51
|
Dendritic cells in hematological malignancies. Crit Rev Oncol Hematol 2016; 108:86-96. [DOI: 10.1016/j.critrevonc.2016.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 09/05/2016] [Accepted: 10/18/2016] [Indexed: 01/17/2023] Open
|
52
|
Jelinek T, Hajek R. PD-1/PD-L1 inhibitors in multiple myeloma: The present and the future. Oncoimmunology 2016; 5:e1254856. [PMID: 28123899 DOI: 10.1080/2162402x.2016.1254856] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/22/2016] [Accepted: 10/24/2016] [Indexed: 01/05/2023] Open
Abstract
The introduction of PD-1/PD-L1 pathway inhibitors has marked a significant milestone in the treatment of various types of solid tumors. The current situation in multiple myeloma (MM) is rather unclear, as distinct research groups have reported discordant results. This discrepancy dominantly concerns the expression of PD-1/PD-L1 molecules as well as the identification of the responsible immune effector cell population. The results of monotherapy with PD-1/PD-L1 inhibitors have been unsatisfactory in MM, suggesting that a combination approach is needed. The most logical partners are immunomodulatory agents as they possess many synergistic effects. We are also proposing other rational and promising combinations (e.g., daratumumab, ibrutinib, anti-CD137) that warrant further investigation.
Collapse
Affiliation(s)
- T Jelinek
- Faculty of Science, University of Ostrava, Czech Republic; Department of Haematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Czech Republic; Centro de Investigacion Medica Aplicada (CIMA), Clinica Universidad de Navarra, IDISNA, Pamplona, Spain
| | - R Hajek
- Department of Haematooncology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava , Czech Republic
| |
Collapse
|
53
|
Abstract
Our initial understanding of immune-regulatory cells was based on the discovery of suppressor cells that assure peripheral T-cell tolerance and promote immune homeostasis. Research has particularly focused on the importance of regulatory T cells (Tregs) for immune modulation, e.g. directing host responses to tumours or inhibiting autoimmunity development. However, recent studies report the discovery of self-reactive pro-inflammatory T cells-termed anti-regulatory T cells (anti-Tregs)-that target immune-suppressive cells. Thus, regulatory cells can now be defined as both cells that suppress immune reactions as well as effector cells that counteract the effects of suppressor cells and support immune reactions. Self-reactive anti-Tregs have been described that specifically recognize human leukocyte antigen-restricted epitopes derived from proteins that are normally expressed by regulatory immune cells, including indoleamine 2,3-dioxygenase (IDO), tryptophan 2,6-dioxygenase (TDO), programmed death-ligand 1 (PD-L1), and forkhead box P3 (Foxp3). These proteins are highly expressed in professional antigen-presenting cells under various physiological conditions, such as inflammation and stress. Therefore, self-reactive T cells that recognize such targets may be activated due to the strong activation signal given by their cognate targets. The current review describes the existing knowledge regarding these self-reactive anti-Tregs, providing examples of antigen-specific anti-Tregs and discussing their possible roles in immune homeostasis and their potential future clinical applications.
Collapse
Affiliation(s)
- Mads Hald Andersen
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, 2730, Herlev, Denmark. .,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
54
|
Al-Hujaily EM, Oldham RAA, Hari P, Medin JA. Development of Novel Immunotherapies for Multiple Myeloma. Int J Mol Sci 2016; 17:E1506. [PMID: 27618026 PMCID: PMC5037783 DOI: 10.3390/ijms17091506] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Revised: 08/24/2016] [Accepted: 09/01/2016] [Indexed: 12/12/2022] Open
Abstract
Multiple myeloma (MM) is a disorder of terminally differentiated plasma cells characterized by clonal expansion in the bone marrow (BM). It is the second-most common hematologic malignancy. Despite significant advances in therapeutic strategies, MM remains a predominantly incurable disease emphasizing the need for the development of new treatment regimens. Immunotherapy is a promising treatment modality to circumvent challenges in the management of MM. Many novel immunotherapy strategies, such as adoptive cell therapy and monoclonal antibodies, are currently under investigation in clinical trials, with some already demonstrating a positive impact on patient survival. In this review, we will summarize the current standards of care and discuss major new approaches in immunotherapy for MM.
Collapse
Affiliation(s)
- Ensaf M Al-Hujaily
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Robyn A A Oldham
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
| | - Parameswaran Hari
- Department of Medicine, Division of Hematology/Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Jeffrey A Medin
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada.
- The Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada.
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
- Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
55
|
Touzeau C, Moreau P. Pomalidomide in the management of relapsed multiple myeloma. Future Oncol 2016; 12:1975-83. [DOI: 10.2217/fon-2016-0184] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Pomalidomide, a very potent member of the immunomodulatory drug family, is considered a standard of care for patients with relapsed and refractory myeloma, who have previously been treated with bortezomib and lenalidomide. Pomalidomide induces both direct myeloma cell death, and indirect antimyeloma response through its impact on the microenvironment (modulation of immune response, inhibition of angiogenesis, inhibition of bone resorption). Pomalidomide in combination with dexamethasone is an approved regimen in Europe and USA based on the results of a Phase III randomized trial. In order to improve response rate and patient survival, pomalidomide is currently being assessed in triplet combinations with other antimyeloma agents. The present review addresses current knowledge regarding the clinical use of pomalidomide in relapsed myeloma patients.
