1
|
Maxwell M, Yan D, Rivest B, Boone A, Cardia J, Noessner E. INTASYL self-delivering RNAi decreases TIGIT expression, enhancing NK cell cytotoxicity: a potential application to increase the efficacy of NK adoptive cell therapy against cancer. Cancer Immunol Immunother 2024; 73:239. [PMID: 39358647 PMCID: PMC11447204 DOI: 10.1007/s00262-024-03835-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/06/2024] [Indexed: 10/04/2024]
Abstract
Natural killer (NK) cells are frontline defenders against cancer and are capable of recognizing and eliminating tumor cells without prior sensitization or antigen presentation. Due to their unique HLA mismatch tolerance, they are ideal for adoptive cell therapy (ACT) because of their ability to minimize graft-versus-host-disease risk. The therapeutic efficacy of NK cells is limited in part by inhibitory immune checkpoint receptors, which are upregulated upon interaction with cancer cells and the tumor microenvironment. Overexpression of inhibitory receptors reduces NK cell-mediated cytotoxicity by impairing the ability of NK cells to secrete effector cytokines and cytotoxic granules. T-cell immunoreceptor with immunoglobulin and ITIM domains (TIGIT), a well-known checkpoint receptor involved in T-cell exhaustion, has recently been implicated in the exhaustion of NK cells. Overcoming TIGIT-mediated inhibition of NK cells may allow for a more potent antitumor response following ACT. Here, we describe a novel approach to TIGIT inhibition using self-delivering RNAi compounds (INTASYL™) that incorporates the features of RNAi and antisense technology. INTASYL compounds demonstrate potent activity and stability, are rapidly and efficiently taken up by cells, and can be easily incorporated into cell product manufacturing. INTASYL PH-804, which targets TIGIT, suppresses TIGIT mRNA and protein expression in NK cells, resulting in increased cytotoxic capacity and enhanced tumor cell killing in vitro. Delivering PH-804 to NK cells before ACT has emerged as a promising strategy to counter TIGIT inhibition, thereby improving the antitumor response. This approach offers the potential for more potent off-the-shelf products for adoptive cell therapy, particularly for hematological malignancies.
Collapse
Affiliation(s)
- Melissa Maxwell
- Phio Pharmaceuticals, 11 Apex Dr., Ste 300A PMB 2006, Marlborough, MA, 01752, USA.
| | - Dingxue Yan
- Phio Pharmaceuticals, 11 Apex Dr., Ste 300A PMB 2006, Marlborough, MA, 01752, USA
| | - Brianna Rivest
- Phio Pharmaceuticals, 11 Apex Dr., Ste 300A PMB 2006, Marlborough, MA, 01752, USA
| | - Andrew Boone
- Phio Pharmaceuticals, 11 Apex Dr., Ste 300A PMB 2006, Marlborough, MA, 01752, USA
| | - James Cardia
- Phio Pharmaceuticals, 11 Apex Dr., Ste 300A PMB 2006, Marlborough, MA, 01752, USA
| | - Elfriede Noessner
- Immunoanalytics-Tissue Control of Immunocytes, Helmholtz Zentrum Munich, Feodor-Lynen-Str. 21, 81377, Munich, Germany
| |
Collapse
|
2
|
Li C, Yu X, Han X, Lian C, Wang Z, Shao S, Shao F, Wang H, Ma S, Liu J. Innate immune cells in tumor microenvironment: A new frontier in cancer immunotherapy. iScience 2024; 27:110750. [PMID: 39280627 PMCID: PMC11399700 DOI: 10.1016/j.isci.2024.110750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024] Open
Abstract
Innate immune cells, crucial in resisting infections and initiating adaptive immunity, play diverse and significant roles in tumor development. These cells, including macrophages, granulocytes, dendritic cells (DCs), innate lymphoid cells, and innate-like T cells, are pivotal in the tumor microenvironment (TME). Innate immune cells are crucial components of the TME, based on which various immunotherapy strategies have been explored. Immunotherapy strategies, such as novel immune checkpoint inhibitors, STING/CD40 agonists, macrophage-based surface backpack anchoring, ex vivo polarization approaches, DC-based tumor vaccines, and CAR-engineered innate immune cells, aim to enhance their anti-tumor potential and counteract cancer-induced immunosuppression. The proximity of innate immune cells to tumor cells in the TME also makes them excellent drug carriers. In this review, we will first provide a systematic overview of innate immune cells within the TME and then discuss innate cell-based therapeutic strategies. Furthermore, the research obstacles and perspectives within the field will also be addressed.
Collapse
Affiliation(s)
- Changhui Li
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Xinyu Yu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Xinyan Han
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Chen Lian
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
| | - Zijin Wang
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Fangwei Shao
- National Key Laboratory of Biobased Transportation Fuel Technology, ZJU-UIUC Institute, Zhejiang University, Hangzhou 310027, China
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Shenglin Ma
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou 310002, China
| | - Jian Liu
- Department of Respiratory and Critical Care Medicine, the Second Affiliated Hospital, and Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310029, China
- Edinburgh Medical School: Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, UK
- Biomedical and Heath Translational Research Center of Zhejiang Province, Haining, China
- Cancer Center, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
3
|
Selim MA, Suef RA, Saied E, Abdel-Maksoud MA, Almutairi SM, Aufy M, Mousa AA, Mansour MTM, Farag MMS. Peripheral NK cell phenotypic alteration and dysfunctional state post hepatitis B subviral particles stimulation in CHB patients: evading immune surveillance. Front Immunol 2024; 15:1427519. [PMID: 39328404 PMCID: PMC11424423 DOI: 10.3389/fimmu.2024.1427519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/15/2024] [Indexed: 09/28/2024] Open
Abstract
Background The relationship between chronic hepatitis B (CHB) infection and natural killer (NK) cell dysfunction is well-established, but the specific role of HBV viral antigens in driving NK cell impairment in patients with CHB remains unclear. This study investigates the modulatory effects of hepatitis B virus subviral particles (HBVsvp, a representative model for HBsAg) on the phenotypic regulation (activating and inhibitory receptors), cytokine production and cytotoxic potential of peripheral blood mononuclear cell-derived natural killer cells (PBMCs-derived NK cell), which contributes to NK cell dysfunction in CHB infection, potentially serving as an effective HBV immune evasion strategy by the virus. Methods NK cells were isolated from peripheral blood of patients with CHB (n=5) and healthy individuals (n=5), stimulated with HBVsvp. Subsequent flow cytometric characterization involved assessing changes in activating (NKp46 and NKG2D) and inhibitory (CD94) receptors expression, quantifying TNF-α and IFN- γ cytokine secretion, and evaluating the cytotoxic response against HepG2.2.15 cells with subsequent HBVsvp quantification. Results In CHB patients, in vitro exposure of PBMCs-derived NK cell with HBVsvp (represent HBsAg model) significantly reduced NK cell-activating receptors expression (P = 0.022), increased expression of CD94 + NK cells (p = 0.029), accompanied with a reduced TNF-α - IFN-γ cytokine levels, and impaired cytotoxic capacity (evidenced by increased cell proliferation and elevated HBVsvp levels in co-cultures with HepG2.2.15 cells in a time-dependent), relative to healthy donors. Conclusion These findings suggest that HBVsvp may induce dysfunctional NK cell responses characterized by phenotypic imbalance with subsequent reduction in cytokine and cytotoxic levels, indicating HBVsvp immunosuppressive effect that compromises antiviral defense in CHB patients. These data enhance our understanding of NK cell interactions with HBsAg and highlight the potential for targeting CD94 inhibitory receptors to restore NK cell function as an immunotherapeutic approach. Further clinical research is needed to validate these observations and establish their utility as reliable biomarkers.
Collapse
Affiliation(s)
- Mohamed A Selim
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Reda A Suef
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Ebrahim Saied
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Mostafa A Abdel-Maksoud
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Saeedah Musaed Almutairi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed Aufy
- Department of Pharmaceutical Sciences, Division of Pharmacology and Toxicology, University of Vienna, Vienna, Austria
| | - Adel A Mousa
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
| | - Mohamed T M Mansour
- Virology and Immunology Department, National Cancer Institute, Cairo University and Childern's Cancer Hospital, Cairo, Egypt
| | - Mohamed M S Farag
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Cairo, Egypt
- Biomedical Research Department, Armed Forces College of Medicine (AFCM), Cairo, Egypt
- The Regional Centre for Mycology and Biotechnology, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
4
|
Douka S, Papamoschou V, Raimo M, Mastrobattista E, Caiazzo M. Harnessing the Power of NK Cell Receptor Engineering as a New Prospect in Cancer Immunotherapy. Pharmaceutics 2024; 16:1143. [PMID: 39339180 PMCID: PMC11434712 DOI: 10.3390/pharmaceutics16091143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Natural killer (NK) cells have recently gained popularity as an alternative for cancer immunotherapy. Adoptive cell transfer employing NK cells offers a safer therapeutic option compared to T-cell-based therapies, due to their significantly lower toxicity and the availability of diverse autologous and allogeneic NK cell sources. However, several challenges are associated with NK cell therapies, including limited in vivo persistence, the immunosuppressive and hostile tumor microenvironment (TME), and the lack of effective treatments for solid tumors. To address these limitations, the modification of NK cells to stably produce cytokines has been proposed as a strategy to enhance their persistence and proliferation. Additionally, the overexpression of activating receptors and the blockade of inhibitory receptors can restore the NK cell functions hindered by the TME. To further improve tumor infiltration and the elimination of solid tumors, innovative approaches focusing on the enhancement of NK cell chemotaxis through the overexpression of chemotactic receptors have been introduced. This review highlights the latest advancements in preclinical and clinical studies investigating the engineering of activating, inhibitory, and chemotactic NK cell receptors; discusses recent progress in cytokine manipulation; and explores the potential of combining the chimeric antigen receptor (CAR) technology with NK cell receptors engineering.
