1
|
Jiang C, Sun H, Jiang Z, Tian W, Cang S, Yu J. Targeting the CD47/SIRPα pathway in malignancies: recent progress, difficulties and future perspectives. Front Oncol 2024; 14:1378647. [PMID: 39040441 PMCID: PMC11261161 DOI: 10.3389/fonc.2024.1378647] [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: 01/30/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
Since its initial report in 2015, CD47 has garnered significant attention as an innate immune checkpoint, raising expectations to become the next "PD-1." The optimistic early stages of clinical development spurred a flurry of licensing deals for CD47-targeted molecules and company mergers or acquisitions for related assets. However, a series of setbacks unfolded recently, starting with the July 2023 announcement of discontinuing the phase 3 ENHANCE study on Magrolimab plus Azacitidine for higher-risk myelodysplastic syndromes (MDS). Subsequently, in August 2023, the termination of the ASPEN-02 program, assessing Evorpacept in combination with Azacitidine in MDS patients, was disclosed due to insufficient improvement compared to Azacitidine alone. These setbacks have cast doubt on the feasibility of targeting CD47 in the industry. In this review, we delve into the challenges of developing CD47-SIRPα-targeted drugs, analyze factors contributing to the mentioned setbacks, discuss future perspectives, and explore potential solutions for enhancing CD47-SIRPα-targeted drug development.
Collapse
Affiliation(s)
- Chenyang Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Oncology, Henan Key Laboratory for Precision Medicine in Cancer, Henan Provincial People’s Hospital, Henan University People’s Hospital and Zhengzhou University, Zhengzhou, Henan, China
| | - Hao Sun
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wenzhi Tian
- ImmuneOnco Biopharmaceuticals (Shanghai) Inc., Shanghai, China
| | - Shundong Cang
- Department of Oncology, Henan Key Laboratory for Precision Medicine in Cancer, Henan Provincial People’s Hospital, Henan University People’s Hospital and Zhengzhou University, Zhengzhou, Henan, China
| | - Jifeng Yu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
2
|
Rakoczy K, Kaczor J, Sołtyk A, Szymańska N, Stecko J, Drąg-Zalesińska M, Kulbacka J. The Immune Response of Cancer Cells in Breast and Gynecologic Neoplasms. Int J Mol Sci 2024; 25:6206. [PMID: 38892394 PMCID: PMC11172873 DOI: 10.3390/ijms25116206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Cancer diseases constitute a major health problem which leads to the death of millions of people annually. They are unique among other diseases because cancer cells can perfectly adapt to the environment that they create themselves. This environment is usually highly hostile and for normal cells it would be hugely difficult to survive, however neoplastic cells not only can survive but also manage to proliferate. One of the reasons is that they can alter immunological pathways which allow them to be flexible and change their phenotype to the one needed in specific conditions. The aim of this paper is to describe some of these immunological pathways that play significant roles in gynecologic neoplasms as well as review recent research in this field. It is of high importance to possess extensive knowledge about these processes, as greater understanding leads to creating more specialized therapies which may prove highly effective in the future.
Collapse
Affiliation(s)
- Katarzyna Rakoczy
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.)
| | - Justyna Kaczor
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.)
| | - Adam Sołtyk
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.)
| | - Natalia Szymańska
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.)
| | - Jakub Stecko
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland; (K.R.); (J.K.); (A.S.); (N.S.); (J.S.)
