1
|
Zhang ZF, Zhang Y, Chen YW, Cao GS, Zheng XD, Sun R, Peng H, Tian ZG, Sun HY. CD200R blockade enhances anti-tumor immunity by unleashing NK and CD8 + T cells in tumor. Acta Pharmacol Sin 2025:10.1038/s41401-025-01556-0. [PMID: 40329005 DOI: 10.1038/s41401-025-01556-0] [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: 11/09/2024] [Accepted: 03/30/2025] [Indexed: 05/08/2025]
Abstract
Immune checkpoint inhibitors have revolutionized cancer therapy, but a large proportion of patients do not respond well to current checkpoint immunotherapies. CD200R (also known as OX2R) is a transmembrane glycoprotein of the immunoglobulin superfamily that is mainly expressed on myeloid and lymphoid-derived immunocompetent cells such as myeloid cells, natural killer (NK), and CD8+ T cells. In this study, we investigated the therapeutic potential and cellular mechanisms of targeting CD200R in tumor immunotherapy. We established 4 subcutaneous tumor mouse models using MC38 (colon cancer), MCA205 (fibrosarcoma), LLC (lung cancer), and EO771 (mammary cancer) cell lines. We found that CD200R was highly expressed on tumor-infiltrating NK and CD8+ T cells with exhausted phenotypes in the four subcutaneous tumor mouse models. Either genetic ablation or antibody blockade of CD200R retarded tumor growth and prolonged the survival of tumor-bearing mice by preventing or reversing exhaustion of both NK cells and CD8+ T cells. The combined therapy of CD200R antibody with anti-PD-1/anti-PD-L1 synergistically inhibited tumor growth. By depletion of NK or/and CD8+ T cells, we demonstrated that both cell types contributed to the anti-tumor efficacy of CD200R blockade in tumor-bearing mice. Further, the blockade of human CD200R significantly enhanced human NK cell function and inhibited human tumor growth in PBMC-reconstituted xenograft mice. Our results demonstrate that CD200R is a potential immune checkpoint molecule that can suppress the tumoricidal activities of NK and CD8+ T cells, and could thus be exploited as a therapeutic target in the future.
Collapse
Affiliation(s)
- Zheng-Feng Zhang
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Yu Zhang
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Ya-Wen Chen
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Guo-Shuai Cao
- Hefei TG ImmunoPharma Corporation Limited, Hefei, 230027, China
| | - Xiao-Dong Zheng
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Rui Sun
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hui Peng
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Zhi-Gang Tian
- National Key Laboratory of Immune Response and Immunotherapy, The Institute of Immunology, Biomedical Sciences and Health Laboratory of Anhui Province, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Hao-Yu Sun
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
González-Guerrero L, Castellet H, Martínez C, González N, Guijarro F, Lloveras N, Pratcorona M, Gich I, Berenguer-Molins P, Perera-Bel J, Zamora L, Mascaró M, Sampol A, Garcia-Guiñón A, Vives S, Tormo M, Arnan M, Villamor N, Nomdedéu JF. CD200 in acute myeloid leukemia: marked upregulation in CEBPA biallelic mutated cases. Diagn Pathol 2025; 20:56. [PMID: 40307896 PMCID: PMC12042300 DOI: 10.1186/s13000-025-01655-w] [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: 10/22/2024] [Accepted: 04/20/2025] [Indexed: 05/02/2025] Open
Abstract
CD200 is a glycoprotein that binds with its receptor CD200R, providing immunosuppressive signals to T and NK cells. CD200 is expressed by normal stem cells and progenitors committed to B-lymphopoiesis and myeloid development. CD200 biological relevance in acute leukemias is only partially understood.The study included a consecutive series of four hundred thirty-one patients with acute myeloid leukemia (AML). Immunophenotype was established by multiparametric flow cytometry, and the genetic diagnosis was performed by PCR-based methods and a targeted resequencing method covering 42 genes.66% of AML patients expressed CD200 being significantly associated with CD34 reactivity. The frequency of CD200 positivity was higher in cases with core-binding factor genetic lesions such as RUNX1-RUNX1T1 (81.3%) fusions and CBFB-MHY11 (63.2%) rearrangements and also with biallelic CEBPA mutations (100%). The molecular AML group with the lowest CD200 reactivity (19.1%) corresponded to AML with NPM1 mutations. RNA seq showed no uniform pattern of infiltrating cells in CEBPA mutated AML. Deconvolution analysis may be used to assess the immunoregulatory mechanisms of AML.CD200 expression could help identify the more immature compartment and, combined with other markers, single out CEPA-mutated AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/immunology
- Nucleophosmin
- Male
- Female
- Middle Aged
- Adult
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Mutation
- CCAAT-Enhancer-Binding Proteins/genetics
- Aged
- Up-Regulation
- Young Adult
- Adolescent
- Immunophenotyping
- Biomarkers, Tumor/genetics
- Aged, 80 and over
Collapse
Affiliation(s)
- Laura González-Guerrero
- Department of Hematology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona. IIB Sant Pau, Institut Josep Carreras, Sant Quintí, 89, Barcelona, 08041, Spain
| | - Helena Castellet
- Department of Hematology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona. IIB Sant Pau, Institut Josep Carreras, Sant Quintí, 89, Barcelona, 08041, Spain
| | - Clara Martínez
- Department of Hematology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona. IIB Sant Pau, Institut Josep Carreras, Sant Quintí, 89, Barcelona, 08041, Spain
| | - Nuria González
- Department of Hematology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona. IIB Sant Pau, Institut Josep Carreras, Sant Quintí, 89, Barcelona, 08041, Spain
| | - Francesca Guijarro
- Unitat d'Hematopatologia, Servei d'Anatomia Patológica, Hospital Clínic. IDIBAPS, Barcelona, Spain
| | - Natalia Lloveras
- Laboratori Hematologia. Hospital Dr. Josep Trueta. ICO Girona, Girona, Spain
| | - Marta Pratcorona
- Department of Hematology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona. IIB Sant Pau, Institut Josep Carreras, Sant Quintí, 89, Barcelona, 08041, Spain
| | - Ignasi Gich
- Epidemiology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Pau Berenguer-Molins
- Bioniformatics Unit (BU), MARData, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Júlia Perera-Bel
- Bioniformatics Unit (BU), MARData, Hospital del Mar Research Institute (HMRIB), Barcelona, Spain
| | - Lurdes Zamora
- Hematology Department, Hospital Germans Trias i Pujol. ICO Badalona. Institut Josep Carreras, Badalona, Spain
| | - Martí Mascaró
- Hematology Department, Hospital de Son Llàtzer, Palma de Mallorca, Spain
| | - Antonia Sampol
- Hematology Department, Hospital de Son Espases, Palma de Mallorca, Spain
| | | | - Susana Vives
- Hematology Department, Hospital Germans Trias i Pujol. ICO Badalona. Institut Josep Carreras, Badalona, Spain
| | - Mar Tormo
- Laboratorio de Hematología. Hospital Clínico de Valencia, Universidad de Valencia, Valencia, Spain
| | - Montserrat Arnan
- Clinical Hematology Department, ICO-Hospital Duran i Reynals. Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Neus Villamor
- Unitat d'Hematopatologia, Servei d'Anatomia Patológica, Hospital Clínic. IDIBAPS, Barcelona, Spain
| | - Josep F Nomdedéu
- Department of Hematology, Hospital de la Santa Creu i Sant Pau. Universitat Autònoma de Barcelona. IIB Sant Pau, Institut Josep Carreras, Sant Quintí, 89, Barcelona, 08041, Spain.
| |
Collapse
|
3
|
Guo Y, Liu Z, Tian M, Liu X, Li N, Ding K, Liu H, Fu R. Monocytes from patients with myelodysplastic syndrome inhibit natural killer cell-mediated antitumor function through the CD200/CD200R pathway. Int Immunopharmacol 2025; 152:114394. [PMID: 40049088 DOI: 10.1016/j.intimp.2025.114394] [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: 11/28/2024] [Revised: 02/26/2025] [Accepted: 02/27/2025] [Indexed: 03/24/2025]
Abstract
BACKGROUND Reports on the expression of CD200 in monocytes are scarce, and the role of monocytes in patients with myelodysplastic syndrome (MDS) remains unclear. Additionally, monocytes have been implicated in suppressing NK cell function. Therefore, this study aimed to explore the possible mechanism by which monocytes regulate NK cell function through CD200 in patients with MDS. METHODS We collected samples from patients with MDS, those with acute myeloid leukemia, and healthy controls. We detected the expression of CD200 on the surface of monocytes and its receptor CD200R on the surface of NK cells using flow cytometry, explored the effect of the CD200/CD200R pathway on activating STAT3 and ERK of NK cells, and studied the effect of blocking CD200/CD200R pathway on NK cells. RESULTS The expression of CD200 on the surface of monocytes and CD200R on the surface of NK cells in patients with MDS was higher than those in healthy controls. After adding CD200 monoclonal antibody to the co-culture system of monocytes and NK cells, the expression of activated receptors CD107a, CD226, and NKG2D on NK cells significantly increased. We then used siRNA to silence CD200R expression in NK-92 cells and found that the blockade of CD200R enhanced the phosphorylation levels of ERK and STAT3. CONCLUSIONS Our study found that elevated CD200 expression on monocytes in patients with MDS correlates with poor prognosis, suggesting CD200 as a potential prognostic marker. Blocking CD200 enhances NK cell activation and cytotoxicity, indicating that CD200 blockade therapy could enhance antitumor responses in patients with MDS.
Collapse
Affiliation(s)
- Yixuan Guo
- 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, Tianjin 300052, PR China; Tianjin Institute of Hematology, Tianjin 300052, PR China
| | - 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, Tianjin 300052, PR China; Tianjin Institute of Hematology, Tianjin 300052, PR China.
| | - Mengyue Tian
- 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, Tianjin 300052, PR China; Tianjin Institute of Hematology, Tianjin 300052, PR China
| | - Xiaohan 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, Tianjin 300052, PR China; Tianjin Institute of Hematology, Tianjin 300052, PR China
| | - Nianbin Li
- 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, Tianjin 300052, PR China; Tianjin Institute of Hematology, 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, Tianjin 300052, PR China; Tianjin Institute of Hematology, 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, Tianjin 300052, PR China; Tianjin Institute of Hematology, 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, Tianjin 300052, PR China; Tianjin Institute of Hematology, Tianjin 300052, PR China.
| |
Collapse
|
4
|
Wang Y, Yang X, Liu Y, Li Y. A review of common immunotherapy and nano immunotherapy for acute myeloid leukemia. Front Immunol 2025; 16:1505247. [PMID: 40129984 PMCID: PMC11931025 DOI: 10.3389/fimmu.2025.1505247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025] Open
Abstract
Acute myeloid leukemia (AML) is a highly aggressive hematological malignancy. Traditional chemotherapy methods not only bring serious side effects, but also lead to high recurrence rate and drug resistance in some patients. However, as an emerging therapeutic strategy, immunotherapy has shown great potential in the field of AML treatment in recent years. At present, common immunotherapy methods for AML include monoclonal antibodies, CAR-T cell therapy, and immune checkpoint inhibitors. With the deepening of research and technological progress, especially the application of nanotechnology in medicine, new immunotherapy is expected to become one of the important means for the treatment of acute myeloid leukemia in the future.
Collapse
Affiliation(s)
- Yaoyao Wang
- Department of Pediatrics of Yantai Affiliated Hospital, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, China
| | - Xiancong Yang
- Laboratory Department, Qilu Hospital of ShanDong University Dezhou Hospital, Dezhou, Shandong, China
| | - Yalin Liu
- Department of Pediatrics of Yantai Affiliated Hospital, The Second Clinical Medical College of Binzhou Medical University, Yantai, Shandong, China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
5
|
Zhang M, Yang Y, Liu J, Guo L, Guo Q, Liu W. Bone marrow immune cells and drug resistance in acute myeloid leukemia. Exp Biol Med (Maywood) 2025; 250:10235. [PMID: 40008144 PMCID: PMC11851207 DOI: 10.3389/ebm.2025.10235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
In recent years, the relationship between the immunosuppressive niche of the bone marrow and therapy resistance in acute myeloid leukemia (AML) has become a research focus. The abnormal number and function of immunosuppressive cells, including regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs), along with the dysfunction and exhaustion of immunological effector cells, including cytotoxic T lymphocytes (CTLs), dendritic cells (DCs) and natural killer cells (NKs), can induce immune escape of leukemia cells and are closely linked to therapy resistance in leukemia. This article reviews the research progress on the relationship between immune cells in the marrow microenvironment and chemoresistance in AML, aiming to provide new ideas for the immunotherapy of AML.