Collapse
Affiliation(s)
- Stefania Douka
- Pharmaceutics Division, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Vasilis Papamoschou
- Pharmaceutics Division, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Monica Raimo
- Glycostem Therapeutics B.V., Kloosterstraat 9, 5349 AB Oss, The Netherlands;
| | - Enrico Mastrobattista
- Pharmaceutics Division, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
| | - Massimiliano Caiazzo
- Pharmaceutics Division, Faculty of Science, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, 3584 CG Utrecht, The Netherlands
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
5
|
Feng Z, Fu J, Tang L, Bao C, Liu H, Liu K, Yang T, Yuan JH, Zhou CB, Zhang C, Xu R, Wang FS. HBeAg induces neutrophils activation impairing NK cells function in patients with chronic hepatitis B. Hepatol Int 2024; 18:1122-1134. [PMID: 38829576 DOI: 10.1007/s12072-024-10689-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 04/21/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND The role of neutrophils in hepatitis B virus (HBV) infection has been a subject of debate due to their involvement in antiviral responses and immune regulation. This study aimed to elucidate the neutrophil characteristics in patients with chronic hepatitis B (CHB). METHODS Through flow cytometry and ribonucleic acid-sequencing analysis, the phenotypes and counts of neutrophils were analyzed in patients with CHB. Moreover, the effects of HBeAg on neutrophils and the corresponding pattern recognition receptors were identified. Simultaneously, the cross-talk between neutrophils and natural killer (NK) cells was investigated. RESULTS Neutrophils were activated in patients with CHB, characterized by higher expression levels of programmed death-ligand 1 (PD-L1), cluster of differentiation 86, and interleukin-8, and lower levels of CXC motif chemokine receptor (CXCR) 1 and CXCR2. Hepatitis B e antigen (HBeAg) partially induces neutrophil activation through the Toll-like receptor 2 (TLR2). A consistent upregulation of the TLR2 and HBeAg expression was observed in patients with CHB. Notably, the genes encoding molecules pivotal for NK-cell function upon NK receptor engagement enriched in neutrophils after HBeAg activation. The HBeAg-activated neutrophils demonstrated the ability to decrease the production of interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) in NK cells, while the PD-1 and PD-L1 pathways partially mediated the immunosuppression. CONCLUSIONS The immunosuppression of neutrophils induced by HBeAg suggests a novel pathogenic mechanism contributing to immune tolerance in patients with CHB.
Collapse
Affiliation(s)
- Zhiqian Feng
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Junliang Fu
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Lili Tang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Chunmei Bao
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Honghong Liu
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Kai Liu
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Tao Yang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Jin-Hong Yuan
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Chun-Bao Zhou
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Chao Zhang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China
| | - Ruonan Xu
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Han J, Son H, Jung D, Kim KY, Jin C, Hwang H, Kang SS, Mitalipov S, An HJ, Lee Y, Kang E. Comparison of Natural Killer Cells Differentiated from Various Pluripotent Stem Cells. Int J Mol Sci 2024; 25:8209. [PMID: 39125777 PMCID: PMC11311719 DOI: 10.3390/ijms25158209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/03/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
Allogeneic natural killer (NK) cell therapy has been effective in treating cancer. Many studies have tested NK cell therapy using human pluripotent stem cells (hPSCs). However, the impacts of the origin of PSC-NK cells on competence are unclear. In this study, several types of hPSCs, including human-induced PSCs (hiPSCs) generated from CD34+, CD3-CD56+, and CD56- cells in umbilical cord blood (UCB), three lines of human embryonic stem cells (hESCs, ES-1. ES-2 and ES-3) and MHC I knockout (B2M-KO)-ESCs were used to differentiate into NK cells and their capacities were analyzed. All PSC types could differentiate into NK cells. Among the iPSC-derived NK cells (iPSC-NKs) and ESC-derived NK cells (ES-NKs), 34+ iPSCs and ES-3 had a higher growth rate and cytotoxicity, respectively, ES-3 also showed better efficacy than 34+ iPSCs. B2M-KO was similar to the wild type. These results suggest that the screening for differentiation of PSCs into NK cells prior to selecting the PSC lines for the development of NK cell immunotherapy is an essential process for universal allotransplantation, including the chimeric antigen receptor (CAR).
Collapse
Affiliation(s)
- Jongsuk Han
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (J.H.); (H.S.); (C.J.); (H.H.)
| | - Hyeongbin Son
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (J.H.); (H.S.); (C.J.); (H.H.)
| | - Daun Jung
- Department of Pathology, CHA Bundang Medical Center, CHA University, Sungnam-si 13496, Gyeonggi-do, Republic of Korea; (D.J.); (K.-Y.K.); (H.-J.A.)
| | - Ki-Yeon Kim
- Department of Pathology, CHA Bundang Medical Center, CHA University, Sungnam-si 13496, Gyeonggi-do, Republic of Korea; (D.J.); (K.-Y.K.); (H.-J.A.)
| | - Chaeyeon Jin
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (J.H.); (H.S.); (C.J.); (H.H.)
| | - Hyeonwook Hwang
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (J.H.); (H.S.); (C.J.); (H.H.)
| | - Soon-Suk Kang
- Cell Therapy 3 Center, CHA Advanced Research Institute, CHA Bundang Medical Center, Sungnam-si 13488, Gyeonggi-do, Republic of Korea;
| | - Shoukhrat Mitalipov
- Center for Embryonic Cell and Gene Therapy, Oregon Health and Science University, Portland, OR 97239, USA;
| | - Hee-Jung An
- Department of Pathology, CHA Bundang Medical Center, CHA University, Sungnam-si 13496, Gyeonggi-do, Republic of Korea; (D.J.); (K.-Y.K.); (H.-J.A.)
| | - Yeonmi Lee
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (J.H.); (H.S.); (C.J.); (H.H.)
- Cell Therapy 3 Center, CHA Advanced Research Institute, CHA Bundang Medical Center, Sungnam-si 13488, Gyeonggi-do, Republic of Korea;
| | - Eunju Kang
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea; (J.H.); (H.S.); (C.J.); (H.H.)
- Cell Therapy 3 Center, CHA Advanced Research Institute, CHA Bundang Medical Center, Sungnam-si 13488, Gyeonggi-do, Republic of Korea;
- Department of Biochemistry, School of Medicine, CHA University, Seongnam-si 13488, Gyeonggi-do, Republic of Korea
| |
Collapse
|
7
|
Liu Z, Wang H, Liu H, Ding K, Shen H, Zhao X, Fu R. Targeting NKG2D/NKG2DL axis in multiple myeloma therapy. Cytokine Growth Factor Rev 2024; 76:1-11. [PMID: 38378397 DOI: 10.1016/j.cytogfr.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
Immune effector cells in patients with multiple myeloma (MM) are at the forefront of many immunotherapy treatments, and several methods have been developed to fully utilise the antitumour potential of immune cells. T and NK cell-derived immune lymphocytes both expressed activating NK receptor group 2 member D(NKG2D). This receptor can identify eight distinct NKG2D ligands (NKG2DL), including major histocompatibility complex class I (MHC) chain-related protein A and B (MICA and MICB). Their binding to NKG2D triggers effector roles in T and NK cells. NKG2DL is polymorphic in MM cells. The decreased expression of NKG2DL on the cell surface is explained by multiple mechanisms of tumour immune escape. In this review, we discuss the mechanisms by which the NKG2D/NKG2DL axis regulates immune effector cells and strategies for promoting NKG2DL expression and inhibiting its release in multiple myeloma and propose therapeutic strategies that increase the expression of NKG2DL in MM cells while enhancing the activation and killing function of NK cells.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, 154 Anshan Street, Heping District, Tianjin 300052, PR China.
| | - Hao Wang
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, 154 Anshan Street, Heping District, Tianjin 300052, PR China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, 154 Anshan Street, Heping District, Tianjin 300052, PR China
| | - Kai Ding
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, 154 Anshan Street, Heping District, Tianjin 300052, PR China
| | - Hongli Shen
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, 154 Anshan Street, Heping District, Tianjin 300052, PR China
| | - Xianghong Zhao
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, 154 Anshan Street, Heping District, Tianjin 300052, PR China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin 300052, PR China; Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, 154 Anshan Street, Heping District, Tianjin 300052, PR China.
| |
Collapse
|
8
|
Wang QM, Lian GY, Sheng SM, Xu J, Ye LL, Min C, Guo SF. Exosomal lncRNA NEAT1 Inhibits NK-Cell Activity to Promote Multiple Myeloma Cell Immune Escape via an EZH2/PBX1 Axis. Mol Cancer Res 2024; 22:125-136. [PMID: 37889101 DOI: 10.1158/1541-7786.mcr-23-0282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/16/2023] [Accepted: 10/24/2023] [Indexed: 10/28/2023]
Abstract
Exosomal long noncoding RNAs (lncRNA) derived from cancer cells are implicated in various processes, including cancer cell proliferation, metastasis, and immunomodulation. We investigated the role and underlying mechanism of exosome-transmitted lncRNA NEAT1 in the immune escape of multiple myeloma cells from natural killer (NK) cells. Multiple myeloma cells and samples from patients with multiple myeloma were obtained. The effects of multiple myeloma cell-derived exosomes (multiple myeloma exosomes) and exosomal NEAT1 on the functions of NK cells were evaluated using EdU staining, CCK-8, flow cytometry, and ELISA. Chromatin and RNA immunoprecipitation were performed to identify interactions between NEAT1, enhancer of Zeste Homolog 2 (EZH2), and pre-B-cell leukemia transcription factor 1 (PBX1). A xenograft tumor model was constructed to verify the effects of exosomal NEAT1 on tumor growth. qRT-PCR, Western blot analysis, and IHC were conducted to detect related genes. NEAT1 levels were upregulated in multiple myeloma tumor tissues, multiple myeloma cells, and multiple myeloma exosomes. Multiple myeloma exosomes suppressed cell proliferation, promoted apoptosis, reduced natural killer group 2, member D (NKG2D)-positive cells, and the production of TNFα) and interferon-gamma (IFN-γ) in NK cells, whereas NEAT1-silenced exosomes had little effect. NEAT1 silenced PBX1 by recruiting EZH2. PBX1 knockdown abrogated the effects of NEAT1-silenced exosomes on NK and multiple myeloma cells. NEAT1-silenced exosomes inhibited tumor growth in mice, decreased Ki67 and PD-L1, and increased NKG2D, TNFα, and IFNγ in tumor tissues. In summary, multiple myeloma cell-derived exosomal NEAT1 suppressed NK-cell activity by downregulating PBX1, promoting multiple myeloma cell immune escape. This study suggests a potential strategy for treating multiple myeloma. IMPLICATIONS This study reveals that exosomal NEAT1 regulates EZH2/PBX1 axis to inhibit NK-cell activity, thereby promoting multiple myeloma cell immune escape, which offers a novel therapeutic potential for multiple myeloma.
