1
|
Li Z, Han B, Qi M, Li Y, Duan Y, Yao Y. Modulating macrophage-mediated programmed cell removal: An attractive strategy for cancer therapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189172. [PMID: 39151808 DOI: 10.1016/j.bbcan.2024.189172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 08/11/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024]
Abstract
Macrophage-mediated programmed cell removal (PrCR) is crucial for the identification and elimination of needless cells that maintain tissue homeostasis. The efficacy of PrCR depends on the balance between pro-phagocytic "eat me" signals and anti-phagocytic "don't eat me" signals. Recently, a growing number of studies have shown that tumourigenesis and progression are closely associated with PrCR. In the tumour microenvironment, PrCR activated by the "eat me" signal is counterbalanced by the "don't eat me" signal of CD47/SIRPα, resulting in tumour immune escape. Therefore, targeting exciting "eat me" signalling while simultaneously suppressing "don't eat me" signalling and eventually inducing macrophages to produce effective PrCR will be a very attractive antitumour strategy. Here, we comprehensively review the functions of PrCR-activating signal molecules (CRT, PS, Annexin1, SLAMF7) and PrCR-inhibiting signal molecules (CD47/SIRPα, MHC-I/LILRB1, CD24/Siglec-10, SLAMF3, SLAMF4, PD-1/PD-L1, CD31, GD2, VCAM1), the interactions between these molecules, and Warburg effect. In addition, we highlight the molecular regulatory mechanisms that affect immune system function by exciting or suppressing PrCR. Finally, we review the research advances in tumour therapy by activating PrCR and discuss the challenges and potential solutions to smooth the way for tumour treatment strategies that target PrCR.
Collapse
Affiliation(s)
- Zhenzhen Li
- Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Bingqian Han
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Menghui Qi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yinchao Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yongtao Duan
- Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China; Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China.
| | - Yongfang Yao
- Henan International Joint Laboratory of Prevention and Treatment of Pediatric Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou University, Zhengzhou 450018, China; School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
2
|
Lu Y, Fu W, Xing W, Wu H, Zhang C, Xu D. Transcriptional regulation mechanism of PARP1 and its application in disease treatment. Epigenetics Chromatin 2024; 17:26. [PMID: 39118189 PMCID: PMC11308664 DOI: 10.1186/s13072-024-00550-w] [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: 04/08/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024] Open
Abstract
Poly (ADP-ribose) polymerase 1 (PARP1) is a multifunctional nuclear enzyme that catalyzes poly-ADP ribosylation in eukaryotic cells. In addition to maintaining genomic integrity, this nuclear enzyme is also involved in transcriptional regulation. PARP1 can trigger and maintain changes in the chromatin structure and directly recruit transcription factors. PARP1 also prevents DNA methylation. However, most previous reviews on PARP1 have focused on its involvement in maintaining genome integrity, with less focus on its transcriptional regulatory function. This article comprehensively reviews the transcriptional regulatory function of PARP1 and its application in disease treatment, providing new ideas for targeting PARP1 for the treatment of diseases other than cancer.
Collapse
Affiliation(s)
- Yu Lu
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Beijing, 100850, P.R. China
- Hebei University, Baoding, Hebei, P.R. China
| | - Wenliang Fu
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Beijing, 100850, P.R. China
| | - Weiwei Xing
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Beijing, 100850, P.R. China
| | - Haowei Wu
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Beijing, 100850, P.R. China
| | - Chao Zhang
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Beijing, 100850, P.R. China.
| | - Donggang Xu
- Beijing Institute of Basic Medical Sciences, No. 27 Taiping Road, Beijing, 100850, P.R. China.