| | - Małgorzata Drąg-Zalesińska
- Department of Human Morphology and Embryology, Division of Histology and Embryology, Faculty of Medicine, Wroclaw Medical University, T. Chalubińskiego 6a, 50-368 Wroclaw, Poland;
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211a, 50-556 Wroclaw, Poland
- Department of Immunology and Bioelectrochemistry, State Research Institute Centre for Innovative Medicine Santariškių g. 5, LT-08406 Vilnius, Lithuania
| |
Collapse
|
3
|
Wu F, Pang H, Li F, Hua M, Song C, Tang J. Progress in cancer research on the regulator of phagocytosis CD47, which determines the fate of tumor cells (Review). Oncol Lett 2024; 27:256. [PMID: 38646501 PMCID: PMC11027102 DOI: 10.3892/ol.2024.14389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/21/2024] [Indexed: 04/23/2024] Open
Abstract
Cluster of differentiation 47 (CD47) is a transmembrane protein that is widely and moderately expressed on the surface of various cells and can have an essential role in mediating cell proliferation, migration, phagocytosis, apoptosis, immune homeostasis and other related responses by binding to its ligands, integrins, thrombospondin-1 and signal regulatory protein α. The poor prognosis of cancer patients is closely associated with high expression of CD47 in glioblastoma, ovarian cancer, breast cancer, bladder cancer, colon cancer and hepatocellular carcinoma. Upregulation of CD47 expression facilitates the growth of numerous types of tumor cells, while downregulation of its expression promotes phagocytosis of tumor cells by macrophages, thereby limiting tumor growth. In addition, blocking CD47 activates the cyclic GMP-AMP (cGAMP) synthase/cGAMP/interferon gene stimulating factor signaling pathway and initiates an adaptive immune response that kills tumor cells. The present review describes the structure, function and interactions of CD47 with its ligands, as well as its regulation of phagocytosis and tumor cell fate. It summarizes the therapeutics, mechanisms of action, research advances and challenges of targeting CD47. In addition, this paper provides an overview of the latest therapeutic options for targeting CD47, such as chimeric antigen receptor (CAR) T-cells, CAR macrophages and nanotechnology-based delivery systems, which are essential for future clinical research on targeting CD47.
Collapse
Affiliation(s)
- Fan Wu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Hongyuan Pang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Fan Li
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Mengqing Hua
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Chuanwang Song
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Jie Tang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| |
Collapse
|
4
|
Luo X, Mo J, Zhang M, Huang W, Bao Y, Zou R, Yao L, Yuan L. CD47-a novel prognostic predicator in epithelial ovarian cancer and correlations with clinicopathological and gene mutation features. World J Surg Oncol 2024; 22:44. [PMID: 38317230 PMCID: PMC10845810 DOI: 10.1186/s12957-024-03308-6] [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: 10/09/2023] [Accepted: 01/13/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND Epithelial ovarian cancer (EOC) is insensitive to immunotherapy due to its poor immunogenicity; thus, suitable biomarkers need to be identified for better prognostic stratification and individualized treatment. CD47 is a novel immunotherapy target; however, its impact on EOC prognosis is controversial and correlation with genetic features is unclear. The aim of this study was to investigate the prognostic significance of CD47 and its correlations with biological behaviors and genetic features of EOC. METHODS Immunohistochemistry (IHC) and next-generation sequencing (NGS) were performed to examine expressions of CD47, PD-L1, and genomic mutations in the tissue samples of 75 EOC patients. Various clinicopathologic and genomic features were then evaluated to determine their correlation with CD47 expression. Kaplan-Meier analysis and Cox regression analysis were used to identify independent prognostic factors. Risk score modeling was then established, and the predictive capacity of this model was further confirmed by nomogram analysis. RESULTS CD47 was mainly expressed in the tumor cell membrane and cytoplasm, and the rate of high CD47 expression was 63.7%. CD47 expression was associated with various clinicopathological factors, including FIGO stage, CA125 and HE4 value, presence of multidisciplinary surgeries, presence and volume of ascites, lymph-node metastasis, Ki-67 index and platinum-resistant, as well as genetic characteristics like BRCA mutation, HRD status, and TP53 mutation in EOC. Patients with high CD47 expression showed worse prognosis than the low-expression group. Cox regression analysis demonstrated that CA125, CD47, and BRCA mutation were independent factors for EOC prognosis. Patients were then categorized into high-risk and low-risk subgroups based on the risk score of the aforementioned independent factors, and the prognosis of the high-risk group was worse than those of the low-risk group. The nomogram showed adequate discrimination with a concordance index of 0.777 (95% CI, 0.732-0.822). The calibration curve showed good consistency. CONCLUSION CD47 correlated with various malignant biology and genetic characteristics of EOC and may play pivotal and multifaceted roles in the tumor microenvironment of EOC Finally, we constructed a reliable prediction model centered on CD47 and integrated CA125 and BRCA to better guide high-risk population management.