Collapse
Affiliation(s)
- Cyrille Touzeau
- Department of Hematology, University hospital, Nantes, France INSERM UMR892, CNRS UMR6299, University of Nantes, France
| | - Philippe Moreau
- Department of Hematology, University hospital, Nantes, France INSERM UMR892, CNRS UMR6299, University of Nantes, France
| |
Collapse
|
56
|
Ahmad SM, Borch TH, Hansen M, Andersen MH. PD-L1-specific T cells. Cancer Immunol Immunother 2016; 65:797-804. [PMID: 26724936 PMCID: PMC11028888 DOI: 10.1007/s00262-015-1783-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 12/13/2015] [Indexed: 12/21/2022]
Abstract
Recently, there has been an increased focus on the immune checkpoint protein PD-1 and its ligand PD-L1 due to the discovery that blocking the PD-1/PD-L1 pathway with monoclonal antibodies elicits striking clinical results in many different malignancies. We have described naturally occurring PD-L1-specific T cells that recognize both PD-L1-expressing immune cells and malignant cells. Thus, PD-L1-specific T cells have the ability to modulate adaptive immune reactions by reacting to regulatory cells. Thus, utilization of PD-L1-derived T cell epitopes may represent an attractive vaccination strategy for targeting the tumor microenvironment and for boosting the clinical effects of additional anticancer immunotherapy. This review summarizes present information about PD-L1 as a T cell antigen, depicts the initial findings about the function of PD-L1-specific T cells in the adjustment of immune responses, and discusses future opportunities.
Collapse
Affiliation(s)
- Shamaila Munir Ahmad
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Troels Holz Borch
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Morten Hansen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730, Herlev, Denmark
| | - Mads Hald Andersen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Herlev Ringvej 75, 2730, Herlev, Denmark.
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
57
|
Hartmann S, Bhola NE, Grandis JR. HGF/Met Signaling in Head and Neck Cancer: Impact on the Tumor Microenvironment. Clin Cancer Res 2016; 22:4005-13. [PMID: 27370607 DOI: 10.1158/1078-0432.ccr-16-0951] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 06/08/2016] [Indexed: 12/21/2022]
Abstract
Studies to date have revealed several major molecular alterations that contribute to head and neck squamous cell carcinoma (HNSCC) initiation, progression, metastatic spread, and therapeutic failure. The EGFR is the only FDA-approved therapeutic target, yet responses to cetuximab have been limited. Activation and cross-talk of cellular receptors and consequent activation of different signaling pathways contribute to limited activity of blockade of a single pathway. The hepatocyte growth factor (HGF) receptor, Met, has been implicated in HNSCC tumorigenesis and EGFR inhibitor resistance. HGF, the sole ligand of Met, is overexpressed in the tumor microenvironment. The role of HGF/Met signaling in proliferation, metastasis, and angiogenesis has been investigated in HNSCC, leading to clinical trials with various Met inhibitors and HGF antibodies. However, the role of the HGF/Met signaling axis in mediating the tumor microenvironment has been relatively understudied in HNSCC. In this review, we discuss the functional roles of Met and HGF in HNSCC with a focus on the tumor microenvironment and the immune system. Clin Cancer Res; 22(16); 4005-13. ©2016 AACR.
Collapse
Affiliation(s)
- Stefan Hartmann
- Department of Otolaryngology, University of California San Francisco, San Francisco, California. Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Neil E Bhola
- Department of Otolaryngology, University of California San Francisco, San Francisco, California
| | - Jennifer R Grandis
- Department of Otolaryngology, University of California San Francisco, San Francisco, California.
| |
Collapse
|
58
|
Perales MA, Sauter CS, Armand P. Reprint of: Fast Cars and No Brakes: Autologous Stem Cell Transplantation as a Platform for Novel Immunotherapies. Biol Blood Marrow Transplant 2016; 22:S9-S14. [PMID: 26899275 DOI: 10.1016/j.bbmt.2016.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 10/14/2015] [Indexed: 11/24/2022]
Abstract
Autologous stem cell transplantation (ASCT) is indicated in a number of hematologic malignancies, including multiple myeloma, non-Hodgkin lymphoma, and Hodgkin lymphoma. Relapse, however, remains 1 of the main causes of post-ASCT failure, and several strategies are being investigated to decrease the risk of relapse of progression. Recent advances in the treatment of hematological malignancies have included adoptive transfer of genetically modified T cells that express chimeric antigen receptors or T cell receptors, as well the use of checkpoint inhibitors. Early clinical results in non-transplantation patients have been very promising. This review will focus on the use of gene-modified T cells and checkpoint inhibitors in stem cell transplantation.
