| Peer-Reviewed

Potential Treatment for Multiple Myeloma: Immune Checkpoint Inhibitors

Received: 3 September 2021    Accepted: 17 September 2021    Published: 26 September 2021
Views:       Downloads:
Abstract

Multiple myeloma (MM) is a malignant disease in which monoclonal plasma cells proliferate abnormally. The poor prognosis of MM is always closely related to the immunosuppression of T cells. Restore the immune function of suppressed T cells may reverse the immunodeficiency in the MM microenvironment and improve prognosis. In recent years, immunosuppressive receptors such as programmed cell death receptor-1 (PD-1), T cells immunoglobulin and ITIM domain (TIGIT), Lymphocyte-Activation Gene-3 (LAG-3), T cell immunoglobulin and mucin domain–containing molecule 3 (Tim-3), Leukocyte immunoglobulin-like receptor B1 (LILRB1) and Cytotoxic T- lymphocyte antigen 4 (CTLA-4) have been discovered playing a key role in the tumor immunodeficiency microenvironment. For patients with solid tumors and some leukemia, immunotherapy targeting such receptors can significantly improve the T cells immunodeficiency. However, similar positive results were not found in MM patients, which is related to the complex immunosuppressive mechanism. At present, the understanding of immunosuppressive receptors in MM is insufficient. In this review, this paper summarized part of the studies on PD-1, TCLA-4, Tim-3, TIGIT and other popular immunosuppressive receptors in MM, in order to attract more attention, and in-depth research on the immunotherapy also to promote the immunotherapy of MM from basic research to clinical transformation as soon as possible.

Published in International Journal of Immunology (Volume 9, Issue 3)
DOI 10.11648/j.iji.20210903.14
Page(s) 59-67
Creative Commons

This is an Open Access article, distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution and reproduction in any medium or format, provided the original work is properly cited.

Copyright

Copyright © The Author(s), 2024. Published by Science Publishing Group

Keywords

Multiple Myeloma, Immune Therapy, Immunosuppressive Receptors, T Cell Exhaustion, Immune Dysfunction