Collapse
Affiliation(s)
- Xukai Luo
- Department of Gynecological Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Jiahang Mo
- Institute of Reproduction and Development, Fudan University, Shanghai, 200011, China
| | - Min Zhang
- Department of Gynecological Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Wu Huang
- Department of Gynecological Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Yiting Bao
- Department of Gynecological Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Ruoyao Zou
- Department of Gynecological Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Liangqing Yao
- Department of Gynecological Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China
| | - Lei Yuan
- Department of Gynecological Oncology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
5
|
Cheng Y, Wang P, Liu L. PFKFB3 Regulates the Growth and Migration of Ovarian Cancer Cells through Pyroptosis and Warburg Effect Progression. J Environ Pathol Toxicol Oncol 2024; 43:53-64. [PMID: 39016141 DOI: 10.1615/jenvironpatholtoxicoloncol.2024052948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2024] Open
Abstract
Ovarian cancer is one of the most common malignant tumors in female reproductive organs. Its incidence rate is second only to uterine body cancer and cervical cancer, posing a serious threat to women's health. Herein, we explored that PFKFB3 in cancer progression of ovarian cancer and its underlying mechanism. All the serum samples from ovarian cancer were collected by our hospital. PFKFB3 mRNA expressions in patients with ovarian cancer and ovarian cancer cell lines were up-regulated. PFKFB3 protein expressions in ovarian cancer cells were induced. ovarian cancer patients with high PFKFB3expression had lower survival rate. The PFKFB3gene promoted cell proliferation and EDU cells, and increased cell metastasis of ovarian cancer. Si-PFKFB3 reduced cell proliferation and EDU cells, and decreased cell metastasis of ovarian cancer. PFKFB3 gene up-regulation reduced caspase-3/9 activity levels of ovarian cancer. Si-PFKFB3 also promoted caspase-3/9 activity levels of ovarian cancer. PFKFB3 gene promoted Warburg effect progression of ovarian cancer. PFKFB3 gene reduced NLRP3-induced pyroptosis of ovarian cancer. PFKFB3 suppressed NLRP3 expression. NLRP3 was one target spot for PFKFB3 on pyroptosis of ovarian cancer. Taken together, we conclude that PFKFB3 suppressed NLRP3 axis to reduce pyroptosis and increase Warburg effect progression of ovarian cancer, and provide molecular insight into the mechanisms by which the PFKFB3 regulates pyroptosis of ovarian cancer.
Collapse
Affiliation(s)
- Ye Cheng
- Department of Obstetrics and Gynecology, Wuhan No. 1 Hospital, Wuhan 430030, China
| | - Ping Wang
- Department of Obstetrics and Gynecology, Wuhan No. 1 Hospital, Wuhan 430030, China
| | | |
Collapse
|
6
|
Calvillo-Rodríguez KM, Lorenzo-Anota HY, Rodríguez-Padilla C, Martínez-Torres AC, Scott-Algara D. Immunotherapies inducing immunogenic cell death in cancer: insight of the innate immune system. Front Immunol 2023; 14:1294434. [PMID: 38077402 PMCID: PMC10701401 DOI: 10.3389/fimmu.2023.1294434] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/01/2023] [Indexed: 12/18/2023] Open
Abstract
Cancer immunotherapies include monoclonal antibodies, cytokines, oncolytic viruses, cellular therapies, and other biological and synthetic immunomodulators. These are traditionally studied for their effect on the immune system's role in eliminating cancer cells. However, some of these therapies have the unique ability to directly induce cytotoxicity in cancer cells by inducing immunogenic cell death (ICD). Unlike general immune stimulation, ICD triggers specific therapy-induced cell death pathways, based on the release of damage-associated molecular patterns (DAMPs) from dying tumour cells. These activate innate pattern recognition receptors (PRRs) and subsequent adaptive immune responses, offering the promise of sustained anticancer drug efficacy and durable antitumour immune memory. Exploring how onco-immunotherapies can trigger ICD, enhances our understanding of their mechanisms and potential for combination strategies. This review explores the complexities of these immunotherapeutic approaches that induce ICD, highlighting their implications for the innate immune system, addressing challenges in cancer treatment, and emphasising the pivotal role of ICD in contemporary cancer research.