Collapse
Affiliation(s)
- Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| | - Craig S Sauter
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Philippe Armand
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| |
Collapse
|
59
|
Atanackovic D, Radhakrishnan SV, Bhardwaj N, Luetkens T. Chimeric Antigen Receptor (CAR) therapy for multiple myeloma. Br J Haematol 2016; 172:685-98. [DOI: 10.1111/bjh.13889] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Djordje Atanackovic
- Multiple Myeloma Program, Hematology and Hematologic Malignancies; University of Utah/Huntsman Cancer Institute; Salt Lake City UT USA
| | - Sabarinath V. Radhakrishnan
- Multiple Myeloma Program, Hematology and Hematologic Malignancies; University of Utah/Huntsman Cancer Institute; Salt Lake City UT USA
| | - Neelam Bhardwaj
- Multiple Myeloma Program, Hematology and Hematologic Malignancies; University of Utah/Huntsman Cancer Institute; Salt Lake City UT USA
| | - Tim Luetkens
- Multiple Myeloma Program, Hematology and Hematologic Malignancies; University of Utah/Huntsman Cancer Institute; Salt Lake City UT USA
| |
Collapse
|
60
|
Ayed AO, Chang LJ, Moreb JS. Immunotherapy for multiple myeloma: Current status and future directions. Crit Rev Oncol Hematol 2015; 96:399-412. [DOI: 10.1016/j.critrevonc.2015.06.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Revised: 04/26/2015] [Accepted: 06/15/2015] [Indexed: 01/01/2023] Open
|
61
|
Willenbacher W, Willenbacher E, Zelle-Rieser C, Biedermann R, Weger R, Jöhrer K, Brunner A. Bone marrow microenvironmental CD4 + and CD8 + lymphocyte infiltration patterns define overall- and progression free survival in standard risk multiple myeloma--an analysis from the Austrian Myeloma Registry. Leuk Lymphoma 2015; 57:1478-81. [PMID: 26413883 DOI: 10.3109/10428194.2015.1099646] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Wolfgang Willenbacher
- a Internal Medicine V - Hematology and Oncology , Medical University of Innsbruck , Innsbruck , Austria ;,b Area 4 Health Technology Assessment and Bioinformatics, ONCOTYROL - Center for Personalized Cancer Medicine , Innsbruck , Austria
| | - Ella Willenbacher
- a Internal Medicine V - Hematology and Oncology , Medical University of Innsbruck , Innsbruck , Austria
| | | | - Rainer Biedermann
- d Department of Orthopedic Surgery , Medical University of Innsbruck , Innsbruck , Austria
| | - Roman Weger
- b Area 4 Health Technology Assessment and Bioinformatics, ONCOTYROL - Center for Personalized Cancer Medicine , Innsbruck , Austria
| | - Karin Jöhrer
- c Tyrolean Cancer Research Institute , Innsbruck , Austria
| | - Andrea Brunner
- e Department of Pathology, Division of General Pathology , Medical University of Innsbruck , Innsbruck , Austria
| |
Collapse
|
62
|
Mechanisms of Drug Resistance in Relapse and Refractory Multiple Myeloma. BIOMED RESEARCH INTERNATIONAL 2015; 2015:341430. [PMID: 26649299 PMCID: PMC4663284 DOI: 10.1155/2015/341430] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 09/24/2015] [Accepted: 10/21/2015] [Indexed: 12/11/2022]
Abstract
Multiple myeloma (MM) is a hematological malignancy that remains incurable because most patients eventually relapse or become refractory to current treatments. Although the treatments have improved, the major problem in MM is resistance to therapy. Clonal evolution of MM cells and bone marrow microenvironment changes contribute to drug resistance. Some mechanisms affect both MM cells and microenvironment, including the up- and downregulation of microRNAs and programmed death factor 1 (PD-1)/PD-L1 interaction. Here, we review the pathogenesis of MM cells and bone marrow microenvironment and highlight possible drug resistance mechanisms. We also review a potential molecular targeting treatment and immunotherapy for patients with refractory or relapse MM.
Collapse
|
63
|
Oldreive CE, Skowronska A, Davies NJ, Parry H, Agathanggelou A, Krysov S, Packham G, Rudzki Z, Cronin L, Vrzalikova K, Murray P, Odintsova E, Pratt G, Taylor AMR, Moss P, Stankovic T. T-cell number and subtype influence the disease course of primary chronic lymphocytic leukaemia xenografts in alymphoid mice. Dis Model Mech 2015; 8:1401-12. [PMID: 26398941 PMCID: PMC4631786 DOI: 10.1242/dmm.021147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 08/10/2015] [Indexed: 01/28/2023] Open
Abstract
Chronic lymphocytic leukaemia (CLL) cells require microenvironmental support for their proliferation. This can be recapitulated in highly immunocompromised hosts in the presence of T cells and other supporting cells. Current primary CLL xenograft models suffer from limited duration of tumour cell engraftment coupled with gradual T-cell outgrowth. Thus, a greater understanding of the interaction between CLL and T cells could improve their utility. In this study, using two distinct mouse xenograft models, we investigated whether xenografts recapitulate CLL biology, including natural environmental interactions with B-cell receptors and T cells, and whether manipulation of autologous T cells can expand the duration of CLL engraftment. We observed that primary CLL xenografts recapitulated both the tumour phenotype and T-cell repertoire observed in patients and that engraftment was significantly shorter for progressive tumours. A reduction in the number of patient T cells that were injected into the mice to 2-5% of the initial number or specific depletion of CD8(+) cells extended the limited xenograft duration of progressive cases to that characteristic of indolent disease. We conclude that manipulation of T cells can enhance current CLL xenograft models and thus expand their utility for investigation of tumour biology and pre-clinical drug assessment.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cell Proliferation
- Cell Survival
- Cells, Cultured
- Coculture Techniques
- Cytotoxicity, Immunologic
- Graft Survival
- Heterografts
- Humans
- Immunocompromised Host
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Lymphocyte Activation
- Lymphocyte Depletion
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Mice, Inbred NOD
- Mice, SCID
- Neoplasm Transplantation
- Phenotype
- Spleen/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/pathology
- Time Factors
- Tumor Microenvironment
Collapse
Affiliation(s)
- Ceri E Oldreive
- School of Cancer Sciences, Department of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Anna Skowronska
- School of Cancer Sciences, Department of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Nicholas J Davies
- School of Cancer Sciences, Department of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Helen Parry
- School of Cancer Sciences, Department of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Angelo Agathanggelou
- School of Cancer Sciences, Department of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Sergey Krysov
- CRUK Centre, Cancer Sciences Unit, University of Southampton, Southampton, SO16 6YD, UK
| | - Graham Packham
- CRUK Centre, Cancer Sciences Unit, University of Southampton, Southampton, SO16 6YD, UK
| | - Zbigniew Rudzki
- Department of Pathology, Heart of England Hospital, Birmingham, B9 5SS, UK
| | - Laura Cronin
- School of Biosciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Katerina Vrzalikova
- School of Cancer Sciences, Department of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Paul Murray
- School of Cancer Sciences, Department of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Elena Odintsova
- School of Cancer Sciences, Department of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Guy Pratt
- School of Cancer Sciences, Department of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - A Malcolm R Taylor
- School of Cancer Sciences, Department of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Paul Moss
- School of Cancer Sciences, Department of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Tatjana Stankovic
- School of Cancer Sciences, Department of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
64
|
Fast Cars and No Brakes: Autologous Stem Cell Transplantation as a Platform for Novel Immunotherapies. Biol Blood Marrow Transplant 2015; 22:17-22. [PMID: 26485445 DOI: 10.1016/j.bbmt.2015.10.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Accepted: 10/14/2015] [Indexed: 01/21/2023]
Abstract
Autologous stem cell transplantation (ASCT) is indicated in a number of hematologic malignancies, including multiple myeloma, non-Hodgkin lymphoma, and Hodgkin lymphoma. Relapse, however, remains 1 of the main causes of post-ASCT failure, and several strategies are being investigated to decrease the risk of relapse of progression. Recent advances in the treatment of hematological malignancies have included adoptive transfer of genetically modified T cells that express chimeric antigen receptors or T cell receptors, as well the use of checkpoint inhibitors. Early clinical results in nontransplantation patients have been very promising. This review will focus on the use of gene-modified T cells and checkpoint inhibitors in stem cell transplantation.