Collapse
Affiliation(s)
- Miao Zhang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - You Yang
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Jing Liu
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Ling Guo
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Qulian Guo
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Wenjun Liu
- Department of Pediatrics (Hematological Oncology), Children Hematological Oncology and Birth Defects Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- Sichuan Clinical Research Center for Birth Defects, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
6
|
Rady M, Mostafa M, Dida G, Sabet F, Abou-Aisha K, Watzl C. Adoptive NK cell therapy in AML: progress and challenges. Clin Exp Med 2025; 25:41. [PMID: 39820676 PMCID: PMC11748472 DOI: 10.1007/s10238-025-01559-5] [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: 11/12/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Adoptive cell therapy (ACT) using natural killer (NK) cells has emerged as a promising therapeutic strategy for acute myeloid leukemia (AML), addressing challenges such as chemotherapy resistance and high relapse rates. Over the years, clinical trials and studies have explored various sources of NK cells, including ex vivo expanded NK cell lines, CAR-NK cells, peripheral blood-derived NK cells, and umbilical cord blood-derived NK cells. These therapies have demonstrated varying degrees of therapeutic efficacy, ranging from transient anti-leukemia activity to sustained remission in select patient groups. Toxicity profiles have generally shown favorable safety outcomes, with minimal incidence of severe adverse effects such as cytokine release syndrome (CRS) or graft-versus-host disease (GVHD). However, persistent challenges remain, including limited NK cell persistence, relapse, and heterogeneity in patient responses. This review provides a comprehensive analysis of clinical outcomes and toxicity profiles provided from clinical trials, clinical studies and case reports conducted in the last 15 years to judge on the efficacy, safety and applicability of using NK cells for ACT of AML. Our review highlights the significant potential of NK cell-based therapies for AML, while addressing the technical and biological challenges that must be overcome to enhance their efficacy and safety.
Collapse
Affiliation(s)
- Mona Rady
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo, Egypt.
- Faculty of Biotechnology, German International University, New Administrative Capital, Egypt.
| | - Maha Mostafa
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo, Egypt
| | - Gabriel Dida
- University of South Wales, Pontypridd, Wales, UK
- Department of Health Systems Management and Public Health, Technical University of Kenya, Nairobi, Kenya
| | - Fatima Sabet
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo, Egypt
| | - Khaled Abou-Aisha
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC), New Cairo, Egypt
| | - Carsten Watzl
- Immunology Department, Leibniz Research Center for Working Environment and Human Factors at TU Dortmund (IfADo), Dortmund, Germany
| |
Collapse
|
7
|
Baek S, Cui K. Targeting CD200 in Breast Cancer: Opportunities and Challenges in Immunotherapeutic Strategies. Int J Mol Sci 2024; 26:115. [PMID: 39795972 PMCID: PMC11719565 DOI: 10.3390/ijms26010115] [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: 09/24/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
One of the key factors that contribute to tumor progression and resistance is the immunosuppressive microenvironment of the tumor. CD200 is a recently identified cell surface glycoprotein recognized as an important molecule in breast cancer for its versatile modulation of the immune response via its receptor, CD200R. The interaction between CD200 and CD200R suppresses the immune activities against tumor cells and allows them to be undetected and, in doing so, to escape from the destructive capability of the immune cells. Here, we review recent advances and future trends in CD200-targeted therapies for cancer treatments. We also discuss molecular pathways that include variable expressions across different cancer types and their importance in treatment options.
Collapse
Affiliation(s)
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
8
|
Zhu C, Feng X, Tong L, Mu P, Wang F, Quan W, Dong Y, Zhu X. Prediction of acute myeloid leukemia prognosis based on autophagy features and characterization of its immune microenvironment. Front Immunol 2024; 15:1489171. [PMID: 39650664 PMCID: PMC11621098 DOI: 10.3389/fimmu.2024.1489171] [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: 08/31/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Background Autophagy promotes the survival of acute myeloid leukemia (AML) cells by removing damaged organelles and proteins and protecting them from stress-induced apoptosis. Although many studies have identified candidate autophagy genes associated with AML prognosis, there are still great challenges in predicting the survival prognosis of AML patients. Therefore, it is necessary to identify more novel autophagy gene markers to improve the prognosis of AML by utilizing information at the molecular level. Methods In this study, the Random Forest, SVM and XGBoost algorithms were utilized to identify autophagy genes linked to prognosis, respectively. Subsequently, six autophagy genes (TSC2, CALCOCO2, BAG3, UBQLN4, ULK1 and DAPK1) that were significantly associated with patients' overall survival (OS) were identified using Lasso-Cox regression analysis. A prediction model incorporating these autophagy genes was then developed. In addition, the immunological microenvironment analysis of autophagy genes was performed in this study. Results The experimental results showed that the predictive model had good predictive ability. After adjusting for clinicopathologic parameters, this feature proved an independent prognostic predictor and was validated in an external AML sample set. Analysis of differentially expressed genes in patients in the high-risk and low-risk groups showed that these genes were enriched in immune-related pathways such as humoral immune response, T cell differentiation in thymus and lymphocyte differentiation. Then immune infiltration analysis of autophagy genes in patients showed that the cellular abundance of T cells CD4+ memory activated, NK cells activated and T cells CD4+ in the high-risk group was significantly lower than that in the low-risk group. Conclusion This study systematically analyzed autophagy-related genes (ARGs) and developed prognostic predictors related to OS for patients with AML, thus more accurately assessing the prognosis of AML patients. This not only helps to improve the prognostic assessment and therapeutic outcome of patients, but may also provide new help for future research and clinical applications.
Collapse
Affiliation(s)
- Chaoqun Zhu
- School of Computer and Control Engineering, Yantai University, Yantai, Shandong, China
| | - Xiangyan Feng
- Department of Hematology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, Shandong, China
| | - Lanxin Tong
- Guangzhou Dublin International College of Life Sciences and Technology, South China Agricultural University, Guangzhou, Guangdong, China
| | - Peizheng Mu
- School of Computer and Control Engineering, Yantai University, Yantai, Shandong, China
| | - Fei Wang
- School of Computer and Control Engineering, Yantai University, Yantai, Shandong, China
| | - Wei Quan
- School of Computer and Control Engineering, Yantai University, Yantai, Shandong, China
| | - Yucui Dong
- Department of Immunology, Binzhou Medical University, Yantai, Shandong, China
| | - Xiao Zhu
- School of Computer and Control Engineering, Yantai University, Yantai, Shandong, China
| |
Collapse
|
9
|
Dunbar ZT, González-Ochoa S, Kanagasabai T, Ivanova A, Shanker A. Differential Effector Function of Tissue-Specific Natural Killer Cells against Lung Tumors. J Innate Immun 2024; 16:573-594. [PMID: 39561728 PMCID: PMC11644122 DOI: 10.1159/000542078] [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: 06/03/2024] [Accepted: 10/10/2024] [Indexed: 11/21/2024] Open
Abstract
INTRODUCTION Natural killer (NK) cells are innate lymphoid cells capable of directly killing target cells while modulating immune effector responses. Despite their multifunctional capacities, a limited understanding of their plasticity and heterogeneity has impeded progress in developing effective NK cell-based cancer therapies. In this study, we investigated NK cell tissue heterogeneity in relation to their phenotype and effector functions against lung tumors. METHODS Using hanging drop tumor spheroid and subcutaneously established LL/2 (LLC1) lung tumor models, we examined NK cell receptor diversity and its correlation with tissue-specific cytotoxicity through multiparametric flow cytometry, fluorescence imaging, and cytotoxicity assays. RESULTS We identified distinct patterns of cell surface receptors expression on tissue-specific NK cells that are crucial for antitumor activity. Linear regression mathematical analyses further revealed significant positive correlations between activation-associated cell surface receptors and cytotoxic capacity in NK cells from tissues such as the liver and bone marrow. CONCLUSION These findings underscore the differential effector capacities of NK cells from distinct tissues, even prior to exposure to LL/2 tumor cells. This highlights the significance of tissue-specific NK cell heterogeneity and its impact on their antitumor cytotoxicity. Recognizing these distinct tissue-specific receptor expression patterns will be instrumental in developing more efficacious NK cell-based cancer treatments.
Collapse
Affiliation(s)
- Zerick Terrell Dunbar
- Department of Microbiology, Immunology and Physiology, School of Medicine, Meharry Medical College, Nashville, TN, USA
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN, USA
| | - Salvador González-Ochoa
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Thanigaivelan Kanagasabai
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Alla Ivanova
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
| | - Anil Shanker
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN, USA
- Host-Tumor Interactions Research Program, Vanderbilt-Ingram Comprehensive Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
10
|
Raphael I, Mujeeb AA, Ampudia-Mesias E, Sever RE, McClellan B, Frederico SC, Sneiderman CT, Mirji A, Daba A, Puerta-Martinez F, Nisnboym M, Edwards WB, Graner M, Moertel CL, Castro MG, Kohanbash G, Olin MR. CD200 depletion in glioma enhances antitumor immunity and induces tumor rejection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.08.611922. [PMID: 39314283 PMCID: PMC11418997 DOI: 10.1101/2024.09.08.611922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
High-grade gliomas are a major health challenge with poor prognosis and high morbidity. Immune-checkpoint inhibitors (ICI) have emerged as promising therapeutic options for several malignancies yet show little efficacy against central nervous system (CNS) tumors. CD200 is a newly recognized immune checkpoint that modulates immune homeostasis. CD200 protein is expressed by a variety of cells, including immune cells and stromal cells, and is overexpressed by many tumors. The shedding of CD200 from tumor cells can create an immunosuppressive environment that dampens anti-tumor immunity by modulating cytolytic activity and cytokine expression both within and outside the tumor microenvironment (TME). While it is well-accepted that CD200 induces a pro-tumorigenic environment through its ability to suppress the immune response, we sought to determine the role of glioma-specific expression of CD200. We show that CD200 is expressed across glioma types, is shed from tumor cells, and increases over time in the serum of patients undergoing immunotherapy. Using CD200 knockout (KO) glioma models, we demonstrated that glioma cell-derived CD200 promotes tumor growth in vivo and in vitro. Notably, CD200 KO gliomas are spontaneously rejected by their host, a process that required a fully functional immune system, including NK and T-cells. Moreover, we report that glioma-derived or brain-injected soluble CD200 contributes to the suppression of antigen-specific CD8 T-cells in the draining lymph nodes (dLNs). Our work provides new mechanistic insights regarding CD200-mediated immunosuppression by gliomas. Statement of significance We demonstrate mechanisms of the druggable glioma-derived CD200 checkpoint on tumor growth and immune suppression.
Collapse
|
11
|
Sun W, Hu S, Wang X. Advances and clinical applications of immune checkpoint inhibitors in hematological malignancies. Cancer Commun (Lond) 2024; 44:1071-1097. [PMID: 39073258 PMCID: PMC11492363 DOI: 10.1002/cac2.12587] [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: 11/30/2023] [Revised: 06/09/2024] [Accepted: 06/25/2024] [Indexed: 07/30/2024] Open
Abstract
Immune checkpoints are differentially expressed on various immune cells to regulate immune responses in tumor microenvironment. Tumor cells can activate the immune checkpoint pathway to establish an immunosuppressive tumor microenvironment and inhibit the anti-tumor immune response, which may lead to tumor progression by evading immune surveillance. Interrupting co-inhibitory signaling pathways with immune checkpoint inhibitors (ICIs) could reinvigorate the anti-tumor immune response and promote immune-mediated eradication of tumor cells. As a milestone in tumor treatment, ICIs have been firstly used in solid tumors and subsequently expanded to hematological malignancies, which are in their infancy. Currently, immune checkpoints have been investigated as promising biomarkers and therapeutic targets in hematological malignancies, and novel immune checkpoints, such as signal regulatory protein α (SIRPα) and tumor necrosis factor-alpha-inducible protein 8-like 2 (TIPE2), are constantly being discovered. Numerous ICIs have received clinical approval for clinical application in the treatment of hematological malignancies, especially when used in combination with other strategies, including oncolytic viruses (OVs), neoantigen vaccines, bispecific antibodies (bsAb), bio-nanomaterials, tumor vaccines, and cytokine-induced killer (CIK) cells. Moreover, the proportion of individuals with hematological malignancies benefiting from ICIs remains lower than expected due to multiple mechanisms of drug resistance and immune-related adverse events (irAEs). Close monitoring and appropriate intervention are needed to mitigate irAEs while using ICIs. This review provided a comprehensive overview of immune checkpoints on different immune cells, the latest advances of ICIs and highlighted the clinical applications of immune checkpoints in hematological malignancies, including biomarkers, targets, combination of ICIs with other therapies, mechanisms of resistance to ICIs, and irAEs, which can provide novel insight into the future exploration of ICIs in tumor treatment.
Collapse
Affiliation(s)
- Wenyue Sun
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
| | - Shunfeng Hu
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
| | - Xin Wang
- Department of HematologyShandong Provincial HospitalShandong UniversityJinanShandongP. R. China
- Department of HematologyShandong Provincial Hospital Affiliated to Shandong First Medical UniversityJinanShandongP. R. China
- Taishan Scholars Program of Shandong ProvinceJinanShandongP. R. China
- Branch of National Clinical Research Center for Hematologic DiseasesJinanShandongP. R. China
- National Clinical Research Center for Hematologic Diseasesthe First Affiliated Hospital of Soochow UniversitySuzhouJiangsuP. R. China
| |
Collapse
|
12
|
Chandra DJ, Alber B, Saultz JN. The Immune Resistance Signature of Acute Myeloid Leukemia and Current Immunotherapy Strategies. Cancers (Basel) 2024; 16:2615. [PMID: 39123343 PMCID: PMC11311077 DOI: 10.3390/cancers16152615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/29/2024] [Accepted: 07/11/2024] [Indexed: 08/12/2024] Open
Abstract
Acute myeloid leukemia (AML) is a complex hematopoietic clonal disorder with limited curative options beyond stem cell transplantation. The success of transplant is intimately linked with the graft versus leukemia effect from the alloreactive donor immune cells including, T and NK cells. The immune system plays a dynamic role in leukemia survival and resistance. Despite our growing understanding of the immune microenvironment, responses to immune-based therapies differ greatly between patients. Herein, we review the biology of immune evasion mechanisms in AML, discuss the current landscape of immunotherapeutic strategies, and discuss the implications of therapeutic targets. This review focuses on T and NK cell-based therapy, including modified and non-modified NK cells, CAR-T and CAR-NK cells, antibodies, and checkpoint blockades. Understanding the complex interchange between immune tolerance and the emergence of tumor resistance will improve patient outcomes.