Collapse
Affiliation(s)
- Kenny Misael Calvillo-Rodríguez
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
| | - Helen Yarimet Lorenzo-Anota
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
- The Institute for Obesity Research, Tecnológico de Monterrey, Monterrey, NL, Mexico
| | - Cristina Rodríguez-Padilla
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
| | - Ana Carolina Martínez-Torres
- Laboratorio de Inmunología y Virología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
| | - Daniel Scott-Algara
- Département d'Immunologie, Unité de Biologie Cellulaire des Lymphocytes, Pasteur Institute, Paris, France
| |
Collapse
|
7
|
CHEN QIUQIANG, GUO XUEJUN, MA WENXUE. Opportunities and challenges of CD47-targeted therapy in cancer immunotherapy. Oncol Res 2023; 32:49-60. [PMID: 38188674 PMCID: PMC10767231 DOI: 10.32604/or.2023.042383] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/09/2023] [Indexed: 01/09/2024] Open
Abstract
Cancer immunotherapy has emerged as a promising strategy for the treatment of cancer, with the tumor microenvironment (TME) playing a pivotal role in modulating the immune response. CD47, a cell surface protein, has been identified as a crucial regulator of the TME and a potential therapeutic target for cancer therapy. However, the precise functions and implications of CD47 in the TME during immunotherapy for cancer patients remain incompletely understood. This comprehensive review aims to provide an overview of CD47's multifaced role in TME regulation and immune evasion, elucidating its impact on various types of immunotherapy outcomes, including checkpoint inhibitors and CAR T-cell therapy. Notably, CD47-targeted therapies offer a promising avenue for improving cancer treatment outcomes, especially when combined with other immunotherapeutic approaches. The review also discusses current and potential CD47-targeted therapies being explored for cancer treatment and delves into the associated challenges and opportunities inherent in targeting CD47. Despite the demonstrated effectiveness of CD47-targeted therapies, there are potential problems, including unintended effects on healthy cells, hematological toxicities, and the development if resistance. Consequently, further research efforts are warranted to fully understand the underlying mechanisms of resistance and to optimize CD47-targeted therapies through innovative combination approaches, ultimately improving cancer treatment outcomes. Overall, this comprehensive review highlights the significance of CD47 as a promising target for cancer immunotherapy and provides valuable insight into the challenges and opportunities in developing effective CD47-targeted therapies for cancer treatment.
Collapse
Affiliation(s)
- QIUQIANG CHEN
- Key Laboratory for Translational Medicine, The First Affiliated Hospital, Huzhou University School of Medicine, Huzhou, 313000, China
| | - XUEJUN GUO
- Department of Hematology, Puyang Youtian General Hospital, Puyang, 457001, China
| | - WENXUE MA
- Department of Medicine, Moores Cancer Center, Sanford Stem Cell Institute, University of California San Diego, La Jolla, San Diego, 92093, USA
| |
Collapse
|
8
|
Aizaz M, Khan A, Khan F, Khan M, Musad Saleh EA, Nisar M, Baran N. The cross-talk between macrophages and tumor cells as a target for cancer treatment. Front Oncol 2023; 13:1259034. [PMID: 38033495 PMCID: PMC10682792 DOI: 10.3389/fonc.2023.1259034] [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: 07/15/2023] [Accepted: 10/17/2023] [Indexed: 12/02/2023] Open
Abstract
Macrophages represent an important component of the innate immune system. Under physiological conditions, macrophages, which are essential phagocytes, maintain a proinflammatory response and repair damaged tissue. However, these processes are often impaired upon tumorigenesis, in which tumor-associated macrophages (TAMs) protect and support the growth, proliferation, and invasion of tumor cells and promote suppression of antitumor immunity. TAM abundance is closely associated with poor outcome of cancer, with impediment of chemotherapy effectiveness and ultimately a dismal therapy response and inferior overall survival. Thus, cross-talk between cancer cells and TAMs is an important target for immune checkpoint therapies and metabolic interventions, spurring interest in it as a therapeutic vulnerability for both hematological cancers and solid tumors. Furthermore, targeting of this cross-talk has emerged as a promising strategy for cancer treatment with the antibody against CD47 protein, a critical macrophage checkpoint recognized as the "don't eat me" signal, as well as other metabolism-focused strategies. Therapies targeting CD47 constitute an important milestone in the advancement of anticancer research and have had promising effects on not only phagocytosis activation but also innate and adaptive immune system activation, effectively counteracting tumor cells' evasion of therapy as shown in the context of myeloid cancers. Targeting of CD47 signaling is only one of several possibilities to reverse the immunosuppressive and tumor-protective tumor environment with the aim of enhancing the antitumor response. Several preclinical studies identified signaling pathways that regulate the recruitment, polarization, or metabolism of TAMs. In this review, we summarize the current understanding of the role of macrophages in cancer progression and the mechanisms by which they communicate with tumor cells. Additionally, we dissect various therapeutic strategies developed to target macrophage-tumor cell cross-talk, including modulation of macrophage polarization, blockade of signaling pathways, and disruption of physical interactions between leukemia cells and macrophages. Finally, we highlight the challenges associated with tumor hypoxia and acidosis as barriers to effective cancer therapy and discuss opportunities for future research in this field.