Collapse
Affiliation(s)
- Qing-Ming Wang
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Hematology, Nanchang, Jiangxi, China
| | - Guang-Yu Lian
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong SAR, China
| | | | - Jing Xu
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Hematology, Nanchang, Jiangxi, China
| | - Long-Long Ye
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
- Jiangxi Provincial Key Laboratory of Hematology, Nanchang, Jiangxi, China
| | - Chao Min
- Nanchang University, Nanchang, Jiangxi, China
| | | |
Collapse
|
9
|
Shao D, Bai T, Zhu B, Guo X, Dong K, Shi J, Huang Q, Kong J. Construction and Mechanism of IL-15-Based Coactivated Polymeric Micelles for NK Cell Immunotherapy. Adv Healthc Mater 2024; 13:e2302589. [PMID: 37897328 DOI: 10.1002/adhm.202302589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/12/2023] [Indexed: 10/30/2023]
Abstract
Natural killer (NK) cells are an important contributor to cancer immunotherapy, but their antitumor efficacy remains suboptimal. While cytokine-based priming shows promise in enhancing NK-cell activity, its clinical translation faces many challenges, including coactivation of multiple cytokines, poor pharmacokinetics, and limited mechanistic understanding. Here, this work develops a polymeric micelle-based IL-15/IL-2 codelivery system (IL-15/2-PEG-PTMC) for NK-cell activation. In vivo studies demonstrate that half-life of IL-15 and IL-2 and the recruitment of NK cell within tumor tissue are significantly increased after PEG-PTMC loading. Coupled with the coactivation effect of IL-15 and IL-2 conferred by this system, it noticeably delays the growth of tumors compared to conventional NK-cell activation approach, that is free IL-15 and IL-2. It is also surprisingly found that cholesterol metabolism is highly involved in the NK cell activation by IL-15/2-PEG-PTMC. Following stimulation with IL-15/2-PEG-PTMC or IL-15, NK cells undergo a series of cholesterol metabolism reprogramming, which elevates the cholesterol levels on NK cell membrane. This in turn promotes the formation of lipid rafts and activates immune synapses, effectively contributing to the enhancement of NK cell's antitumor activity. It is believed that it will open a new avenue for improving the efficacy of NK cell immunotherapy by regulating cholesterol metabolism.
Collapse
Affiliation(s)
- Dongyan Shao
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Ting Bai
- Key Laboratory of Textile Fiber and Products, Ministry of Education, Wuhan Textile University, Wuhan, 430200, China
| | - Bobo Zhu
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Xiaojia Guo
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Kai Dong
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Junling Shi
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Qingsheng Huang
- Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jie Kong
- Shaanxi Key Laboratory of Macromolecular Science and Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, 710072, China
| |
Collapse
|
10
|
Meng X, Xu H, Liang Y, Liang M, Song W, Zhou B, Shi J, Du M, Gao Y. Enhanced CT-based radiomics model to predict natural killer cell infiltration and clinical prognosis in non-small cell lung cancer. Front Immunol 2024; 14:1334886. [PMID: 38283362 PMCID: PMC10811188 DOI: 10.3389/fimmu.2023.1334886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Background Natural killer (NK) cells are crucial for tumor prognosis; however, their role in non-small-cell lung cancer (NSCLC) remains unclear. The current detection methods for NSCLC are inefficient and costly. Therefore, radiomics represent a promising alternative. Methods We analyzed the radiogenomics datasets to extract clinical, radiological, and transcriptome data. The effect of NK cells on the prognosis of NSCLC was assessed. Tumors were delineated using a 3D Slicer, and features were extracted using pyradiomics. A radiomics model was developed and validated using five-fold cross-validation. A nomogram model was constructed using the selected clinical variables and a radiomic score (RS). The CIBERSORTx database and gene set enrichment analysis were used to explore the correlations of NK cell infiltration and molecular mechanisms. Results Higher infiltration of NK cells was correlated with better overall survival (OS) (P = 0.002). The radiomic model showed an area under the curve of 0.731, with 0.726 post-validation. The RS differed significantly between high and low infiltration of NK cells (P < 0.01). The nomogram, using RS and clinical variables, effectively predicted 3-year OS. NK cell infiltration was correlated with the ICOS and BTLA genes (P < 0.001) and macrophage M0/M2 levels. The key pathways included TNF-α signaling via NF-κB and Wnt/β-catenin signaling. Conclusions Our radiomic model accurately predicted NK cell infiltration in NSCLC. Combined with clinical characteristics, it can predict the prognosis of patients with NSCLC. Bioinformatic analysis revealed the gene expression and pathways underlying NK cell infiltration in NSCLC.
Collapse
Affiliation(s)
- Xiangzhi Meng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haijun Xu
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Liang
- Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mei Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weijian Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Boxuan Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianwei Shi
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minjun Du
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
11
|
Xu F, Li L, Jiang L, Zhang J. Identification of key genes and immune infiltration in multiple myeloma by bioinformatics analysis. Hematology 2023; 28:2264517. [PMID: 37815499 DOI: 10.1080/16078454.2023.2264517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 09/24/2023] [Indexed: 10/11/2023] Open
Abstract
OBJECTIVE Multiple Myeloma (MM) is a hematologic malignant disease with unclear molecular mechanisms. This integrated bioinformatic study aimed to identify key genes, pathways and immune cell infiltration pattern in MM. METHODS Differentially expressed genes (DEGs) from GSE6477 and GSE16558 dataset were filtrated with R package 'limma', whose function were explored by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The key genes were selected from Protein-protein interaction network (PPI) and logistic regression model. The correlation between key genes and survival in MM was evaluated using the survival and survminer package. Additionally, immune filtration analysis was accomplished by CIBERSORT tools. RESULTS 118 DEGs (92 up-regulated and 26 down-regulated) from two GSE datasets were identified, which were closely related with B cell receptor signaling pathway and Epstein-Barr virus infection. Furthermore, CD24 and PTPRC of five hub genes identified in PPI network were further screened out by the logistic regression model. Besides, CD24 and PTPRC expression were significantly correlated to the survival time in MM patients. Finally, MM might cause different infiltrating immune cell compositions, including increased infiltrations of B cells memory, Plasma cells, T cells CD4 memory resting, T cells follicular helper, Tregs, NK cells resting, Macrophages(M0/M1), Dendritic cells resting and Mast cells activating, and lower proportions of B cells naïve, T cells CD4 naïve, Macrophages M2 and Neutrophils. CONCLUSION Targeting CD24 and PTPRC as molecular markers of MM is valuable to MM therapy. Moreover, the immune cell infiltration will provide new insights into MM immunopathology.
Collapse
Affiliation(s)
- Fei Xu
- Department of Hematology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Ling Li
- Department of Hematology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - LiMei Jiang
- Department of Hematology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| | - Jing Zhang
- Institute of Medical Sciences, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, People's Republic of China
| |
Collapse
|
12
|
Liu Z, Xu X, Liu H, Zhao X, Yang C, Fu R. Immune checkpoint inhibitors for multiple myeloma immunotherapy. Exp Hematol Oncol 2023; 12:99. [PMID: 38017516 PMCID: PMC10685608 DOI: 10.1186/s40164-023-00456-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 11/02/2023] [Indexed: 11/30/2023] Open
Abstract
Multiple myeloma (MM) is related to immune disorders, recent studys have revealed that immunotherapy can greatly benefit MM patients. Immune checkpoints can negatively modulate the immune system and are closely associated with immune escape. Immune checkpoint-related therapy has attracted much attention and research in MM. However, the efficacy of those therapies need further improvements. There need more thoughts about the immune checkpoint to translate their use in clinical work. In our review, we aggregated the currently known immune checkpoints and their corresponding ligands, further more we propose various ways of potential translation applying treatment based on immune checkpoints for MM patients.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xintong Xu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xianghong Zhao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Chun Yang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China.
| |
Collapse
|
13
|
Jiang D, Zhang J, Mao Z, Shi J, Ma P. Driving natural killer cell-based cancer immunotherapy for cancer treatment: An arduous journey to promising ground. Biomed Pharmacother 2023; 165:115004. [PMID: 37352703 DOI: 10.1016/j.biopha.2023.115004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 06/06/2023] [Accepted: 06/08/2023] [Indexed: 06/25/2023] Open
Abstract
Immunotherapy represents one of the most effective strategies for cancer treatment. Recently, progress has been made in using natural killer (NK) cells for cancer therapy. NK cells can directly kill tumor cells without pre-sensitization and thus show promise in clinical applications, distinct from the use of T cells. Whereas, research and development on NK cell-based immunotherapy is still in its infancy, and enhancing the therapeutic effects of NK cells remains a key problem to be solved. An incompletely understanding of the mechanisms of action of NK cells, immune resistance in the tumor microenvironment, and obstacles associated with the delivery of therapeutic agents in vivo, represent three mountains that need to be scaled. Here, we firstly describe the mechanisms underlying the development, activity, and maturation of NK cells, and the formation of NK‑cell immunological synapses. Secondly, we discuss strategies for NK cell-based immunotherapy strategies, including adoptive transfer of NK cell therapy and treatment with cytokines, monoclonal antibodies, and immune checkpoint inhibitors targeting NK cells. Finally, we review the use of nanotechnology to overcome immune resistance, including enhancing the anti-tumor efficiency of chimeric antigen receptor-NK, cytokines and immunosuppressive-pathways inhibitors, promoting NK cell homing and developing NK cell-based nano-engagers.
Collapse
Affiliation(s)
- Dandan Jiang
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Jingya Zhang
- Patent Examination Cooperation (Henan) Center of the Patent office, China National Intellectual Property Administration, Henan 450046, China
| | - Zhenkun Mao
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou University, Zhengzhou 450001, China.
| | - Peizhi Ma
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450003, China.
| |
Collapse
|
14
|
Li YM, Li YX, Hu XZ, Li DY, An L, Yuan ZY, Liu ZL, Du KM, Zheng ZZ. Exploration of KIR genes and hematological-related diseases in Chinese Han population. Sci Rep 2023; 13:9773. [PMID: 37328612 PMCID: PMC10276034 DOI: 10.1038/s41598-023-36882-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 06/12/2023] [Indexed: 06/18/2023] Open
Abstract
The function of natural killer (NK) cells has previously been implicated in hematopoietic-related diseases. Killer immunoglobulin-like receptors (KIR) play an important role in NK cells after hematopoietic stem cell transplantation. To explore the immunogenetic predisposition of hematological-related diseases, herein, a multi-center retrospective study in China was conducted, analyzing and comparing 2519 patients with hematopathy (mainly, acute lymphoblastic leukemia, acute myeloid leukemia, aplastic anemia, and myelodysplastic syndrome) to 18,108 individuals without known pathology. Genotyping was performed by polymerase chain reaction with specific sequence primers (PCR-SSP). As a result, we discovered four genes including KIR2DL5 (OR: 0.74, 95% CI 0.59-0.93; Pc = 0.0405), 2DS1 (OR: 0.74, 95% CI 0.59-0.93; Pc = 0.0405), 2DS3 (OR: 0.58, 95% CI 0.41-0.81; Pc = 0.0180), and 3DS1 (OR: 0.74, 95% CI 0.58-0.94; Pc = 0.0405) to be protective factors that significantly reduce the risk of aplastic anemia. Our findings offer new approaches to immunotherapy for hematological-related diseases. As these therapies mature, they are promising to be used alone or in combination with current treatments to help to make blood disorders a manageable disease.