| |
Collapse
|
3
|
Cerchietti L. Genetic mechanisms underlying tumor microenvironment composition and function in diffuse large B-cell lymphoma. Blood 2024; 143:1101-1111. [PMID: 38211334 PMCID: PMC10972714 DOI: 10.1182/blood.2023021002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/18/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024] Open
Abstract
ABSTRACT Cells in the tumor microenvironment (TME) of diffuse large B-cell lymphoma (DLBCL) show enormous diversity and plasticity, with functions that can range from tumor inhibitory to tumor supportive. The patient's age, immune status, and DLBCL treatments are factors that contribute to the shaping of this TME, but evidence suggests that genetic factors, arising principally in lymphoma cells themselves, are among the most important. Here, we review the current understanding of the role of these genetic drivers of DLBCL in establishing and modulating the lymphoma microenvironment. A better comprehension of the relationship between lymphoma genetic factors and TME biology should lead to better therapeutic interventions, especially immunotherapies.
Collapse
Affiliation(s)
- Leandro Cerchietti
- Hematology and Oncology Division, Medicine Department, New York-Presbyterian Hospital, Meyer Cancer Center, Weill Cornell Medicine, Cornell University, New York, NY
| |
Collapse
|
4
|
Huang S, Zhang X, Wei Y, Xiao Y. Checkpoint CD24 function on tumor and immunotherapy. Front Immunol 2024; 15:1367959. [PMID: 38487533 PMCID: PMC10937401 DOI: 10.3389/fimmu.2024.1367959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/12/2024] [Indexed: 03/17/2024] Open
Abstract
CD24 is a protein found on the surface of cells that plays a crucial role in the proliferation, invasion, and spread of cancer cells. It adheres to cell membranes through glycosylphosphatidylinositol (GPI) and is associated with the prognosis and survival rate of cancer patients. CD24 interacts with the inhibitory receptor Siglec-10 that is present on immune cells like natural killer cells and macrophages, leading to the inhibition of natural killer cell cytotoxicity and macrophage-mediated phagocytosis. This interaction helps tumor cells escape immune detection and attack. Although the use of CD24 as a immune checkpoint receptor target for cancer immunotherapy is still in its early stages, clinical trials have shown promising results. Monoclonal antibodies targeting CD24 have been found to be well-tolerated and safe. Other preclinical studies are exploring the use of chimeric antigen receptor (CAR) T cells, antibody-drug conjugates, and gene therapy to target CD24 and enhance the immune response against tumors. In summary, this review focuses on the role of CD24 in the immune system and provides evidence for CD24 as a promising immune checkpoint for cancer immunotherapy.
Collapse
Affiliation(s)
- Shiming Huang
- Department of Radiology, First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
- Graduate School, Chinese PLA Medical School, Beijing, China
- Department of Nuclear Medicine, Characteristic Medical Center of the Chinese People’s Armed Police Force, Tianjin, China
| | - Xiaobo Zhang
- Department of Radiology, First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Yingtian Wei
- Department of Radiology, First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| | - Yueyong Xiao
- Department of Radiology, First Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, China
| |
Collapse
|
5
|
Moon SY, Han M, Ryu G, Shin SA, Lee JH, Lee CS. Emerging Immune Checkpoint Molecules on Cancer Cells: CD24 and CD200. Int J Mol Sci 2023; 24:15072. [PMID: 37894750 PMCID: PMC10606340 DOI: 10.3390/ijms242015072] [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: 09/12/2023] [Revised: 10/04/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Cancer immunotherapy strategies are based on the utilization of immune checkpoint inhibitors to instigate an antitumor immune response. The efficacy of immune checkpoint blockade, directed at adaptive immune checkpoints, has been demonstrated in select cancer types. However, only a limited subset of patients has exhibited definitive outcomes characterized by a sustained response after discontinuation of therapy. Recent investigations have highlighted the significance of immune checkpoint molecules that are overexpressed in cancer cells and inhibit myeloid lineage immune cells within a tumor microenvironment. These checkpoints are identified as potential targets for anticancer immune responses. Notably, the immune checkpoint molecules CD24 and CD200 have garnered attention owing to their involvement in tumor immune evasion. CD24 and CD200 are overexpressed across diverse cancer types and serve as signaling checkpoints by engaging their respective receptors, Siglec-10 and CD200 receptor, which are expressed on tumor-associated myeloid cells. In this review, we summarized and discussed the latest advancements and insights into CD24 and CD200 as emergent immune checkpoint moieties, further delving into their therapeutic potentials for cancer treatment.