Collapse
|
65
|
Abstract
In this issue of Blood, Sehgal et al report on the clinical and pharmacodynamics analysis of pomalidomide dosing strategies in multiple myeloma (MM) and their impact on immune activation and cereblon targets. The particular novelty of this study lies in the direct correlation of immune effects triggered by pomalidomide with clinical responses in MM patients. Results of this study will stimulate many additional studies.
Collapse
|
66
|
Monoclonal antibodies in the treatment of multiple myeloma: current status and future perspectives. Leukemia 2015; 30:526-35. [PMID: 26265184 PMCID: PMC4777772 DOI: 10.1038/leu.2015.223] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 07/21/2015] [Accepted: 07/22/2015] [Indexed: 02/06/2023]
Abstract
The treatment landscape for patients with multiple myeloma (MM) is constantly evolving. Over the past decade, the introduction of novel agents such as proteasome inhibitors and immunomodulatory drugs has led to notable changes in therapeutic strategy, and improvements in survival, yet MM remains incurable in the vast majority of cases. More recently, a targeted approach to MM treatment has emerged, using monoclonal antibodies (mAbs) to target antigens expressed on the surface of MM cells. MAbs tested to date kill MM cells via the host's immune system and/or by promoting apoptosis, and appear to have generally improved tolerability compared with currently available treatments. Due to their distinct mode of action, mAbs are promising both for patients who have exhausted current regimens, and as part of first-line treatments in newly diagnosed patients. This review examines the recent developments in mAb-based therapy for MM, primarily focused on those agents in ongoing clinical testing.
Collapse
|
67
|
Mimura N, Hideshima T, Anderson KC. Novel therapeutic strategies for multiple myeloma. Exp Hematol 2015; 43:732-41. [PMID: 26118499 DOI: 10.1016/j.exphem.2015.04.010] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 04/15/2015] [Indexed: 12/23/2022]
Abstract
Multiple myeloma (MM) is a plasma-cell malignancy which remains incurable despite the recent emergence of multiple novel agents. Importantly, recent genetic and molecular analyses have revealed the complexity and heterogeneity of this disease, highlighting the need for therapeutic strategies to eliminate all clones. Moreover, the bone marrow microenvironment, including stromal cells and immune cells, plays a central role in MM pathogenesis, promoting tumor cell growth, survival, and drug resistance. New classes of agents including proteasome inhibitors, immunomodulatory drugs, monoclonal antibodies, and histone deacetylase inhibitors have shown remarkable efficacy; however, novel therapeutic approaches are still urgently needed to further improve patient outcomes. In this review, we discuss the recent advances and future strategies to ultimately develop MM therapies with curative potential.
Collapse
Affiliation(s)
- Naoya Mimura
- Department of Transfusion Medicine and Cell Therapy, Chiba University Hospital, Chiba, Japan.
| | - Teru Hideshima
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
68
|
Vaccination of multiple myeloma: Current strategies and future prospects. Crit Rev Oncol Hematol 2015; 96:339-54. [PMID: 26123319 DOI: 10.1016/j.critrevonc.2015.06.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 05/06/2015] [Accepted: 06/09/2015] [Indexed: 01/21/2023] Open
Abstract
Tumor immunotherapy holds great promise in controlling multiple myeloma (MM) and may provide an alternative treatment modality to conventional chemotherapy for MM patients. For this reason, a major area of investigation is the development of cancer vaccines to generate myeloma-specific immunity. Several antigens that are able to induce specific T-cell responses are involved in different critical mechanisms for cell differentiation, inhibition of apoptosis, demethylation and proliferation. Strategies under development include infusion of vaccine-primed and ex vivo expanded/costimulated autologous T cells after high-dose melphalan, genetic engineering of autologous T cells with receptors for myeloma-specific epitopes, administration of dendritic cell/plasma cell fusions and administration expanded marrow-infiltrating lymphocytes. In addition, novel immunomodulatory drugs may synergize with immunotherapies. The task ahead is to evaluate these approaches in appropriate clinical settings, and to couple them with strategies to overcome mechanisms of immunoparesis as a means to induce more robust clinically significant immune responses.
