) for metastatic melanoma reveals a substantial subset that experienced complete, lasting tumor regression - and may be cured. Increasing evidence points to mutated gene products as the primary immunological targets of TILs from melanomas. Recent technological advances permit rapid identification of the neoepitopes resulting from these somatic gene mutations and of T cells with reactivity against these targets. Isolation and adoptive transfer of these T cells may improve TIL therapy for melanoma and permit its broader application to non-melanoma tumors. Extension of ACT to other malignancies may also be possible through antigen receptor gene engineering. Tumor regression has been observed following transfer of T cells engineered to express chimeric antigen receptors against CD19 in B-cell malignancies or a T-cell receptor against NY-ESO-1 in synovial cell sarcoma and melanoma. Herein, we review recent clinical trials of TILs and antigen receptor gene therapy for advanced cancers. We discuss lessons from this experience and consider how they might be applied to realize the full curative potential of ACT.
Collapse
|
1164
|
Abstract
Recent clinical success has underscored the potential for immunotherapy based on the adoptive cell transfer (ACT) of engineered T lymphocytes to mediate dramatic, potent, and durable clinical responses. This success has led to the broader evaluation of engineered T-lymphocyte-based adoptive cell therapy to treat a broad range of malignancies. In this review, we summarize concepts, successes, and challenges for the broader development of this promising field, focusing principally on lessons gleaned from immunological principles and clinical thought. We present ACT in the context of integrating T-cell and tumor biology and the broader systemic immune response.
Collapse
Affiliation(s)
- Marco Ruella
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
1165
|
Anderson D, Najafzadeh M, Gopalan R, Ghaderi N, Scally AJ, Britland ST, Jacobs BK, Reynolds PD, Davies J, Wright AL, Al-Ghazal S, Sharpe D, Denyer MC. Sensitivity and specificity of the empirical lymphocyte genome sensitivity (LGS) assay: implications for improving cancer diagnostics. FASEB J 2014; 28:4563-70. [PMID: 25063845 DOI: 10.1096/fj.14-254748] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Lymphocyte responses from 208 individuals: 20 with melanoma, 34 with colon cancer, and 4 with lung cancer (58), 18 with suspected melanoma, 28 with polyposis, and 10 with COPD (56), and 94 healthy volunteers were examined. The natural logarithm of the Olive tail moment (OTM) was plotted for exposure to UVA through 5 different agar depths (100 cell measurements/depth) and analyzed using a repeated measures regression model. Responses of patients with cancer plateaued after treatment with different UVA intensities, but returned toward control values for healthy volunteers. For precancerous conditions and suspected cancers, intermediate responses occurred. ROC analysis of mean log OTMs, for cancers plus precancerous/suspect conditions vs. controls, cancer vs. precancerous/suspect conditions plus controls, and cancer vs. controls, gave areas under the curve of 0.87, 0.89, and 0.93, respectively (P<0.001). Optimization allowed test sensitivity or specificity to approach 100% with acceptable complementary measures. This modified comet assay could represent a stand-alone test or an adjunct to other investigative procedures for detecting cancer.
Collapse
Affiliation(s)
| | | | | | | | - Andrew J Scally
- School of Health Studies, University of Bradford, Bradford, UK
| | - Stephen T Britland
- Faculty of Science and Engineering, University of Wolverhampton, Wolverhampton, UK; and
| | | | | | | | | | | | | | | |
Collapse
|
1166
|
Abstract
Targeting CTLA-4 represents a new type of immunotherapeutic approach, namely immune checkpoint inhibition. Blockade of CTLA-4 by ipilimumab was the first strategy to achieve a significant clinical benefit for late-stage melanoma patients in two phase 3 trials. These results fueled the notion of immunotherapy being the breakthrough strategy for oncology in 2013. Subsequently, many trials have been set up to test various immune checkpoint modulators in malignancies, not only in melanoma. In this review, recent new ideas about the mechanism of action of CTLA-4 blockade, its current and future therapeutic use, and the intensive search for biomarkers for response will be discussed. Immune checkpoint blockade, targeting CTLA-4 and/or PD-1/PD-L1, is currently the most promising systemic therapeutic approach to achieve long-lasting responses or even cure in many types of cancer, not just in patients with melanoma.
Collapse
Affiliation(s)
- Christian U Blank
- Department of Medical Oncology and Division of Immunology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - Alexander Enk
- Department of Dermatology, The University of Heidelberg, Im Neunheimer Feld 440, 69115 Heidelberg, Germany
| |
Collapse
|
1167
|
Malas S, Harrasser M, Lacy KE, Karagiannis SN. Antibody therapies for melanoma: new and emerging opportunities to activate immunity (Review). Oncol Rep 2014; 32:875-86. [PMID: 24969320 PMCID: PMC4121424 DOI: 10.3892/or.2014.3275] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 06/06/2014] [Indexed: 12/21/2022] Open
Abstract
The interface between malignant melanoma and patient immunity has long been recognised and efforts to treat this most lethal form of skin cancer by activating immune responses with cytokine, vaccine and also antibody immunotherapies have demonstrated promise in limited subsets of patients. In the present study, we discuss different antibody immunotherapy approaches evaluated in the context of melanoma, each designed to act on distinct targets and to employ different mechanisms to restrict tumour growth and spread. Monoclonal antibodies recognising melanoma-associated antigens such as CSPG4/MCSP and targeting elements of tumour-associated vasculature (VEGF) have constituted long-standing translational approaches aimed at reducing melanoma growth and metastasis. Recent insights into mechanisms of immune regulation and tumour-immune cell interactions have helped to identify checkpoint molecules on immune (CTLA4, PD-1) and tumour (PD-L1) cells as promising therapeutic targets. Checkpoint blockade with antibodies to activate immune responses and perhaps to counteract melanoma-associated immunomodulatory mechanisms led to the first clinical breakthrough in the form of an anti-CTLA4 monoclonal antibody. Novel modalities to target key mechanisms of immune suppression and to redirect potent effector cell subsets against tumours are expected to improve clinical outcomes and to provide previously unexplored avenues for therapeutic interventions.
Collapse
Affiliation(s)
- Sadek Malas
- St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine and NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals, King's College London, London SE1 9RT, UK
| | - Micaela Harrasser
- St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine and NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals, King's College London, London SE1 9RT, UK
| | - Katie E Lacy
- St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine and NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals, King's College London, London SE1 9RT, UK
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, Division of Genetics and Molecular Medicine and NIHR Biomedical Research Centre at Guy's and St. Thomas' Hospitals, King's College London, London SE1 9RT, UK
| |
Collapse
|
1168
|
Li S, Symonds ALJ, Miao T, Sanderson I, Wang P. Modulation of antigen-specific T-cells as immune therapy for chronic infectious diseases and cancer. Front Immunol 2014; 5:293. [PMID: 24987395 PMCID: PMC4060297 DOI: 10.3389/fimmu.2014.00293] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2014] [Accepted: 06/04/2014] [Indexed: 12/22/2022] Open
Abstract
T-cell responses are induced by antigen presenting cells (APC) and signals from the microenvironment. Antigen persistence and inflammatory microenvironments in chronic infections and cancer can induce a tolerant state in T-cells resulting in hyporesponsiveness, loss of effector function, and weak biochemical signaling patterns in response to antigen stimulation. Although the mechanisms of T-cell tolerance induced in chronic infection and cancer may differ from those involved in tolerance to self-antigen, the impaired proliferation and production of IL-2 in response to antigen stimulation are hallmarks of all tolerant T cells. In this review, we will summarize the evidence that the immune responses change from non-self to “self”-like in chronic infection and cancer, and will provide an overview of strategies for re-balancing the immune response of antigen-specific T cells in chronic infection and cancer without affecting the homeostasis of the immune system.