Collapse
Affiliation(s)
- Ye-Mo Li
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | - Yu-Xia Li
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | - Xiao-Zhuang Hu
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | - Dai-Yang Li
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | - Lin An
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | - Zhi-Yang Yuan
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | | | - Ke-Ming Du
- Shanghai Tissuebank Biotechnology Co, Ltd, Shanghai, China
| | | |
Collapse
|
15
|
Wang B, Wang W, Li Q, Guo T, Yang S, Shi J, Yuan W, Chu Y. High Expression of Microtubule-associated Protein TBCB Predicts Adverse Outcome and Immunosuppression in Acute Myeloid Leukemia. J Cancer 2023; 14:1707-1724. [PMID: 37476188 PMCID: PMC10355208 DOI: 10.7150/jca.84215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 06/03/2023] [Indexed: 07/22/2023] Open
Abstract
Acute myeloid leukemia (AML) is a devastating blood cancer with high heterogeneity and ill-fated outcome. Despite numerous advances in AML treatment, the prognosis remains poor for a significant proportion of patients. Consequently, it is necessary to accurately and comprehensively identify biomarkers as soon as possible to enhance the efficacy of diagnosis, prognosis and treatment of AML. In this study, we aimed to identify prognostic markers of AML by analyzing the cohorts from TCGA-LAML database and GEO microarray datasets. Interestingly, the transcriptional level of microtubule-associated protein TBCB in AML patients was noticeably increased when compared with normal individuals, and this was verified in two independent cohorts (GSE9476 and GSE13159) and with our AML patients. Furthermore, univariate and multivariate regression analysis revealed that high TBCB expression was an independent poor prognostic factor for AML. GO and GSEA enrichment analysis hinted that immune-related signaling pathways were enriched in up-regulated DEGs between two populations separated by the median expression level of TBCB. By constructing a protein-protein interaction network, we obtained six hub genes, all of which are immune-related molecules, and their expression levels were positively linked to that of TBCB. In addition, the high expression of three hub genes was significantly associated with a poor prognosis in AML. Moreover, we found that the tumor microenvironment in AML with high TBCB expression tended to be infiltrated by NK cells, especially CD56bright NK cells. The transcriptional levels of NK cell inhibitory receptors and their ligands were positively related to that of TBCB, and their high expression levels also predicted poor prognosis in AML. Notably, we found that the down-regulation of TBCB suppressed cell proliferation in AML cell lines by enhancing the apoptosis and cell cycle arrest. Finally, drug sensitivity prediction illustrated that cells with high TBCB expression were more responsive to ATRA and midostaurin but resistant to cytarabine, dasatinib, and imatinib. In conclusion, our findings shed light on the feasibility of TBCB as a potential predictor of poor outcome and to be an alternative target of treatment in AML.
Collapse
Affiliation(s)
- Bichen Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Wenjun Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
- Regenerative Medicine Clinic, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Qiaoli Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
- Regenerative Medicine Clinic, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Tengxiao Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Shuang Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Jun Shi
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
- Regenerative Medicine Clinic, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Weiping Yuan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| | - Yajing Chu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin 301600, China
| |
Collapse
|
16
|
Li YR, Dunn ZS, Yu Y, Li M, Wang P, Yang L. Advancing cell-based cancer immunotherapy through stem cell engineering. Cell Stem Cell 2023; 30:592-610. [PMID: 36948187 PMCID: PMC10164150 DOI: 10.1016/j.stem.2023.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 01/04/2023] [Accepted: 02/22/2023] [Indexed: 03/24/2023]
Abstract
Advances in cell-based therapy, particularly CAR-T cell therapy, have transformed the treatment of hematological malignancies. Although an important step forward for the field, autologous CAR-T therapies are hindered by high costs, manufacturing challenges, and limited efficacy against solid tumors. With ongoing progress in gene editing and culture techniques, engineered stem cells and their application in cell therapy are poised to address some of these challenges. Here, we review stem cell-based immunotherapy approaches, stem cell sources, gene engineering and manufacturing strategies, therapeutic platforms, and clinical trials, as well as challenges and future directions for the field.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zachary Spencer Dunn
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Yanqi Yu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Miao Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA; Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
17
|
Gao Y, Zhang X, Jiang T, Zhou H, Liu H, Hu Y, Cao J. Inhibition of hepatic natural killer cell function via the TIGIT receptor in schistosomiasis-induced liver fibrosis. PLoS Pathog 2023; 19:e1011242. [PMID: 36930687 PMCID: PMC10022799 DOI: 10.1371/journal.ppat.1011242] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Schistosomiasis is a zoonotic parasitic disease. Schistosoma japonicum eggs deposited in the liver tissue induce egg granuloma formation and liver fibrosis, seriously threatening human health. Natural killer (NK) cells kill activated hepatic stellate cells (HSCs) or induce HSC apoptosis and inhibit the progression of liver fibrosis. However, the function of NK cells in liver fibrosis caused by S. japonicum infection is significantly inhibited. The mechanism of this inhibition remains unclear. Twenty mice were percutaneously infected with S. japonicum cercariae. Before infection and 2, 4, 6, and 8 weeks after infection, five mice were euthanized and dissected at each time point. Hepatic NK cells were isolated and transcriptome sequenced. The sequencing results showed that Tigit expression was high at 4-6 weeks post infection. This phenomenon was verified by reverse transcription quantitative PCR (RT-qPCR) and flow cytometry. NK cells derived from Tigit-/- and wild-type (WT) mice were co-cultured with HSCs. It was found that Tigit-/- NK cells induced apoptosis in a higher proportion of HSCs than WT NK cells. Schistosomiasis infection models of Tigit-/- and WT mice were established. The proportion and killing activity of hepatic NK cells were significantly higher in Tigit-/- mice than in WT mice. The degree of liver fibrosis in Tigit-/- mice was significantly lower than that in WT mice. NK cells were isolated from Tigit-/- and WT mice and injected via the tail vein into WT mice infected with S. japonicum. The degree of liver fibrosis in mice that received NK cell infusion reduced significantly, but there was no significant difference between mice that received NK cells from Tigit-/- and WT mice, respectively. Our findings indicate that Tigit knockout enhanced the function of NK cells and reduced the degree of liver fibrosis in schistosomiasis, thus providing a novel strategy for treating hepatic fibrosis induced by schistosomiasis.
Collapse
Affiliation(s)
- Yuan Gao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
| | - Xiaocheng Zhang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
| | - Tingting Jiang
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
| | - Hao Zhou
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
| | - Hua Liu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
| | - Yuan Hu
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
- * E-mail: (YH); (JC)
| | - Jianping Cao
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research); Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China; World Health Organization Collaborating Center for Tropical Diseases, Shanghai, China
- The School of Global Health, Chinese Center for Tropical Diseases Research, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (YH); (JC)
| |
Collapse
|
18
|
MiR-30c facilitates natural killer cell cytotoxicity to lung cancer through targeting GALNT7. Genes Genomics 2023; 45:247-260. [PMID: 36040682 DOI: 10.1007/s13258-022-01306-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/11/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND MicroRNAs (miRNAs) have been reported to play important roles in regulating natural killer (NK) cell cytotoxicity to cancer cells. OBJECTIVE This study aimed to investigate the effects and potential mechanism of miR-30c in regulating NK cell cytotoxicity to lung cancer cells. METHODS Primary NK cells were derived from the peripheral blood of lung cancer and normal participants. Exosomes were isolated and validated via transmission electron microscopy and nanoparticle tracking analysis. The levels of miR-30c, polypeptide N-acetylgalactosaminyltransferase 7 (GALNT7) and proteins in PI3K/AKT pathway were determined using quantitative real-time polymerase chain reaction or western blot. Tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ) levels and the cytotoxicity of effector NK cells to target lung cancer cells were measured via enzyme linked immunosorbent assay, cell apoptosis or xenograft experiments. The relationship between miR-30c and GALNT7 was analyzed by luciferase activity, RNA pull-down and RNA immunoprecipitation assays. And a xenograft mice model was established to verify the effect of miR-30c in regulating NK cell cytotoxicity to lung cancer cells in vivo. RESULTS NK cell-derived exosomes carrying miR-30c, and miR-30c level was significantly downregulated in primary NK cells of lung cancer patients. MiR-30c overexpression promoted TNF-α and IFN-γ secretion and enhanced the cytotoxicity of interleukin 2 (IL-2)-treated NK cells to lung cancer cells, while knockdown of miR-30c played an opposite effect in regulating the cytotoxicity of NK cells to lung cancer cells. GALNT7 was a target of miR-30c and was negatively regulated by miR-30c. Besides, miR-30c targeted GALNT7 to exert its function in regulating NK cell cytotoxicity. Furthermore, GALNT7 prompted the activation of PI3K/AKT pathway in NK cells. Additionally, miR-30c overexpression enhanced NK cell cytotoxicity to lung cancer cells and inhibited tumor growth in vivo. CONCLUSION miR-30c enhanced NK cell cytotoxicity to lung cancer cells via decreasing GALNT7 and inactivating the PI3K/AKT pathway, suggesting that regulating miR-30c expression maybe a promising approach for enhancing NK cell-based antitumor therapies.
Collapse
|
19
|
Immunotherapy targeting inhibitory checkpoints: The role of NK and other innate lymphoid cells. Semin Immunol 2022; 61-64:101660. [PMID: 36370672 DOI: 10.1016/j.smim.2022.101660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 12/14/2022]
Abstract
Monoclonal antibodies that target specific ligand-receptor signaling pathways and act as immune checkpoint inhibitors have been designed to remove the brakes in T cells and restore strong and long-term antitumor-immunity. Of note, many of these inhibitory receptors are also expressed by Innate Lymphoid Cells (ILCs), suggesting that also blockade of inhibitory pathways in innate lymphocytes has a role in the response to the treatment with checkpoint inhibitors. ILCs comprise cytotoxic NK cells and "helper" subsets and are important cellular components in the tumor microenvironment. In addition to killing tumor cells, ILCs release inflammatory cytokines, thus contributing to shape adaptive cell activation in the context of immunotherapy. Therefore, ILCs play both a direct and indirect role in the response to checkpoint blockade. Understanding the impact of ILC-mediated response on the treatment outcome would contribute to enhance immunotherapy efficacy, as still numerous patients resist or relapse.
Collapse
|
20
|
Liu Z, Wang H, Li Y, Meng N, Liu H, Ding K, Fu R. PIM2 kinase regulates the expression of TIGIT and energy metabolism on NK cell in multiple myeloma patients.. [DOI: 10.21203/rs.3.rs-2159151/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Abstract
Background: PIM2 kinase play a vital role in the generation of plasma cell and bone loss in multiple myeloma(MM), which highly related to the tumor progression and as a potential therapy target in MM. In immune cell,PIM2 kinase involved in the regulation of lymphocyte like T cell and B cell, However, its role in NK cells remains unclear.
Methods: Single-cell RNA sequencing data were analysed the expression of PIM2 kinase in NK cells from MM patients and healthy donors.Immune checkpoint expression, cell apoptosis, and NK cell function had been evaluated through flow cytometry.Then, NCBI, UCSC, JASPAR and GEPIA database were used to predict promoter of TIGIT.NK-92 cells with ETS-1 knockdown were established by using sh-RNA. Kinase functional assay (ADP-Glo) were used to confirm PIM2 inhibitor from 160 kinds of natural flavonoids compound.Samples treated with or without drugs were analyzed using mass spectrometry and RNA-seq. The oxygen consumption rate (OCR), and the extracellular acidification rate (ECAR) were measured by assay kit.