References
[1] Mortaz E, Tabarsi P, Mansouri D, et al. Cancers Related to Immunodeficiencies: Update and Perspectives. Front Immunol. 2016; 7: 365.
[2] Tan J, Chen S, Huang J, et al. Increased exhausted CD8+T cells with programmed death-1, T-cell immunoglobulin and mucin-domain-containing-3 phenotype in patients with multiple myeloma. Asia-Pacific Journal of Clinical Oncology. 2018; 14 (5): e266-e74.
[3] Cooke RE, Quinn KM, Quach H, et al. Conventional Treatment for Multiple Myeloma Drives Premature Aging Phenotypes and Metabolic Dysfunction in T Cells. Front Immunol. 2020; 11: 2153.
[4] Elhelbawy NG, Nassar AAH, Eltorgoman AEA, et al. Immunological microenvironment gene expression in patients with diffuse large B cell non Hodgkin lymphoma. Biochem Biophys Rep. 2020; 21: 100731.
[5] Meng F, Li L, Lu F, et al. Overexpression of TIGIT in NK and T Cells Contributes to Tumor Immune Escape in Myelodysplastic Syndromes. Front Oncol. 2020; 10: 1595.
[6] Panda A, Rosenfeld JA, Singer EA, et al. Genomic and immunologic correlates of LAG-3 expression in cancer. Oncoimmunology. 2020; 9 (1): 1756116.
[7] Jones RB, Ndhlovu LC, Barbour JD, et al. Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. J Exp Med. 2008; 205 (12): 2763-79.
[8] Kang X, Kim J, Deng M, et al. Inhibitory leukocyte immunoglobulin-like receptors: Immune checkpoint proteins and tumor sustaining factors. Cell Cycle. 2016; 15 (1): 25-40.
[9] Takahashi T TT, Yamazaki S, et al.. Immunologic self-tolerance maintained by CD25 (+)CD4 (+) regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4.. journal of Experiment Medicine. 2000.
[10] Liu X, Ranganathan R, Jiang S, et al. A Chimeric Switch-Receptor Targeting PD1 Augments the Efficacy of Second-Generation CAR T Cells in Advanced Solid Tumors. Cancer Res. 2016; 76 (6): 1578-90.
[11] Yan Z, Yao ZH, Yao SN, et al. Combining PD-1 Inhibitor Nivolumab with Radiotherapy Successfully Treated a Patient with Refractory Primary Mediastinal Large B-Cell Lymphoma: A Case Report and Literature Review. Cancer Manag Res. 2020; 12: 6311-6.
[12] Pan B, Shang L, Liu C, et al. PD-1 antibody and ruxolitinib enhances graft-versus-lymphoma effect without increasing acute graft-versus-host disease in mice. Am J Transplant. 2020.
[13] Hui E, Cheung J, Zhu J, et al. T cell costimulatory receptor CD28 is a primary target for PD-1-mediated inhibition. Science. 2017; 355 (6332): 1428-33.
[14] Francisco LM, Sage PT, Sharpe AH. The PD-1 pathway in tolerance and autoimmunity. Immunol Rev. 2010; 236: 219-42.
[15] Tan J, Chen S, Lu Y, et al. Higher PD-1 expression concurrent with exhausted CD8+ T cells in patients with de novo acute myeloid leukemia. Chin J Cancer Res. 2017; 29 (5): 463-70.
[16] Passiglia F, Bironzo P, Righi L, et al. A Prospective Phase II Single-arm Study of Niraparib Plus Dostarlimab in Patients With Advanced Non-small-cell Lung Cancer and/or Malignant Pleural Mesothelioma, Positive for PD-L1 Expression and Germline or Somatic Mutations in the DNA Repair Genes: Rationale and Study Design. Clin Lung Cancer. 2020.
[17] Willenbacher W, Willenbacher E, Zelle-Rieser C, et al. 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. 2016; 57 (6): 1478-81.