Collapse
Affiliation(s)
- Daniel J. Chandra
- Division of Hematology/Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA;
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA;
| | - Bernhard Alber
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA;
| | - Jennifer N. Saultz
- Division of Hematology/Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA;
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA;
| |
Collapse
|
13
|
Kaito Y, Imai Y. Evolution of natural killer cell-targeted therapy for acute myeloid leukemia. Int J Hematol 2024; 120:34-43. [PMID: 38693419 DOI: 10.1007/s12185-024-03778-0] [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: 03/15/2024] [Revised: 04/04/2024] [Accepted: 04/14/2024] [Indexed: 05/03/2024]
Abstract
In hematologic oncology, acute myeloid leukemia (AML) presents a significant challenge due to its complex genetic landscape and resistance to conventional therapies. Despite advances in treatment, including intensive chemotherapy and hematopoietic stem cell transplantation (HSCT), the prognosis for many patients with AML remains poor. Recently, immunotherapy has emerged as a promising approach to improve outcomes by augmenting existing treatments. Natural killer (NK) cells, a subset of innate lymphoid cells, have garnered attention for their potent cytotoxic capabilities against AML cells. In this review, we discuss the role of NK cells in AML immunosurveillance, their dysregulation in patients with AML, and various therapeutic strategies leveraging NK cells in AML treatment. We explore the challenges and prospects associated with NK cell therapy, including approaches to enhance NK cell function, overcome immune evasion mechanisms, and optimize treatment efficacy. Finally, we emphasize the importance of further research to validate and refine patient-first NK cell-based immunotherapies for AML.
Collapse
Affiliation(s)
- Yuta Kaito
- Department of Hematology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-Ku, Tokyo, 113-8602, Japan.
| | - Yoichi Imai
- Department of Hematology and Oncology, Dokkyo Medical University, Tochigi, Japan
| |
Collapse
|
14
|
Bode K, Wei S, Gruber I, Li J, Kissler S, Yi P. Beta cells deficient for Renalase counteract autoimmunity by shaping natural killer cell activity. Front Immunol 2024; 15:1403752. [PMID: 38975343 PMCID: PMC11225407 DOI: 10.3389/fimmu.2024.1403752] [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: 03/19/2024] [Accepted: 05/14/2024] [Indexed: 07/09/2024] Open
Abstract
Type 1 diabetes (T1D) arises from autoimmune-mediated destruction of insulin-producing pancreatic beta cells. Recent advancements in the technology of generating pancreatic beta cells from human pluripotent stem cells (SC-beta cells) have facilitated the exploration of cell replacement therapies for treating T1D. However, the persistent threat of autoimmunity poses a significant challenge to the survival of transplanted SC-beta cells. Genetic engineering is a promising approach to enhance immune resistance of beta cells as we previously showed by inactivating the Renalase (Rnls) gene. Here, we demonstrate that Rnls loss of function in beta cells shapes autoimmunity by mediating a regulatory natural killer (NK) cell phenotype important for the induction of tolerogenic antigen-presenting cells. Rnls-deficient beta cells mediate cell-cell contact-independent induction of hallmark anti-inflammatory cytokine Tgfβ1 in NK cells. In addition, surface expression of regulatory NK immune checkpoints CD47 and Ceacam1 is markedly elevated on beta cells deficient for Rnls. Altered glucose metabolism in Rnls mutant beta cells is involved in the upregulation of CD47 surface expression. These findings are crucial to better understand how genetically engineered beta cells shape autoimmunity, giving valuable insights for future therapeutic advancements to treat and cure T1D.
Collapse
Affiliation(s)
- Kevin Bode
- Section for Immunobiology, Joslin Diabetes Center, Boston, MA, United States
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Siying Wei
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Isabella Gruber
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, United States
| | - Jian Li
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Stephan Kissler
- Section for Immunobiology, Joslin Diabetes Center, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA, United States
| | - Peng Yi
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA, United States
- Department of Medicine, Harvard Medical School, Boston, MA, United States
- Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA, United States
| |
Collapse
|
15
|
Perzolli A, Koedijk JB, Zwaan CM, Heidenreich O. Targeting the innate immune system in pediatric and adult AML. Leukemia 2024; 38:1191-1201. [PMID: 38459166 PMCID: PMC11147779 DOI: 10.1038/s41375-024-02217-7] [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: 01/29/2024] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 03/10/2024]
Abstract
While the introduction of T cell-based immunotherapies has improved outcomes in many cancer types, the development of immunotherapies for both adult and pediatric AML has been relatively slow and limited. In addition to the need to identify suitable target antigens, a better understanding of the immunosuppressive tumor microenvironment is necessary for the design of novel immunotherapy approaches. To date, most immune characterization studies in AML have focused on T cells, while innate immune lineages such as monocytes, granulocytes and natural killer (NK) cells, received less attention. In solid cancers, studies have shown that innate immune cells, such as macrophages, myeloid-derived suppressor cells and neutrophils are highly plastic and may differentiate into immunosuppressive cells depending on signals received in their microenvironment, while NK cells appear to be functionally impaired. Hence, an in-depth characterization of the innate immune compartment in the TME is urgently needed to guide the development of immunotherapeutic interventions for AML. In this review, we summarize the current knowledge on the innate immune compartment in AML, and we discuss how targeting its components may enhance T cell-based- and other immunotherapeutic approaches.
Collapse
Affiliation(s)
- Alicia Perzolli
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - Joost B Koedijk
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - C Michel Zwaan
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
- Department of Pediatric Oncology, Erasmus MC/Sophia Children's Hospital, 3015 GD, Rotterdam, The Netherlands
| | - Olaf Heidenreich
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands.
- Wolfson Childhood Cancer Research Centre, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK.
| |
Collapse
|
16
|
Sayitoglu EC, Luca BA, Boss AP, Thomas BC, Freeborn RA, Uyeda MJ, Chen PP, Nakauchi Y, Waichler C, Lacayo N, Bacchetta R, Majeti R, Gentles AJ, Cepika AM, Roncarolo MG. AML/T cell interactomics uncover correlates of patient outcomes and the key role of ICAM1 in T cell killing of AML. Leukemia 2024; 38:1246-1255. [PMID: 38724673 PMCID: PMC11147760 DOI: 10.1038/s41375-024-02255-1] [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: 10/11/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 05/21/2024]
Abstract
T cells are important for the control of acute myeloid leukemia (AML), a common and often deadly malignancy. We observed that some AML patient samples are resistant to killing by human-engineered cytotoxic CD4+ T cells. Single-cell RNA-seq of primary AML samples and CD4+ T cells before and after their interaction uncovered transcriptional programs that correlate with AML sensitivity or resistance to CD4+ T cell killing. Resistance-associated AML programs were enriched in AML patients with poor survival, and killing-resistant AML cells did not engage T cells in vitro. Killing-sensitive AML potently activated T cells before being killed, and upregulated ICAM1, a key component of the immune synapse with T cells. Without ICAM1, killing-sensitive AML became resistant to killing by primary ex vivo-isolated CD8+ T cells in vitro, and engineered CD4+ T cells in vitro and in vivo. While AML heterogeneity implies that multiple factors may determine their sensitivity to T cell killing, these data show that ICAM1 acts as an immune trigger, allowing T cell killing, and could play a role in AML patient survival in vivo.
Collapse
Affiliation(s)
- Ece Canan Sayitoglu
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Bogdan A Luca
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Allison Paige Boss
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Benjamin Craig Thomas
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Robert Arthur Freeborn
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Molly Javier Uyeda
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Pauline Ping Chen
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Yusuke Nakauchi
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Colin Waichler
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Norman Lacayo
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Rosa Bacchetta
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Center for Definitive and Curative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Ravindra Majeti
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Andrew J Gentles
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Alma-Martina Cepika
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Maria Grazia Roncarolo
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Stanford Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Center for Definitive and Curative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
17
|
Bode K, Wei S, Gruber I, Kissler S, Yi P. Beta Cells Deficient for Renalase Counteract Autoimmunity by Shaping Natural Killer Cell Activity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582816. [PMID: 38496417 PMCID: PMC10942322 DOI: 10.1101/2024.02.29.582816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Type 1 diabetes (T1D) arises from autoimmune-mediated destruction of insulin-producing pancreatic beta cells. Recent advancements in the technology of generating pancreatic beta cells from human pluripotent stem cells (SC-beta cells) have facilitated the exploration of cell replacement therapies for treating T1D. However, the persistent threat of autoimmunity poses a significant challenge to the survival of transplanted SC-beta cells. Genetic engineering is a promising approach to enhance immune resistance of beta cells as we previously showed by inactivating of the Renalase (Rnls) gene. Here we demonstrate that Rnls loss-of-function in beta cells shape autoimmunity by mediating a regulatory Natural Killer (NK) cell phenotype important for the induction of tolerogenic antigen presenting cells. Rnls-deficient beta cells mediate cell-cell-contact-independent induction of hallmark anti-inflammatory cytokine Tgfβ1 in NK cells. In addition, surface expression of key regulatory NK immune checkpoints CD47 and Ceacam1 are markedly elevated on beta cells deficient for Rnls. Enhanced glucose metabolism in Rnls mutant beta cells is responsible for upregulation of CD47 surface expression. These findings are crucial to a better understand how genetically engineered beta cells shape autoimmunity giving valuable insights for future therapeutic advancements to treat and cure T1D.
Collapse
Affiliation(s)
- Kevin Bode
- Section for Immunobiology, Joslin Diabetes Center, Boston, MA 02215
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215
- Department of Medicine, Harvard Medical School, Boston MA 02115
| | - Siying Wei
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215
- Department of Medicine, Harvard Medical School, Boston MA 02115
| | - Isabella Gruber
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215
| | - Stephan Kissler
- Section for Immunobiology, Joslin Diabetes Center, Boston, MA 02215
- Department of Medicine, Harvard Medical School, Boston MA 02115
- Diabetes Program, Harvard Stem Cell Institute, Cambridge MA 02138
| | - Peng Yi
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA 02215
- Department of Medicine, Harvard Medical School, Boston MA 02115
- Diabetes Program, Harvard Stem Cell Institute, Cambridge MA 02138
| |
Collapse
|
18
|
Lin M, Chen D, Shao Z, Liu Q, Hao Z, Xin Z, Chen Y, Wu W, Chen X, He T, Wu D, Wu P. Inflammatory dendritic cells restrain CD11b +CD4 + CTLs via CD200R in human NSCLC. Cell Rep 2024; 43:113767. [PMID: 38354085 DOI: 10.1016/j.celrep.2024.113767] [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: 07/26/2023] [Revised: 12/12/2023] [Accepted: 01/25/2024] [Indexed: 02/16/2024] Open
Abstract
CD4+ cytotoxic T lymphocytes (CD4+ CTLs) are suggested to play a crucial role in inflammatory diseases, including cancer, but their characteristics in human non-small cell lung cancer (NSCLC) remain unknown. Here, using the cell surface marker CD11b, we identify CD11b+CD4+ CTLs as a cytotoxic subset of CD4+ T cells in multiple tissues of NSCLC patients. In addition, tumor-infiltrating CD11b+CD4+ CTLs show a dysfunctional phenotype with elevated expression of CD200 receptor (CD200R), a negatively immunomodulatory receptor. CD4+ regulatory T (Treg) cells restrain the anti-tumor role of CD11b+CD4+ CTLs via CD200. Mechanistically, inflammatory dendritic cells promote the CD200R expression of CD11b+CD4+ CTLs by secreting interleukin-1β (IL-1β). Finally, we demonstrate that CD200 blockade can revive the tumor-killing role of CD11b+CD4+ CTLs and prolong the survival of tumor-bearing mice. Taken together, our study identifies CD11b+CD4+ CTLs in NSCLC with decreased cytotoxicity that can be reinvigorated by CD200 blockade, suggesting that targeting CD200 is a promising immunotherapy strategy in NSCLC.