Collapse
Affiliation(s)
- Sun Young Moon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| | - Minjoo Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| | - Gyoungah Ryu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| | - Seong-Ah Shin
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| | - Jun Hyuck Lee
- Research Unit of Cryogenic Novel Material, Korea Polar Research Institute, Incheon 21990, Republic of Korea;
- Department of Polar Sciences, University of Science and Technology, Incheon 21990, Republic of Korea
| | - Chang Sup Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Gyeongsang National University, Jinju 52828, Republic of Korea; (S.Y.M.); (M.H.); (G.R.); (S.-A.S.)
| |
Collapse
|
6
|
Aroldi A, Mauri M, Ramazzotti D, Villa M, Malighetti F, Crippa V, Cocito F, Borella C, Bossi E, Steidl C, Scollo C, Voena C, Chiarle R, Mologni L, Piazza R, Gambacorti‐Passerini C. Effects of blocking CD24 and CD47 'don't eat me' signals in combination with rituximab in mantle-cell lymphoma and chronic lymphocytic leukaemia. J Cell Mol Med 2023; 27:3053-3064. [PMID: 37654003 PMCID: PMC10568669 DOI: 10.1111/jcmm.17868] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/03/2023] [Accepted: 07/12/2023] [Indexed: 09/02/2023] Open
Abstract
Mantle-cell lymphoma (MCL) is a B-cell non-Hodgkin Lymphoma (NHL) with a poor prognosis, at high risk of relapse after conventional treatment. MCL-associated tumour microenvironment (TME) is characterized by M2-like tumour-associated macrophages (TAMs), able to interact with cancer cells, providing tumour survival and resistance to immuno-chemotherapy. Likewise, monocyte-derived nurse-like cells (NLCs) present M2-like profile and provide proliferation signals to chronic lymphocytic leukaemia (CLL), a B-cell malignancy sharing with MCL some biological and phenotypic features. Antibodies against TAMs targeted CD47, a 'don't eat me' signal (DEMs) able to quench phagocytosis by TAMs within TME, with clinical effectiveness when combined with Rituximab in pretreated NHL. Recently, CD24 was found as valid DEMs in solid cancer. Since CD24 is expressed during B-cell differentiation, we investigated and identified consistent CD24 in MCL, CLL and primary human samples. Phagocytosis increased when M2-like macrophages were co-cultured with cancer cells, particularly in the case of paired DEMs blockade (i.e. anti-CD24 + anti-CD47) combined with Rituximab. Similarly, unstimulated CLL patients-derived NLCs provided increased phagocytosis when DEMs blockade occurred. Since high levels of CD24 were associated with worse survival in both MCL and CLL, anti-CD24-induced phagocytosis could be considered for future clinical use, particularly in association with other agents such as Rituximab.