[18] Li Y, Chen S, Yang L, et al. Change in expression pattern of TCR-CD3 complex in patients with multiple myeloma. Hematology. 2011; 16 (3): 143-50.
[19] Gordon SR, Maute RL, Dulken BW, et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature. 2017; 545 (7655): 495-9.
[20] Ray A, Das DS, Song Y, et al. Targeting PD1–PDL1 immune checkpoint in plasmacytoid dendritic cell interactions with T cells, natural killer cells and multiple myeloma cells. Leukemia. 2015; 29 (6): 1441-4.
[21] Benson DM, Jr., Bakan CE, Mishra A, et al. The PD-1/PD-L1 axis modulates the natural killer cell versus multiple myeloma effect: a therapeutic target for CT-011, a novel monoclonal anti-PD-1 antibody. Blood. 2010; 116 (13): 2286-94.
[22] Hallett WH, Jing W, Drobyski WR, et al. Immunosuppressive effects of multiple myeloma are overcome by PD-L1 blockade. Biol Blood Marrow Transplant. 2011; 17 (8): 1133-45.
[23] Lesokhin AM, Ansell SM, Armand P, et al. Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. J Clin Oncol. 2016; 34 (23): 2698-704.
[24] Badros A, Hyjek E, Ma N, et al. Pembrolizumab, pomalidomide, and low-dose dexamethasone for relapsed/refractory multiple myeloma. Blood. 2017; 130 (10): 1189-97.
[25] Rizvi NA, Hellmann MD, Snyder A, et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015; 348 (6230): 124-8.
[26] Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014; 515 (7528): 568-71.
[27] Herbst RS, Soria JC, Kowanetz M, et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature. 2014; 515 (7528): 563-7.
[28] Braga WMT, da Silva BR, de Carvalho AC, et al. FOXP3 and CTLA4 overexpression in multiple myeloma bone marrow as a sign of accumulation of CD4+ T regulatory cells. Cancer Immunology, Immunotherapy. 2014; 63 (11): 1189-97.
[29] Zhu Y, Wu S. [Immune Characteristics of Small Cell Lung Cancer]. Zhongguo Fei Ai Za Zhi. 2020; 23 (10): 889-96.
[30] Gao X, Yang J, Chen Y. Identification of a four immune-related genes signature based on an immunogenomic landscape analysis of clear cell renal cell carcinoma. J Cell Physiol. 2020; 235 (12): 9834-50.
[31] Aref S, El Agdar M, El Sebaie A, et al. Prognostic Value of CD200 Expression and Soluble CTLA-4 Concentrations in Intermediate and High-Risk Myelodysplastic Syndrome Patients. Asian Pac J Cancer Prev. 2020; 21 (8): 2225-30.
[32] Tcvetkov N, Gusak A, Morozova E, et al. Immune checkpoints bone marrow expression as the predictor of clinical outcome in myelodysplastic syndrome. Leuk Res Rep. 2020; 14: 100215.
[33] Hodi FS ODS, McDermott DF, et al. Improved survival with ipilimumab in patients with metastatic melanoma. he New England journal of medicine. 2010.
[34] Chau V, Bilusic M. Pembrolizumab in Combination with Axitinib as First-Line Treatment for Patients with Renal Cell Carcinoma (RCC): Evidence to Date. Cancer Manag Res. 2020; 12: 7321-30.
[35] Pinheiro FD, Teixeira AF, de Brito BB, et al. Immunotherapy - new perspective in lung cancer. World J Clin Oncol. 2020; 11 (5): 250-9.
[36] Allison MFKaJp. CD28 and CTLA-4 Have Opposing Effects on the Response of T ceils to Stimulation. Journal of Experiment Medicine. 1995.
[37] Brian T. JAB. Control of peripheral T-cell tolerance and autoimmunity via the CTLA-4 and PD-1 pathways. Bluestone - Immunological reviews. 2008.