Collapse
Affiliation(s)
- Mingjie Lin
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Di Chen
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Zheyu Shao
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Qinyuan Liu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Zhixing Hao
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Zhongwei Xin
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yongyuan Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Wenxuan Wu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Gastrointestinal Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xiaoke Chen
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Teng He
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Infectious Disease, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Dang Wu
- Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China.
| | - Pin Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Key Laboratory of Tumor Microenvironment and Immune Therapy of Zhejiang Province, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
19
|
Gonzalez-Montes Y, Osca-Gelis G, Rodriguez-Romanos R, Villavicencio A, González-Bártulos M, Llopis F, Clapes V, Oriol A, Sureda A, Escoda L, Sarrà J, Garzó A, Lloveras N, Gómez B, Granada I, Gallardo D. CD200 genotype is associated with clinical outcome of patients with multiple myeloma. Front Immunol 2024; 15:1252445. [PMID: 38455039 PMCID: PMC10917927 DOI: 10.3389/fimmu.2024.1252445] [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: 07/03/2023] [Accepted: 01/30/2024] [Indexed: 03/09/2024] Open
Abstract
Immune dysfunction in patients with MM affects both the innate and adaptive immune system. Molecules involved in the immune response pathways are essential to determine the ability of cancer cells to escape from the immune system surveillance. However, few data are available concerning the role of immune checkpoint molecules in predicting the myeloma control and immunological scape as mechanism of disease progression. We retrospectively analyzed the clinical impact of the CD200 genotype (rs1131199 and rs2272022) in 291 patients with newly diagnosed MM. Patients with a CD200 rs1131199 GG genotype showed a median overall survival (OS) significantly lower than those with CC+CG genotype (67.8 months versus 94.4 months respectively; p: 0.022) maintaining significance in the multivariate analysis. This effect was specially detected in patients not receiving an autologous stem cell transplant (auto-SCT) (p < 0.001). In these patients the rs1131199 GG genotype negatively influenced in the mortality not related with the progression of MM (p: 0.02) mainly due to infections events.
Collapse
Affiliation(s)
- Yolanda Gonzalez-Montes
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Gemma Osca-Gelis
- Hospital Cancer Registry Unit, Catalan Institute of Oncology, Girona, Spain
- Research Group on Statistics, Econometrics and Health (GRECS), Universitat de Girona, Girona, Spain
- Center CIBER of Epidemiology and Public Health (CIBERESP), Girona, Spain
| | - Rocío Rodriguez-Romanos
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Alicia Villavicencio
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Marta González-Bártulos
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Francesca Llopis
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Victòria Clapes
- Clinical Hematology Department, Institut Català d’Oncologia, L’Hospitalet de Llobregat, Institut d’Investigaciò Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, Barcelona, Spain
| | - Albert Oriol
- Hematology Department, Institut Català d’Oncologia, Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Barcelona, Spain
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d’Oncologia, L’Hospitalet de Llobregat, Institut d’Investigaciò Biomèdica de Bellvitge (IDIBELL), Universitat de Barcelona, Barcelona, Spain
| | - Lourdes Escoda
- Hematology Department, Institut Català d’Oncologia, Hospital Joan XXIII, Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Josep Sarrà
- Hematology Department, Institut Català d’Oncologia, Hospital Joan XXIII, Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Ana Garzó
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Natàlia Lloveras
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Beatriz Gómez
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| | - Isabel Granada
- Hematology Department, Institut Català d’Oncologia, Hospital Germans Trias i Pujol, Josep Carreras Research Institute, Barcelona, Spain
| | - David Gallardo
- Hematology Department, Institut Català d’Oncologia, Hospital Dr. Josep Trueta, Institut d’Investigació Biomèdica de Girona (IDIBGI), Josep Carreras Research Institute, Universitat de Girona, Girona, Spain
| |
Collapse
|
20
|
Barshidi A, Ardeshiri K, Ebrahimi F, Alian F, Shekarchi AA, Hojjat-Farsangi M, Jadidi-Niaragh F. The role of exhausted natural killer cells in the immunopathogenesis and treatment of leukemia. Cell Commun Signal 2024; 22:59. [PMID: 38254135 PMCID: PMC10802000 DOI: 10.1186/s12964-023-01428-2] [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: 09/28/2023] [Accepted: 12/08/2023] [Indexed: 01/24/2024] Open
Abstract
The immune responses to cancer cells involve both innate and acquired immune cells. In the meantime, the most attention has been drawn to the adaptive immune cells, especially T cells, while, it is now well known that the innate immune cells, especially natural killer (NK) cells, play a vital role in defending against malignancies. While the immune cells are trying to eliminate malignant cells, cancer cells try to prevent the function of these cells and suppress immune responses. The suppression of NK cells in various cancers can lead to the induction of an exhausted phenotype in NK cells, which will impair their function. Recent studies have shown that the occurrence of this phenotype in various types of leukemic malignancies can affect the prognosis of the disease, and targeting these cells may be considered a new immunotherapy method in the treatment of leukemia. Therefore, a detailed study of exhausted NK cells in leukemic diseases can help both to understand the mechanisms of leukemia progression and to design new treatment methods by creating a deeper understanding of these cells. Here, we will comprehensively review the immunobiology of exhausted NK cells and their role in various leukemic malignancies. Video Abstract.
Collapse
Affiliation(s)
- Asal Barshidi
- Department of Biological Sciences, Faculty of Sciences, University of Kurdistan, Sanandaj, Iran
| | - Keivan Ardeshiri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Farbod Ebrahimi
- Nanoparticle Process Technology, Faculty of Engineering, University of Duisburg-Essen, Duisburg, Germany
| | - Fatemeh Alian
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Ali Akbar Shekarchi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
21
|
Li L, Li A, Jin H, Li M, Jia Q. Inhibitory receptors and checkpoints on NK cells: Implications for cancer immunotherapy. Pathol Res Pract 2024; 253:155003. [PMID: 38042093 DOI: 10.1016/j.prp.2023.155003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
With the success of immunosuppressive checkpoint in tumor therapy, the corresponding adverse response and drug resistance defects have been exposed. T cells and NK cells are the body's immune system of the two substantial main forces. in recent years, study of T cell checkpoints appeared a certain block, such as PD-1 the effect not benign, on the distribution of NK cell surface excitatory and inhibitory receptors under normal conditions to maintain steady, could be targeted in the tumor treatment blockade have therapeutic effect. This paper reviews the function of NK cells and the effects of corresponding receptors in various types of tumors, providing a direction for the selection of appropriate gate control sites for future treatment.
Collapse
Affiliation(s)
- Lingfei Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Ang Li
- Department of Cardiology, 2nd Medical Center of PLA General Hospital, Beijing, China
| | - Hai Jin
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China.
| | - Mingyang Li
- State Key Laboratory of Cancer Biology, Department of Pathology, Xijing Hospital and School of Basic Medicine, Fourth Military Medical University, Xi'an, China.
| | - Qingge Jia
- Department of Reproductive Medicine, Xi'an International Medical Center Hospital, Northwest University, Xi'an, China.
| |
Collapse
|
22
|
Bakhtiyaridovvombaygi M, Yazdanparast S, Mikanik F, Izadpanah A, Parkhideh S, Shahbaz Ghasabeh A, Roshandel E, Hajifathali A, Gharehbaghian A. Cytokine-Induced Memory-Like NK Cells: Emerging strategy for AML immunotherapy. Biomed Pharmacother 2023; 168:115718. [PMID: 37857247 DOI: 10.1016/j.biopha.2023.115718] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease developed from the malignant expansion of myeloid precursor cells in the bone marrow and peripheral blood. The implementation of intensive chemotherapy and hematopoietic stem cell transplantation (HSCT) has improved outcomes associated with AML, but relapse, along with suboptimal outcomes, is still a common scenario. In the past few years, exploring new therapeutic strategies to optimize treatment outcomes has occurred rapidly. In this regard, natural killer (NK) cell-based immunotherapy has attracted clinical interest due to its critical role in immunosurveillance and their capabilities to target AML blasts. NK cells are cytotoxic innate lymphoid cells that mediate anti-viral and anti-tumor responses by producing pro-inflammatory cytokines and directly inducing cytotoxicity. Although NK cells are well known as short-lived innate immune cells with non-specific responses that have limited their clinical applications, the discovery of cytokine-induced memory-like (CIML) NK cells could overcome these challenges. NK cells pre-activated with the cytokine combination IL-12/15/18 achieved a long-term life span with adaptive immunity characteristics, termed CIML-NK cells. Previous studies documented that using CIML-NK cells in cancer treatment is safe and results in promising outcomes. This review highlights the current application, challenges, and opportunities of CIML-NK cell-based therapy in AML.
Collapse
Affiliation(s)
- Mehdi Bakhtiyaridovvombaygi
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Yazdanparast
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Mikanik
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amirhossein Izadpanah
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sahar Parkhideh
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amin Shahbaz Ghasabeh
- Department of Hematology and Blood Bank, School of Allied Medical Science, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Elham Roshandel
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ahmad Gharehbaghian
- Department of Hematology and Blood Bank, School of Allied Medical Science, Shahid Beheshti University of Medical Science, Tehran, Iran; Pediatric Congenital Hematologic Disorders Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Guarnera L, Bravo-Perez C, Visconte V. Immunotherapy in Acute Myeloid Leukemia: A Literature Review of Emerging Strategies. Bioengineering (Basel) 2023; 10:1228. [PMID: 37892958 PMCID: PMC10604866 DOI: 10.3390/bioengineering10101228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/05/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
In the last twenty years, we have witnessed a paradigm shift in the treatment and prognosis of acute myeloid leukemia (AML), thanks to the introduction of new efficient drugs or approaches to refine old therapies, such as Gemtuzumab Ozogamicin, CPX 3-5-1, hypomethylating agents, and Venetoclax, the optimization of conditioning regimens in allogeneic hematopoietic stem cell transplantation and the improvement of supportive care. However, the long-term survival of non-M3 and non-core binding factor-AML is still dismal. For this reason, the expectations for the recently developed immunotherapies, such as antibody-based therapy, checkpoint inhibitors, and chimeric antigen receptor strategies, successfully tested in other hematologic malignancies, were very high. The inherent characteristics of AML blasts hampered the development of these treatments, and the path of immunotherapy in AML has been bumpy. Herein, we provide a detailed review of potential antigenic targets, available data from pre-clinical and clinical trials, and future directions of immunotherapies in AML.
Collapse
Affiliation(s)
- Luca Guarnera
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.B.-P.); (V.V.)
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Carlos Bravo-Perez
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.B.-P.); (V.V.)
- Department of Hematology and Medical Oncology, Hospital Universitario Morales Meseguer, University of Murcia, IMIB-Pascual Parrilla, CIBERER—Instituto de Salud Carlos III, 30005 Murcia, Spain
| | - Valeria Visconte
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (C.B.-P.); (V.V.)
| |
Collapse
|
24
|
Sayitoglu EC, Luca BA, Boss AP, Thomas BC, Freeborn RA, Uyeda MJ, Chen PP, Nakauchi Y, Waichler C, Lacayo N, Bacchetta R, Majeti R, Gentles AJ, Cepika AM, Roncarolo MG. AML/T cell interactomics uncover correlates of patient outcomes and the key role of ICAM1 in T cell killing of AML. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.21.558911. [PMID: 37790561 PMCID: PMC10542521 DOI: 10.1101/2023.09.21.558911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
T cells are important for the control of acute myeloid leukemia (AML), a common and often deadly malignancy. We observed that some AML patient samples are resistant to killing by human engineered cytotoxic CD4 + T cells. Single-cell RNA-seq of primary AML samples and CD4 + T cells before and after their interaction uncovered transcriptional programs that correlate with AML sensitivity or resistance to CD4 + T cell killing. Resistance-associated AML programs were enriched in AML patients with poor survival, and killing-resistant AML cells did not engage T cells in vitro . Killing-sensitive AML potently activated T cells before being killed, and upregulated ICAM1 , a key component of the immune synapse with T cells. Without ICAM1, killing-sensitive AML became resistant to killing to primary ex vivo -isolated CD8 + T cells in vitro , and engineered CD4 + T cells in vitro and in vivo . Thus, ICAM1 on AML acts as an immune trigger, allowing T cell killing, and could affect AML patient survival in vivo . SIGNIFICANCE AML is a common leukemia with sub-optimal outcomes. We show that AML transcriptional programs correlate with susceptibility to T cell killing. Killing resistance-associated AML programs are enriched in patients with poor survival. Killing-sensitive, but not resistant AML activate T cells and upregulate ICAM1 that binds to LFA-1 on T cells, allowing immune synapse formation which is critical for AML elimination. GRAPHICAL ABSTRACT
Collapse
|
25
|
Batuello C, Mason EF. Diagnostic Utility of CD200 Immunohistochemistry in Distinguishing EBV-Positive Large B-Cell Lymphoma From Classic Hodgkin Lymphoma. Am J Clin Pathol 2023; 160:284-291. [PMID: 37227967 PMCID: PMC10472740 DOI: 10.1093/ajcp/aqad053] [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: 11/22/2022] [Accepted: 04/12/2023] [Indexed: 05/27/2023] Open
Abstract
OBJECTIVES Epstein-Barr virus-positive large B-cell lymphoma (EBV+ LBCL) is a heterogeneous group of diseases that may resemble classic Hodgkin lymphoma (CHL) both morphologically and immunophenotypically. However, these diseases are treated with different therapies and carry distinct prognoses. We examined CD200 expression by immunohistochemistry in EBV+ LBCL and evaluated its diagnostic utility in the differential diagnosis with CHL. METHODS CD200 immunohistochemistry was performed on archival material from 20 cases of CHL (11 EBV+, 9 EBV-), 11 cases of EBV+ LBCL, and 10 cases of diffuse large B-cell lymphoma, not otherwise specified (DLBCL NOS). Staining pattern and intensity (0-3+ scale) were recorded. RESULTS CD200 positivity was seen in Reed-Sternberg cells in 19 (95%) of 20 cases of CHL, predominantly in a strong (3+, 15/19) and diffuse (>50% of cells, 17/19) pattern. In contrast, CD200 was negative in 8 (73%) of 11 cases of EBV+ LBCL; the 3 positive cases showed 1 to 2+ staining in less than 50% of lesional cells. All cases of DLBCL NOS were negative for CD200. CONCLUSIONS CD200 may be a useful immunophenotypic marker in differentiating EBV+ LBCL from CHL, with negative to partial/weak staining favoring a diagnosis of EBV+ LBCL and strong diffuse staining favoring a diagnosis of CHL.