Collapse
Affiliation(s)
- Andrea Aroldi
- Hematology DivisionSan Gerardo HospitalMonzaItaly
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Mario Mauri
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Daniele Ramazzotti
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Matteo Villa
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | | | - Valentina Crippa
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | | | | | - Elisa Bossi
- Hematology DivisionSan Gerardo HospitalMonzaItaly
| | - Carolina Steidl
- Lymphoma Unit, Department of Onco‐HematologyIRCCS San Raffaele Scientific InstituteMilanItaly
| | - Chiara Scollo
- Transfusion Medicine UnitSan Gerardo HospitalMonzaItaly
| | - Claudia Voena
- Department of Molecular Biotechnology and Health SciencesUniversity of TorinoTorinoItaly
| | - Roberto Chiarle
- Department of Molecular Biotechnology and Health SciencesUniversity of TorinoTorinoItaly
- Department of PathologyBoston Children's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Division of HematopathologyEuropean Institute of Oncology (IEO) IRCCSMilanItaly
| | - Luca Mologni
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Rocco Piazza
- Hematology DivisionSan Gerardo HospitalMonzaItaly
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| | - Carlo Gambacorti‐Passerini
- Hematology DivisionSan Gerardo HospitalMonzaItaly
- Department of Medicine and SurgeryUniversity of Milano‐BicoccaMonzaItaly
| |
Collapse
|
7
|
Gu Y, Zhou G, Tang X, Shen F, Ding J, Hua K. The biological roles of CD24 in ovarian cancer: old story, but new tales. Front Immunol 2023; 14:1183285. [PMID: 37359556 PMCID: PMC10288981 DOI: 10.3389/fimmu.2023.1183285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/25/2023] [Indexed: 06/28/2023] Open
Abstract
CD24 is a glycosylphosphatidylinositol linked molecular which expressed in diverse malignant tumor cells, particular in ovarian carcinoma cells and ovarian carcinoma stem cells. The CD24 expression is associated with increased metastatic potential and poor prognosis of malignancies. CD24 on the surface of tumor cells could interact with Siglec-10 on the surface of immune cells, to mediate the immune escape of tumor cells. Nowadays, CD24 has been identified as a promising focus for targeting therapy of ovarian cancer. However, the roles of CD24 in tumorigenesis, metastasis, and immune escape are still not clearly demonstrated systematically. In this review, we i) summarized the existing studies on CD24 in diverse cancers including ovarian cancer, ii) illustrated the role of CD24-siglec10 signaling pathway in immune escape, iii) reviewed the existing immunotherapeutic strategies (targeting the CD24 to restore the phagocytic effect of Siglec-10 expressing immune cells) based on the above mechanisms and evaluated the priorities in the future research. These results might provide support for guiding the CD24 immunotherapy as the intervention upon solid tumors.
Collapse
Affiliation(s)
- Yuanyuan Gu
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Guannan Zhou
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
- Changning Maternity and Infant Health Hospital, East China Normal University, Shanghai, China
| | - Xue Tang
- Department of Laboratory Medicine, Division of Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Fang Shen
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Jingxin Ding
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| | - Keqin Hua
- Department of Gynecology, The Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Department of Gynecology, Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, China
| |
Collapse
|
8
|
Chen K, Dai M, Luo Q, Wang Y, Shen W, Liao Y, Zhou Y, Cheng W. PARP1 controls the transcription of CD24 by ADP-ribosylating the RNA helicase DDX5 in pancreatic cancer. Int J Biochem Cell Biol 2023; 155:106358. [PMID: 36584909 DOI: 10.1016/j.biocel.2022.106358] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/11/2022] [Accepted: 12/27/2022] [Indexed: 12/29/2022]
Abstract
The PARP1 protein plays a key role in DNA damage repair and ADP-ribosylation to regulate gene expression. Strategies to target PARP1 have rapidly been developed for cancer treatment. However, the role of the innate immune response in targeted anti-PARP1 therapy remains poorly understood. In this work, we aimed to elucidate the regulatory mechanism underlying the immunogenicity of PARP1 and explore efficient therapeutic strategies to enhance the antitumor effect of PARP inhibitors. The relationships between PARP1 expression and immunosuppressive factors were examined by qRTPCR and immunoblot analysis. DNA pull-down, chromatin immunoprecipitation-quantitative PCR (ChIPqPCR) and luciferase reporter assays were employed to reveal the mechanism by which the expression of the immune checkpoint regulator CD24 is regulated by PARP1. Phagocytosis assays and pancreatic cancer animal models were applied to evaluate the therapeutic effect of simultaneous disruption of PARP1 and the antiphagocytic factor CD24. Upregulation of the innate immunosuppressive factor CD24 was observed in pancreatic cancer during PARP1 inhibition. The activating effect of targeting CD24 on macrophage phagocytosis was verified. Then, we showed that PARP1 attenuated the transcription of CD24 by ADP-ribosylating the transcription factor DDX5 in pancreatic cancer. Combined blockade of PARP1 and the antiphagocytic factor CD24 elicited a synergetic antitumor effect in pancreatic cancer. Our research provided evidence that combination treatment with PARP inhibitors and CD24 blocking monoclonal antibodies (mAbs) could be an effective strategy to improve the clinical therapeutic response in pancreatic cancer.