[38] Zhang H, Dutta P, Liu J, et al. Tumour cell-intrinsic CTLA4 regulates PD-L1 expression in non-small cell lung cancer. J Cell Mol Med. 2019; 23 (1): 535-42.
[39] Hellmann MD, Rizvi NA, Goldman JW, et al. Nivolumab plus ipilimumab as first-line treatment for advanced non-small-cell lung cancer (CheckMate 012): results of an open-label, phase 1, multicohort study. The Lancet Oncology. 2017; 18 (1): 31-41.
[40] Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med. 2015; 372 (21): 2006-17.
[41] Tang J, Chen Q, Zhang F, et al. [Peripheral blood exosomes from patients with multiple myeloma mediate bortezomib resistance in cultured multiple myeloma cells]. Nan Fang Yi Ke Da Xue Xue Bao. 2019; 39 (4): 485-9.
[42] Chalmers AW, Patel S, Boucher K, et al. Phase I Trial of Targeted EGFR or ALK Therapy with Ipilimumab in Metastatic NSCLC with Long-Term Follow-Up. Target Oncol. 2019; 14 (4): 417-21.
[43] Dougall WC, Kurtulus S, Smyth MJ, et al. TIGIT and CD96: new checkpoint receptor targets for cancer immunotherapy. Immunological Reviews. 2017; 276 (1): 112-20.
[44] Guillerey C, Ferrari de Andrade L, Vuckovic S, et al. Immunosurveillance and therapy of multiple myeloma are CD226 dependent. J Clin Invest. 2015; 125 (5): 2077-89.
[45] Hoogi S, Eisenberg V, Mayer S, et al. A TIGIT-based chimeric co-stimulatory switch receptor improves T-cell anti-tumor function. J Immunother Cancer. 2019; 7 (1): 243.
[46] Kitsou M, Ayiomamitis GD, Zaravinos A. High expression of immune checkpoints is associated with the TIL load, mutation rate and patient survival in colorectal cancer. Int J Oncol. 2020; 57 (1): 237-48.
[47] Xu D, Zhao E, Zhu C, et al. TIGIT and PD-1 may serve as potential prognostic biomarkers for gastric cancer. Immunobiology. 2020; 225 (3).
[48] Simone A. Minnie RDK, Kate H. Gartlan, Ping Zhang, Andrew N. Wilkinson. Myeloma escape after stem cell transplantation is aconsequence of T-cell exhaustion and is prevented by TIGIT blockade. blood. 2018.
[49] Guillerey C, Harjunpaa H, Carrie N, et al. TIGIT immune checkpoint blockade restores CD8 (+) T-cell immunity against multiple myeloma. Blood. 2018; 132 (16): 1689-94.
[50] Johnston RJ, Comps-Agrar L, Hackney J, et al. The immunoreceptor TIGIT regulates antitumor and antiviral CD8 (+) T cell effector function. Cancer Cell. 2014; 26 (6): 923-37.
[51] Zhang Q, Bi J, Zheng X, et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat Immunol. 2018; 19 (7): 723-32.
[52] He W, Zhang H, Han F, et al. CD155T/TIGIT Signaling Regulates CD8 (+) T-cell Metabolism and Promotes Tumor Progression in Human Gastric Cancer. Cancer Res. 2017; 77 (22): 6375-88.
[53] Wang J, Sanmamed MF, Datar I, et al. Fibrinogen-like Protein 1 Is a Major Immune Inhibitory Ligand of LAG-3. Cell. 2019; 176 (1-2): 334-47 e12.
[54] Turnis ME, Andrews LP, Vignali DA. Inhibitory receptors as targets for cancer immunotherapy. Eur J Immunol. 2015; 45 (7): 1892-905.
[55] Zarour HM. Reversing T-cell Dysfunction and Exhaustion in Cancer. Clin Cancer Res. 2016; 22 (8): 1856-64.
[56] Chen PL, Roh W, Reuben A, et al. Analysis of Immune Signatures in Longitudinal Tumor Samples Yields Insight into Biomarkers of Response and Mechanisms of Resistance to Immune Checkpoint Blockade. Cancer Discov. 2016; 6 (8): 827-37.