Collapse
|
69
|
Benson DM, Cohen AD, Jagannath S, Munshi NC, Spitzer G, Hofmeister CC, Efebera YA, Andre P, Zerbib R, Caligiuri MA. A Phase I Trial of the Anti-KIR Antibody IPH2101 and Lenalidomide in Patients with Relapsed/Refractory Multiple Myeloma. Clin Cancer Res 2015; 21:4055-61. [PMID: 25999435 DOI: 10.1158/1078-0432.ccr-15-0304] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 04/25/2015] [Indexed: 02/05/2023]
Abstract
PURPOSE Natural killer (NK) cells may play an important role in the immune response to multiple myeloma; however, multiple myeloma cells express killer immunoglobulin-like receptor (KIR) ligands to prevent NK cell cytotoxicity. Lenalidomide can expand and activate NK cells in parallel with its direct effects against multiple myeloma; however, dexamethasone may impair these favorable immunomodulatory properties. IPH2101, a first-in-class antiinhibitory KIR antibody, has acceptable safety and tolerability in multiple myeloma as a single agent. The present work sought to characterize lenalidomide and IPH2101 as a novel, steroid-sparing, dual immune therapy for multiple myeloma. EXPERIMENTAL DESIGN A phase I trial enrolled 15 patients in three cohorts. Lenalidomide was administered per os at 10 mg on cohort 1 and 25 mg on cohorts 2 and 3 days 1 to 21 on a 28-day cycle with IPH2101 given intravenously on day 1 of each cycle at 0.2 mg/kg in cohort 1, 1 mg/kg in cohort 2, and 2 mg/kg in cohort 3. No corticosteroids were utilized. The primary endpoint was safety, and secondary endpoints included clinical activity, pharmacokinetics (PK), and pharmacodynamics (PD). RESULTS The biologic endpoint of full KIR occupancy was achieved across the IPH2101 dosing interval. PD and PK of IPH2101 with lenalidomide were similar to data from a prior single-agent IPH2101 trial. Five serious adverse events (SAE) were reported. Five objective responses occurred. No autoimmunity was seen. CONCLUSIONS These findings suggest that lenalidomide in combination with antiinhibitory KIR therapy warrants further investigation in multiple myeloma as a steroid-sparing, dual immune therapy. This trial was registered at www.clinicaltrials.gov (reference: NCT01217203).
Collapse
Affiliation(s)
- Don M Benson
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio.
| | - Adam D Cohen
- University of Pennsylvania Abramson Cancer Center, Philadelphia, Pennsylvania
| | | | - Nikhil C Munshi
- Dana Farber Cancer Institute, Boston, Massachusetts. Boston VA Healthcare System, Harvard Medical School, Boston, Massachusetts
| | - Gary Spitzer
- St. Francis Hospital, Greenville, South Carolina
| | | | - Yvonne A Efebera
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | | | | |
Collapse
|
70
|
Görgün G, Samur MK, Cowens KB, Paula S, Bianchi G, Anderson JE, White RE, Singh A, Ohguchi H, Suzuki R, Kikuchi S, Harada T, Hideshima T, Tai YT, Laubach JP, Raje N, Magrangeas F, Minvielle S, Avet-Loiseau H, Munshi NC, Dorfman DM, Richardson PG, Anderson KC. Lenalidomide Enhances Immune Checkpoint Blockade-Induced Immune Response in Multiple Myeloma. Clin Cancer Res 2015; 21:4607-18. [PMID: 25979485 DOI: 10.1158/1078-0432.ccr-15-0200] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 05/01/2015] [Indexed: 12/31/2022]
Abstract
PURPOSE PD-1/PD-L1 signaling promotes tumor growth while inhibiting effector cell-mediated antitumor immune responses. Here, we assessed the impact of single and dual blockade of PD-1/PD-L1, alone or in combination with lenalidomide, on accessory and immune cell function as well as multiple myeloma cell growth in the bone marrow (BM) milieu. EXPERIMENTAL DESIGN Surface expression of PD-1 on immune effector cells, and PD-L1 expression on CD138(+) multiple myeloma cells and myeloid-derived suppressor cells (MDSC) were determined in BM from newly diagnosed (ND) multiple myeloma and relapsed/refractory (RR) multiple myeloma versus healthy donor (HD). We defined the impact of single and dual blockade of PD-1/PD-L1, alone and with lenalidomide, on autologous anti-multiple myeloma immune response and tumor cell growth. RESULTS Both ND and RR patient multiple myeloma cells have increased PD-L1 mRNA and surface expression compared with HD. There is also a significant increase in PD-1 expression on effector cells in multiple myeloma. Importantly, PD-1/PD-L1 blockade abrogates BM stromal cell (BMSC)-induced multiple myeloma growth, and combined blockade of PD-1/PD-L1 with lenalidomide further inhibits BMSC-induced tumor growth. These effects are associated with induction of intracellular expression of IFNγ and granzyme B in effector cells. Importantly, PD-L1 expression in multiple myeloma is higher on MDSC than on antigen-presenting cells, and PD-1/PD-L1 blockade inhibits MDSC-mediated multiple myeloma growth. Finally, lenalidomide with PD-1/PD-L1 blockade inhibits MDSC-mediated immune suppression. CONCLUSIONS Our data therefore demonstrate that checkpoint signaling plays an important role in providing the tumor-promoting, immune-suppressive microenvironment in multiple myeloma, and that PD-1/PD-L1 blockade induces anti-multiple myeloma immune response that can be enhanced by lenalidomide, providing the framework for clinical evaluation of combination therapy.