Collapse
Affiliation(s)
- Suling Li
- Bioscience, Brunel University , London , UK
| | - Alistair L J Symonds
- Blizard Institute (BICMS), Barts and the London School of Medicine and Dentistry , London , UK
| | - Tizong Miao
- Blizard Institute (BICMS), Barts and the London School of Medicine and Dentistry , London , UK
| | - Ian Sanderson
- Blizard Institute (BICMS), Barts and the London School of Medicine and Dentistry , London , UK
| | - Ping Wang
- Blizard Institute (BICMS), Barts and the London School of Medicine and Dentistry , London , UK
| |
Collapse
|
1169
|
Voo KS, Bover L, Harline ML, Weng J, Sugimoto N, Liu YJ. Targeting of TLRs inhibits CD4+ regulatory T cell function and activates lymphocytes in human peripheral blood mononuclear cells. THE JOURNAL OF IMMUNOLOGY 2014; 193:627-34. [PMID: 24928999 DOI: 10.4049/jimmunol.1203334] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Accumulating evidence suggests elements within tumors induce exhaustion of effector T cells and infiltration of immunosuppressive regulatory T cells (Tregs), thus preventing the development of durable antitumor immunity. Therefore, the discovery of agents that simultaneously block Treg suppressive function and reinvigorate effector function of lymphocytes is key to the development of effective cancer immunotherapy. Previous studies have shown that TLR ligands (TLRLs) could modulate the function of these T cell targets; however, those studies relied on cell-free or accessory cell-based assay systems that do not accurately reflect in vivo responses. In contrast, we used a human PBMC-based proliferation assay system to simultaneously monitor the effect of TLRLs on T cells (CD4(+), CD8(+), Tregs), B cells, and NK cells, which gave different and even conflicting results. We found that the TLR7/8L:CL097 could simultaneously activate CD8(+) T cells, B cells, and NK cells plus block Treg suppression of T cells and B cells. The TLRLs TLR1/2L:Pam3CSK4, TLR5L:flagellin, TLR4L:LPS, and TLR8/7L:CL075 also blocked Treg suppression of CD4(+) or CD8(+) T cell proliferation, but not B cell proliferation. Besides CL097, TLR2L:PGN, CL075, and TLR9L:CpG-A, CpG-B, and CpG-C) were strong activators of NK cells. Importantly, we found that Pam3CSK4 could: 1) activate CD4(+) T cell proliferation, 2) inhibit the expansion of IL-10(+) naturally occurring FOXP3(+) Tregs and induction of IL-10(+) CD4(+) Tregs (IL-10-producing type 1 Treg), and 3) block naturally occurring FOXP3(+) Tregs suppressive function. Our results suggest these agents could serve as adjuvants to enhance the efficacy of current immunotherapeutic strategies in cancer patients.
Collapse
Affiliation(s)
- Kui Shin Voo
- Department of Genomic Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030;
| | - Laura Bover
- Department of Genomic Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Megan Lundell Harline
- Department of Genomic Medicine, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030
| | - Jinsheng Weng
- Department of Lymphoma and Myeloma, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030; and
| | | | | |
Collapse
|
1170
|
Moon EK, Wang LC, Dolfi DV, Wilson CB, Ranganathan R, Sun J, Kapoor V, Scholler J, Puré E, Milone MC, June CH, Riley JL, Wherry EJ, Albelda SM. Multifactorial T-cell hypofunction that is reversible can limit the efficacy of chimeric antigen receptor-transduced human T cells in solid tumors. Clin Cancer Res 2014; 20:4262-73. [PMID: 24919573 DOI: 10.1158/1078-0432.ccr-13-2627] [Citation(s) in RCA: 334] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE Immunotherapy using vaccines or adoptively transferred tumor-infiltrating lymphocytes (TIL) is limited by T-cell functional inactivation within the solid tumor microenvironment. The purpose of this study was to determine whether a similar tumor-induced inhibition occurred with genetically modified cytotoxic T cells expressing chimeric antigen receptors (CAR) targeting tumor-associated antigens. EXPERIMENTAL DESIGN Human T cells expressing CAR targeting mesothelin or fibroblast activation protein and containing CD3ζ and 4-1BB cytoplasmic domains were intravenously injected into immunodeficient mice bearing large, established human mesothelin-expressing flank tumors. CAR TILs were isolated from tumors at various time points and evaluated for effector functions and status of inhibitory pathways. RESULTS CAR T cells were able to traffic into tumors with varying efficiency and proliferate. They were able to slow tumor growth, but did not cause regressions or cures. The CAR TILs underwent rapid loss of functional activity that limited their therapeutic efficacy. This hypofunction was reversible when the T cells were isolated away from the tumor. The cause of the hypofunction seemed to be multifactorial and was associated with upregulation of intrinsic T-cell inhibitory enzymes (diacylglycerol kinase and SHP-1) and the expression of surface inhibitory receptors (PD1, LAG3, TIM3, and 2B4). CONCLUSIONS Advanced-generation human CAR T cells are reversibly inactivated within the solid tumor microenvironment of some tumors by multiple mechanisms. The model described here will be an important tool for testing T cell-based strategies or systemic approaches to overcome this tumor-induced inhibition. Our results suggest that PD1 pathway antagonism may augment human CAR T-cell function.
Collapse
MESH Headings
- Animals
- BALB 3T3 Cells
- Cytotoxicity, Immunologic/immunology
- Endopeptidases
- Female
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/immunology
- GPI-Linked Proteins/metabolism
- Gelatinases/genetics
- Gelatinases/immunology
- Gelatinases/metabolism
- Humans
- Immunotherapy, Adoptive
- Lymphocytes, Tumor-Infiltrating/immunology
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Mesothelin
- Mesothelioma/immunology
- Mesothelioma/metabolism
- Mesothelioma/therapy
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Serine Endopeptidases/genetics
- Serine Endopeptidases/immunology
- Serine Endopeptidases/metabolism
- Signal Transduction
- T-Lymphocytes/immunology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Edmund K Moon
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine,
| | - Liang-Chuan Wang
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | | | - Caleph B Wilson
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine; and
| | | | - Jing Sun
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - Veena Kapoor
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| | - John Scholler
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine; and
| | - Ellen Puré
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Michael C Milone
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine; and
| | - Carl H June
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, Perelman School of Medicine; and
| | - James L Riley
- Department of Microbiology and Institute for Immunology
| | - E John Wherry
- Department of Microbiology and Institute for Immunology
| | - Steven M Albelda
- Division of Pulmonary, Allergy, and Critical Care, Department of Medicine
| |
Collapse
|
1171
|
Gyorki DE, Spillane J, Speakman D, Shackleton M, Henderson MA. Current management of advanced melanoma: a transformed landscape. ANZ J Surg 2014; 84:612-7. [PMID: 24842394 DOI: 10.1111/ans.12673] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/10/2014] [Indexed: 12/12/2022]
Abstract
The prognosis for patients with stage IV melanoma has historically been extremely poor and there have until recently been no effective treatment options. The last 3 years have seen a seismic shift in the management of these patients with the entry to the clinic of a number of novel agents with proven efficacy. These agents fall into two main classes: molecular-targeted therapy and immunotherapy. Molecular therapies have primarily targeted the mitogen-activated protein kinase pathway, most notably with oral inhibitors targetting oncogenic BRAF. Immunotherapy agents such as ipilimumab, and more recently antibodies against PD-1 boost the host immune response against the melanoma. It is important for surgeons to be aware of these advances for a number of reasons. Firstly, to be able to inform their patients of the general options available in the event of disease progression. Secondly, these agents are currently being assessed in the adjuvant setting and are likely to demonstrate efficacy for earlier stages of disease. Finally, it is important for surgeons to be able to advocate on their patients' behalf to minimize the lag time between publication of these promising results and the availability of these agents in the clinic. Furthermore, patients with advanced melanoma should be offered participation in clinical trials in order to refine the indications for these agents to maximize their chance of benefit.