Collapse
Affiliation(s)
- Christopher Batuello
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, US
| | - Emily F Mason
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, US
| |
Collapse
|
26
|
Zhi L, Wang X, Gao Q, He W, Shang C, Guo C, Niu Z, Zhu W, Zhang X. Intrinsic and extrinsic factors determining natural killer cell fate: Phenotype and function. Biomed Pharmacother 2023; 165:115136. [PMID: 37453199 DOI: 10.1016/j.biopha.2023.115136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/26/2023] [Accepted: 07/07/2023] [Indexed: 07/18/2023] Open
Abstract
Natural killer (NK) cells are derived from hematopoietic stem cells. They belong to the innate lymphoid cell family, which is an important part of innate immunity. This family plays a role in the body mainly through the release of perforin, granzyme, and various cytokines and is involved in cytotoxicity and cytokine-mediated immune regulation. NK cells involved in normal immune regulation and the tumor microenvironment (TME) can exhibit completely different states. Here, we discuss the growth, development, and function of NK cells in regard to intrinsic and extrinsic factors. Intrinsic factors are those that influence NK cells to promote cell maturation and exert their effector functions under the control of internal metabolism and self-related genes. Extrinsic factors include the metabolism of the TME and the influence of related proteins on the "fate" of NK cells. This review targets the potential of NK cell metabolism, cellular molecules, regulatory genes, and other mechanisms involved in immune regulation. We further discuss immune-mediated tumor therapy, which is the trend of current research.
Collapse
Affiliation(s)
- Lingtong Zhi
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province 453003, PR China
| | - Xing Wang
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province 453003, PR China
| | - Qing Gao
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province 453003, PR China
| | - Wenhui He
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province 453003, PR China
| | - Chongye Shang
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province 453003, PR China
| | - Changjiang Guo
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province 453003, PR China
| | - Zhiyuan Niu
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province 453003, PR China
| | - Wuling Zhu
- Henan Province Engineering Research Center of Innovation for Synthetic Biology, School of Life Sciences and Technology, Xinxiang Medical University, Xinxiang, Henan Province 453003, PR China.
| | - Xuan Zhang
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan 453003, PR China.
| |
Collapse
|
27
|
Christodoulou I, Solomou EE. A Panorama of Immune Fighters Armored with CARs in Acute Myeloid Leukemia. Cancers (Basel) 2023; 15:cancers15113054. [PMID: 37297016 DOI: 10.3390/cancers15113054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/28/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Acute myeloid leukemia (AML) is a devastating disease. Intensive chemotherapy is the mainstay of treatment but results in debilitating toxicities. Moreover, many treated patients will eventually require hematopoietic stem cell transplantation (HSCT) for disease control, which is the only potentially curative but challenging option. Ultimately, a subset of patients will relapse or have refractory disease, posing a huge challenge to further therapeutic decisions. Targeted immunotherapies hold promise for relapsed/refractory (r/r) malignancies by directing the immune system against cancer. Chimeric antigen receptors (CARs) are important components of targeted immunotherapy. Indeed, CAR-T cells have achieved unprecedented success against r/r CD19+ malignancies. However, CAR-T cells have only achieved modest outcomes in clinical studies on r/r AML. Natural killer (NK) cells have innate anti-AML functionality and can be engineered with CARs to improve their antitumor response. CAR-NKs are associated with lower toxicities than CAR-T cells; however, their clinical efficacy against AML has not been extensively investigated. In this review, we cite the results from clinical studies of CAR-T cells in AML and describe their limitations and safety concerns. Moreover, we depict the clinical and preclinical landscape of CAR used in alternative immune cell platforms with a specific focus on CAR-NKs, providing insight into the future optimization of AML.
Collapse
Affiliation(s)
- Ilias Christodoulou
- Department of Internal Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Internal Medicine, University of Patras Medical School, 26500 Rion, Greece
| | - Elena E Solomou
- Department of Internal Medicine, University of Patras Medical School, 26500 Rion, Greece
| |
Collapse
|
28
|
D’Silva SZ, Singh M, Pinto AS. NK cell defects: implication in acute myeloid leukemia. Front Immunol 2023; 14:1112059. [PMID: 37228595 PMCID: PMC10203541 DOI: 10.3389/fimmu.2023.1112059] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/25/2023] [Indexed: 05/27/2023] Open
Abstract
Acute Myeloid Leukemia (AML) is a complex disease with rapid progression and poor/unsatisfactory outcomes. In the past few years, the focus has been on developing newer therapies for AML; however, relapse remains a significant problem. Natural Killer cells have strong anti-tumor potential against AML. This NK-mediated cytotoxicity is often restricted by cellular defects caused by disease-associated mechanisms, which can lead to disease progression. A stark feature of AML is the low/no expression of the cognate HLA ligands for the activating KIR receptors, due to which these tumor cells evade NK-mediated lysis. Recently, different Natural Killer cell therapies have been implicated in treating AML, such as the adoptive NK cell transfer, Chimeric antigen receptor-modified NK (CAR-NK) cell therapy, antibodies, cytokine, and drug treatment. However, the data available is scarce, and the outcomes vary between different transplant settings and different types of leukemia. Moreover, remission achieved by some of these therapies is only for a short time. In this mini-review, we will discuss the role of NK cell defects in AML progression, particularly the expression of different cell surface markers, the available NK cell therapies, and the results from various preclinical and clinical trials.
Collapse
Affiliation(s)
- Selma Z. D’Silva
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Meenakshi Singh
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Andrea S. Pinto
- Transplant Immunology and Immunogenetics Lab, Advanced Centre for Treatment, Education and Research in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| |
Collapse
|
29
|
Čelakovská J, Čermáková E, Boudková P, Andrýs C, Krejsek J. Evaluation of Leukocytes, B and T Lymphocytes, and expression of CD200 and CD23 on B lymphocytes in Patients with Atopic Dermatitis on Dupilumab Therapy-Pilot Study. Dermatol Ther (Heidelb) 2023; 13:1171-1192. [PMID: 37097547 PMCID: PMC10149535 DOI: 10.1007/s13555-023-00918-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 03/16/2023] [Indexed: 04/26/2023] Open
Abstract
BACKGROUND There are a lot of studies that describe the change in quantity of T cells in patients with atopic dermatitis (AD) compared with healthy subjects. Other components of lymphocytes such as B cells are not examined as well as T cells. OBJECTIVE We focus on immunophenotyping of B cells with their subsets (memory, naïve, switched, non-switched) and the expression of CD23 and CD200 markers in patients with AD with and without dupilumab therapy. We also evaluate the count of leukocytes and their subsets, T lymphocytes (CD4+, CD8+), natural killer (NK) cells, and T regulatory cells. METHODS A total of 45 patients suffering from AD were examined: 32 patients without dupilumab treatment (10 men, 22 women, average age 35 years), 13 patients with dupilumab treatment (7 men, 6 women, average age 43.4 years), and 30 subjects as a control group (10 men, 20 women, average age 44.7 years). Immunophenotype was examined by flow cytometry in which monoclonal antibodies with fluorescent molecules were used. We compared the absolute and relative count of leukocytes and their subsets, T lymphocytes (CD4+ , CD8+), NK cells, T regulatory cells, absolute and relative count of B lymphocytes (memory, naïve, non-switched, switched, transient), and expression of CD23 and CD200 activation markers on B cells and on their subsets in patients with AD and control group. For statistical analysis we used nonparametric Kruskal-Wallis one-factor analysis of variance with post hoc by Dunn's test with Bonferroni modification of significance level. RESULTS In patients with AD with and without dupilumab therapy we confirmed the significantly higher count of neutrophils, monocytes, and eosinophils; there was no difference in absolute count of B cells, NK cells and transitional B cells compared with control subjects. We confirmed higher expression of activation marker CD23 on total, memory, naïve, non-switched, and switched B lymphocytes and higher expression of CD200 on total B lymphocytes in both groups of patients with AD compared with controls. In patients without dupilumab therapy we confirmed significantly higher count of relative monocytes, relative eosinophils, and higher expression of CD200 on memory, naïve, and non-switched B lymphocytes compared with controls. In patients with dupilumab therapy we confirmed significantly higher expression of CD200 on switched B lymphocytes, higher count of relative CD4+ T lymphocytes, and lower count of absolute CD8+ T lymphocytes compared with controls. CONCLUSION This pilot study shows higher expression of CD23 on B lymphocytes and on their subsets in patients with AD with and without dupilumab therapy. The higher expression of CD200 on switched B lymphocytes is confirmed only in patients with AD with dupilumab therapy.
Collapse
Affiliation(s)
- Jarmila Čelakovská
- Department of Dermatology and Venereology, Faculty Hospital and Medical Faculty of Charles University, 50002, Hradec Králové, Czech Republic.
| | - Eva Čermáková
- Department of Medical Biophysics, Medical Faculty of Charles University, 50002, Hradec Králové, Czech Republic
| | - Petra Boudková
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, 50002, Hradec Králové, Czech Republic
| | - Ctirad Andrýs
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, 50002, Hradec Králové, Czech Republic
| | - Jan Krejsek
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, 50002, Hradec Králové, Czech Republic
| |
Collapse
|
30
|
Holicek P, Truxova I, Rakova J, Salek C, Hensler M, Kovar M, Reinis M, Mikyskova R, Pasulka J, Vosahlikova S, Remesova H, Valentova I, Lysak D, Holubova M, Kaspar P, Prochazka J, Kasikova L, Spisek R, Galluzzi L, Fucikova J. Type I interferon signaling in malignant blasts contributes to treatment efficacy in AML patients. Cell Death Dis 2023; 14:209. [PMID: 36964168 PMCID: PMC10039058 DOI: 10.1038/s41419-023-05728-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 03/26/2023]
Abstract
While type I interferon (IFN) is best known for its key role against viral infection, accumulating preclinical and clinical data indicate that robust type I IFN production in the tumor microenvironment promotes cancer immunosurveillance and contributes to the efficacy of various antineoplastic agents, notably immunogenic cell death inducers. Here, we report that malignant blasts from patients with acute myeloid leukemia (AML) release type I IFN via a Toll-like receptor 3 (TLR3)-dependent mechanism that is not driven by treatment. While in these patients the ability of type I IFN to stimulate anticancer immune responses was abolished by immunosuppressive mechanisms elicited by malignant blasts, type I IFN turned out to exert direct cytostatic, cytotoxic and chemosensitizing activity in primary AML blasts, leukemic stem cells from AML patients and AML xenograft models. Finally, a genetic signature of type I IFN signaling was found to have independent prognostic value on relapse-free survival and overall survival in a cohort of 132 AML patients. These findings delineate a clinically relevant, therapeutically actionable and prognostically informative mechanism through which type I IFN mediates beneficial effects in patients with AML.
Collapse
Affiliation(s)
- Peter Holicek
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | | | | | - Cyril Salek
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
- Institute of Clinical and Experimental Hematology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | | | - Marek Kovar
- Laboratory of Tumor Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Milan Reinis
- Laboratory of Immunological and Tumour Models, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Romana Mikyskova
- Laboratory of Immunological and Tumour Models, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | | | | | - Hana Remesova
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Iva Valentova
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Daniel Lysak
- Department of Hematology and Oncology, Faculty Hospital in Pilsen, Pilsen, Czech Republic
| | - Monika Holubova
- Biomedical Center, Medical Faculty in Pilsen, Charles University, Pilsen, Czech Republic
| | - Petr Kaspar
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Prochazka
- Czech Centre for Phenogenomics, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | | | - Radek Spisek
- Sotio Biotech, Prague, Czech Republic
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| | - Jitka Fucikova
- Sotio Biotech, Prague, Czech Republic.
- Department of Immunology, Charles University, 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic.
| |
Collapse
|
31
|
Liang ZX, Liu HS, Xiong L, Zeng ZW, Zheng XB, Kang L, Lan P, Wu XR. GAS6 From CD200+ Adipose-Derived Stem Cells Mitigates Colonic Inflammation in a Macrophage-Dependent Manner. J Crohns Colitis 2023; 17:289-301. [PMID: 36006655 DOI: 10.1093/ecco-jcc/jjac123] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Stem cell therapy is a promising cell-based treatment modality for inflammatory bowel diseases [IBD], but its application is limited by the nature of cell heterogeneity. METHODS Single-cell RNA-sequencing was performed on the adipose-derived stem cells [ADSCs]. The in vitro immunomodulatory effect of ADSCs was evaluated by co-culturing with human CD4+ T cells or macrophages. The in vivo therapeutic value of ADSCs was assessed using a murine colitis model induced by dextran sulphate sodium [DSS] or 2,4,6-trinitrobenzene sulphonic acid [TNBS]. RESULTS CD200+ ADSCs were identified as a novel subpopulation of ADSCs, based on gene ontology analysis of immunoregulatory functions. The immunoregulatory functions of these cells were further confirmed by co-culturing with CD4+ T cells or macrophages. Administration of CD200+ ADSCs effectively reduced intestinal inflammation in IBD mice models. Furthermore, we found CD200+ ADSCs-derived GAS6 exerted protective effects on experimental colitis by promoting macrophage M2 polarization via the Mer/PI3K/Akt/GSK3β signalling pathway. CONCLUSIONS This study uncovered the heterogeneity in ADSCs, in which CD200+ ADSCs presents as an alternative to conventional treatment of IBD.