Collapse
Affiliation(s)
- Güllü Görgün
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| | - Mehmet K Samur
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Department of Biostatistics and Computational Biology, Harvard School of Public Health, Boston, Massachusetts
| | - Kristen B Cowens
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Steven Paula
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Giada Bianchi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Julie E Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Randie E White
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ahaana Singh
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Hiroto Ohguchi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Rikio Suzuki
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Shohei Kikuchi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Takeshi Harada
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Teru Hideshima
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Yu-Tzu Tai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Jacob P Laubach
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Noopur Raje
- Massachusetts General Hospital, Boston, Massachusetts
| | - Florence Magrangeas
- Inserm UMR892, CNRS 6299, Université de Nantes, Nantes, France. Centre Hospitalier Universitaire de Nantes, Unité Mixte de Genomique du Cancer, Nantes, France
| | - Stephane Minvielle
- Inserm UMR892, CNRS 6299, Université de Nantes, Nantes, France. Centre Hospitalier Universitaire de Nantes, Unité Mixte de Genomique du Cancer, Nantes, France
| | | | - Nikhil C Munshi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts. Boston VA Health Care System, Boston, Massachusetts
| | - David M Dorfman
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Paul G Richardson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kenneth C Anderson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
71
|
Guillerey C, Ferrari de Andrade L, Vuckovic S, Miles K, Ngiow SF, Yong MCR, Teng MWL, Colonna M, Ritchie DS, Chesi M, Bergsagel PL, Hill GR, Smyth MJ, Martinet L. Immunosurveillance and therapy of multiple myeloma are CD226 dependent. J Clin Invest 2015; 125:2077-89. [PMID: 25893601 DOI: 10.1172/jci77181] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Accepted: 03/12/2015] [Indexed: 12/20/2022] Open
Abstract
Multiple myeloma (MM) is an age-dependent hematological malignancy. Evaluation of immune interactions that drive MM relies on in vitro experiments that do not reflect the complex cellular stroma involved in MM pathogenesis. Here we used Vk*MYC transgenic mice, which spontaneously develop MM, and demonstrated that the immune system plays a critical role in the control of MM progression and the response to treatment. We monitored Vk*MYC mice that had been crossed with Cd226 mutant mice over a period of 3 years and found that CD226 limits spontaneous MM development. The CD226-dependent anti-myeloma immune response against transplanted Vk*MYC MM cells was mediated both by NK and CD8+ T cells through perforin and IFN-γ pathways. Moreover, CD226 expression was required for optimal antimyeloma efficacy of cyclophosphamide (CTX) and bortezomib (Btz), which are both standardly used to manage MM in patients. Activation of costimulatory receptor CD137 with mAb (4-1BB) exerted strong antimyeloma activity, while inhibition of coinhibitory receptors PD-1 and CTLA-4 had no effect. Taken together, the results of this study provide in vivo evidence that CD226 is important for MM immunosurveillance and indicate that specific immune components should be targeted for optimal MM treatment efficacy. As progressive immunosuppression associates with MM development, strategies aimed to increase immune functions may have important therapeutic implications in MM.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/therapeutic use
- Antigens, Differentiation, T-Lymphocyte/genetics
- Antigens, Differentiation, T-Lymphocyte/immunology
- Antigens, Differentiation, T-Lymphocyte/physiology
- Antineoplastic Agents/therapeutic use
- Boronic Acids/therapeutic use
- Bortezomib
- CD8-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen/antagonists & inhibitors
- Crosses, Genetic
- Cyclophosphamide/therapeutic use
- Disease Progression
- Genes, myc
- Genetic Predisposition to Disease
- Immunologic Surveillance/immunology
- Immunotherapy
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/physiology
- Killer Cells, Natural/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Multiple Myeloma/drug therapy
- Multiple Myeloma/genetics
- Multiple Myeloma/immunology
- Neoplasm Proteins/deficiency
- Neoplasm Proteins/genetics
- Neoplasm Proteins/immunology
- Neoplasm Proteins/physiology
- Neoplasm Transplantation
- Pore Forming Cytotoxic Proteins/deficiency
- Pore Forming Cytotoxic Proteins/genetics
- Pore Forming Cytotoxic Proteins/physiology
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Pyrazines/therapeutic use
- Receptors, Virus/deficiency
- Receptors, Virus/genetics
- Receptors, Virus/physiology
- Tumor Burden
- Tumor Necrosis Factor Receptor Superfamily, Member 9/antagonists & inhibitors
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
Collapse
|
72
|
Bhaskar S, Benson DM. Current and future immunotherapeutic approaches to multiple myeloma therapy. Int J Hematol Oncol 2015. [DOI: 10.2217/ijh.14.43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Multiple myeloma (MM) is a hematologic malignancy associated with heterogeneous treatment and survival outcomes due in part to the ability of MM to evade and suppress the immune system. Research has focused on finding ways to modulate and enhance immunity while targeting the bone marrow microenvironment. Contemporary therapies include immunomodulatory drugs, proteasome inhibitors and autologous and allogeneic stem cell transplant and have improved outcomes for patients with MM. Future therapies, including monoclonal antibodies, chimeric antigen receptor cells and MM vaccines, show promise to further improved outcomes, particularly when used in combination with existing therapies. This review covers the mechanism of action of currently available and future therapies and explores ways in which treatment may be more specifically directed in the future.