Collapse
Affiliation(s)
- David E Gyorki
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia; Department of Surgery, University of Melbourne, Parkville, Victoria, Australia
| | | | | | | | | |
Collapse
|
1172
|
Kong YCM, Flynn JC. Opportunistic Autoimmune Disorders Potentiated by Immune-Checkpoint Inhibitors Anti-CTLA-4 and Anti-PD-1. Front Immunol 2014; 5:206. [PMID: 24904570 PMCID: PMC4032988 DOI: 10.3389/fimmu.2014.00206] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 04/25/2014] [Indexed: 12/16/2022] Open
Abstract
To improve the efficacy of immunotherapy for cancer and autoimmune diseases, recent ongoing and completed clinical trials have focused on specific targets to redirect the immune network toward eradicating a variety of tumors and ameliorating the self-destructive process. In a previous review, both systemic immunomodulators and monoclonal antibodies (mAbs), anti-CTLA-4, and anti-CD52, were discussed regarding therapeutics and autoimmune sequelae, as well as predisposing factors known to exacerbate immune-related adverse events (irAEs). This review will focus on immune-checkpoint inhibitors, and the data from most clinical trials involve blockade with anti-CTLA-4 such as ipilimumab. However, despite the mild to severe irAEs observed with ipilimumab in ~60% of patients, overall survival (OS) averaged ~22-25% at 3-5 years. To boost OS, other mAbs targeting programed death-1 and its ligand are undergoing clinical trials as monotherapy or dual therapy with anti-CTLA-4. Therapeutic combinations may generate different spectrum of opportunistic autoimmune disorders. To simulate clinical scenarios, we have applied regulatory T cell perturbation to murine models combined to examine the balance between thyroid autoimmunity and tumor-specific immunity.
Collapse
Affiliation(s)
- Yi-Chi M Kong
- Department of Immunology and Microbiology, Wayne State University School of Medicine , Detroit, MI , USA
| | - Jeffrey C Flynn
- Department of Orthopaedic Surgery, Providence Hospital and Medical Centers , Southfield, MI , USA
| |
Collapse
|
1173
|
Systemic Therapy for Merkel Cell Carcinoma: What's on the Horizon? Cancers (Basel) 2014; 6:1180-94. [PMID: 24840048 PMCID: PMC4074823 DOI: 10.3390/cancers6021180] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/08/2014] [Accepted: 05/09/2014] [Indexed: 12/23/2022] Open
Abstract
Merkel cell carcinoma is an aggressive neuroendocrine skin cancer that usually affects elderly patients. Despite being uncommon, incidence has been steadily increasing over the last two decades, likely due to increased awareness, better diagnostic methods and aging of the population. It is currently one of the most lethal cutaneous malignancies, with a five-year overall survival of approximately 50%. With the better understanding of the molecular pathways that lead to the development of Merkel cell carcinoma, there has been an increasing excitement and optimism surrounding novel targeted therapies, in particular to immunotherapy. Some of the concepts surrounding the novel targeted therapies and currently ongoing clinical trials are reviewed here.
Collapse
|
1174
|
Mockler MB, Conroy MJ, Lysaght J. Targeting T cell immunometabolism for cancer immunotherapy; understanding the impact of the tumor microenvironment. Front Oncol 2014; 4:107. [PMID: 24904823 PMCID: PMC4032940 DOI: 10.3389/fonc.2014.00107] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2014] [Accepted: 04/28/2014] [Indexed: 12/20/2022] Open
Abstract
The immune system has a key role to play in controlling cancer initiation and progression. T cell activation, which is central to anti-tumor immune responses, coincides with changes in cellular metabolism. Naïve T cells predominantly require an ATP generating metabolic profile, whereas proliferating effector T cells require anabolic metabolic profiles that promote rapid growth and proliferation. Furthermore, specific T cell subsets require distinct energetic and biosynthetic pathways to match their functional requirements. The often hostile tumor microenvironment can affect T cell immune responses by altering the resulting cellular metabolism. Tailoring immune responses by manipulating cellular metabolic pathways may provide an exciting new option for cancer immunotherapy. T cell responses might also be skewed via metabolic manipulation to treat the complications of obesity-associated inflammation, which is a rapidly growing global health problem and a major risk factor for many malignancies. In this review, the diverse metabolic requirements of T cells in anti-tumor immunity are discussed, as well as the profound influence of the tumor microenvironment and the possible avenues for manipulation to enhance anti-tumor immunity.
Collapse
Affiliation(s)
- Mary B Mockler
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin , Dublin , Ireland
| | - Melissa J Conroy
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin , Dublin , Ireland
| | - Joanne Lysaght
- Department of Surgery, Trinity Centre for Health Sciences, St. James's Hospital, Trinity College Dublin , Dublin , Ireland
| |
Collapse
|
1175
|
Wood LM, Paterson Y. Attenuated Listeria monocytogenes: a powerful and versatile vector for the future of tumor immunotherapy. Front Cell Infect Microbiol 2014; 4:51. [PMID: 24860789 PMCID: PMC4026700 DOI: 10.3389/fcimb.2014.00051] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 04/04/2014] [Indexed: 12/17/2022] Open
Abstract
For over a century, inactivated or attenuated bacteria have been employed in the clinic as immunotherapies to treat cancer, starting with the Coley's vaccines in the 19th century and leading to the currently approved bacillus Calmette-Guérin vaccine for bladder cancer. While effective, the inflammation induced by these therapies is transient and not designed to induce long-lasting tumor-specific cytolytic T lymphocyte (CTL) responses that have proven so adept at eradicating tumors. Therefore, in order to maintain the benefits of bacteria-induced acute inflammation but gain long-lasting anti-tumor immunity, many groups have constructed recombinant bacteria expressing tumor-associated antigens (TAAs) for the purpose of activating tumor-specific CTLs. One bacterium has proven particularly adept at inducing powerful anti-tumor immunity, Listeria monocytogenes (Lm). Lm is a gram-positive bacterium that selectively infects antigen-presenting cells wherein it is able to efficiently deliver tumor antigens to both the MHC Class I and II antigen presentation pathways for activation of tumor-targeting CTL-mediated immunity. Lm is a versatile bacterial vector as evidenced by its ability to induce therapeutic immunity against a wide-array of TAAs and specifically infect and kill tumor cells directly. It is for these reasons, among others, that Lm-based immunotherapies have delivered impressive therapeutic efficacy in preclinical models of cancer for two decades and are now showing promise clinically. In this review, we will provide an overview of the history leading up to the development of current Lm-based immunotherapies, the advantages and mechanisms of Lm as a therapeutic vaccine vector, the preclinical experience with Lm-based immunotherapies targeting a number of malignancies, and the recent findings from clinical trials along with concluding remarks on the future of Lm-based tumor immunotherapies.
Collapse
Affiliation(s)
- Laurence M Wood
- Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center Abilene, TX, USA
| | - Yvonne Paterson
- Microbiology, Perelman School of Medicine, University of Pennsylvania Philadelphia, PA, USA ; University of Pennsylvania School of Nursing Philadelphia, PA, USA
| |
Collapse
|
1176
|
Bauer CA, Kim EY, Marangoni F, Carrizosa E, Claudio NM, Mempel TR. Dynamic Treg interactions with intratumoral APCs promote local CTL dysfunction. J Clin Invest 2014; 124:2425-40. [PMID: 24812664 DOI: 10.1172/jci66375] [Citation(s) in RCA: 180] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 03/20/2014] [Indexed: 12/29/2022] Open
Abstract
Tregs control various functions of effector T cells; however, where and how Tregs exert their immunomodulatory effects remain poorly understood. Here we developed a murine model of adoptive T cell therapy and found that Tregs induce a dysfunctional state in tumor-infiltrating CTLs that resembles T cell exhaustion and is characterized by low expression of effector cytokines, inefficient cytotoxic granule release, and coexpression of coinhibitory receptors PD-1 and TIM-3. Induction of CTL dysfunction was an active process, requiring local TCR signals in tumor tissue. Tregs infiltrated tumors only subsequent to Ag-dependent activation and expansion in tumor-draining LNs; however, Tregs also required local Ag reencounter within tumor tissue to induce CTL dysfunction and prevent tumor rejection. Multiphoton intravital microscopy revealed that in contrast to CTLs, Tregs only rarely and briefly interrupted their migration in tumor tissue in an Ag-dependent manner and formed unstable tethering-interactions with CD11c+ APCs, coinciding with a marked reduction of CD80 and CD86 on APCs. Activation of CTLs by Treg-conditioned CD80/86lo DCs promoted enhanced expression of both TIM-3 and PD-1. Based on these data, we propose that Tregs locally change the costimulatory landscape in tumor tissue through transient, Ag-dependent interactions with APCs, thus inducing CTL dysfunction by altering the balance of costimulatory and coinhibitory signals these cells receive.