Collapse
Affiliation(s)
- Kang Chen
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China; Translational Medicine Laboratory of Pancreas Disease of Hunan Normal University, Changsha 410005, Hunan Province, China
| | - Manxiong Dai
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China; Translational Medicine Laboratory of Pancreas Disease of Hunan Normal University, Changsha 410005, Hunan Province, China
| | - Quanneng Luo
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China; Translational Medicine Laboratory of Pancreas Disease of Hunan Normal University, Changsha 410005, Hunan Province, China
| | - Yi Wang
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China; Translational Medicine Laboratory of Pancreas Disease of Hunan Normal University, Changsha 410005, Hunan Province, China
| | - Weitao Shen
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yan Liao
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China; Translational Medicine Laboratory of Pancreas Disease of Hunan Normal University, Changsha 410005, Hunan Province, China
| | - Yiying Zhou
- Department of Clinical Pathology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China
| | - Wei Cheng
- Department of Hepatobiliary Surgery, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha 410005, Hunan Province, China; Xiangyue Hospital Affiliated to Hunan Institute of Parasitic Diseases, National Clinical Center for Schistosomiasis Treatment, Yueyang 414000, Hunan Province, China; Translational Medicine Laboratory of Pancreas Disease of Hunan Normal University, Changsha 410005, Hunan Province, China.
| |
Collapse
|
9
|
Christian SL. CD24 as a Potential Therapeutic Target in Patients with B-Cell Leukemia and Lymphoma: Current Insights. Onco Targets Ther 2022; 15:1391-1402. [PMID: 36425299 PMCID: PMC9680537 DOI: 10.2147/ott.s366625] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/10/2022] [Indexed: 01/12/2024] Open
Abstract
CD24 is a highly glycosylated glycophosphatidylinositol (GPI)-anchored protein that is expressed in many types of differentiating cells and some mature cells of the immune system as well as the central nervous system. CD24 has been extensively used as a biomarker for developing B cells as its expression levels change over the course of B cell development. Functionally, engagement of CD24 induces apoptosis in developing B cells and restricts cell growth in more mature cell types. Interestingly, CD24 is also expressed on many hematological and solid tumors. As such, it has been investigated as a therapeutic target in many solid tumors including ovarian, colorectal, pancreatic, lung and others. Most of the B-cell leukemias and lymphomas studied to date express CD24 but its role as a therapeutic target in these malignancies has, thus far, been understudied. Here, I review what is known about CD24 biology with a focus on B cell development and activation followed by a brief overview of how CD24 is being targeted in solid tumors. This is followed by an assessment of the value of CD24 as a therapeutic target in B cell leukemia and lymphoma in humans, including an evaluation of the challenges in using CD24 as a target considering its pattern of expression on normal cells.