Result: The PIM2 kinase was highly expressed in the NK cells from MM patients based on single-cell sequencing analysis and confirmed in clinical sample by PCR and flow cytometry.Inhibition of PIM2 kinase can increase the function of NK cells and down regulation TIGIT expression. Mechanism, we confirmed that ETS-1 which was directly binding to the promoter of TIGIT was up-regulated by PIM2 kinase, which can lead the strengthened transcription of TIGIT on NK cells.Furthermore, two novel natural flavonoids compound named Kaempferol and Quercetin dihydrate as PIM2 kinase inhibitors exhibiting higher efficiency at low dose in MM cells,while influence the expression of TIGIT and energy metabolism on NK-92 cells. For in vitro experiment,PIM2 kinase inhibitors can activate NK cell killing function and decrease TIGIT expression,while promoted the apoptosis of MM cells irrespective of adding BMSCs or not in co-culture systems BMSCs.
Conclusion: PIM2 kinase involved in the regulation of NK cell.Inhibiting PIM2 kinase could down-regulate the expression of TIGIT and improve energy metabolism to enhance NK cell anti myeloma cell.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rong Fu
- Tianjin Medical University General Hospital
| |
Collapse
|
21
|
Li X, Chen M, Wan Y, Zhong L, Han X, Chen X, Xiao F, Liu J, Zhang Y, Zhu D, Xiang J, Liu J, Huang H, Hou J. Single-cell transcriptome profiling reveals the key role of ZNF683 in natural killer cell exhaustion in multiple myeloma. Clin Transl Med 2022; 12:e1065. [PMID: 36245253 PMCID: PMC9574488 DOI: 10.1002/ctm2.1065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 01/28/2023] Open
Abstract
BACKGROUNDS Decreased cytotoxicity of natural killer (NK) cells has been shown in multiple myeloma (MM). However, the underlying molecular mechanisms remain unclear. Here, by using single-cell RNA sequencing analysis and in vitro experiments, we aim to uncover and validate molecularly distinctive insights into identifying regulators for NK cell exhaustion and provide potential targets for novel immune therapies in MM. METHODS Single-cell RNA sequencing was conducted in the bone marrow and peripheral blood samples from 10 newly diagnosed MM patients and three healthy volunteers. Based on the cluster-defining differentially expressed genes, we named and estimated functional states of each cluster via bioinformatics analyses. Functional significance of key findings obtained from sequencing analysis was examined in a series of in vitro experiments, including luciferase reporter assay, lentiviral expression vector construction, NK cell transfection, RT-qPCR, flow cytometry, and cytotoxicity assay. RESULTS We classified NK cells into seven distinct clusters and confirmed that a subset of ZNF683+ NK cells were enriched in MM patients with 'exhausted' transcriptomic profile, featuring as decreased expression of activating receptors and cytolytic molecules, as well as increased expression of inhibitory receptors. Next, we found a significant downregulation of SH2D1B gene that encodes EAT-2, an adaptor protein of activating receptor SLAMF7, in ZNF683+ NK cells from MM patients versus healthy volunteers. We further proved that ZNF683 transfection in NK cells significantly downregulated SH2D1B expression via directly binding to the promoter of SH2D1B, leading to NK cell cytotoxic activity impairment and exhausted phenotypes acquisition. In contrast, ZNF683 knockout in NK cells from MM patients increased cytotoxic activity and reversed NK cell exhaustion. CONCLUSIONS In summary, our findings uncover an important mechanism of ZNF683+ NK cell exhaustion and suggest that transcriptional suppressor ZNF683 as a potential useful therapeutic target in immunotherapy of MM.
Collapse
Affiliation(s)
- Xin Li
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Mengping Chen
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yike Wan
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lu Zhong
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaofeng Han
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaotong Chen
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Fei Xiao
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jia Liu
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yiwei Zhang
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Di Zhu
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jing Xiang
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Junling Liu
- Department of Biochemistry and Molecular Cell BiologyShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Honghui Huang
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jian Hou
- Department of HematologyRen Ji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
22
|
Li C, Liang H, Bian S, Hou X, Ma Y. Construction of a Prognosis Model of the Pyroptosis-Related Gene in Multiple Myeloma and Screening of Core Genes. ACS OMEGA 2022; 7:34608-34620. [PMID: 36188246 PMCID: PMC9521030 DOI: 10.1021/acsomega.2c04212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/02/2022] [Indexed: 06/16/2023]
Abstract
Pyroptosis is an important factor affecting the proliferation, invasion, and metastasis of tumor cells. However, in multiple myeloma (MM), there are few studies on whether the occurrence of pyroptosis is related to the occurrence and prognosis of the disease. Based on the Gene Expression Omnibus and Cancer Genome Atlas database search dataset, this study identified pyroptosis-related genes with a specific prognosis, constructed and verified the prediction model by stepwise Cox regression analysis and time receiver operating characteristic curve analysis, and predicted specific functions by single-sample gene set enrichment analysis and the Kyoto Encyclopedia of Genes and Genomes. Dataset analysis identified key genes, which were used to construct a risk scoring system for the prognosis of MM. The entire test set and external verification set verified the results. The expression levels of related genes in the clinical samples were detected using fluorescence quantitative PCR. A prognostic gene model based on six pyroptosis-related genes (CYCS, NLRP9, AIM2, NOD2, CHMP3, and GSDME) was constructed. The model has an excellent prognostic ability and can be popularized in the external validation set. The predictive prognostic nomogram integrating clinical information can effectively evaluate the risk score of each patient and predict their survival. After sample validation, our study found three potential key pyroptosis-related genes in multiple myeloma. GSDME, NOD2, and CHMP3 were significantly different between MM and healthy subjects, suggesting that they are pyroptosis-related protective genes. This study shows that the key pyroptosis-related gene in the model can be used as a marker for predicting the prognosis of myeloma, which may provide a basis for clinical individualized stratification therapy.
Collapse
Affiliation(s)
- Can Li
- Department
of Hematology, The Second Clinical Medical College of Shanxi Medical
University, Shanxi Medical University, 030000 Taiyuan, China
| | - Hongzheng Liang
- Department
of Hematology, The Second Clinical Medical College of Shanxi Medical
University, Shanxi Medical University, 030000 Taiyuan, China
| | - Sicheng Bian
- Harbin
Medical University, 23 Youzheng Street, NanGang District, Harbin 150001, PR China
| | - Xiaoxu Hou
- Department
of Hematology, The Second Clinical Medical College of Shanxi Medical
University, Shanxi Medical University, 030000 Taiyuan, China
| | - Yanping Ma
- Department
of Hematology, The Second Clinical Medical College of Shanxi Medical
University, Shanxi Medical University, 030000 Taiyuan, China
| |
Collapse
|
23
|
Kosta A, Mekhloufi A, Lucantonio L, Zingoni A, Soriani A, Cippitelli M, Gismondi A, Fazio F, Petrucci MT, Santoni A, Stabile H, Fionda C. GAS6/TAM signaling pathway controls MICA expression in multiple myeloma cells. Front Immunol 2022; 13:942640. [PMID: 35967396 PMCID: PMC9368199 DOI: 10.3389/fimmu.2022.942640] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
NKG2D ligands play a relevant role in Natural Killer (NK) cell -mediated immune surveillance of multiple myeloma (MM). Different levels of regulation control the expression of these molecules at cell surface. A number of oncogenic proteins and miRNAs act as negative regulators of NKG2D ligand transcription and translation, but the molecular mechanisms sustaining their basal expression in MM cells remain poorly understood. Here, we evaluated the role of the growth arrest specific 6 (GAS6)/TAM signaling pathway in the regulation of NKG2D ligand expression and MM recognition by NK cells. Our data showed that GAS6 as well as MERTK and AXL depletion in MM cells results in MICA downregulation and inhibition of NKG2D-mediated NK cell degranulation. Noteworthy, GAS6 derived from bone marrow stromal cells (BMSCs) also increases MICA expression at both protein and mRNA level in human MM cell lines and in primary malignant plasma cells. NF-kB activation is required for these regulatory mechanisms since deletion of a site responsive for this transcription factor compromises the induction of mica promoter by BMSCs. Accordingly, knockdown of GAS6 reduces the capability of BMSCs to activate NF-kB pathway as well as to enhance MICA expression in MM cells. Taken together, these results shed light on molecular mechanism underlying NKG2D ligand regulation and identify GAS6 protein as a novel autocrine and paracrine regulator of basal expression of MICA in human MM cells.
Collapse
Affiliation(s)
- Andrea Kosta
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Abdelilah Mekhloufi
- Department of Biomedical Engineering, Emory University, Atlanta, GA, United States
| | - Lorenzo Lucantonio
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Marco Cippitelli
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Angela Gismondi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesca Fazio
- Division of Hematology, Department of Translational Medicine and Precision, Sapienza University of Rome, Rome, Italy
| | - Maria Teresa Petrucci
- Division of Hematology, Department of Translational Medicine and Precision, Sapienza University of Rome, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Neuromed, Pozzilli, Italy
- Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Helena Stabile
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- *Correspondence: Cinzia Fionda, ; Helena Stabile,
| | - Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- *Correspondence: Cinzia Fionda, ; Helena Stabile,
| |
Collapse
|
24
|
Stalker ME, Mark TM. Clinical Management of Triple-Class Refractory Multiple Myeloma: A Review of Current Strategies and Emerging Therapies. Curr Oncol 2022; 29:4464-4477. [PMID: 35877215 PMCID: PMC9315521 DOI: 10.3390/curroncol29070355] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022] Open
Abstract
Major progress has been made in the upfront treatment of multiple myeloma, but the disease ultimately relapses and leads to death in the vast majority of those afflicted. New treatment strategies and modalities are necessary to treat myeloma in relapse, particularly in cases of triple-refractory status defined by disease progression during or shortly after treatment with immunomodulatory agents, proteasome inhibitors, and anti-CD38 monoclonal antibody therapy. In this manuscript, we review recent promising developments in the treatment of triple-class refractory myeloma including bispecific antibodies and T cell engagers, chimeric antigen receptor cellular therapies, as well as chemotherapeutics with novel mechanisms of action.