Collapse
|
1177
|
Wong RM, Ianculescu I, Sharma S, Gage DL, Olevsky OM, Kotova S, Kostic MN, Grundfest WS, Hou D, Cameron RB. Immunotherapy for Malignant Pleural Mesothelioma. Current Status and Future Prospects. Am J Respir Cell Mol Biol 2014; 50:870-875. [DOI: 10.1165/rcmb.2013-0472tr] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Raymond M. Wong
- Pacific Meso Center at the Pacific Heart, Lung & Blood Institute, Los Angeles, California
| | - Irina Ianculescu
- Pacific Meso Center at the Pacific Heart, Lung & Blood Institute, Los Angeles, California
| | | | | | | | - Svetlana Kotova
- Division of Thoracic Surgery, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California; and
| | | | | | - Dongmei Hou
- Division of Thoracic Surgery and Comprehensive Mesothelioma Program, University of California, Los Angeles, CA
| | - Robert B. Cameron
- Division of Thoracic Surgery, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California; and
- Division of Thoracic Surgery and Comprehensive Mesothelioma Program, University of California, Los Angeles, CA
| |
Collapse
|
1178
|
Stinchcombe TE. Unmet needs in squamous cell carcinoma of the lung: potential role for immunotherapy. Med Oncol 2014; 31:960. [PMID: 24748366 PMCID: PMC4006124 DOI: 10.1007/s12032-014-0960-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 04/02/2014] [Indexed: 12/15/2022]
Abstract
Squamous cell carcinoma of the lung accounts for 20–30 % of non-small cell lung cancers (NSCLC). Despite the differences in disease characteristics between squamous and non-squamous NSCLC, both have historically been treated similarly in the clinic. Recently approved drugs have revealed differences in activity and safety profiles across histologic subtypes and have applicability in treating non-squamous, but not typically squamous, NSCLC. Exploration of immune checkpoints—co-inhibitory molecules used to regulate immune responses—has resulted in novel immunotherapies designed to interrupt signaling through the cytotoxic T lymphocyte-associated antigen-4 or programmed cell death protein-1 pathways on lymphocytes. Modulation of these pathways can lead to restored antitumor immune responses, and preliminary evidence shows that agents targeting these pathways have activity in lung cancer, including squamous NSCLC.
Collapse
Affiliation(s)
- Thomas E Stinchcombe
- Multi-disciplinary Thoracic Oncology Program, University of North Carolina UNC Chapel Hill, Physicians Office Bldg. CB# 7305, 170 Manning Drive, 3rd Floor, Chapel Hill, NC, 27599-7305, USA,
| |
Collapse
|
1179
|
Shtivelman E, Davies MA, Hwu P, Yang J, Lotem M, Oren M, Flaherty KT, Fisher DE. Pathways and therapeutic targets in melanoma. Oncotarget 2014; 5:1701-52. [PMID: 24743024 PMCID: PMC4039128 DOI: 10.18632/oncotarget.1892] [Citation(s) in RCA: 165] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 04/07/2014] [Indexed: 02/07/2023] Open
Abstract
This review aims to summarize the current knowledge of molecular pathways and their clinical relevance in melanoma. Metastatic melanoma was a grim diagnosis, but in recent years tremendous advances have been made in treatments. Chemotherapy provided little benefit in these patients, but development of targeted and new immune approaches made radical changes in prognosis. This would not have happened without remarkable advances in understanding the biology of disease and tremendous progress in the genomic (and other "omics") scale analyses of tumors. The big problems facing the field are no longer focused exclusively on the development of new treatment modalities, though this is a very busy area of clinical research. The focus shifted now to understanding and overcoming resistance to targeted therapies, and understanding the underlying causes of the heterogeneous responses to immune therapy.
Collapse
Affiliation(s)
| | | | - Patrick Hwu
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James Yang
- National Cancer Institute, NIH, Washington DC, USA
| | - Michal Lotem
- Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Moshe Oren
- The Weizmann Institute of Science, Rehovot, Israel
| | | | - David E. Fisher
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| |
Collapse
|
1180
|
Karyampudi L, Lamichhane P, Scheid AD, Kalli KR, Shreeder B, Krempski JW, Behrens MD, Knutson KL. Accumulation of memory precursor CD8 T cells in regressing tumors following combination therapy with vaccine and anti-PD-1 antibody. Cancer Res 2014; 74:2974-85. [PMID: 24728077 DOI: 10.1158/0008-5472.can-13-2564] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Immunosuppression in the tumor microenvironment blunts vaccine-induced immune effectors. PD-1/B7-H1 is an important inhibitory axis in the tumor microenvironment. Our goal in this study was to determine the effect of blocking this inhibitory axis during and following vaccination against breast cancer. We observed that using anti-PD-1 antibody and a multipeptide vaccine (consisting of immunogenic peptides derived from breast cancer antigens, neu, legumain, and β-catenin) as a combination therapy regimen for the treatment of breast cancer-bearing mice prolonged the vaccine-induced progression-free survival period. This prolonged survival was associated with increase in number of Tc1 and Tc2 CD8 T cells with memory precursor phenotype, CD27+IL-7RhiT-betlo, and decrease in number of PD-1+ dendritic cells (DC) in regressing tumors and enhanced antigen reactivity of tumor-infiltrating CD8 T cells. It was also observed that blockade of PD-1 on tumor DCs enhanced IL-7R expression on CD8 T cells. Taken together, our results suggest that PD-1 blockade enhances breast cancer vaccine efficacy by altering both CD8 T cell and DC components of the tumor microenvironment. Given the recent success of anti-PD-1 monotherapy, our results are encouraging for developing combination therapies for the treatment of patients with cancer in which anti-PD-1 monotherapy alone may be ineffective (i.e., PD-L1-negative tumors).
Collapse
Affiliation(s)
- Lavakumar Karyampudi
- Authors' Affiliations: Cancer Vaccines and Immune Therapies Program, Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida; and Departments of Immunology and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Purushottam Lamichhane
- Authors' Affiliations: Cancer Vaccines and Immune Therapies Program, Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida; and Departments of Immunology and Oncology, Mayo Clinic, Rochester, MinnesotaAuthors' Affiliations: Cancer Vaccines and Immune Therapies Program, Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida; and Departments of Immunology and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Adam D Scheid
- Authors' Affiliations: Cancer Vaccines and Immune Therapies Program, Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida; and Departments of Immunology and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Kimberly R Kalli
- Authors' Affiliations: Cancer Vaccines and Immune Therapies Program, Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida; and Departments of Immunology and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Barath Shreeder
- Authors' Affiliations: Cancer Vaccines and Immune Therapies Program, Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida; and Departments of Immunology and Oncology, Mayo Clinic, Rochester, Minnesota
| | - James W Krempski
- Authors' Affiliations: Cancer Vaccines and Immune Therapies Program, Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida; and Departments of Immunology and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Marshall D Behrens
- Authors' Affiliations: Cancer Vaccines and Immune Therapies Program, Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida; and Departments of Immunology and Oncology, Mayo Clinic, Rochester, Minnesota
| | - Keith L Knutson
- Authors' Affiliations: Cancer Vaccines and Immune Therapies Program, Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida; and Departments of Immunology and Oncology, Mayo Clinic, Rochester, MinnesotaAuthors' Affiliations: Cancer Vaccines and Immune Therapies Program, Vaccine & Gene Therapy Institute of Florida, Port St. Lucie, Florida; and Departments of Immunology and Oncology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
1181
|
Fehres CM, Unger WWJ, Garcia-Vallejo JJ, van Kooyk Y. Understanding the biology of antigen cross-presentation for the design of vaccines against cancer. Front Immunol 2014; 5:149. [PMID: 24782858 PMCID: PMC3986565 DOI: 10.3389/fimmu.2014.00149] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/21/2014] [Indexed: 12/19/2022] Open
Abstract
Antigen cross-presentation, the process in which exogenous antigens are presented on MHC class I molecules, is crucial for the generation of effector CD8(+) T cell responses. Although multiple cell types are being described to be able to cross-present antigens, in vivo this task is mainly carried out by certain subsets of dendritic cells (DCs). Aspects such as the internalization route, the pathway of endocytic trafficking, and the simultaneous activation through pattern-recognition receptors have a determining influence in how antigens are handled for cross-presentation by DCs. In this review, we will summarize new insights in factors that affect antigen cross-presentation of human DC subsets, and we will discuss the possibilities to exploit antigen cross-presentation for immunotherapy against cancer.