Collapse
Affiliation(s)
- Zhen-Xing Liang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Hua-Shan Liu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li Xiong
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zi-Wei Zeng
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Bin Zheng
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Liang Kang
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ping Lan
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| | - Xian-Rui Wu
- Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Bioland Laboratory, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
32
|
Shao A, Owens DM. The immunoregulatory protein CD200 as a potentially lucrative yet elusive target for cancer therapy. Oncotarget 2023; 14:96-103. [PMID: 36738455 PMCID: PMC9899099 DOI: 10.18632/oncotarget.28354] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
CD200 is an immunoregulatory cell surface ligand with proven pro-tumorigenic credentials via its ability to suppress CD200 receptor (CD200R)-expressing anti-tumor immune function. This definitive role for the CD200-CD200R axis in regulating an immunosuppressive tumor microenvironment has garnered increasing interest in CD200 as a candidate target for immune checkpoint inhibition therapy. However, while the CD200 blocking antibody samalizumab is still in the early stages of clinical testing, alternative mechanisms for the pro-tumorigenic role of CD200 have recently emerged that extend beyond direct suppression of anti-tumor T cell responses and, as such, may not be susceptible to CD200 antibody blockade. Herein, we will summarize the current understanding of CD200 expression and function in the tumor microenvironment as well as alternative strategies for potential neutralization of multiple CD200 mechanisms in human cancers.
Collapse
Affiliation(s)
- Anqi Shao
- 1Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - David M. Owens
- 1Department of Dermatology, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, New York, NY 10032, USA,2Department of Pathology and Cell Biology, Columbia University Irving Medical Center, Vagelos College of Physicians and Surgeons, New York, NY 10032, USA,Correspondence to:David M. Owens, email:
| |
Collapse
|
33
|
Choe D, Choi D. Cancel cancer: The immunotherapeutic potential of CD200/CD200R blockade. Front Oncol 2023; 13:1088038. [PMID: 36756156 PMCID: PMC9900175 DOI: 10.3389/fonc.2023.1088038] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023] Open
Abstract
Immune checkpoint molecules function to inhibit and regulate immune response pathways to prevent hyperactive immune activity from damaging healthy tissues. In cancer patients, targeting these key molecules may serve as a valuable therapeutic mechanism to bolster immune function and restore the body's natural defenses against tumors. CD200, an immune checkpoint molecule, is a surface glycoprotein that is widely but not ubiquitously expressed throughout the body. By interacting with its inhibitory receptor CD200R, CD200 suppresses immune cell activity within the tumor microenvironment, creating conditions that foster tumor growth. Targeting the CD200/CD200R pathway, either through the use of monoclonal antibodies or peptide inhibitors, has shown to be effective in boosting anti-tumor immune activity. This review will explore CD200 and the protein's expression and role within the tumor microenvironment, blood endothelial cells, and lymph nodes. This paper will also discuss the advantages and challenges of current strategies used to target CD200 and briefly summarize relevant preclinical/clinical studies investigating the immunotherapeutic efficacy of CD200/CD200R blockade.
Collapse
|
34
|
Boudková P, Čelakovská J, Čermáková E, Andrýs C, Krejsek J. Immunological Parameters in Patients Suffering from Atopic Dermatitis and Either Treated or Non-Treated with Dupilumab. ACTA MEDICA (HRADEC KRALOVE) 2023; 66:47-54. [PMID: 37930093 DOI: 10.14712/18059694.2023.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
OBJECTIVE The aim of the study is to analyze the absolute count of leukocytes, neutrophils, monocytes, eosinophils, T cells, natural killer cells, B cells and to evaluate the expression of functionally important CD23 and CD200 molecules on B cells in patients suffering from atopic dermatitis (AD), (with and without dupilumab therapy). MATERIALS AND METHODS We examined 45 patients suffering from AD - 32 patients without dupilumab treatment (10 men, 22 women, average age 35.0 years), 13 patients with dupilumab treatment (7 men, 6 women, average age 43.4 years) and 30 healthy control (10 men, 20 women, average age 44.7 years). Immunophenotype was examined by flow cytometry (Navios Flow Cytometer - Beckman Coulter). The blood count was examined with a Sysmex XN 3000, Sysmex SP10, microscope DI60 for digital morphology evaluating cell division and microscope Olympus BX40. We compared the absolute count of leukocytes and their subsets, T cells (CD4, CD8), natural killers cells, absolute and relative count of B lymphocytes and expression of surface molecules CD23 and CD200 on B cells in AD patients and in control group. Non-parametric Kruskal-Wallis one-factor analysis of variance with post-hoc (follow-up multiple comparison) and Dunn's test with Bonferroni modification of significance level were used for statistical analysis. RESULTS We confirmed the significantly higher number of neutrophils, monocytes and eosinophils and higher expression of CD23 and CD200 on B cells in peripheral blood of AD patients (either with or without dupilumab) therapy. We demonstrated the lower number of CD8+ T cells. CONCLUSION We demonstrated the difference in the count of white blood cells populations in patients suffering from AD compared with healthy control. There were a differences in the expression of immunoregulatory molecules CD23 and CD200 on B cells in AD patients (either with or without dupilumab therapy) in comparison to healthy controls.
Collapse
Affiliation(s)
- Petra Boudková
- Department of Clinical Immunology and Allergology, Faculty Hospital and Faculty of Medicine, Charles University, Hradec Králové, Czech Republic.
| | - Jarmila Čelakovská
- Department of Dermatology and Venereology Faculty Hospital and Faculty of Medicine, Charles University, Hradec Králové, Czech Republic
| | - Eva Čermáková
- Department of Medical Biophysics, Faculty of Medicine, Charles University, Hradec Králové, Czech Republic
| | - Ctirad Andrýs
- Department of Clinical Immunology and Allergology, Faculty Hospital and Faculty of Medicine, Charles University, Hradec Králové, Czech Republic
| | - Jan Krejsek
- Department of Clinical Immunology and Allergology, Faculty Hospital and Faculty of Medicine, Charles University, Hradec Králové, Czech Republic
| |
Collapse
|
35
|
Serroukh Y, Hébert J, Busque L, Mercier F, Rudd CE, Assouline S, Lachance S, Delisle JS. Blasts in context: the impact of the immune environment on acute myeloid leukemia prognosis and treatment. Blood Rev 2023; 57:100991. [PMID: 35941029 DOI: 10.1016/j.blre.2022.100991] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 01/28/2023]
Abstract
Acute myeloid leukemia (AML) is a cancer that originates from the bone marrow (BM). Under physiological conditions, the bone marrow supports the homeostasis of immune cells and hosts memory lymphoid cells. In this review, we summarize our present understanding of the role of the immune microenvironment on healthy bone marrow and on the development of AML, with a focus on T cells and other lymphoid cells. The types and function of different immune cells involved in the AML microenvironment as well as their putative role in the onset of disease and response to treatment are presented. We also describe how the immune context predicts the response to immunotherapy in AML and how these therapies modulate the immune status of the bone marrow. Finally, we focus on allogeneic stem cell transplantation and summarize the current understanding of the immune environment in the post-transplant bone marrow, the factors associated with immune escape and relevant strategies to prevent and treat relapse.
Collapse
Affiliation(s)
- Yasmina Serroukh
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Erasmus Medical center Cancer Institute, University Medical Center Rotterdam, Department of Hematology, Rotterdam, the Netherlands; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada.
| | - Josée Hébert
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada; The Quebec Leukemia Cell Bank, Canada
| | - Lambert Busque
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - François Mercier
- Division of Hematology and Experimental Medicine, Department of Medicine, McGill University, 3755 Côte-Sainte-Catherine Road, Montreal, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte-Sainte-Catherine Road, Montreal, Canada
| | - Christopher E Rudd
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - Sarit Assouline
- Division of Hematology and Experimental Medicine, Department of Medicine, McGill University, 3755 Côte-Sainte-Catherine Road, Montreal, Canada; Lady Davis Institute for Medical Research, Jewish General Hospital, 3755 Côte-Sainte-Catherine Road, Montreal, Canada
| | - Silvy Lachance
- Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| | - Jean-Sébastien Delisle
- Centre de recherche de l'Hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, Canada; Department of Medicine, Université de Montréal, Montreal, Canada; Institute for Hematology-Oncology, Transplantation, Cell and Gene Therapy, Hôpital Maisonneuve-Rosemont, Montreal, Canada
| |
Collapse
|
36
|
Rahmani S, Yazdanpanah N, Rezaei N. Natural killer cells and acute myeloid leukemia: promises and challenges. Cancer Immunol Immunother 2022; 71:2849-2867. [PMID: 35639116 PMCID: PMC10991240 DOI: 10.1007/s00262-022-03217-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 04/26/2022] [Indexed: 10/18/2022]
Abstract
Acute myeloid leukemia (AML) is considered as one of the most malignant conditions of the bone marrow. Over the past few decades, despite substantial progresses in the management of AML, relapse remission remains a major problem. Natural killer cells (NK cells) are known as a unique component of the innate immune system. Due to swift tumor detection, distinct cytotoxic action, and extensive immune interaction, NK cells have been used in various cancer settings for decades. It has been a growing knowledge of therapeutic magnitudes ranging from adoptive NK cell transfer to chimeric antigen receptor NK cells, aiming to achieve better therapeutic responses in patients with AML. In this article, the potentials of NK cells for treatment of AML are highlighted, and challenges for such therapeutic methods are discussed. In addition, the clinical application of NK cells, mainly in patients with AML, is pictured according to the existing evidence.
Collapse
Affiliation(s)
- Shayan Rahmani
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Niloufar Yazdanpanah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, Tehran, 14194, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Morgan HJ, Rees E, Lanfredini S, Powell KA, Gore J, Gibbs A, Lovatt C, Davies GE, Olivero C, Shorning BY, Tornillo G, Tonks A, Darley R, Wang EC, Patel GK. CD200 ectodomain shedding into the tumor microenvironment leads to NK cell dysfunction and apoptosis. J Clin Invest 2022; 132:150750. [PMID: 36074574 PMCID: PMC9621138 DOI: 10.1172/jci150750] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/01/2022] [Indexed: 11/24/2022] Open
Abstract
The basis of immune evasion, a hallmark of cancer, can differ even when cancers arise from one cell type such as in the human skin keratinocyte carcinomas: basal and squamous cell carcinoma. Here we showed that the basal cell carcinoma tumor-initiating cell surface protein CD200, through ectodomain shedding, was responsible for the near absence of NK cells within the basal cell carcinoma tumor microenvironment. In situ, CD200 underwent ectodomain shedding by metalloproteinases MMP3 and MMP11, which released biologically active soluble CD200 into the basal cell carcinoma microenvironment. CD200 bound its cognate receptor on NK cells to suppress MAPK pathway signaling that in turn blocked indirect (IFN-γ release) and direct cell killing. In addition, reduced ERK phosphorylation relinquished negative regulation of PPARγ-regulated gene transcription and led to membrane accumulation of the Fas/FADD death receptor and its ligand, FasL, which resulted in activation-induced apoptosis. Blocking CD200 inhibition of MAPK or PPARγ signaling restored NK cell survival and tumor cell killing, with relevance to many cancer types. Our results thus uncover a paradigm for CD200 as a potentially novel and targetable NK cell-specific immune checkpoint, which is responsible for NK cell-associated poor outcomes in many cancers.
Collapse
Affiliation(s)
- Huw J Morgan
- European Cancer Stem Cell Research Institute, School of Biosciences
| | - Elise Rees
- European Cancer Stem Cell Research Institute, School of Biosciences
| | | | - Kate A Powell
- European Cancer Stem Cell Research Institute, School of Biosciences
| | - Jasmine Gore
- European Cancer Stem Cell Research Institute, School of Biosciences
| | - Alex Gibbs
- European Cancer Stem Cell Research Institute, School of Biosciences
| | - Charlotte Lovatt
- European Cancer Stem Cell Research Institute, School of Biosciences
| | - Gemma E Davies
- European Cancer Stem Cell Research Institute, School of Biosciences
| | - Carlotta Olivero
- European Cancer Stem Cell Research Institute, School of Biosciences
| | - Boris Y Shorning
- European Cancer Stem Cell Research Institute, School of Biosciences
| | - Giusy Tornillo
- European Cancer Stem Cell Research Institute, School of Biosciences
| | - Alex Tonks
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, and
| | - Richard Darley
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, and
| | - Eddie Cy Wang
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Girish K Patel
- European Cancer Stem Cell Research Institute, School of Biosciences
| |
Collapse
|
38
|
Liao KL, Watt KD. Mathematical Modeling and Analysis of CD200-CD200R in Cancer Treatment. Bull Math Biol 2022; 84:82. [PMID: 35792958 DOI: 10.1007/s11538-022-01039-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: 10/08/2021] [Accepted: 06/01/2022] [Indexed: 11/26/2022]
Abstract
CD200 is a cell membrane protein that binds to its receptor, CD200 receptor (CD200R). The CD200 positive tumor cells inhibit the cellular functions of M1 and M2 macrophages and dendritic cells (DCs) through the CD200-CD200R complex, resulting in downregulation of Interleukin-10 and Interleukin-12 productions and affecting the activation of cytotoxic T lymphocytes. In this work, we provide two ordinary differential equation models, one complete model and one simplified model, to investigate how the binding affinities of CD200R and the populations of M1 and M2 macrophages affect the functions of the CD200-CD200R complex in tumor growth. Our simulations demonstrate that (i) the impact of the CD200-CD200R complex on tumor promotion or inhibition highly depends on the binding affinity of the CD200R on M2 macrophages and DCs to the CD200 on tumor cells, and (ii) a stronger binding affinity of the CD200R on M1 macrophages or DCs to the CD200 on tumor cells induces a higher tumor cell density in the CD200 positive tumor. Thus, the CD200 blockade would be an efficient treatment method in this case. Moreover, the simplified model shows that the binding affinity of CD200R on macrophages is the major factor to determine the treatment efficacy of CD200 blockade when the binding affinities of CD200R on M1 and M2 macrophages are significantly different to each other. On the other hand, both the binding affinity of CD200R and the population of macrophages are the major factors to determine the treatment efficacy of CD200 blockade when the binding affinities of CD200R on M1 and M2 macrophages are close to each other. We also analyze the simplified model to investigate the dynamics of the positive and trivial equilibria of the CD200 positive tumor case and the CD200 deficient tumor case. The bifurcation diagrams show that when M1 macrophages dominate the population, the tumor cell density of the CD200 positive tumor is higher than the one of CD200 deficient tumor. Moreover, the dynamics of tumor cell density change from tumor elimination to tumor persistence to oscillation, as the maximal proliferation rate of tumor cells increases.