Collapse
Affiliation(s)
| | - Tomer M. Mark
- Department of Medicine, Division of Hematology, University of Colorado, Aurora, CO 80045, USA
- Correspondence:
| |
Collapse
|
25
|
Annese T, Tamma R, Ribatti D. Update in TIGIT Immune-Checkpoint Role in Cancer. Front Oncol 2022; 12:871085. [PMID: 35656508 PMCID: PMC9152184 DOI: 10.3389/fonc.2022.871085] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/14/2022] [Indexed: 12/12/2022] Open
Abstract
The in-depth characterization of cross-talk between tumor cells and T cells in solid and hematological malignancies will have to be considered to develop new therapeutical strategies concerning the reactivation and maintenance of patient-specific antitumor responses within the patient tumor microenvironment. Activation of immune cells depends on a delicate balance between activating and inhibitory signals mediated by different receptors. T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT) is an inhibitory receptor expressed by regulatory T cells (Tregs), activated T cells, and natural killer (NK) cells. TIGIT pathway regulates T cell-mediated tumor recognition in vivo and in vitro and represents an exciting target for checkpoint blockade immunotherapy. TIGIT blockade as monotherapy or in combination with other inhibitor receptors or drugs is emerging in clinical trials in patients with cancer. The purpose of this review is to update the role of TIGIT in cancer progression, looking at TIGIT pathways that are often upregulated in immune cells and at possible therapeutic strategies to avoid tumor aggressiveness, drug resistance, and treatment side effects. However, in the first part, we overviewed the role of immune checkpoints in immunoediting, the TIGIT structure and ligands, and summarized the key immune cells that express TIGIT.
Collapse
Affiliation(s)
- Tiziana Annese
- Department of Medicine and Surgery, Libera Università del Mediterraneo (LUM) Giuseppe Degennaro University, Bari, Italy.,Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Roberto Tamma
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Section of Human Anatomy and Histology, University of Bari Medical School, Bari, Italy
| |
Collapse
|
26
|
Allegra A, Casciaro M, Lo Presti E, Musolino C, Gangemi S. Harnessing Unconventional T Cells and Innate Lymphoid Cells to Prevent and Treat Hematological Malignancies: Prospects for New Immunotherapy. Biomolecules 2022; 12:biom12060754. [PMID: 35740879 PMCID: PMC9221132 DOI: 10.3390/biom12060754] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/21/2022] [Accepted: 05/26/2022] [Indexed: 11/18/2022] Open
Abstract
Unconventional T cells and innate lymphoid cells (ILCs) make up a heterogeneous set of cells that characteristically show prompt responses toward specific antigens. Unconventional T cells recognize non-peptide antigens, which are bound and presented by diverse non-polymorphic antigen-presenting molecules and comprise γδ T cells, MR1-restricted mucosal-associated invariant T cells (MAITs), and natural killer T cells (NKTs). On the other hand, ILCs lack antigen-specific receptors and act as the innate counterpart to the T lymphocytes found in the adaptive immune response. The alteration of unconventional T cells and ILCs in frequency and functionality is correlated with the onset of several autoimmune diseases, allergy, inflammation, and tumor. However, depending on the physio-pathological framework, unconventional T cells may exhibit either protective or pathogenic activity in a range of neoplastic diseases. Nonetheless, experimental models and clinical studies have displayed that some unconventional T cells are potential therapeutic targets, as well as prognostic and diagnostic markers. In fact, cell-mediated immune response in tumors has become the focus in immunotherapy against neoplastic disease. This review concentrates on the present knowledge concerning the function of unconventional T cell sets in the antitumor immune response in hematological malignancies, such as acute and chronic leukemia, multiple myeloma, and lymphoproliferative disorders. Moreover, we discuss the possibility that modulating the activity of unconventional T cells could be useful in the treatment of hematological neoplasms, in the prevention of specific conditions (such as graft versus host disease), and in the formulation of an effective anticancer vaccine therapy. The exact knowledge of the role of these cells could represent the prerequisite for the creation of a new form of immunotherapy for hematological neoplasms.
Collapse
Affiliation(s)
- Alessandro Allegra
- Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, Division of Hematology, University of Messina, 98125 Messina, Italy; (A.A.); (C.M.)
| | - Marco Casciaro
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy;
- Correspondence: ; Tel.: +39-090-221-2013
| | - Elena Lo Presti
- National Research Council (CNR)—Institute for Biomedical Research and Innovation (IRIB), 90146 Palermo, Italy;
| | - Caterina Musolino
- Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, Division of Hematology, University of Messina, 98125 Messina, Italy; (A.A.); (C.M.)
| | - Sebastiano Gangemi
- Department of Clinical and Experimental Medicine, School and Operative Unit of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy;
| |
Collapse
|
27
|
Zhang L, Liu M, Song B, Miao W, Zhan R, Yang S, Han Z, Cai H, Xu X, Zhao Y, Han Z, Guo T, Yao J, Huang Q. Decoding the multidimensional signatures of resident and expanded natural killer cells generated from perinatal blood. Am J Cancer Res 2022; 12:2132-2145. [PMID: 35693070 PMCID: PMC9185604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/14/2022] [Indexed: 06/15/2023] Open
Abstract
Natural killer (NK) cells are lymphocytes and play a pivotal role in innate and adaptive immune responses against infections and malignancies. Longitudinal studies have indicated the feasibility of perinatal blood for large-scale NK cell generation, yet the systematic and detailed comparations of the signatures of resident and expanded NK cells (rNKs, eNKs) are largely obscure. Herein, we harvested rNKs from umbilical cord blood (rUC-NKs) and placental blood (rP-NKs) as well as the corresponding eNKs (eUC-NKs, eP-NKs). Furthermore, the biological properties and transcriptomic signatures including cellular subpopulations, cytotoxicity, gene expression profiling, genetic characteristics, signaling pathways and gene set-related biological process were investigated. The enriched rNKs and eNKs exhibited diversity in biomarker expression pattern, and eNKs with higher percentages of NKG2D+, NKG2A+, NKp44+ and NKp46+ subsets. rNKs or eNKs with different origins showed more similarities in transcriptomic signatures than those with the same origin. Our data revealed multifaceted similarities and differences of the indicated rNKs and pNKs both at the cellular and molecular levels. Our findings provide new references for further dissecting the efficacy and molecular mechanisms of rNKs and eNKs, which will collectively benefit the fundamental and translational studies of NK cell-based immunotherapy.
Collapse
Affiliation(s)
- Leisheng Zhang
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province & NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial HospitalLanzhou 730000, Gansu, China
- Center for Cellular Therapies, The First Affiliated Hospital of Shandong First Medical UniversityJinan 250014, Shandong, China
- Key Laboratory of Radiation Technology and Biophysics, Hefei Institute of Physical Science, Chinese Academy of SciencesHefei 230031, Anhui, China
- Institute of Health-Biotech, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd.Tianjin 301701, China
| | - Min Liu
- Veterinary Bureau, Department agriculture and Animal Husbandry of Inner Mongolia Autonomous RegionHohhot 010011, China
| | - Baoquan Song
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Wenjing Miao
- National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Rucai Zhan
- Center for Cellular Therapies, The First Affiliated Hospital of Shandong First Medical UniversityJinan 250014, Shandong, China
| | - Sijun Yang
- Jiangxi Research Center of Stem Cell Engineering, Jiangxi Health-Biotech Stem Cell Technology Co., Ltd.Shangrao 334000, Jiangxi, China
| | - Zhihai Han
- Jiangxi Research Center of Stem Cell Engineering, Jiangxi Health-Biotech Stem Cell Technology Co., Ltd.Shangrao 334000, Jiangxi, China
| | - Hui Cai
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province & NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial HospitalLanzhou 730000, Gansu, China
| | - Xianghong Xu
- Department of Biotherapy Center, Gansu Provincial HospitalLanzhou 730000, Gansu, China
| | - Yixiao Zhao
- College of Food Science and Engineering, Northwest Agriculture & Forestry UniversityYangling 712100, Shaanxi, China
| | - Zhongchao Han
- Institute of Health-Biotech, Health-Biotech (Tianjin) Stem Cell Research Institute Co., Ltd.Tianjin 301701, China
- Jiangxi Research Center of Stem Cell Engineering, Jiangxi Health-Biotech Stem Cell Technology Co., Ltd.Shangrao 334000, Jiangxi, China
- Stem Cell Bank of Guizhou Province, Guizhou Health-Biotech Biotechnology Co., Ltd.Guiyang 550000, Guizhou, China
| | - Tiankang Guo
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province & NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial HospitalLanzhou 730000, Gansu, China
| | - Jun Yao
- School and Hospital of Stomatology, Fujian Medical UniversityFuzhou 350002, Fujian, China
| | - Qing Huang
- Key Laboratory of Radiation Technology and Biophysics, Hefei Institute of Physical Science, Chinese Academy of SciencesHefei 230031, Anhui, China
| |
Collapse
|
28
|
Venglar O, Bago JR, Motais B, Hajek R, Jelinek T. Natural Killer Cells in the Malignant Niche of Multiple Myeloma. Front Immunol 2022; 12:816499. [PMID: 35087536 PMCID: PMC8787055 DOI: 10.3389/fimmu.2021.816499] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells represent a subset of CD3- CD7+ CD56+/dim lymphocytes with cytotoxic and suppressor activity against virus-infected cells and cancer cells. The overall potential of NK cells has brought them to the spotlight of targeted immunotherapy in solid and hematological malignancies, including multiple myeloma (MM). Nonetheless, NK cells are subjected to a variety of cancer defense mechanisms, leading to impaired maturation, chemotaxis, target recognition, and killing. This review aims to summarize the available and most current knowledge about cancer-related impairment of NK cell function occurring in MM.
Collapse
Affiliation(s)
- Ondrej Venglar
- Faculty of Science, University of Ostrava, Ostrava, Czechia.,Faculty of Medicine, University of Ostrava, Ostrava, Czechia.,Hematooncology Clinic, University Hospital Ostrava, Ostrava, Czechia
| | - Julio Rodriguez Bago
- Faculty of Medicine, University of Ostrava, Ostrava, Czechia.,Hematooncology Clinic, University Hospital Ostrava, Ostrava, Czechia
| | - Benjamin Motais
- Faculty of Science, University of Ostrava, Ostrava, Czechia.,Faculty of Medicine, University of Ostrava, Ostrava, Czechia
| | - Roman Hajek
- Faculty of Medicine, University of Ostrava, Ostrava, Czechia.,Hematooncology Clinic, University Hospital Ostrava, Ostrava, Czechia
| | - Tomas Jelinek
- Faculty of Medicine, University of Ostrava, Ostrava, Czechia.,Hematooncology Clinic, University Hospital Ostrava, Ostrava, Czechia
| |
Collapse
|
29
|
Jiang W, Li F, Jiang Y, Li S, Liu X, Xu Y, Li B, Feng X, Zheng C. Tim-3 Blockade Elicits Potent Anti-Multiple Myeloma Immunity of Natural Killer Cells. Front Oncol 2022; 12:739976. [PMID: 35280800 PMCID: PMC8913933 DOI: 10.3389/fonc.2022.739976] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 02/07/2022] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) is still an incurable plasma cell tumor. Natural killer (NK) cells are characterized by efficient anti-tumor activity, and their activity is one basis of cancer immunotherapeutic strategies. Tim-3, one of the immune checkpoint molecules, negatively regulates NK cell activity. To evaluate roles of the Tim-3 pathway blocking in the regulation of NK cell mediated- anti-MM activity in vitro and in vivo, anti-Tim-3 and/or anti-its ligand (HMGB1, CEACAM1 or Galetin-9) antibodies were applied respectively to block the Tim-3 pathway in the present study. Our results showed that Tim-3 was highly expressed on NK cells, in particular on in vitro expanded NK (exNK) cells. NK cells with Tim-3 blockade displayed a significantly higher degranulation and cytolytic activity against both human MM cell lines and primary MM cells, compared to the isotype control antibody-treated NK cells. The increased NK cell cytolytic activity by Tim-3 blocking was associated with up-regulation of cytotoxicity-related molecules, including perforin, granzyme B, TNF-α and IFN-γ. Ligand (HMGB1, CEACAM1 or Galetin-9) expression on MM cells was at different levels, and accordingly, the improvement in NK cell-mediated killing activity by different ligand blocking were also varying. Tim-3 blocking showed much more efficient enhancement of NK cell cytolytic activity than its ligand blockings. More importantly, exNK cells with Tim-3 blockade significantly inhibited MM tumor growth and prolonged the survival of MM-bearing NOD/SCID mice. Our results also showed that NK cells from peripheral blood and bone marrow of MM patients expressed much higher levels of Tim-3 than their counterparts from controls. Taken together, Tim-3 may be an important target molecule used for developing an antibody and/or NK cell based immunotherapeutic strategies for MM.