Collapse
Affiliation(s)
- Cynthia M Fehres
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| | - Wendy W J Unger
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| | - Juan J Garcia-Vallejo
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| |
Collapse
|
1182
|
The pharmacological impact of ATP-binding cassette drug transporters on vemurafenib-based therapy. Acta Pharm Sin B 2014; 4:105-11. [PMID: 26579371 PMCID: PMC4590304 DOI: 10.1016/j.apsb.2013.12.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/02/2013] [Accepted: 12/10/2013] [Indexed: 01/06/2023] Open
Abstract
Melanoma is the most serious type of skin cancer and one of the most common cancers in the world. Advanced melanoma is often resistant to conventional therapies and has high potential for metastasis and low survival rates. Vemurafenib is a small molecule inhibitor of the BRAF serine-threonine kinase recently approved by the United States Food and Drug Administration to treat patients with metastatic and unresectable melanomas that carry an activating BRAF (V600E) mutation. Many clinical trials evaluating other therapeutic uses of vemurafenib are still ongoing. The ATP-binding cassette (ABC) transporters are membrane proteins with important physiological and pharmacological roles. Collectively, they transport and regulate levels of physiological substrates such as lipids, porphyrins and sterols. Some of them also remove xenobiotics and limit the oral bioavailability and distribution of many chemotherapeutics. The overexpression of three major ABC drug transporters is the most common mechanism for acquired resistance to anticancer drugs. In this review, we highlight some of the recent findings related to the effect of ABC drug transporters such as ABCB1 and ABCG2 on the oral bioavailability of vemurafenib, problems associated with treating melanoma brain metastases and the development of acquired resistance to vemurafenib in cancers harboring the BRAF (V600E) mutation.
Collapse
Key Words
- ABC transporter
- ABC, ATP-binding cassette
- AML, acute myeloid leukemia
- BBB, blood–brain barrier
- CNS, central nervous system
- CSCs, cancer stem cells
- Drug resistance
- GI, gastrointestinal
- MAPK, mitogen-activated protein kinase
- MDR, multidrug resistance
- Melanoma
- NBDs, nucleotide-binding domains
- P-glycoprotein
- PFS, longer progression-free survival
- PKIs, protein kinase inhibitors
- TKIs, tyrosine kinase inhibitors
- TMDs, transmembrane domains
- Vemurafenib
Collapse
|
1183
|
Sun S, Fei X, Mao Y, Wang X, Garfield DH, Huang O, Wang J, Yuan F, Sun L, Yu Q, Jin X, Wang J, Shen K. PD-1(+) immune cell infiltration inversely correlates with survival of operable breast cancer patients. Cancer Immunol Immunother 2014; 63:395-406. [PMID: 24514954 PMCID: PMC11029035 DOI: 10.1007/s00262-014-1519-x] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 01/10/2014] [Indexed: 01/21/2023]
Abstract
The programmed death-1 (PD-1) molecule is mainly expressed on functionally "exhausted" CD8(+) T cells, dampening the host antitumor immune response. We evaluated the ratio between effective and regulatory T cells (Tregs) and PD-1 expression as a prognostic factor for operable breast cancer patients. A series of 218 newly diagnosed invasive breast cancer patients who had undergone primary surgery at Ruijin Hospital were identified. The influence of CD8(+) cytotoxic T lymphocytes, FOXP3(+) (Treg cell marker), and PD-1(+) immune cell counts on prognosis was analyzed utilizing immunohistochemistry. Both PD-1(+) immune cells and FOXP3(+) Tregs counts were significantly associated with unfavorable prognostic factors. In bivariate, but not multivariate analysis, high tumor infiltrating PD-1(+) cell counts correlated with significantly shorter patient survival. Our results suggest a prognostic value of the PD-1(+) immune cell population in such breast cancer patients. Targeting the PD-1 pathway may be a feasible approach to treating patients with breast cancer.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antigens, Differentiation, T-Lymphocyte/analysis
- Apoptosis/immunology
- Breast Neoplasms/immunology
- Breast Neoplasms/mortality
- Breast Neoplasms/pathology
- Breast Neoplasms/radiotherapy
- Breast Neoplasms/surgery
- CD8-Positive T-Lymphocytes/chemistry
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Carcinoma, Ductal, Breast/immunology
- Carcinoma, Ductal, Breast/mortality
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Ductal, Breast/radiotherapy
- Carcinoma, Ductal, Breast/surgery
- Carcinoma, Lobular/immunology
- Carcinoma, Lobular/mortality
- Carcinoma, Lobular/pathology
- Carcinoma, Lobular/radiotherapy
- Carcinoma, Lobular/surgery
- Combined Modality Therapy
- Disease-Free Survival
- Female
- Follow-Up Studies
- Forkhead Transcription Factors/analysis
- Humans
- Kaplan-Meier Estimate
- Lymphocyte Count
- Lymphocytes, Tumor-Infiltrating/chemistry
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Mastectomy
- Middle Aged
- Prognosis
- Programmed Cell Death 1 Receptor/analysis
- Radiotherapy, Adjuvant
- Survival Analysis
- T-Lymphocyte Subsets/chemistry
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/pathology
- T-Lymphocytes, Cytotoxic/chemistry
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/pathology
- T-Lymphocytes, Regulatory/chemistry
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Shenyou Sun
- Shanghai Ruijin Hospital, Comprehensive Breast Health Center, Shanghai, 200025 China
- Linyi People’s Hospital, General Surgery Ward 3, Linyi, 276003 Shandong China
| | - Xiaochun Fei
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai, 200025 China
| | - Yan Mao
- Shanghai Ruijin Hospital, Comprehensive Breast Health Center, Shanghai, 200025 China
| | - Xiumin Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - David H. Garfield
- Shanghai Ruijin Hospital, Comprehensive Breast Health Center, Shanghai, 200025 China
- Promed Cancer Centers-Shanghai, Shanghai, 200020 China
| | - Ou Huang
- Shanghai Ruijin Hospital, Comprehensive Breast Health Center, Shanghai, 200025 China
| | - Jinglong Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Fei Yuan
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai, 200025 China
| | - Long Sun
- Shanghai Ruijin Hospital, Comprehensive Breast Health Center, Shanghai, 200025 China
| | - Qixiang Yu
- Shanghai Ruijin Hospital, Comprehensive Breast Health Center, Shanghai, 200025 China
| | - Xiaolong Jin
- Department of Pathology, Shanghai Ruijin Hospital, Shanghai, 200025 China
| | - Jianhua Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Kunwei Shen
- Shanghai Ruijin Hospital, Comprehensive Breast Health Center, Shanghai, 200025 China
- Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2 Road, Shanghai, China
| |
Collapse
|
1184
|
Gros A, Robbins PF, Yao X, Li YF, Turcotte S, Tran E, Wunderlich JR, Mixon A, Farid S, Dudley ME, Hanada KI, Almeida JR, Darko S, Douek DC, Yang JC, Rosenberg SA. PD-1 identifies the patient-specific CD8⁺ tumor-reactive repertoire infiltrating human tumors. J Clin Invest 2014; 124:2246-59. [PMID: 24667641 DOI: 10.1172/jci73639] [Citation(s) in RCA: 865] [Impact Index Per Article: 78.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Accepted: 01/23/2014] [Indexed: 12/13/2022] Open
Abstract
Adoptive transfer of tumor-infiltrating lymphocytes (TILs) can mediate regression of metastatic melanoma; however, TILs are a heterogeneous population, and there are no effective markers to specifically identify and select the repertoire of tumor-reactive and mutation-specific CD8⁺ lymphocytes. The lack of biomarkers limits the ability to study these cells and develop strategies to enhance clinical efficacy and extend this therapy to other malignancies. Here, we evaluated unique phenotypic traits of CD8⁺ TILs and TCR β chain (TCRβ) clonotypic frequency in melanoma tumors to identify patient-specific repertoires of tumor-reactive CD8⁺ lymphocytes. In all 6 tumors studied, expression of the inhibitory receptors programmed cell death 1 (PD-1; also known as CD279), lymphocyte-activation gene 3 (LAG-3; also known as CD223), and T cell immunoglobulin and mucin domain 3 (TIM-3) on CD8⁺ TILs identified the autologous tumor-reactive repertoire, including mutated neoantigen-specific CD8⁺ lymphocytes, whereas only a fraction of the tumor-reactive population expressed the costimulatory receptor 4-1BB (also known as CD137). TCRβ deep sequencing revealed oligoclonal expansion of specific TCRβ clonotypes in CD8⁺PD-1⁺ compared with CD8⁺PD-1- TIL populations. Furthermore, the most highly expanded TCRβ clonotypes in the CD8⁺ and the CD8⁺PD-1⁺ populations recognized the autologous tumor and included clonotypes targeting mutated antigens. Thus, in addition to the well-documented negative regulatory role of PD-1 in T cells, our findings demonstrate that PD-1 expression on CD8⁺ TILs also accurately identifies the repertoire of clonally expanded tumor-reactive cells and reveal a dual importance of PD-1 expression in the tumor microenvironment.