Collapse
Affiliation(s)
- Kang-Ling Liao
- Department of Mathematics, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada.
| | - Kenton D Watt
- Department of Mathematics, University of Manitoba, Winnipeg, MB, R3T 2N2, Canada
| |
Collapse
|
39
|
Bhattacharjee R, Ghosh S, Nath A, Basu A, Biswas O, Patil CR, Kundu CN. Theragnostic strategies harnessing the self-renewal pathways of stem-like cells in the acute myeloid leukemia. Crit Rev Oncol Hematol 2022; 177:103753. [PMID: 35803452 DOI: 10.1016/j.critrevonc.2022.103753] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 06/21/2022] [Accepted: 07/02/2022] [Indexed: 02/07/2023] Open
Abstract
Acute myelogenous leukemia (AML) is a genetically heterogeneous and aggressive cancer of the Hematopoietic Stem/progenitor cells. It is distinguished by the uncontrollable clonal growth of malignant myeloid stem cells in the bone marrow, venous blood, and other body tissues. AML is the most predominant of leukemias occurring in adults (25%) and children (15-20%). The relapse after chemotherapy is a major concern in the treatment of AML. The overall 5-year survival rate in young AML patients is about 40-45% whereas in the elderly patients it is less than 10%. Leukemia stem-like cells (LSCs) having the ability to self-renew indefinitely, repopulate and persist longer in the G0/G1 phase play a crucial role in the AML relapse and refractoriness to chemotherapy. Hence, novel treatment strategies and diagnostic biomarkers targeting LSCs are being increasingly investigated. Through this review, we have explored the signaling modulations in the LSCs as the theragnostic targets. The significance of the self-renewal pathways in overcoming the treatment challenges in AML has been highlighted.
Collapse
Affiliation(s)
- Rahul Bhattacharjee
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Sharad Ghosh
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Arijit Nath
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Asmita Basu
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Ojaswi Biswas
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India
| | - Chandragauda R Patil
- Department of Pharmacology, DIPSAR, Delhi Pharmaceutical Sciences and Research University, New Delhi, India
| | - Chanakya Nath Kundu
- KIIT School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT-DU), Bhubaneswar, Odisha, India.
| |
Collapse
|
40
|
Yang X, Ma L, Zhang X, Huang L, Wei J. Targeting PD-1/PD-L1 pathway in myelodysplastic syndromes and acute myeloid leukemia. Exp Hematol Oncol 2022; 11:11. [PMID: 35236415 PMCID: PMC8889667 DOI: 10.1186/s40164-022-00263-4] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 02/16/2022] [Indexed: 12/14/2022] Open
Abstract
Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are clonal hematopoietic stem cell diseases arising from the bone marrow (BM), and approximately 30% of MDS eventually progress to AML, associated with increasingly aggressive neoplastic hematopoietic clones and poor survival. Dysregulated immune microenvironment has been recognized as a key pathogenic driver of MDS and AML, causing high rate of intramedullary apoptosis in lower-risk MDS to immunosuppression in higher-risk MDS and AML. Immune checkpoint molecules, including programmed cell death-1 (PD-1) and programmed cell death ligand-1 (PD-L1), play important roles in oncogenesis by maintaining an immunosuppressive tumor microenvironment. Recently, both molecules have been examined in MDS and AML. Abnormal inflammatory signaling, genetic and/or epigenetic alterations, interactions between cells, and treatment of patients all have been involved in dysregulating PD-1/PD-L1 signaling in these two diseases. Furthermore, with the PD-1/PD-L1 pathway activated in immune microenvironment, the milieu of BM shift to immunosuppressive, contributing to a clonal evolution of blasts. Nevertheless, numerous preclinical studies have suggested a potential response of patients to PD-1/PD-L1 blocker. Current clinical trials employing these drugs in MDS and AML have reported mixed clinical responses. In this paper, we focus on the recent preclinical advances of the PD-1/PD-L1 signaling in MDS and AML, and available and ongoing outcomes of PD-1/PD-L1 inhibitor in patients. We also discuss the novel PD-1/PD-L1 blocker-based immunotherapeutic strategies and challenges, including identifying reliable biomarkers, determining settings, and exploring optimal combination therapies.
Collapse
Affiliation(s)
- Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Ling Ma
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaoying Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China.,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China
| | - Liang Huang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, Hubei, China. .,Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, 430030, Hubei, China.
| |
Collapse
|
41
|
Archilla-Ortega A, Domuro C, Martin-Liberal J, Muñoz P. Blockade of novel immune checkpoints and new therapeutic combinations to boost antitumor immunity. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:62. [PMID: 35164813 PMCID: PMC8842574 DOI: 10.1186/s13046-022-02264-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 01/18/2022] [Indexed: 12/18/2022]
Abstract
Immunotherapy has emerged as a promising strategy for boosting antitumoral immunity. Blockade of immune checkpoints (ICs), which regulate the activity of cytotoxic T lymphocytes (CTLs) and natural killer (NK) cells has proven clinical benefits. Antibodies targeting CTLA-4, PD-1, and PD-L1 are IC-blockade drugs approved for the treatment of various solid and hematological malignancies. However, a large subset of patients does not respond to current anti-IC immunotherapy. An integrative understanding of tumor-immune infiltrate, and IC expression and function in immune cell populations is fundamental to the design of effective therapies. The simultaneous blockade of newly identified ICs, as well as of previously described ICs, could improve antitumor response. We review the potential for novel combinatory blockade strategies as antitumoral therapy, and their effects on immune cells expressing the targeted ICs. Preclinical evidence and clinical trials involving the blockade of the various ICs are reported. We finally discuss the rationale of IC co-blockade strategy with respect to its downstream signaling in order to improve effective antitumoral immunity and prevent an increased risk of immune-related adverse events (irAEs).
Collapse
|
42
|
Widodo N, Puspitarini S, Widyananda MH, Alamsyah A, Wicaksono ST, Masruri M, Jatmiko YD. Anticancer activity of Caesalpinia sappan by downregulating mitochondrial genes in A549 lung cancer cell line. F1000Res 2022; 11:169. [PMID: 36128561 PMCID: PMC9468624 DOI: 10.12688/f1000research.76187.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/24/2022] [Indexed: 09/05/2024] Open
Abstract
Background: The standardization and mechanism of action of Caesalpinia sappan as an anticancer agent are still lacking. This study aimed to understand the mechanism of action of C,sappan extract as an anticancer agent. Methods: This study was conducted using the A549 lung cancer cell line to understand the mechanism of action of C. sappan extract as an anticancer agent. The cytotoxicity activity, cell cycle progression, apoptosis, protein-related apoptosis (i.e., BCL-2and BAX protein) assays, and RNA sequencing were performed level were measured. Moreover, the antioxidant activity, total flavonoids, and phenolics of C.sappan were also assessed. Results: C.sappan has strong antioxidant activity (22.14 ± 0.93 ppm) total flavonoid content of (529.3 ± 4.56 mgQE/g), and phenolics content of (923.37 ± 5 mgGAE/g). The C.sappan ethanol extract inhibited cancer cell growth and arrested at G0/G1 phase of cell cycle, inducing apoptosis by increasing BAX/BCL-2 protein ratio in A549 lung cancer cell line. Furthermore, results from RNA sequencing analysis showed that C.sappan ethanol extract caused downregulation of genes acting on mitochondrial function including adenosine triphosphate (ATP) production and respiration. Conclusions: This study demonstrated that C.sappan has the ability to inhibit cancer cell growth by inducing apoptosis and mitochondrial dysfunction in A549 cells.
Collapse
Affiliation(s)
- Nashi Widodo
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Sapti Puspitarini
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | | | - Adzral Alamsyah
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Septian Tri Wicaksono
- Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Masruri Masruri
- Chemistry Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Yoga Dwi Jatmiko
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| |
Collapse
|
43
|
Widodo N, Puspitarini S, Widyananda MH, Alamsyah A, Wicaksono ST, Masruri M, Jatmiko YD. Anticancer activity of Caesalpinia sappan by downregulating mitochondrial genes in A549 lung cancer cell line. F1000Res 2022; 11:169. [PMID: 36128561 PMCID: PMC9468624 DOI: 10.12688/f1000research.76187.2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/18/2022] [Indexed: 11/20/2022] Open
Abstract
Background: The standardization and mechanism of action of Caesalpinia sappan as an anticancer agent are still lacking. This study aimed to understand the mechanism of action of C,sappan extract as an anticancer agent. Methods: This study was conducted using the A549 lung cancer cell line to understand the mechanism of action of C. sappan extract as an anticancer agent. The cytotoxicity activity, cell cycle progression, apoptosis, protein-related apoptosis (i.e., BCL-2and BAX protein) assays, and RNA sequencing were performed level were measured. Moreover, the antioxidant activity, total flavonoids, and phenolics of C.sappan were also assessed. Results: C.sappan has strong antioxidant activity (22.14 ± 0.93 ppm) total flavonoid content of (529.3 ± 4.56 mgQE/g), and phenolics content of (923.37 ± 5 mgGAE/g). The C.sappan ethanol extract inhibited cancer cell growth and arrested at G0/G1 phase of cell cycle, inducing apoptosis by increasing BAX/BCL-2 protein ratio in A549 lung cancer cell line. Furthermore, results from RNA sequencing analysis showed that C.sappan ethanol extract caused downregulation of genes acting on mitochondrial function including adenosine triphosphate (ATP) production and respiration. Conclusions: This study demonstrated that C.sappan has the ability to inhibit cancer cell growth by inducing apoptosis and mitochondrial dysfunction in A549 cells.
Collapse
Affiliation(s)
- Nashi Widodo
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Sapti Puspitarini
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | | | - Adzral Alamsyah
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Septian Tri Wicaksono
- Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Masruri Masruri
- Chemistry Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| | - Yoga Dwi Jatmiko
- Biology Department, Faculty of Mathematics and Natural Sciences, Brawijaya University, Malang, Indonesia
| |
Collapse
|
44
|
Lin YZ, Liu SH, Wu WR, Shen YC, Wang YL, Liao CC, Lin PL, Chang H, Liu LC, Cheng WC, Wang SC. miR-4759 suppresses breast cancer through immune checkpoint blockade. Comput Struct Biotechnol J 2022; 20:241-251. [PMID: 35024096 PMCID: PMC8718579 DOI: 10.1016/j.csbj.2021.12.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 12/02/2021] [Accepted: 12/12/2021] [Indexed: 12/24/2022] Open
Abstract
Programmed cell death protein 1 (PD-1)/ programmed cell death protein ligand 1 (PD-L1) is the key immune checkpoint governing evasion of advanced cancer from immune surveillance. Immuno-oncology (IO) therapy targeting PD-1/PD-L1 with traditional antibodies is a promising approach to multiple cancer types but to which the response rate remains moderate in breast cancer, calling for the need of exploring alternative IO targeting approaches. A miRNA-gene network was integrated by a bioinformatics approach and corroborated with The Cancer Genome Atlas (TCGA) to screen miRNAs regulating immune checkpoint genes and associated with patient survival. Here we show the identification of a novel microRNA miR-4759 which repressed RNA expression of the PD-L1 gene. miR-4759 targeted the PD-L1 gene through two binding motifs in the 3′ untranslated region (3′-UTR) of PD-L1. Reconstitution of miR-4759 inhibited PD-L1 expression and sensitized breast cancer cells to killing by immune cells. Treatment with miR-4759 suppressed tumor growth of orthotopic xenografts and promoted tumor infiltration of CD8+ T lymphocytes in immunocompetent mice. In contrast, miR-4759 had no effect to tumor growth in immunodeficient mice. In patients with breast cancer, expression of miR-4759 was preferentially downregulated in tumors compared to normal tissues and was associated with poor overall survival. Together, our results demonstrated miR-4759 as a novel non-coding RNA which promotes anti-tumor immunity of breast cancer.