[57] Andrews LP, Marciscano AE, Drake CG, et al. LAG3 (CD223) as a cancer immunotherapy target. Immunol Rev. 2017; 276 (1): 80-96.
[58] Kim YJ, Won CH, Lee MW, et al. Correlation Between Tumor-Associated Macrophage and Immune Checkpoint Molecule Expression and Its Prognostic Significance in Cutaneous Melanoma. J Clin Med. 2020; 9 (8).
[59] Saleh R, Taha RZ, Toor SM, et al. Expression of immune checkpoints and T cell exhaustion markers in early and advanced stages of colorectal cancer. Cancer Immunol Immunother. 2020; 69 (10): 1989-99.
[60] Radwan SM, Elleboudy NS, Nabih NA, et al. The immune checkpoints Cytotoxic T lymphocyte antigen-4 and Lymphocyte activation gene-3 expression is up-regulated in acute myeloid leukemia. HLA. 2020; 96 (1): 3-12.
[61] Xie N, Sun YY, Jiang HJ, et al. [Lymphocyte activation gene 3 expression on T lymphocyte cell subsets in patients with myelodysplastic syndrome]. Zhonghua Yi Xue Za Zhi. 2020; 100 (24): 1886-9.
[62] Mussetti A, Pellegrinelli A, Cieri N, et al. PD-L1, LAG3, and HLA-DR are increasingly expressed during smoldering myeloma progression. Ann Hematol. 2019; 98 (7): 1713-20.
[63] Lucas F, Pennell M, Huang Y, et al. T Cell Transcriptional Profiling and Immunophenotyping Uncover LAG3 as a Potential Significant Target of Immune Modulation in Multiple Myeloma. Biol Blood Marrow Transplant. 2020; 26 (1): 7-15.
[64] Grosso JF, Kelleher CC, Harris TJ, et al. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. J Clin Invest. 2007; 117 (11): 3383-92.
[65] Deng WW, Mao L, Yu GT, et al. LAG-3 confers poor prognosis and its blockade reshapes antitumor response in head and neck squamous cell carcinoma. Oncoimmunology. 2016; 5 (11): e1239005.
[66] Yu X, Huang X, Chen X, et al. Characterization of a novel anti-human lymphocyte activation gene 3 (LAG-3) antibody for cancer immunotherapy. MAbs. 2019; 11 (6): 1139-48.
[67] Jing W, Gershan JA, Weber J, et al. Combined immune checkpoint protein blockade and low dose whole body irradiation as immunotherapy for myeloma. J Immunother Cancer. 2015; 3 (1): 2.
[68] Legat A, Maby-El Hajjami H, Baumgaertner P, et al. Vaccination with LAG-3Ig (IMP321) and Peptides Induces Specific CD4 and CD8 T-Cell Responses in Metastatic Melanoma Patients--Report of a Phase I/IIa Clinical Trial. Clin Cancer Res. 2016; 22 (6): 1330-40.
[69] Freeman GJ, Casasnovas JM, Umetsu DT, et al. TIM genes: a family of cell surface phosphatidylserine receptors that regulate innate and adaptive immunity. Immunol Rev. 2010; 235 (1): 172-89.
[70] Zhu C, Anderson AC, Schubart A, et al. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. 2005; 6 (12): 1245-52.
[71] Anderson AC AD, Bregoli L, Hastings WD, Kassam N, Lei C, Chandwaskar R, Karman J, Su EW, Hirashima M, Bruce JN, Kane LP, Kuchroo VK, Hafler DA. Promotion of Tissue Inflammation by the Immune Receptor Tim-3 Expressed on Innate Immune Cells. Science 2007 Nov 16. 2007.
[72] Nakayama M, Akiba H, Takeda K, et al. Tim-3 mediates phagocytosis of apoptotic cells and cross-presentation. Blood. 2009; 113 (16): 3821-30.
[73] Holderried TAW, de Vos L, Bawden EG, et al. Molecular and immune correlates of TIM-3 (HAVCR2) and galectin 9 (LGALS9) mRNA expression and DNA methylation in melanoma. Clin Epigenetics. 2019; 11 (1): 161.