Collapse
MESH Headings
- Adoptive Transfer
- Antigens, CD/genetics
- Antigens, CD/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cell Line, Tumor
- Female
- Hepatitis A Virus Cellular Receptor 2
- Humans
- Male
- Melanoma/genetics
- Melanoma/immunology
- Melanoma/pathology
- Melanoma/therapy
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
- Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
- Lymphocyte Activation Gene 3 Protein
Collapse
|
1185
|
Zheng Z, Bu Z, Liu X, Zhang L, Li Z, Wu A, Wu X, Cheng X, Xing X, Du H, Wang X, Hu Y, Ji J. Level of circulating PD-L1 expression in patients with advanced gastric cancer and its clinical implications. Chin J Cancer Res 2014; 26:104-11. [PMID: 24653632 DOI: 10.3978/j.issn.1000-9604.2014.02.08] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 02/10/2014] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE The programmed cell death-1 receptor/programmed cell death-1 ligand (PD-1/PD-L1) pathway plays a crucial role in tumor evasion from host immunity. This study was designed to evaluate the association between circulating PD-L1 expression and prognosis in patients with advanced gastric cancer. METHODS Totally 80 advanced gastric cancer patients and 40 health controls from Beijing Cancer Hospital were enrolled in the present study. Circulating PD-L1 expression was tested by enzyme-linked immunosorbent assay (ELISA). The associations between the expression level of PD-L1 and clinicopathological features and prognosis were analyzed statistically. RESULTS Expression of PD-L1 in advanced gastric cancer patients was significantly up-regulated compared with health people (P=0.006). The expression of PD-L1 was significantly correlated with differentiation and lymph node metastasis (P=0.026 and P=0.041, respectively). Although we didn't find significant difference in all advanced gastric cancer patients with different PD-L1 expression, the adenocarcinoma patients with higher up-regulated PD-L1 expression had much better prognosis than low expression patients (65.6% vs. 44.7%, P=0.028). CONCLUSIONS PD-L1 was elevated in advance gastric cancer patients and may play an important role in tumor immune evasion and patients prognosis.
Collapse
Affiliation(s)
- Zhixue Zheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhaode Bu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xijuan Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lianhai Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ziyu Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Aiwen Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaojiang Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaojing Cheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaofang Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Hong Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaohong Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ying Hu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jiafu Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), 1 Department of Gastrointestinal Surgery, 2 Department of Central Laboratory, 3 Clinical Gastric Cancer Translational Research Laboratory, 4 Biological Tissue Bank, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
1186
|
Tjin EP, Krebbers G, Meijlink KJ, van de Kasteele W, Rosenberg EH, Sanders J, Nederlof PM, van de Wiel BA, Haanen JB, Melief CJ, Vyth-Dreese FA, Luiten RM. Immune-Escape Markers in Relation to Clinical Outcome of Advanced Melanoma Patients Following Immunotherapy. Cancer Immunol Res 2014; 2:538-46. [DOI: 10.1158/2326-6066.cir-13-0097] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
1187
|
Chiu YL, Shan L, Huang H, Haupt C, Bessell C, Canaday DH, Zhang H, Ho YC, Powell JD, Oelke M, Margolick JB, Blankson JN, Griffin DE, Schneck JP. Sprouty-2 regulates HIV-specific T cell polyfunctionality. J Clin Invest 2014; 124:198-208. [PMID: 24292711 DOI: 10.1172/jci70510] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Accepted: 09/27/2013] [Indexed: 01/03/2023] Open
Abstract
The ability of individual T cells to perform multiple effector functions is crucial for protective immunity against viruses and cancer. This polyfunctionality is frequently lost during chronic infections; however, the molecular mechanisms driving T cell polyfunctionality are poorly understood. We found that human T cells stimulated by a high concentration of antigen lacked polyfunctionality and expressed a transcription profile similar to that of exhausted T cells. One specific pathway implicated by the transcription profile in control of T cell polyfunctionality was the MAPK/ERK pathway. This pathway was altered in response to different antigen concentrations, and polyfunctionality correlated with upregulation of phosphorylated ERK. T cells that were stimulated with a high concentration of antigen upregulated sprouty-2 (SPRY2), a negative regulator of the MAPK/ERK pathway. The clinical relevance of SPRY2 was confirmed by examining SPRY2 expression in HIV-specific T cells, where high levels of SPRY2 were seen in HIV-specific T cells and inhibition of SPRY2 expression enhanced the HIV-specific polyfunctional response independently of the PD-1 pathway. Our findings indicate that increased SPRY2 expression during chronic viral infection reduces T cell polyfunctionality and identify SPRY2 as a potential target for immunotherapy.
Collapse
|
1188
|
TGF-β upregulates CD70 expression and induces exhaustion of effector memory T cells in B-cell non-Hodgkin's lymphoma. Leukemia 2014; 28:1872-84. [PMID: 24569779 DOI: 10.1038/leu.2014.84] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 01/17/2014] [Accepted: 02/03/2014] [Indexed: 12/16/2022]
Abstract
Transforming growth factor beta (TGF-β) has an important role in mediating T-cell suppression in B-cell non-Hodgkin lymphoma (NHL). However, the underlying mechanism responsible for TGF-β-mediated inhibition of effector memory T (Tm) cells is largely unknown. As reported here, we show that exhaustion is a major mechanism by which TGF-β inhibits Tm cells, and TGF-β mediated exhaustion is associated with upregulation of CD70. We found that TGF-β upregulates CD70 expression on effector Tm cells while it preferentially induces Foxp3 expression in naive T cells. CD70 induction by TGF-β is Smad3-dependent and involves IL-2/Stat5 signaling. CD70+ T cells account for TGF-β-induced exhaustion of effector Tm cells. Both TGF-β-induced and preexisting intratumoral CD70+ effector Tm cells from B-cell NHL have an exhausted phenotype and express higher levels of PD-1 and TIM-3 compared with CD70- T cells. Signaling transduction, proliferation and cytokine production are profoundly decreased in these cells, and they are highly susceptible to apoptosis. Clinically, intratumoral CD70-expressing T cells are prevalent in follicular B-cell lymphoma (FL) biopsy specimens, and increased numbers of intratumoral CD70+ T cells correlate with an inferior patient outcome. These findings confirm TGF-β-mediated effector Tm cell exhaustion as an important mechanism of immune suppression in B-cell NHL.
Collapse
|
1189
|
Shi G, Zhou C, Wang D, Ma W, Liu B, Zhang S. Antitumor enhancement by adoptive transfer of tumor antigen primed, inactivated MHC-haploidentical lymphocytes. Cancer Lett 2014; 343:42-50. [DOI: 10.1016/j.canlet.2013.09.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2013] [Revised: 09/05/2013] [Accepted: 09/09/2013] [Indexed: 01/05/2023]
|
1190
|
Wolchinsky R, Hod-Marco M, Oved K, Shen-Orr SS, Bendall SC, Nolan GP, Reiter Y. Antigen-dependent integration of opposing proximal TCR-signaling cascades determines the functional fate of T lymphocytes. THE JOURNAL OF IMMUNOLOGY 2014; 192:2109-19. [PMID: 24489091 DOI: 10.4049/jimmunol.1301142] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
T cell anergy is a key tolerance mechanism to mitigate unwanted T cell activation against self by rendering lymphocytes functionally inactive following Ag encounter. Ag plays an important role in anergy induction where high supraoptimal doses lead to the unresponsive phenotype. How T cells "measure" Ag dose and how this determines functional output to a given antigenic dose remain unclear. Using multiparametric phospho-flow and mass cytometry, we measured the intracellular phosphorylation-dependent signaling events at a single-cell resolution and studied the phosphorylation levels of key proximal human TCR activation- and inhibition-signaling molecules. We show that the intracellular balance and signal integration between these opposing signaling cascades serve as the molecular switch gauging Ag dose. An Ag density of 100 peptide-MHC complexes/cell was found to be the transition point between dominant activation and inhibition cascades, whereas higher Ag doses induced an anergic functional state. Finally, the neutralization of key inhibitory molecules reversed T cell unresponsiveness and enabled maximal T cell functions, even in the presence of very high Ag doses. This mechanism permits T cells to make integrated "measurements" of Ag dose that determine subsequent functional outcomes.