Collapse
Affiliation(s)
- Shakthi Bhaskar
- Department of Internal Medicine, The Ohio State University College of Medicine, 370 West 9th Avenue, Columbus, OH 43210, USA
| | - Don M Benson
- The Division of Hematology, 898 Biomedical Research Tower, The Ohio State University Comprehensive Cancer Center, 460 W 12th Ave, Columbus, OH 43210, USA
| |
Collapse
|
73
|
Targeting PD1-PDL1 immune checkpoint in plasmacytoid dendritic cell interactions with T cells, natural killer cells and multiple myeloma cells. Leukemia 2015; 29:1441-4. [PMID: 25634684 DOI: 10.1038/leu.2015.11] [Citation(s) in RCA: 170] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
74
|
Carmon L, Avivi I, Kovjazin R, Zuckerman T, Dray L, Gatt ME, Or R, Shapira MY. Phase I/II study exploring ImMucin, a pan-major histocompatibility complex, anti-MUC1 signal peptide vaccine, in multiple myeloma patients. Br J Haematol 2014; 169:44-56. [DOI: 10.1111/bjh.13245] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/24/2014] [Indexed: 01/01/2023]
Affiliation(s)
- Lior Carmon
- Vaxil BioTherapeutics Ltd.; Nes-Ziona Israel
| | - Irit Avivi
- Department of Haematology; Rambam Medical Campus; Haifa Israel
- Technion; Israel Institute of Technology; Haifa Israel
| | | | - Tsila Zuckerman
- Department of Haematology; Rambam Medical Campus; Haifa Israel
- Technion; Israel Institute of Technology; Haifa Israel
| | - Lillian Dray
- Department of Bone Marrow Transplantation & Cancer Immunotherapy; Hadassah Medical Centre; Jerusalem Israel
| | - Moshe E. Gatt
- Department of Bone Marrow Transplantation & Cancer Immunotherapy; Hadassah Medical Centre; Jerusalem Israel
| | - Reuven Or
- Department of Bone Marrow Transplantation & Cancer Immunotherapy; Hadassah Medical Centre; Jerusalem Israel
| | - Michael Y. Shapira
- Department of Bone Marrow Transplantation & Cancer Immunotherapy; Hadassah Medical Centre; Jerusalem Israel
| |
Collapse
|
75
|
Cellular immunotherapy in multiple myeloma: lessons from preclinical models. Biochim Biophys Acta Rev Cancer 2014; 1846:392-404. [PMID: 25109893 DOI: 10.1016/j.bbcan.2014.08.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 07/31/2014] [Accepted: 08/02/2014] [Indexed: 12/20/2022]
Abstract
The majority of multiple myeloma patients relapse with the current treatment strategies, raising the need for alternative therapeutic approaches. Cellular immunotherapy is a rapidly evolving field and currently being translated into clinical trials with encouraging results in several cancer types, including multiple myeloma. Murine multiple myeloma models are of critical importance for the development and refinement of cellular immunotherapy. In this review, we summarize the immune cell changes that occur in multiple myeloma patients and we discuss the cell-based immunotherapies that have been tested in multiple myeloma, with a focus on murine models.
Collapse
|
76
|
Ahmad SM, Svane IM, Andersen MH. The stimulation of PD-L1-specific cytotoxic T lymphocytes can both directly and indirectly enhance antileukemic immunity. Blood Cancer J 2014; 4:e230. [PMID: 25036801 PMCID: PMC4219446 DOI: 10.1038/bcj.2014.50] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- S M Ahmad
- Center for Cancer Immune Therapy (CCIT), Department of Hematology and Oncology, Copenhagen University Hospital, Herlev, Herlev, Denmark
| | - I M Svane
- Center for Cancer Immune Therapy (CCIT), Department of Hematology and Oncology, Copenhagen University Hospital, Herlev, Herlev, Denmark
| | - M H Andersen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology and Oncology, Copenhagen University Hospital, Herlev, Herlev, Denmark
| |
Collapse
|
77
|
Han L, Liu F, Li R, Li Z, Chen X, Zhou Z, Zhang X, Hu T, Zhang Y, Young K, Sun S, Wen J, Zhang M. Role of programmed death ligands in effective T-cell interactions in extranodal natural killer/T-cell lymphoma. Oncol Lett 2014; 8:1461-1469. [PMID: 25202350 PMCID: PMC4156194 DOI: 10.3892/ol.2014.2356] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Accepted: 04/03/2014] [Indexed: 01/15/2023] Open
Abstract
Extranodal natural killer/T-cell lymphoma (ENKL) is marked by a profound cellular immune deficiency that may influence the capacity of T cells to extract an efficient antitumor immune response. It has been confirmed that the B7-CD28 pathway may promote tumor immune evasion by providing a negative regulatory signal. The current study analyzed the expression of programmed death 1 (PD-1)/programmed death ligand (PD-L) in ENKL cell lines and tissues. The functional studies were performed to analyze the functional activity of PD-L1 interacting with effective T cells in ENKL. PD-L1 and PD-L2 mRNA levels in ENKL cell lines were markedly upregulated compared with those in normal natural killer cells. The proteins constitutively expressed in the 30 ENKL specimens were significantly higher than in the 20 rhinitis specimens. In addition, PD-L1 and PD-L2 expression were found to closely correlate with certain clinical histopathological parameters. Furthermore, the count of PD-1+ tumor-infiltrating T lymphocytes was found to negatively correlate with the expression of PD-L1 and PD-L2. The PD-1 expression in the CD4+ and CD8+ T-cell subsets of 20 ENKL patients prior to therapy were significantly higher than that of the 10 healthy volunteers. In the functional studies, the cytokines (interleukin-2 and interferon-γ) secreted by CD8+ T cells were inhibited by PD-L1 expression in SNK-6 cells and this was restored with the presence of the PD-L1 blocking antibody. However no direct effect of PD-L1 was identified on CD8+ T-cell apoptosis and CD8+ T-cell cytotoxicity, as assessed by the proliferation of SNK-6 cells in the presence or absence of the neutralizing anti-PD-L1 antibody. The results of the current study revealed that PD-Ls and PD-1 are aberrantly expressed in ENKL and, furthermore, PD-L1 expression in SNK-6 cells was found to inhibit the activity of CD8+ T-cell cytokine secretion. This indicated that the PD-Ls may prevent effective antitumor immunity in vivo by interacting with tumor T cells, which provides important evidence to delineate the cellular immune deficiency mechanism in ENKL. Therefore, PD-1/PD-Ls are predicted to become novel targets for ENKL immunotherapy.