Collapse
Affiliation(s)
- Muhammad Aizaz
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, China
| | - Aakif Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Faisal Khan
- Centre of Excellence in Molecular Biology, University of the Punjab, Lahore, Pakistan
| | - Maria Khan
- Center of Biotechnology and Microbiology, University of Peshawar, Peshawar, Pakistan
| | - Ebraheem Abdu Musad Saleh
- Department of Chemistry, College of Arts & Science, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Maryum Nisar
- School of Interdisciplinary Engineering & Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Natalia Baran
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
9
|
Chen Y, Zhu X, Liu H, Wang C, Chen Y, Wang H, Fang Y, Wu X, Xu Y, Li C, Lv X, Huang J, Han X, Li R, Hong W, Yu Z, Wei W, Tu J. The application of HER2 and CD47 CAR-macrophage in ovarian cancer. J Transl Med 2023; 21:654. [PMID: 37740183 PMCID: PMC10517545 DOI: 10.1186/s12967-023-04479-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/28/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND The chimeric antigen receptor (CAR)-T therapy has a limited therapeutic effect on solid tumors owing to the limited CAR-T cell infiltration into solid tumors and the inactivation of CAR-T cells by the immunosuppressive tumor microenvironment. Macrophage is an important component of the innate and adaptive immunity, and its unique phagocytic function has been explored to construct CAR macrophages (CAR-Ms) against solid tumors. This study aimed to investigate the therapeutic application of CAR-Ms in ovarian cancer. METHODS In this study, we constructed novel CAR structures, which consisted of humanized anti-HER2 or CD47 scFv, CD8 hinge region and transmembrane domains, as well as the 4-1BB and CD3ζ intracellular domains. We examined the phagocytosis of HER2 CAR-M and CD47 CAR-M on ovarian cancer cells and the promotion of adaptive immunity. Two syngeneic tumor models were used to estimate the in vivo antitumor activity of HER2 CAR-M and CD47 CAR-M. RESULTS We constructed CAR-Ms targeting HER2 and CD47 and verified their phagocytic ability to ovarian cancer cells in vivo and in vitro. The constructed CAR-Ms showed antigen-specific phagocytosis of ovarian cancer cells in vitro and could activate CD8+ cytotoxic T lymphocyte (CTL) to secrete various anti-tumor factors. For the in vivo model, mice with human-like immune systems were used. We found that CAR-Ms enhanced CD8+ T cell activation, affected tumor-associated macrophage (TAM) phenotype, and led to tumor regression. CONCLUSIONS We demonstrated the inhibition effect of our constructed novel HER2 CAR-M and CD47 CAR-M on target antigen-positive ovarian cancer in vitro and in vivo, and preliminarily verified that this inhibitory effect is due to phagocytosis, promotion of adaptive immunity and effect on tumor microenvironment.