Collapse
Affiliation(s)
- Ron Wolchinsky
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | | | | | | | | | | | | |
Collapse
|
1191
|
Pen JJ, Keersmaecker BD, Heirman C, Corthals J, Liechtenstein T, Escors D, Thielemans K, Breckpot K. Interference with PD-L1/PD-1 co-stimulation during antigen presentation enhances the multifunctionality of antigen-specific T cells. Gene Ther 2014; 21:262-71. [PMID: 24401835 DOI: 10.1038/gt.2013.80] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 10/02/2013] [Accepted: 11/25/2013] [Indexed: 01/03/2023]
Abstract
The release of cytokines by T cells strongly defines their functional activity in vivo. The ability to produce multiple cytokines has been associated with beneficial immune responses in cancer and infectious diseases, while their progressive loss is associated with T-cell exhaustion, senescence and anergy. Consequently, strategies that enhance the multifunctional status of T cells are a key for immunotherapy. Dendritic cells (DCs) are professional antigen presenting cells that regulate T-cell functions by providing positive and negative co-stimulatory signals. A key negative regulator of T-cell activity is provided by binding of programmed death-1 (PD-1) receptor on activated T cells, to its ligand PD-L1, expressed on DCs. We investigated the impact of interfering with PD-L1/PD-1 co-stimulation on the multifunctionality of T cells, by expression of the soluble extracellular part of PD-1 (sPD-1) or PD-L1 (sPD-L1) in human monocyte-derived DCs during antigen presentation. Expression, secretion and binding of these soluble molecules after mRNA electroporation were demonstrated. Modification of DCs with sPD-1 or sPD-L1 mRNA resulted in increased levels of the co-stimulatory molecule CD80 and a distinct cytokine profile, characterized by the secretion of IL-10 and TNF-α, respectively. Co-expression in DCs of sPD-1 and sPD-L1 with influenza virus nuclear protein 1 (Flu NP1) stimulated Flu NP1 memory T cells, with a significantly higher number of multifunctional T cells and increased cytokine secretion, while it did not induce regulatory T cells. These data provide a rationale for the inclusion of interfering sPD-1 or sPD-L1 in DC-based immunotherapeutic strategies.
Collapse
Affiliation(s)
- J J Pen
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Brussels, Belgium
| | - B D Keersmaecker
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Brussels, Belgium
| | - C Heirman
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Brussels, Belgium
| | - J Corthals
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Brussels, Belgium
| | - T Liechtenstein
- 1] Division of Infection and Immunity, Rayne Institute, University College London, London, UK [2] Navarrabiomed-FMS, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - D Escors
- 1] Division of Infection and Immunity, Rayne Institute, University College London, London, UK [2] Navarrabiomed-FMS, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - K Thielemans
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Brussels, Belgium
| | - K Breckpot
- Laboratory of Molecular and Cellular Therapy, Department of Immunology-Physiology, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
1192
|
Nagato T, Lee YR, Harabuchi Y, Celis E. Combinatorial immunotherapy of polyinosinic-polycytidylic acid and blockade of programmed death-ligand 1 induce effective CD8 T-cell responses against established tumors. Clin Cancer Res 2014; 20:1223-34. [PMID: 24389326 DOI: 10.1158/1078-0432.ccr-13-2781] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Epitope-based cancer vaccines capable of inducing CD8 T-cell responses to tumor-associated antigens (TAA) expressed by tumor cells have been considered as attractive alternatives for the treatment of some types of cancer. However, reliable TAAs have not been identified for most malignant diseases, limiting the development of epitope-based vaccines. Herein, we report that the combinatorial therapy of polyinosinic-polycytidylic acid (poly-IC) and antiprogrammed death-ligand 1 (PD-L1) monoclonal antibody (mAb) can be implemented with good results for tumors where no known TAAs have been identified. EXPERIMENTAL DESIGN Three cancer mouse models (melanoma, lung, and colon) were used to evaluate therapeutic efficacy and examine the immunologic mechanisms of the poly-IC/anti-PD-L1 mAb therapy. RESULTS The combined administration of poly-IC and anti-PD-L1 mAb into tumor-bearing mice generated potent immune responses resulting in the complete eradication or remarkable reduction of tumor growth. In some instances, the poly-IC/anti-PD-L1 mAb therapy induced long-lasting protection against tumor rechallenges. The results indicate that CD8 T cells but not CD4 T cells or NK cells mediated the therapeutic efficacy of this combinatorial therapy. Experiments using genetically deficient mice indicate that the therapeutic efficacy of this combinatorial therapy depended in part by the participation of type-I IFN, whereas IFN-γ did not seem to play a major role. CONCLUSIONS The overall results suggest that immunotherapy consisting of the combination of poly-IC/anti-PD-L1 mAb could be a promising new approach for treating patients with cancer, especially those instances where no reliable TAAs are available as a therapeutic vaccine.
Collapse
Affiliation(s)
- Toshihiro Nagato
- Authors' Affiliations: Immunology Program, Moffitt Cancer Center; Departments of Oncologic Sciences and Molecular Medicine, University of South Florida, Tampa, Florida; and Department of Otolaryngology-Head and Neck Surgery, Asahikawa Medical University, Asahikawa, Japan
| | | | | | | |
Collapse
|
1193
|
Merelli B, Massi D, Cattaneo L, Mandalà M. Targeting the PD1/PD-L1 axis in melanoma: Biological rationale, clinical challenges and opportunities. Crit Rev Oncol Hematol 2014; 89:140-65. [DOI: 10.1016/j.critrevonc.2013.08.002] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 08/10/2013] [Accepted: 08/15/2013] [Indexed: 12/16/2022] Open
|
1194
|
Reiss KA, Forde PM, Brahmer JR. Harnessing the power of the immune system via blockade of PD-1 and PD-L1: a promising new anticancer strategy. Immunotherapy 2014; 6:459-75. [PMID: 24815784 PMCID: PMC4732706 DOI: 10.2217/imt.14.9] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Cancer cells employ several mechanisms to evade the immune system of their host, thus escaping immune recognition and elimination. Of particular interest is a cancer cell's ability to co-opt the immune system's innate ligands and inhibitory receptors (also known as checkpoints), thus creating an immunosuppressive microenvironment that downregulates T-cell activation and cell signaling. The recent development of the checkpoint inhibitors anti-programmed death-1 and anti-programmed death ligand-1 has generated an enormous amount of interest as a potential new anticancer strategy in solid tumors, particularly in non-small-cell lung cancer, renal cell carcinoma and melanoma. Data suggest significant disease response rates using anti-programmed death-1 and anti-programmed death ligand-1 antibodies, even in heavily pretreated patients. Future directions include optimization of drug delivery sequence and combination of immunotherapy with other therapies including cytotoxic chemotherapy, radiation, antiangiogenic agents and small-molecule tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Kim A Reiss
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Research Building I, Room 186, 401 North Broadway Street, Baltimore, 21287, USA
| | - Patrick M Forde
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Research Building I, Room 186, 401 North Broadway Street, Baltimore, 21287, USA
| | - Julie R Brahmer
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Research Building I, Room G94, 401 North Broadway Street, Baltimore, 21287, USA
| |
Collapse
|
1195
|
Dudek RM, Chuang Y, Leonard JN. Engineered cell-based therapies: a vanguard of design-driven medicine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 844:369-91. [PMID: 25480651 DOI: 10.1007/978-1-4939-2095-2_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Engineered cell-based therapies are uniquely capable of performing sophisticated therapeutic functions in vivo, and this strategy is yielding promising clinical benefits for treating cancer. In this review, we discuss key opportunities and challenges for engineering customized cellular functions using cell-based therapy for cancer as a representative case study. We examine the historical development of chimeric antigen receptor (CAR) therapies as an illustration of the engineering design cycle. We also consider the potential roles that the complementary disciplines of systems biology and synthetic biology may play in realizing safe and effective treatments for a broad range of patients and diseases. In particular, we discuss how systems biology may facilitate both fundamental research and clinical translation, and we describe how the emerging field of synthetic biology is providing novel modalities for building customized cellular functions to overcome existing clinical barriers. Together, these approaches provide a powerful set of conceptual and experimental tools for transforming information into understanding, and for translating understanding into novel therapeutics to establish a new framework for design-driven medicine.