Collapse
Affiliation(s)
- Lijuan Han
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Feifei Liu
- Department of Sports Medicine, Research Center of Sports Medicine, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Ruping Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Zhaoming Li
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Xinfeng Chen
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Zhiyuan Zhou
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Xudong Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Tengpeng Hu
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Ken Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77230-1439, USA
| | - Suke Sun
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Jianguo Wen
- Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| | - Mingzhi Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450000, P.R. China
| |
Collapse
|
78
|
Roehnisch T, Then C, Nagel W, Blumenthal C, Braciak T, Donzeau M, Böhm T, Flaig M, Bourquin C, Oduncu FS. Phage idiotype vaccination: first phase I/II clinical trial in patients with multiple myeloma. J Transl Med 2014; 12:119. [PMID: 24885819 PMCID: PMC4113220 DOI: 10.1186/1479-5876-12-119] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 04/29/2014] [Indexed: 11/24/2022] Open
Abstract
Background Multiple myeloma is characterized by clonal expansion of B cells producing monoclonal immunoglobulins or fragments thereof, which can be detected in the serum and/or urine and are ideal target antigens for patient-specific immunotherapies. Methods Using phage particles as immunological carriers, we employed a novel chemically linked idiotype vaccine in a clinical phase I/II trial including 15 patients with advanced multiple myeloma. Vaccines composed of purified paraproteins linked to phage were manufactured successfully for each patient. Patients received six intradermal immunizations with phage idiotype vaccines in three different dose groups. Results Phage idiotype was well tolerated by all study participants. A subset of patients (80% in the middle dose group) displayed a clinical response indicated by decrease or stabilization of paraprotein levels. Patients exhibiting a clinical response to phage vaccines also raised idiotype-specific immunoglobulins. Induction of a cellular immune response was demonstrated by a cytotoxicity assay and delayed type hypersensitivity tests. Conclusion We present a simple, time- and cost-efficient phage idiotype vaccination strategy, which represents a safe and feasible patient-specific therapy for patients with advanced multiple myeloma and produced promising anti-tumor activity in a subset of patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Fuat S Oduncu
- Division of Hematology and Oncology, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany.
| |
Collapse
|
79
|
Rossille D, Gressier M, Damotte D, Maucort-Boulch D, Pangault C, Semana G, Le Gouill S, Haioun C, Tarte K, Lamy T, Milpied N, Fest T. High level of soluble programmed cell death ligand 1 in blood impacts overall survival in aggressive diffuse large B-Cell lymphoma: results from a French multicenter clinical trial. Leukemia 2014; 28:2367-75. [DOI: 10.1038/leu.2014.137] [Citation(s) in RCA: 245] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 03/24/2014] [Accepted: 04/03/2014] [Indexed: 01/21/2023]
|
80
|
Andersen MH. The targeting of immunosuppressive mechanisms in hematological malignancies. Leukemia 2014; 28:1784-92. [PMID: 24691076 DOI: 10.1038/leu.2014.108] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 02/20/2014] [Accepted: 03/13/2014] [Indexed: 12/20/2022]
Abstract
The adaptive immune system has the capacity to recognize and kill leukemic cells. However, immune tolerance mechanisms that normally protect healthy tissues from autoimmune effects prevent the development of effective antitumor immunity. Tumors use several different immunosuppressive mechanisms to evade otherwise effective T-cell responses. A growing number of immune evasion mechanisms have been characterized mainly in solid tumors. In hematological malignancies, less is known about how different immune escape mechanisms influence tumor immune evasion and the extent of their impact on ongoing immune responses. The present review highlights the potential role of three well-defined immunosuppressive mechanisms in hematological malignancies: (i) inhibitory T-cell pathways (especially programmed death ligand 1/programmed death 1 (PD-L1/PD-1)), (ii) regulatory immune cells, and (iii) metabolic enzymes such as indoeamine-2,3-dioxygenase (IDO). The possible therapeutic targeting of these pathways is also discussed. Exciting new strategies that might affect future antileukemia immunotherapy include monoclonal antibodies that block inhibitory T-cell pathways (PD-1/PD-L1) and the prevention of tryptophan depletion by IDO inhibitors. Furthermore, the clinical effect of several chemotherapeutic drugs may arise from the targeting of immunosuppressive cells. Evidence for a new feedback mechanism to suppress the function of regulatory immune cells was recently provided by the identification and characterization of spontaneous cytotoxic T lymphocyte (CTL) responses against regulatory immune cells. Such specific CTLs may be immensely useful in anticancer immunotherapy (for example, by anticancer vaccination). The targeting of one or more immunosuppressive pathways may be especially interesting in combination with antileukemic immunotherapy in cases in which immunosuppressive mechanisms antagonize the desired effects of the therapy.
Collapse
Affiliation(s)
- M H Andersen
- Department of Hematology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|