Collapse
Affiliation(s)
- Yizhao Chen
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, #81 Meishan Road, Shushan District, Hefei, China
| | - Xiangling Zhu
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, #81 Meishan Road, Shushan District, Hefei, China
| | - Hanze Liu
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Shushan District, Hefei, China
| | - Cunzhi Wang
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Shushan District, Hefei, China
| | - Yu Chen
- Department of Gynecology, Health Science Center, The First Affiliated Hospital of Shenzhen University, #3002 Sungangxi Road, Futian District, Shenzhen, China
| | - Huihui Wang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, #81 Meishan Road, Shushan District, Hefei, China
| | - Yilong Fang
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, #81 Meishan Road, Shushan District, Hefei, China
| | - Xuming Wu
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, #81 Meishan Road, Shushan District, Hefei, China
| | - Yuting Xu
- Department of Gynecology, Health Science Center, The First Affiliated Hospital of Shenzhen University, #3002 Sungangxi Road, Futian District, Shenzhen, China
| | - Chunhua Li
- Department of Gynecology, Health Science Center, The First Affiliated Hospital of Shenzhen University, #3002 Sungangxi Road, Futian District, Shenzhen, China
| | - Xinyue Lv
- Department of Gynecology, Health Science Center, The First Affiliated Hospital of Shenzhen University, #3002 Sungangxi Road, Futian District, Shenzhen, China
| | - Jinghua Huang
- Department of Gynecology, Health Science Center, The First Affiliated Hospital of Shenzhen University, #3002 Sungangxi Road, Futian District, Shenzhen, China
| | - Xintong Han
- Department of Gynecology, Health Science Center, The First Affiliated Hospital of Shenzhen University, #3002 Sungangxi Road, Futian District, Shenzhen, China
| | - Ruilin Li
- Department of Pharmacy, The Third Affiliated Hospital of Anhui Medical University, Hefei First People's Hospital, Hefei, China
| | - Wenming Hong
- Department of Neurosurgery, First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Shushan District, Hefei, China.
| | - Zhiying Yu
- Department of Gynecology, Health Science Center, The First Affiliated Hospital of Shenzhen University, #3002 Sungangxi Road, Futian District, Shenzhen, China.
- Shenzhen Second People's Hospital, #3002 Sungangxi Road, Futian District, Shenzhen, China.
| | - Wei Wei
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, #81 Meishan Road, Shushan District, Hefei, China.
| | - Jiajie Tu
- Institute of Clinical Pharmacology, Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, #81 Meishan Road, Shushan District, Hefei, China.
| |
Collapse
|
10
|
El-Tanani M, Nsairat H, Matalka II, Aljabali AAA, Mishra V, Mishra Y, Naikoo GA, Chava SR, Charbe NB, Tambuwala MM. Impact of exosome therapy on pancreatic cancer and its progression. Med Oncol 2023; 40:225. [PMID: 37405480 PMCID: PMC10322774 DOI: 10.1007/s12032-023-02101-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/19/2023] [Indexed: 07/06/2023]
Abstract
Pancreatic cancer, one of the most aggressive tumors, has a dismal prognosis because of the low rates of early identification, fast progression, difficulties following surgery, and the ineffectiveness of current oncologic therapies. There are no imaging techniques or biomarkers that can accurately identify, categorize, or predict the biological behavior of this tumor. Exosomes are extracellular vesicles that play a crucial rule in the progression, metastasis, and chemoresistance of pancreatic cancer. They have been verified to be potential biomarkers for pancreatic cancer management. Studying the role of exosomes in pancreatic cancer is substantial. Exosomes are secreted by most eukaryotic cells and participated in intercellular communication. The components of exosomes, including proteins, DNA, mRNA, microRNA, long non-coding RNA, circular RNA, etc., play a crucial role in regulating tumor growth, metastasis, and angiogenesis in the process of cancer development, and can be used as a prognostic marker and/or grading basis for tumor patients. Hereby, in this concise review, we intend to summarize exosomes components and isolation, exosome secretion, function, importance of exosomes in the progression of pancreatic cancer and exosomal miRNAs as possible pancreatic cancer biomarkers. Finally, the application potential of exosomes in the treatment of pancreatic cancer, which provides theoretical supports for using exosomes to serve precise tumor treatment in the clinic, will be discussed.
Collapse
Affiliation(s)
- Mohamed El-Tanani
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan.
- Institute of Cancer Therapeutics, University of Bradford, Bradford, BD7 1DP, West Yorkshire, UK.
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE.
| | - Hamdi Nsairat
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, 19328, Jordan
| | - Ismail I Matalka
- Ras Al Khaimah Medical and Health Sciences University, Ras Al Khaimah, UAE
- Department of Pathology and Microbiology, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, P.O. Box 566, Irbid, 21163, Jordan
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144411, India
| | - Yachana Mishra
- Department of Zoology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, India
| | - Gowhar A Naikoo
- Department of Mathematics and Sciences, College of Arts and Applied Sciences, Dhofar University, PC 211, Salalah, Oman
| | | | - Nitin B Charbe
- Department of Pharmaceutics, College of Pharmacy, Center for Pharmacometrics and Systems Pharmacology, University of Florida, Orlando, FL, USA
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln, LN6 7TS, UK.
| |
Collapse
|