Collapse
Affiliation(s)
- Wen Jiang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fanglin Li
- Department of Hematology, The Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yang Jiang
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Institute of Biotherapy for Hematological Malignancies, Shandong University, Shandong University-Karolinska College Collaborative Laboratory for Stem Cell Research, Jinan, China
| | - Shengli Li
- Department of Hematology, Jining NO.1 People’s Hospital, Jining, China
| | - Xiaoli Liu
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Institute of Biotherapy for Hematological Malignancies, Shandong University, Shandong University-Karolinska College Collaborative Laboratory for Stem Cell Research, Jinan, China
| | - Yaqi Xu
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Institute of Biotherapy for Hematological Malignancies, Shandong University, Shandong University-Karolinska College Collaborative Laboratory for Stem Cell Research, Jinan, China
| | - Binggen Li
- R&D Department, Weihai Zhengsheng Biotechnology Co., Ltd, Weihai, China
| | - Xiaoli Feng
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Chengyun Zheng, ; Xiaoli Feng,
| | - Chengyun Zheng
- Department of Hematology, The Second Hospital, Cheeloo College of Medicine, Institute of Biotherapy for Hematological Malignancies, Shandong University, Shandong University-Karolinska College Collaborative Laboratory for Stem Cell Research, Jinan, China
- *Correspondence: Chengyun Zheng, ; Xiaoli Feng,
| |
Collapse
|
30
|
Update on lymphocyte-activation gene 3 (LAG-3) in cancers: from biological properties to clinical applications. Chin Med J (Engl) 2022; 135:1203-1212. [PMID: 35170503 PMCID: PMC9337260 DOI: 10.1097/cm9.0000000000001981] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Immunotherapy that targets checkpoints, especially programmed cell death protein 1 and programmed cell death ligand 1, has revolutionized cancer therapy regimens. The overall response rate to mono-immunotherapy, however, is limited, emphasizing the need to potentiate the efficacy of these regimens. The functions of immune cells are modulated by multiple stimulatory and inhibitory molecules, including lymphocyte activation gene 3 (LAG-3). LAG-3 is co-expressed together with other inhibitory checkpoints and plays key roles in immune suppression. Increasing evidence, particularly in the last 5 years, has shown the potential of LAG-3 blockade in anti-tumor immunity. This review provides an update on the biological properties and clinical applications of LAG-3 in cancers.
Collapse
|
31
|
Expression of Immunomodulatory Checkpoint Molecules in Drug-Resistant Neuroblastoma: An Exploratory Study. Cancers (Basel) 2022; 14:cancers14030751. [PMID: 35159017 PMCID: PMC8833944 DOI: 10.3390/cancers14030751] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Neuroblastoma is a common childhood cancer with poor prognosis. Prior studies suggest that inhibition of molecules called checkpoint proteins, which normally prevent one’s own immune system from attacking itself, has been successfully used for treatment of multiple advanced adult cancers but has yet to be fully explored in neuroblastoma. Cancer can hijack these pathways to prevent the immune system from recognizing and destroying cancer cells. We investigated checkpoint protein expression in pediatric neuroblastoma and its role in drug resistance. We created drug-resistant neuroblastoma cell lines and compared expression of checkpoint proteins between drug-resistant and parental cell lines. In total, 13 checkpoint proteins were expressed by all cell lines regardless of drug resistance. Although PD-L1 and checkpoint proteins do not necessarily impart drug resistance, they may be potential targets for drug therapy. Benchmarking checkpoint proteins provides the basis for future studies identifying targets for directed therapy and biomarkers for cancer detection or prognosis. Abstract Neuroblastoma is a common childhood cancer with poor prognosis when at its advanced stage. Checkpoint molecule inhibition is successful in treating multiple advanced adult cancers. We investigated PD-L1 and other checkpoint molecule expression to determine their roles in drug resistance and usefulness as targets for drug therapy. We developed three doxorubicin-resistant (DoxR) cell lines from parental cell lines. Matrigel in vitro invasion assays were used to compare invasiveness. Western blot assays were used to compare PD-L1 expression. Immuno-oncology checkpoint protein panels were used to compare concentrations of 17 checkpoint molecules both cellular and soluble. PD-L1 and 12 other checkpoint molecules were present in all cell lysates of each cell line without significantly different levels. Three were solubilized in the media of each cell line. PD-L1 is expressed in all DoxR and parental neuroblastoma cells and may be a potential target for drug therapy although its role in drug resistance remains unclear. Benchmarking checkpoint molecules provides the basis for future studies identifying targets for directed therapy and biomarkers for cancer detection or prognosis.
Collapse
|
32
|
Liu R, Luo Q, Luo W, Wan L, Zhu Q, Yin X, Lu X, Song Z, Wei L, Xiang Z, Zou Y. A Soluble NK-CAR Mediates the Specific Cytotoxicity of NK Cells toward the Target CD20 + Lymphoma Cells. Aging Dis 2022; 13:1576-1588. [PMID: 36186137 PMCID: PMC9466963 DOI: 10.14336/ad.2022.0415] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/15/2022] [Indexed: 11/01/2022] Open
Affiliation(s)
- Rongjiao Liu
- Department of Immunology, School of Basic Medical of Central South University, Changsha, Hunan, China.
| | - Qizhi Luo
- Department of Immunology, School of Basic Medical of Central South University, Changsha, Hunan, China.
| | - Weiguang Luo
- Department of Laboratory Medicine, Henan Provincial People's Hospital; People’s Hospital of Zhengzhou University, Zhengzhou, China.
| | - Ling Wan
- Department of Immunology, School of Basic Medical of Central South University, Changsha, Hunan, China.
| | - Quan Zhu
- Department of Immunology, School of Basic Medical of Central South University, Changsha, Hunan, China.
| | - Xiangli Yin
- Department of Immunology, School of Basic Medical of Central South University, Changsha, Hunan, China.
| | - Xiaofang Lu
- Department of Immunology, School of Basic Medical of Central South University, Changsha, Hunan, China.
| | - Zixuan Song
- Department of Immunology, School of Basic Medical of Central South University, Changsha, Hunan, China.
| | - Leiyan Wei
- Department of Immunology, School of Basic Medical of Central South University, Changsha, Hunan, China.
| | - Zhiqing Xiang
- Department of Immunology, School of Basic Medical of Central South University, Changsha, Hunan, China.
| | - Yizhou Zou
- Department of Immunology, School of Basic Medical of Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.
- Correspondence should be addressed to: Dr. Yizhou Zou, Department of Immunology, School of Basic Medical of Central South University, Changsha, Hunan, China. .
| |
Collapse
|
33
|
Lin WW, Ho KW, Su HH, Fang TF, Tzou SC, Chen IJ, Lu YC, Chang MS, Tsai YC, Liu ES, Su YC, Wang YT, Cheng TL, Huang HK. Fibrinogen-Like Protein 1 Serves as an Anti-Inflammatory Agent for Collagen-Induced Arthritis Therapy in Mice. Front Immunol 2021; 12:767868. [PMID: 34975855 PMCID: PMC8716738 DOI: 10.3389/fimmu.2021.767868] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/29/2021] [Indexed: 11/18/2022] Open
Abstract
Fibrinogen-like protein 1 (FGL1) was recently identified as a major ligand of lymphocyte-activation gene-3 (LAG-3) on activated T cells and serves as an immune suppressive molecule for regulation of immune homeostasis. However, whether FGL1 has therapeutic potential for use in the T cell-induced the autoimmune disease, rheumatoid arthritis (RA), is still unknown. Here, we attempted to evaluate the effect of FGL1 protein on arthritis progression. We also evaluated potential adverse events in a collagen-induced arthritis (CIA) mouse model. We first confirmed that soluble Fgl1 protein could specifically bind to surface Lag-3 receptor on 3T3-Lag-3 cells and further inhibit interleukin (IL-2) and interferon gamma (IFNγ) secretion from activated primary mouse T cells by 95% and 43%, respectively. Intraperitoneal administration of Fgl1 protein significantly decreased the inflammatory cytokine level (i.e., IL-1β and IL-6) in local paw tissue, and prevented joint inflammation, cellular infiltration, bone deformation and attenuated collagen-induced arthritis progression in vivo. We further demonstrated that exogenous Fgl1 does not cause obvious adverse events during treatment by monitoring body weight and liver weight, and assessing the morphology of several organs (i.e., heart, liver, spleen, lung and kidney) by pathological studies. We expect that Fgl1 protein may be suitable to serve as a potential therapeutic agent for treatment of RA or even other types of T cell-induced autoimmune or inflammatory diseases in the future.
Collapse
Affiliation(s)
- Wen-Wei Lin
- Department of Laboratory Medicine, School of Post Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Laboratory Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- *Correspondence: Wen-Wei Lin, ; Hsin-Kai Huang,
| | - Kai-Wen Ho
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsiang-Han Su
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tien-Fang Fang
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shey-Cherng Tzou
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - I-Ju Chen
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yun-Chi Lu
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Mu-Shen Chang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yun-Chen Tsai
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - En-Shuo Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Cheng Su
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Yen-Tseng Wang
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biochemistry, School of Post Baccalaureate Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tian-Lu Cheng
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsin-Kai Huang
- Department of Medical Laboratory, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
- *Correspondence: Wen-Wei Lin, ; Hsin-Kai Huang,
| |
Collapse
|
34
|
Janakiram M, Arora N, Bachanova V, Miller JS. Novel Cell and Immune Engagers in Optimizing Tumor- Specific Immunity Post-Autologous Transplantation in Multiple Myeloma. Transplant Cell Ther 2021; 28:61-69. [PMID: 34634499 DOI: 10.1016/j.jtct.2021.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 09/07/2021] [Accepted: 10/03/2021] [Indexed: 11/18/2022]
Abstract
Autologous stem cell transplantation (ASCT) is an important component of treatment of multiple myeloma (MM). The post-ASCT setting offers a unique opportunity to increase myeloma specific immunity through enhancement of T and NK cell responses. The vast array of therapeutics being developed for MM, including cell-based therapies, dendritic vaccines, bispecific antibodies, and IL-15 agonists, provide the opportunity to increase tumor-specific immunity. Maintenance therapies, including immunomodulatory drugs, proteasome inhibitors, and daratumumab, exhibit a significant anti-myeloma response by modulating the immune system. Lenalidomide promotes an antitumoral immune microenvironment, whereas daratumumab can potentially cause NK cell fratricide. Thus, understanding the effects of commonly used maintenance drugs on the immune system is important. In this review, we look at current and emerging therapeutics and their integration post-ASCT in the context of immune reconstitution to improve clinical responses in patients with MM. © 2021 American Society for Transplantation and Cellular Therapy. Published by Elsevier Inc.