[74] Jia K, He Y, Dziadziuszko R, et al. T cell immunoglobulin and mucin-domain containing-3 in non-small cell lung cancer. Transl Lung Cancer Res. 2019; 8 (6): 895-906.
[75] Tan J, Yu Z, Huang J, et al. Increased PD-1+Tim-3+ exhausted T cells in bone marrow may influence the clinical outcome of patients with AML. Biomarker Research. 2020; 8 (1).
[76] Kikushige Y, Shima T, Takayanagi S, et al. TIM-3 is a promising target to selectively kill acute myeloid leukemia stem cells. Cell Stem Cell. 2010; 7 (6): 708-17.
[77] Ndhlovu LC, Lopez-Verges S, Barbour JD, et al. Tim-3 marks human natural killer cell maturation and suppresses cell-mediated cytotoxicity. Blood. 2012; 119 (16): 3734-43.
[78] Gleason MK, Lenvik TR, McCullar V, et al. Tim-3 is an inducible human natural killer cell receptor that enhances interferon gamma production in response to galectin-9. Blood. 2012; 119 (13): 3064-72.
[79] Hui Liu J-QZ, Zeng-Feng Ni, Yu-Hong Liu, Jiao Jiao Correlation of the Expression Alteration of Tim-3 with the T Cell and B Cell Dysfunction in Peripheral Blood of Multiple Myeloma Patients.
[80] Zhongguo Shi Yan Xue Ye Xue Za Zhi 2020 Jun; 28 (3): 881-885 Chinese. 2020.
[81] Liu JF, Wu L, Yang LL, et al. Blockade of TIM3 relieves immunosuppression through reducing regulatory T cells in head and neck cancer. J Exp Clin Cancer Res. 2018; 37 (1): 44.
[82] Ngiow SF, von Scheidt B, Akiba H, et al. Anti-TIM3 antibody promotes T cell IFN-gamma-mediated antitumor immunity and suppresses established tumors. Cancer Res. 2011; 71 (10): 3540-51.
[83] Fourcade J, Sun Z, Benallaoua M, et al. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J Exp Med. 2010; 207 (10): 2175-86.
[84] Pasero C, Gravis G, Guerin M, et al. Inherent and Tumor-Driven Immune Tolerance in the Prostate Microenvironment Impairs Natural Killer Cell Antitumor Activity. Cancer Res. 2016; 76 (8): 2153-65.
[85] Roberti MP, Julia EP, Rocca YS, et al. Overexpression of CD85j in TNBC patients inhibits Cetuximab-mediated NK-cell ADCC but can be restored with CD85j functional blockade. Eur J Immunol. 2015; 45 (5): 1560-9.
[86] Chen H, Chen Y, Deng M, et al. Antagonistic anti-LILRB1 monoclonal antibody regulates antitumor functions of natural killer cells. J Immunother Cancer. 2020; 8 (2).
[87] Lozano E, Díaz T, Mena M-P, et al. Loss of the Immune Checkpoint CD85j/LILRB1 on Malignant Plasma Cells Contributes to Immune Escape in Multiple Myeloma. The Journal of Immunology. 2018; 200 (8): 2581-91.
[88] Kim A, Han CJ, Driver I, et al. LILRB1 Blockade Enhances Bispecific T Cell Engager Antibody-Induced Tumor Cell Killing by Effector CD8 (+) T Cells. J Immunol. 2019; 203 (4): 1076-87.
[89] Goldstein RL, Goyos A, Li CM, et al. AMG 701 induces cytotoxicity of multiple myeloma cells and depletes plasma cells in cynomolgus monkeys. Blood Adv. 2020; 4 (17): 4180-94.
[90] Guo X, Wang H, Zhou J, et al. [Clinical Diagnosis and Treatment Recommendations for Cardiac Adverse Reactions Related to Immune Checkpoint Inhibitor]. Zhongguo Fei Ai Za Zhi. 2019; 22 (10): 627-32.
[91] Wang H, Guo X, Zhou J, et al. [Clinical Diagnosis and Treatment Recommendations for the Pneumonitis Associated with Immune Checkpoint Inhibitor]. Zhongguo Fei Ai Za Zhi. 2019; 22 (10): 621-6.
Cite This Article
  • APA Style