Collapse
Affiliation(s)
- Rachel M Dudek
- Northwestern University, 2145 Sheridan Road, Technological Institute, Rm. E136, Evanston, IL, 60208-3120, USA,
| | | | | |
Collapse
|
1196
|
Wei B, Wang L, Zhao X, Du C, Guo Y, Sun Z. The upregulation of programmed death 1 on peripheral blood T cells of glioma is correlated with disease progression. Tumour Biol 2013; 35:2923-9. [PMID: 24375192 DOI: 10.1007/s13277-013-1376-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Accepted: 10/29/2013] [Indexed: 12/31/2022] Open
Abstract
Glioma is the most common primary brain tumor. Programmed death 1 (PD-1) is a surface receptor expressed on activated and exhausted T cells, which mediate T cell inhibition upon binding with its ligand. In the current study, we investigated the expression of PD-1 on peripheral CD4+ and CD8+ T cells in glioma patients. Percentage of PD-1+ cells was measured by flow cytometry in 86 glioma cases and 62 healthy controls. Results showed that PD-1 expression was significantly increased in both peripheral CD4+ and CD8+ T cells in glioma (p < 0.001 and p < 0.001, respectively). When comparing PD-1 level in glioma patients with different histological types, patients with astrocytomas revealed clearly higher proportion of PD-1 on CD4+ T cells than those with oligodendrogliomas (p < 0.001), ependymomas (p < 0.001), or pilocytic astrocytomas (p < 0.001). Also, patients with the highest tumor grade (IV) demonstrated the most elevated expression of PD-1 on both CD4+ and CD8+ T cells. Interestingly, cases with tumor grade III and IV had downregulated PD-1 level on peripheral CD4+ T cells after surgery, whereas only grade IV patients showed decreased proportion of PD-1 on CD8+ T cells after treatment. In addition, no correlation between PD-1 expression and progression to secondary glioblastoma was observed. These data suggested PD-1 may act as a positive regulator in the pathogenesis and progression of glioma.
Collapse
Affiliation(s)
- Bo Wei
- The Second Division of Neurosurgery, The China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun, Jilin, 130033, China
| | | | | | | | | | | |
Collapse
|
1197
|
T cell avidity and tumor immunity: problems and solutions. CANCER MICROENVIRONMENT 2013; 7:1-9. [PMID: 24357332 DOI: 10.1007/s12307-013-0143-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/05/2013] [Indexed: 12/16/2022]
Abstract
A potent T cell response is an important component of durable anti-tumor immunity. The quality of the T cell response can, in-part, be measured by the avidity of the T cell for its tumor antigen-expressing target. While convention suggests that raising the avidity of the responding T cells may make for a more potent anti-tumor immune response, the threshold for effective tumor immunity remains unclear, as do some of the adverse effects of an inappropriately high avidity response. In this review, we discuss the relationship between T cell avidity and anti-tumor immunity, considering both experimental model systems as well as human clinical trials.
Collapse
|
1198
|
Legat A, Speiser DE, Pircher H, Zehn D, Fuertes Marraco SA. Inhibitory Receptor Expression Depends More Dominantly on Differentiation and Activation than "Exhaustion" of Human CD8 T Cells. Front Immunol 2013; 4:455. [PMID: 24391639 PMCID: PMC3867683 DOI: 10.3389/fimmu.2013.00455] [Citation(s) in RCA: 195] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 11/29/2013] [Indexed: 01/02/2023] Open
Abstract
Under conditions of chronic antigen stimulation, such as persistent viral infection and cancer, CD8 T cells may diminish effector function, which has been termed "exhaustion." Expression of inhibitory Receptors (iRs) is often regarded as a hallmark of "exhaustion." Here we studied the expression of eight different iRs by CD8 T cells of healthy humans, including CTLA-4, PD1, TIM3, LAG3, 2B4, BTLA, CD160, and KLRG1. We show that many iRs are expressed upon activation, and with progressive differentiation to effector cells, even in absence of long-term ("chronic") antigenic stimulation. In particular, we evaluated the direct relationship between iR expression and functionality in CD8 T cells by using anti-CD3 and anti-CD28 stimulation to stimulate all cells and differentiation subsets. We observed a striking up-regulation of certain iRs following the cytokine production wave, in agreement with the notion that iRs function as a negative feedback mechanism. Intriguingly, we found no major impairment of cytokine production in cells positive for a broad array of iRs, as previously shown for PD1 in healthy donors. Rather, the expression of the various iRs strongly correlated with T cell differentiation or activation states, or both. Furthermore, we analyzed CD8 T cells from lymph nodes (LNs) of melanoma patients. Interestingly, we found altered iR expression and lower cytokine production by T cells from metastatic LNs, but also from non-metastatic LNs, likely due to mechanisms which are not related to exhaustion. Together, our data shows that expression of iRs per se does not mark dysfunctional cells, but is rather tightly linked to activation and differentiation. This study highlights the importance of considering the status of activation and differentiation for the study and the clinical monitoring of CD8 T cells.
Collapse
Affiliation(s)
- Amandine Legat
- Clinical Tumor Biology and Immunotherapy Unit, Department of Oncology, Ludwig Center for Cancer Research, Lausanne University Hospital (CHUV) , Lausanne , Switzerland
| | - Daniel E Speiser
- Clinical Tumor Biology and Immunotherapy Unit, Department of Oncology, Ludwig Center for Cancer Research, Lausanne University Hospital (CHUV) , Lausanne , Switzerland
| | - Hanspeter Pircher
- Department of Immunology, Institute of Medical Microbiology and Hygiene, University of Freiburg , Freiburg , Germany
| | - Dietmar Zehn
- Swiss Vaccine Research Institute (SVRI) , Epalinges , Switzerland ; Division of Immunology and Allergy, Department of Medicine, Lausanne University Hospital (CHUV) , Lausanne , Switzerland
| | - Silvia A Fuertes Marraco
- Clinical Tumor Biology and Immunotherapy Unit, Department of Oncology, Ludwig Center for Cancer Research, Lausanne University Hospital (CHUV) , Lausanne , Switzerland
| |
Collapse
|
1199
|
Stärck L, Popp K, Pircher H, Uckert W. Immunotherapy with TCR-Redirected T Cells: Comparison of TCR-Transduced and TCR-Engineered Hematopoietic Stem Cell–Derived T Cells. THE JOURNAL OF IMMUNOLOGY 2013; 192:206-13. [DOI: 10.4049/jimmunol.1202591] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
1200
|
Lidsky ME, Speicher PJ, Jiang B, Tsutsui M, Tyler DS. Isolated limb infusion as a model to test new agents to treat metastatic melanoma. J Surg Oncol 2013; 109:357-65. [PMID: 24522940 DOI: 10.1002/jso.23502] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 10/24/2013] [Indexed: 02/06/2023]
Abstract
The limb model of in-transit disease can expand our understanding of treating melanoma because of the ease of obtaining tissue biopsies for correlative studies and the availability of preclinical animal models that allow validation of novel therapeutic strategies. This review will focus on regional therapy for in-transit melanoma as a platform to investigate novel therapeutic approaches to improve regional disease control, and help us develop insights to more rationally design systemic therapy trials.
Collapse
Affiliation(s)
- Michael E Lidsky
- Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | | | | | | | | |
Collapse
|