Collapse
Affiliation(s)
- Murali Janakiram
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.
| | - Nivedita Arora
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Veronika Bachanova
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Jeffrey S Miller
- Blood and Marrow Transplant Program, Department of Medicine, University of Minnesota, Minneapolis, Minnesota; Division of Hematology, Oncology, and Transplantation, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
35
|
Szudy-Szczyrek A, Ahern S, Kozioł M, Majowicz D, Szczyrek M, Krawczyk J, Hus M. Therapeutic Potential of Innate Lymphoid Cells for Multiple Myeloma Therapy. Cancers (Basel) 2021; 13:4806. [PMID: 34638291 PMCID: PMC8507621 DOI: 10.3390/cancers13194806] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 01/08/2023] Open
Abstract
Innate lymphoid cells (ILCs) are a recently identified family of lymphocyte-like cells lacking a specific antigen receptor. They are part of the innate immune system. They play a key role in tissue homeostasis and also control inflammatory and neoplastic processes. In response to environmental stimuli, ILCs change their phenotype and functions, and influence the activity of other cells in the microenvironment. ILC dysfunction can lead to a wide variety of diseases, including cancer. ILC can be divided into three subgroups: ILC Group 1, comprising NK cells and ILC1; Group 2, including ILC2 alone; and Group 3, containing Lymphoid Tissue inducers (LTi) and ILC3 cells. While Group 1 ILCs mainly exert antitumour activity, Group 2 and Group 3 ILCs are protumorigenic in nature. A growing body of preclinical and clinical data support the role of ILCs in the pathogenesis of multiple myeloma (MM). Therefore, targeting ILCs may be of clinical benefit. In this manuscript, we review the available data on the role of ILCs in MM immunology and therapy.
Collapse
Affiliation(s)
- Aneta Szudy-Szczyrek
- Chair and Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland; (M.K.); (D.M.)
| | - Sean Ahern
- Department of Haematology, University Hospital Galway, H91 TK33 Galway, Ireland; (S.A.); (J.K.)
- National University of Ireland, H91 TK33 Galway, Ireland
| | - Magdalena Kozioł
- Chair and Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland; (M.K.); (D.M.)
| | - Daria Majowicz
- Chair and Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland; (M.K.); (D.M.)
| | - Michał Szczyrek
- Chair and Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Janusz Krawczyk
- Department of Haematology, University Hospital Galway, H91 TK33 Galway, Ireland; (S.A.); (J.K.)
- National University of Ireland, H91 TK33 Galway, Ireland
| | - Marek Hus
- Chair and Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland; (M.K.); (D.M.)
| |
Collapse
|
36
|
The EHA Research Roadmap: Immune-based Therapies for Hematological Malignancies. Hemasphere 2021; 5:e642. [PMID: 34522844 PMCID: PMC8432635 DOI: 10.1097/hs9.0000000000000642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 08/16/2021] [Indexed: 11/27/2022] Open
Abstract
In 2016, the European Hematology Association (EHA) published the EHA Roadmap for European Hematology Research1 aiming to highlight achievements in the diagnostics and treatment of blood disorders, and to better inform European policy makers and other stakeholders about the urgent clinical and scientific needs and priorities in the field of hematology. Each section was coordinated by 1-2 section editors who were leading international experts in the field. In the 5 years that have followed, advances in the field of hematology have been plentiful. As such, EHA is pleased to present an updated Research Roadmap, now including 11 sections, each of which will be published separately. The updated EHA Research Roadmap identifies the most urgent priorities in hematology research and clinical science, therefore supporting a more informed, focused, and ideally a more funded future for European Hematology Research. the 11 EHA Research Roadmap sections include normal hematopoiesis; malignant lymphoid diseases; malignant myeloid diseases; anemias and related diseases; platelet disorders; blood coagulation and hemostatic disorders; transfusion medicine; infections in hematology; hematopoietic stem cell transplantation; CAR-T and Other cell-based immune therapies; and gene therapy.
Collapse
|
37
|
Abramson HN. Immunotherapy of Multiple Myeloma: Promise and Challenges. Immunotargets Ther 2021; 10:343-371. [PMID: 34527606 PMCID: PMC8437262 DOI: 10.2147/itt.s306103] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/25/2021] [Indexed: 12/16/2022] Open
Abstract
Whereas the treatment of MM was dependent solely on alkylating agents and corticosteroids during the prior three decades, the landscape of therapeutic measures to treat the disease began to expand enormously early in the current century. The introduction of new classes of small-molecule drugs, such as proteasome blockers (bortezomib and carfilzomib), immunomodulators (lenalidomide and pomalidomide), nuclear export inhibitors (selinexor), and histone deacetylase blockers (panobinostat), as well as the application of autologous stem cell transplantation (ASCT), resulted in a seismic shift in how the disease is treated. The picture changed dramatically once again starting with the 2015 FDA approval of two monoclonal antibodies (mAbs) - the anti-CD38 daratumumab and the anti-SLAMF7 elotuzumab. Daratumumab, in particular, has had a great impact on MM therapy and today is often included in various regimens to treat the disease, both in newly diagnosed cases and in the relapse/refractory setting. Recently, other immunotherapies have been added to the arsenal of drugs available to fight this malignancy. These include isatuximab (also anti-CD38) and, in the past year, the antibody-drug conjugate (ADC) belantamab mafodotin and the chimeric antigen receptor (CAR) T-cell product idecabtagene vicleucel (ide-cel). While the accumulated benefits of these newer agents have resulted in a doubling of the disease's five-year survival rate to more than 5 years and improved quality of life, the disease remains incurable. Almost without exception patients experience relapse and/or become refractory to the drugs used, making the search for innovative therapies all the more essential. This review covers the current scope of anti-myeloma immunotherapeutic agents, both those in clinical use and on the horizon, including naked mAbs, ADCs, bi- and multi-targeted mAbs, and CAR T-cells. Emphasis is placed on the benefits of each along with the challenges that need to be overcome if MM is to be considered curable in the future.
Collapse
Affiliation(s)
- Hanley N Abramson
- Wayne State University, Department of Pharmaceutical Sciences, Detroit, MI, 48201, USA
| |
Collapse
|
38
|
Uckun FM. Overcoming the Immunosuppressive Tumor Microenvironment in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13092018. [PMID: 33922005 PMCID: PMC8122391 DOI: 10.3390/cancers13092018] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/07/2021] [Accepted: 04/20/2021] [Indexed: 02/07/2023] Open
Abstract
Simple Summary This article provides a comprehensive review of new and emerging treatment strategies against multiple myeloma that employ precision medicines and/or drugs capable of improving the ability of the immune system to prevent or slow down the progression of multiple myeloma. These rationally designed new treatment methods have the potential to change the therapeutic landscape in multiple myeloma and improve the long-term survival outcome. Abstract SeverFigurel cellular elements of the bone marrow (BM) microenvironment in multiple myeloma (MM) patients contribute to the immune evasion, proliferation, and drug resistance of MM cells, including myeloid-derived suppressor cells (MDSCs), tumor-associated M2-like, “alternatively activated” macrophages, CD38+ regulatory B-cells (Bregs), and regulatory T-cells (Tregs). These immunosuppressive elements in bidirectional and multi-directional crosstalk with each other inhibit both memory and cytotoxic effector T-cell populations as well as natural killer (NK) cells. Immunomodulatory imide drugs (IMiDs), protease inhibitors (PI), monoclonal antibodies (MoAb), adoptive T-cell/NK cell therapy, and inhibitors of anti-apoptotic signaling pathways have emerged as promising therapeutic platforms that can be employed in various combinations as part of a rationally designed immunomodulatory strategy against an immunosuppressive tumor microenvironment (TME) in MM. These platforms provide the foundation for a new therapeutic paradigm for achieving improved survival of high-risk newly diagnosed as well as relapsed/refractory MM patients. Here we review the scientific rationale and clinical proof of concept for each of these platforms.
Collapse
Affiliation(s)
- Fatih M. Uckun
- Norris Comprehensive Cancer Center and Childrens Center for Cancer and Blood Diseases, University of Southern California Keck School of Medicine (USC KSOM), Los Angeles, CA 90027, USA;
- Department of Developmental Therapeutics, Immunology, and Integrative Medicine, Drug Discovery Institute, Ares Pharmaceuticals, St. Paul, MN 55110, USA
- Reven Pharmaceuticals, Translational Oncology Program, Golden, CO 80401, USA
| |
Collapse
|
39
|
Rubio MT, Dhuyser A, Nguyen S. Role and Modulation of NK Cells in Multiple Myeloma. HEMATO 2021. [DOI: 10.3390/hemato2020010] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
Abstract
Myeloma tumor cells are particularly dependent on their microenvironment and sensitive to cellular antitumor immune response, including natural killer (NK) cells. These later are essential innate lymphocytes implicated in the control of viral infections and cancers. Their cytotoxic activity is regulated by a balance between activating and inhibitory signals resulting from the complex interaction of surface receptors and their respective ligands. Myeloma disease evolution is associated with a progressive alteration of NK cell number, phenotype and cytotoxic functions. We review here the different therapeutic approaches that could restore or enhance NK cell functions in multiple myeloma. First, conventional treatments (immunomodulatory drugs-IMids and proteasome inhibitors) can enhance NK killing of tumor cells by modulating the expression of NK receptors and their corresponding ligands on NK and myeloma cells, respectively. Because of their ability to kill by antibody-dependent cell cytotoxicity, NK cells are important effectors involved in the efficacy of anti-myeloma monoclonal antibodies targeting the tumor antigens CD38, CS1 or BCMA. These complementary mechanisms support the more recent therapeutic combination of IMids or proteasome inhibitors to monoclonal antibodies. We finally discuss the ongoing development of new NK cell-based immunotherapies, such as ex vivo expanded killer cell immunoglobulin-like receptors (KIR)-mismatched NK cells, chimeric antigen receptors (CAR)-NK cells, check point and KIR inhibitors.
Collapse
|