    Jiaxin Wang, Jiaxiong Tan, Yuhong Lu. (2021). Potential Treatment for Multiple Myeloma: Immune Checkpoint Inhibitors. International Journal of Immunology, 9(3), 59-67. https://doi.org/10.11648/j.iji.20210903.14

    Copy | Download

    ACS Style

    Jiaxin Wang; Jiaxiong Tan; Yuhong Lu. Potential Treatment for Multiple Myeloma: Immune Checkpoint Inhibitors. Int. J. Immunol. 2021, 9(3), 59-67. doi: 10.11648/j.iji.20210903.14

    Copy | Download

    AMA Style

    Jiaxin Wang, Jiaxiong Tan, Yuhong Lu. Potential Treatment for Multiple Myeloma: Immune Checkpoint Inhibitors. Int J Immunol. 2021;9(3):59-67. doi: 10.11648/j.iji.20210903.14

    Copy | Download

  • @article{10.11648/j.iji.20210903.14,
      author = {Jiaxin Wang and Jiaxiong Tan and Yuhong Lu},
      title = {Potential Treatment for Multiple Myeloma: Immune Checkpoint Inhibitors},
      journal = {International Journal of Immunology},
      volume = {9},
      number = {3},
      pages = {59-67},
      doi = {10.11648/j.iji.20210903.14},
      url = {https://doi.org/10.11648/j.iji.20210903.14},
      eprint = {https://article.sciencepublishinggroup.com/pdf/10.11648.j.iji.20210903.14},
      abstract = {Multiple myeloma (MM) is a malignant disease in which monoclonal plasma cells proliferate abnormally. The poor prognosis of MM is always closely related to the immunosuppression of T cells. Restore the immune function of suppressed T cells may reverse the immunodeficiency in the MM microenvironment and improve prognosis. In recent years, immunosuppressive receptors such as programmed cell death receptor-1 (PD-1), T cells immunoglobulin and ITIM domain (TIGIT), Lymphocyte-Activation Gene-3 (LAG-3), T cell immunoglobulin and mucin domain–containing molecule 3 (Tim-3), Leukocyte immunoglobulin-like receptor B1 (LILRB1) and Cytotoxic T- lymphocyte antigen 4 (CTLA-4) have been discovered playing a key role in the tumor immunodeficiency microenvironment. For patients with solid tumors and some leukemia, immunotherapy targeting such receptors can significantly improve the T cells immunodeficiency. However, similar positive results were not found in MM patients, which is related to the complex immunosuppressive mechanism. At present, the understanding of immunosuppressive receptors in MM is insufficient. In this review, this paper summarized part of the studies on PD-1, TCLA-4, Tim-3, TIGIT and other popular immunosuppressive receptors in MM, in order to attract more attention, and in-depth research on the immunotherapy also to promote the immunotherapy of MM from basic research to clinical transformation as soon as possible.},
     year = {2021}
    }
    

    Copy | Download

  • TY  - JOUR
    T1  - Potential Treatment for Multiple Myeloma: Immune Checkpoint Inhibitors
    AU  - Jiaxin Wang
    AU  - Jiaxiong Tan
    AU  - Yuhong Lu
    Y1  - 2021/09/26
    PY  - 2021
    N1  - https://doi.org/10.11648/j.iji.20210903.14
    DO  - 10.11648/j.iji.20210903.14
    T2  - International Journal of Immunology
    JF  - International Journal of Immunology
    JO  - International Journal of Immunology
    SP  - 59
    EP  - 67
    PB  - Science Publishing Group
    SN  - 2329-1753
    UR  - https://doi.org/10.11648/j.iji.20210903.14
    AB  - Multiple myeloma (MM) is a malignant disease in which monoclonal plasma cells proliferate abnormally. The poor prognosis of MM is always closely related to the immunosuppression of T cells. Restore the immune function of suppressed T cells may reverse the immunodeficiency in the MM microenvironment and improve prognosis. In recent years, immunosuppressive receptors such as programmed cell death receptor-1 (PD-1), T cells immunoglobulin and ITIM domain (TIGIT), Lymphocyte-Activation Gene-3 (LAG-3), T cell immunoglobulin and mucin domain–containing molecule 3 (Tim-3), Leukocyte immunoglobulin-like receptor B1 (LILRB1) and Cytotoxic T- lymphocyte antigen 4 (CTLA-4) have been discovered playing a key role in the tumor immunodeficiency microenvironment. For patients with solid tumors and some leukemia, immunotherapy targeting such receptors can significantly improve the T cells immunodeficiency. However, similar positive results were not found in MM patients, which is related to the complex immunosuppressive mechanism. At present, the understanding of immunosuppressive receptors in MM is insufficient. In this review, this paper summarized part of the studies on PD-1, TCLA-4, Tim-3, TIGIT and other popular immunosuppressive receptors in MM, in order to attract more attention, and in-depth research on the immunotherapy also to promote the immunotherapy of MM from basic research to clinical transformation as soon as possible.
    VL  - 9
    IS  - 3
    ER  - 

    Copy | Download

Author Information
  • Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China

  • Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China

  • Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, China

  • Sections