Collapse
Affiliation(s)
- You-Zhe Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Shu-Hsuan Liu
- Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan
| | - Wan-Rong Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Yi-Chun Shen
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Yuan-Liang Wang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 40402, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung 40447, Taiwan
| | - Chien-Ching Liao
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Pei-Le Lin
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Han Chang
- Division of Molecular Pathology, Department of Pathology, China Medical University Hospital, Taichung 40447, Taiwan
| | - Liang-Chih Liu
- Department of Surgery, China Medical University Hospital, Taichung 40447, Taiwan
| | - Wei-Chung Cheng
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 40402, Taiwan.,Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan.,Cancer Biology and Drug Discovery Ph.D. Program, China Medical University, Taichung 40402, Taiwan
| | - Shao-Chun Wang
- Graduate Institute of Biomedical Sciences, College of Medicine, China Medical University, Taichung 40402, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung 40447, Taiwan.,Center for Molecular Medicine, China Medical University Hospital, Taichung 40447, Taiwan.,Department of Cancer Biology, University of Cincinnati, Cincinnati, OH 45267, USA.,Cancer Biology and Drug Discovery Ph.D. Program, China Medical University, Taichung 40402, Taiwan.,Department of Biotechnology, Asia University, Taichung 41354, Taiwan
| |
Collapse
|
45
|
Immunosuppression and outcomes in adult patients with de novo acute myeloid leukemia with normal karyotypes. Proc Natl Acad Sci U S A 2021; 118:2116427118. [PMID: 34845035 PMCID: PMC8673586 DOI: 10.1073/pnas.2116427118] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 11/18/2022] Open
Abstract
Acute myeloid leukemia (AML) patients rarely have long first remissions (LFRs; >5 y) after standard-of-care chemotherapy, unless classified as favorable risk at presentation. Identification of the mechanisms responsible for long vs. more typical, standard remissions may help to define prognostic determinants for chemotherapy responses. Using exome sequencing, RNA-sequencing, and functional immunologic studies, we characterized 28 normal karyotype (NK)-AML patients with >5 y first remissions after chemotherapy (LFRs) and compared them to a well-matched group of 31 NK-AML patients who relapsed within 2 y (standard first remissions [SFRs]). Our combined analyses indicated that genetic-risk profiling at presentation (as defined by European LeukemiaNet [ELN] 2017 criteria) was not sufficient to explain the outcomes of many SFR cases. Single-cell RNA-sequencing studies of 15 AML samples showed that SFR AML cells differentially expressed many genes associated with immune suppression. The bone marrow of SFR cases had significantly fewer CD4+ Th1 cells; these T cells expressed an exhaustion signature and were resistant to activation by T cell receptor stimulation in the presence of autologous AML cells. T cell activation could be restored by removing the AML cells or blocking the inhibitory major histocompatibility complex class II receptor, LAG3. Most LFR cases did not display these features, suggesting that their AML cells were not as immunosuppressive. These findings were confirmed and extended in an independent set of 50 AML cases representing all ELN 2017 risk groups. AML cell-mediated suppression of CD4+ T cell activation at presentation is strongly associated with unfavorable outcomes in AML patients treated with standard chemotherapy.
Collapse
|
46
|
Staub RB, Marcondes NA, Rotta LN. CD200 expression in hematopoietic neoplasms: Beyond a marker for diagnosis of B-cell neoplasms. Crit Rev Oncol Hematol 2021; 167:103509. [PMID: 34688895 DOI: 10.1016/j.critrevonc.2021.103509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 08/31/2021] [Accepted: 10/10/2021] [Indexed: 10/20/2022] Open
Abstract
CD200 (OX-2) is expressed in myeloid cells, B cells, subsets of T cells and on other normal and neoplastic non-hematopoietic cells. It interacts with CD200R and has a suppressive effect on T cells immune mediated response. We aimed to review CD200 expression and its role in the immune evasion of non-B cell hematopoietic neoplasms. In acute myeloid leukemia, CD200 seems to be related to the worst outcome, even in diseases of good prognosis, possibly due to an immunosuppressive effect. In plasma cell myeloma studies, while some have associated CD200 expression with worst prognosis possibly due to its suppressive effect on monocyte and T cell-mediated immune response, in others CD200 appeared to be a marker of a better outcome, or even showed no impact in event-free survival (EFS). Few studies have evaluated CD200 expression in T cell neoplasms; however, it appears to be a good immunophenotypic marker for angioimmunoblastic T cell lymphoma. In conclusion, CD200 appears to be involved in the immune evasion of malignant cells, which could affect the survival of these patients.
Collapse
Affiliation(s)
- Renata B Staub
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | | | - Liane N Rotta
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil; Departamento de Métodos Diagnósticos, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
47
|
Zam W, Assaad A. Chimeric antigen receptor T-cells (CARs) in cancer treatment. Curr Mol Pharmacol 2021; 15:532-546. [PMID: 34382510 DOI: 10.2174/1874467214666210811150255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/27/2021] [Accepted: 05/17/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Cancer is one of the leading causes of death worldwide. Chemotherapy, radiation therapy, and stem cell transplantation were the main cancer treatment approaches for several years but due to their limited effectiveness, there was a constant search for new therapeutic approaches. Cancer immunotherapy that utilizes and enhances the normal capacity of the patient's immune system was used to fight against cancer. Genetically engineered T-cells that express chimeric antigen receptors (CARs) showed remarkable anti-tumor activity against hematologic malignancies and is now being investigated in a variety of solid tumors. The use of this therapy in the last few years has been successful, achieving a great success in improving the quality of life and prolonging the survival time of patients with a reduction in remission rates. However, many challenges still need to be resolved in order for this technology to gain widespread adoption. <P> Objective: This review summarizes various experimental approaches towards the use of CAR T-cells in hematologic malignancies and solid tumors. <P> Conclusion: Finally, we address the challenges posed by CAR T-cells and discuss strategies for improving the performance of these T cells in fighting cancers.
Collapse
Affiliation(s)
- Wissam Zam
- Department of Analytical and Food Chemistry, Faculty of Pharmacy, Al-Wadi International University, Homs. Syrian Arab Republic
| | - Amany Assaad
- 2. Department of Analytical and Food Chemistry, Faculty of Pharmacy,Tartous University, Tartous. Syrian Arab Republic
| |
Collapse
|
48
|
Herbrich S, Baran N, Cai T, Weng C, Aitken MJL, Post SM, Henderson J, Shi C, Richard-Carpentier G, Sauvageau G, Baggerly K, Al-Atrash G, Davis RE, Daver N, Zha D, Konopleva M. Overexpression of CD200 is a Stem Cell-Specific Mechanism of Immune Evasion in AML. J Immunother Cancer 2021; 9:e002968. [PMID: 34326171 PMCID: PMC8323398 DOI: 10.1136/jitc-2021-002968] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) stem cells (LSCs) are capable of surviving current standard chemotherapy and are the likely source of deadly, relapsed disease. While stem cell transplant serves as proof-of-principle that AML LSCs can be eliminated by the immune system, the translation of existing immunotherapies to AML has been met with limited success. Consequently, understanding and exploiting the unique immune-evasive mechanisms of AML LSCs is critical. METHODS Analysis of stem cell datasets and primary patient samples revealed CD200 as a putative stem cell-specific immune checkpoint overexpressed in AML LSCs. Isogenic cell line models of CD200 expression were employed to characterize the interaction of CD200+ AML with various immune cell subsets both in vitro and in peripheral blood mononuclear cell (PBMC)-humanized mouse models. CyTOF and RNA-sequencing were performed on humanized mice to identify novel mechanisms of CD200-mediated immunosuppression. To clinically translate these findings, we developed a fully humanized CD200 antibody (IgG1) that removed the immunosuppressive signal by blocking interaction with the CD200 receptor while also inducing a potent Fc-mediated response. Therapeutic efficacy of the CD200 antibody was evaluated using both humanized mice and patient-derived xenograft models. RESULTS Our results demonstrate that CD200 is selectively overexpressed in AML LSCs and is broadly immunosuppressive by impairing cytokine secretion in both innate and adaptive immune cell subsets. In a PBMC-humanized mouse model, CD200+ leukemia progressed rapidly, escaping elimination by T cells, compared with CD200- AML. T cells from mice with CD200+ AML were characterized by an abundance of metabolically quiescent CD8+ central and effector memory cells. Mechanistically, CD200 expression on AML cells significantly impaired OXPHOS metabolic activity in T cells from healthy donors. Importantly, CD200 antibody therapy could eliminate disease in the presence of graft-versus-leukemia in immune competent mice and could significantly improve the efficacy of low-intensity azacitidine/venetoclax chemotherapy in immunodeficient hosts. CONCLUSIONS Overexpression of CD200 is a stem cell-specific marker that contributes to immunosuppression in AML by impairing effector cell metabolism and function. CD200 antibody therapy is capable of simultaneously reducing CD200-mediated suppression while also engaging macrophage activity. This study lays the groundwork for CD200-targeted therapeutic strategies to eliminate LSCs and prevent AML relapse.
Collapse
Affiliation(s)
- Shelley Herbrich
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tianyu Cai
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Connie Weng
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marisa J L Aitken
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sean M Post
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jared Henderson
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chunhua Shi
- Oncology Research for Biologics and Immunotherapy Translation (ORBIT) platform, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Guy Sauvageau
- University of Montreal Institute for Research in Immunology and Cancer, Montreal, Quebec, Canada
| | - Keith Baggerly
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gheath Al-Atrash
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - R Eric Davis
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dongxing Zha
- Oncology Research for Biologics and Immunotherapy Translation (ORBIT) platform, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
49
|
Khan IZ, Del Guzzo CA, Shao A, Cho J, Du R, Cohen AO, Owens DM. The CD200-CD200R axis promotes squamous cell carcinoma metastasis via regulation of cathepsin K. Cancer Res 2021; 81:5021-5032. [PMID: 34183355 DOI: 10.1158/0008-5472.can-20-3251] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 04/05/2021] [Accepted: 06/24/2021] [Indexed: 11/16/2022]
Abstract
The CD200-CD200R immunoregulatory signaling axis plays an etiological role in the survival and spread of numerous cancers primarily through suppression of anti-tumor immune surveillance. Our previous work outlined a pro-metastatic role for the CD200-CD200R axis in cutaneous squamous cell carcinoma (cSCC) that is independent of direct T cell suppression but modulates the function of infiltrating myeloid cells. To identify effectors of the CD200-CD200R axis important for cSCC metastasis, we conducted RNA-Seq profiling of infiltrating CD11B+Cd200R+ cells isolated from CD200+ versus CD200-null cSCCs and identified the cysteine protease cathepsin K (Ctsk) to be highly upregulated in CD200+ cSCCs. CD11B+Cd200R+ cells expressed phenotypic markers associated with myeloid-derived suppressor cell-like cells and tumor-associated macrophages and were the primary source of Ctsk expression in cSCC. A Cd200R+ myeloid cell-cSCC co-culture system showed that induction of Ctsk was dependent on engagement of the CD200-CD200R axis, indicating that Ctsk is a target gene of this pathway in the cSCC tumor microenvironment. Inhibition of Ctsk, but not matrix metalloproteinases (MMP), significantly blocked cSCC cell migration in vitro. Finally, targeted CD200 disruption in tumor cells and Ctsk pharmacological inhibition significantly reduced cSCC metastasis in vivo. Collectively, these findings support the conclusion that CD200 stimulates cSCC invasion and metastasis via induction of Ctsk in CD200R+ infiltrating myeloid cells.
Collapse
Affiliation(s)
| | | | | | | | - Rong Du
- Dermatology, Columbia University
| | | | | |
Collapse
|
50
|
Kaweme NM, Zhou F. Optimizing NK Cell-Based Immunotherapy in Myeloid Leukemia: Abrogating an Immunosuppressive Microenvironment. Front Immunol 2021; 12:683381. [PMID: 34220833 PMCID: PMC8247591 DOI: 10.3389/fimmu.2021.683381] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Accepted: 05/31/2021] [Indexed: 12/12/2022] Open
Abstract
Natural killer (NK) cells are prominent cytotoxic and cytokine-producing components of the innate immune system representing crucial effector cells in cancer immunotherapy. Presently, various NK cell-based immunotherapies have contributed to the substantial improvement in the reconstitution of NK cells against advanced-staged and high-risk AML. Various NK cell sources, including haploidentical NK cells, adaptive NK cells, umbilical cord blood NK cells, stem cell-derived NK cells, chimeric antigen receptor NK cells, cytokine-induced memory-like NK cells, and NK cell lines have been identified. Devising innovative approaches to improve the generation of therapeutic NK cells from the aforementioned sources is likely to enhance NK cell expansion and activation, stimulate ex vivo and in vivo persistence of NK cells and improve conventional treatment response of myeloid leukemia. The tumor-promoting properties of the tumor microenvironment and downmodulation of NK cellular metabolic activity in solid tumors and hematological malignancies constitute a significant impediment in enhancing the anti-tumor effects of NK cells. In this review, we discuss the current NK cell sources, highlight ongoing interventions in enhancing NK cell function, and outline novel strategies to circumvent immunosuppressive factors in the tumor microenvironment to improve the efficacy of NK cell-based immunotherapy and expand their future success in treating myeloid leukemia.
Collapse
Affiliation(s)
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, China
| |
Collapse
|