Collapse
Affiliation(s)
- Sherri L Christian
- Department of Biochemistry, Memorial University of Newfoundland, St. John’s, NL, Canada
- Beatrice Hunter Cancer Research Institute, Halifax, NS, Canada
| |
Collapse
|
10
|
Gao Q, Chen X, Cherian S, Roshal M. Mature B‐ and plasma‐cell flow cytometric analysis: A review of the impact of targeted therapy. CYTOMETRY PART B: CLINICAL CYTOMETRY 2022; 104:224-242. [PMID: 36321879 DOI: 10.1002/cyto.b.22097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022]
Abstract
Flow cytometry has been indispensable in diagnosing B cell lymphoma and plasma cell neoplasms. The advances in novel multicolor flow cytometry have also made this technology a robust tool for monitoring minimal/measurable residual disease in chronic lymphocytic leukemia and multiple myeloma. However, challenges using conventional gating strategies to isolate neoplastic B or plasma cells are emerging due to the rapidly increasing number of antibody therapeutics targeting single or multiple classic B/plasma cell-lineage markers, such as CD19, CD20, and CD22 in B cells and CD38 in plasma cells. This review is the first of a two-part series that summarizes the most current targeted therapies used in B and plasma cell neoplasms and proposes detailed alternative approaches to overcome post-targeted therapy analysis challenges by flow cytometry. The second review in this series (Chen et al.) focuses on challenges encountered in the use of targeted therapy in precursor B cell neoplasms.
Collapse
Affiliation(s)
- Qi Gao
- Hematopathology Service, Department of Pathology and Laboratory Medicine Memorial Sloan Kettering Cancer Center New York New York USA
| | - Xueyan Chen
- Department of Laboratory Medicine and Pathology University of Washington Seattle WA USA
| | - Sindu Cherian
- Department of Laboratory Medicine and Pathology University of Washington Seattle WA USA
| | - Mikhail Roshal
- Hematopathology Service, Department of Pathology and Laboratory Medicine Memorial Sloan Kettering Cancer Center New York New York USA
| |
Collapse
|
11
|
Tsioulos G, Grigoropoulos I, Moschopoulos CD, Shapira S, Poulakou G, Antoniadou A, Boumpas D, Arber N, Tsiodras S. Insights into CD24 and Exosome Physiology and Potential Role in View of Recent Advances in COVID-19 Therapeutics: A Narrative Review. Life (Basel) 2022; 12:1472. [PMID: 36294907 PMCID: PMC9604962 DOI: 10.3390/life12101472] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/09/2022] [Accepted: 09/17/2022] [Indexed: 08/30/2023] Open
Abstract
Cluster of differentiation (CD) 24, a long-known protein with multifaceted functions, has gained attention as a possible treatment for Coronavirus Disease 19 (COVID-19) due to its known anti-inflammatory action. Extracellular vesicles (EVs), such as exosomes and microvesicles, may serve as candidate drug delivery platforms for novel therapeutic approaches in COVID-19 and various other diseases due to their unique characteristics. In the current review, we describe the physiology of CD24 and EVs and try to elucidate their role, both independently and as a combination, in COVID-19 therapeutics. CD24 may act as an important immune regulator in diseases with complex physiologies characterized by excessive inflammation. Very recent data outline a possible therapeutic role not only in COVID-19 but also in other similar disease states, e.g., acute respiratory distress syndrome (ARDS) and sepsis where immune dysregulation plays a key pathophysiologic role. On the other hand, CD24, as well as other therapeutic molecules, can be administered with the use of exosomes, exploiting their unique characteristics to create a novel drug delivery platform as outlined in recent clinical efforts. The implications for human therapeutics in general are huge with regard to pharmacodynamics, pharmacokinetics, safety, and efficacy that will be further elucidated in future randomized controlled trials (RCTs).
Collapse
Affiliation(s)
- Georgios Tsioulos
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Ioannis Grigoropoulos
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Charalampos D. Moschopoulos
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Shiran Shapira
- Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| | - Garyfallia Poulakou
- 3rd Department of Internal Medicine, Medical School, Sotiria General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Anastasia Antoniadou
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Dimitrios Boumpas
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Nadir Arber
- Integrated Cancer Prevention Center, Tel Aviv Medical Center, Tel Aviv 6423906, Israel
| | - Sotirios Tsiodras
- 4th Department of Internal Medicine, Medical School, University General Hospital Attikon, National and Kapodistrian University of Athens, 12462 Athens, Greece
| |
Collapse
|