1
|
Lin P, Gao R, Yang W, Fang Z, Wang Z, Yu M, Xu L, Ma Z, Fang J, Yu W. Platelet membrane-cloaked biomimetic nanoparticles for targeted acute lung injury therapy. Colloids Surf B Biointerfaces 2025; 250:114542. [PMID: 39893893 DOI: 10.1016/j.colsurfb.2025.114542] [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: 10/25/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/04/2025]
Abstract
Acute lung injury (ALI) is a medical condition characterized by significant morbidity and elevated mortality rates; however, to date, there are no clinically approved pharmacological interventions that are both safe and effective for its treatment. In the pathophysiology of ALI, a robust inflammatory response is a critical factor. Dexamethasone (Dex), a potent glucocorticoid, is commonly employed in clinical settings to manage inflammatory conditions. However, the frequent or high-dose administration of corticosteroids can result in significant adverse effects and long-term complications. In this study, we have developed a biomimetic anti-inflammatory nanosystem, designated PM-LPs@Dex, aimed at treating ALI. This system leverages the inherent affinity of platelets for sites of inflammation, alongside the advantageous drug encapsulation properties of liposomes (LPs). By harnessing the suitable physicochemical characteristics of LPs and the distinctive biological functions of platelet membranes (PM), PM-LPs@Dex is capable of stable and sustained drug release in vitro. Experimental results regarding cellular uptake and biodistribution reveal that PM-LPs@Dex is preferentially internalized by inflammatory cells and exhibits enhanced accumulation in inflamed lung tissue compared to LPs@Dex. Pharmacokinetic studies displayed that PM-LPs@Dex showed prolonged circulation time in blood. Additionally, pharmacodynamic assessments demonstrate that PM-LPs@Dex significantly mitigates the severity of ALI, as evidenced by reductions in pulmonary edema, tissue pathology, bronchoalveolar lavage cell counts, protein concentration, and levels of inflammatory cytokines. Notably, PM-LPs@Dex also exhibits favorable biocompatibility. This research is anticipated to contribute novel strategies for the safe and effective targeted management of inflammatory diseases.
Collapse
Affiliation(s)
- Peihong Lin
- School of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People' Hospital), Hangzhou Medical College, Hangzhou 310013, China
| | - Rui Gao
- School of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People' Hospital), Hangzhou Medical College, Hangzhou 310013, China
| | - Wenjing Yang
- School of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People' Hospital), Hangzhou Medical College, Hangzhou 310013, China
| | - Zhengyu Fang
- School of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People' Hospital), Hangzhou Medical College, Hangzhou 310013, China
| | - Zhouru Wang
- School of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People' Hospital), Hangzhou Medical College, Hangzhou 310013, China
| | - Mengdie Yu
- School of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People' Hospital), Hangzhou Medical College, Hangzhou 310013, China
| | - Lihua Xu
- Department of Pharmacy, The First People's Hospital of Xiaoshan District, Hangzhou 310013, China
| | - Zhen Ma
- School of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People' Hospital), Hangzhou Medical College, Hangzhou 310013, China
| | - Jie Fang
- School of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People' Hospital), Hangzhou Medical College, Hangzhou 310013, China.
| | - Wenying Yu
- School of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People' Hospital), Hangzhou Medical College, Hangzhou 310013, China.
| |
Collapse
|
2
|
Yu WB, Ye ZH, Shi JJ, Deng WQ, Chen J, Lu JJ. Dual blockade of GSTK1 and CD47 improves macrophage-mediated phagocytosis on cancer cells. Biochem Pharmacol 2025; 236:116898. [PMID: 40147800 DOI: 10.1016/j.bcp.2025.116898] [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: 12/09/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
CD47 is a crucial anti-phagocytic signal in regulating macrophage responses and its manipulation offers the therapeutic potential in cancer treatment. However, in many cases, blockade of CD47 by itself is insufficient to activate macrophage effectively, indicating other unidentified phagocytosis-regulating factors to resist the macrophage activity. In this study, a genome-wide human CRISPR-Cas9 library was developed for comprehensive screening of phagocytosis-regulating factors in the context of CD47 blockade. The screening results identified GSTK1 as a potential anti-phagocytic signal counteracting the efficacy of CD47-based phagocytosis. The disruption of GSTK1 significantly increased the phagocytosis rate of cancer cells by macrophages in combination with anti-CD47 antibody. Further mechanism investigation unveiled that GSTK1 blockade increased the membrane exposure of calreticulin in different cancer cells, which might be the primary mechanism driving enhanced macrophage-mediated phagocytosis. To this end, siGSTK1-loaded nanoparticles (siGSTK1-LNPs) were designed to suppress the GSTK1 expression efficiently. The comparable phagocytosis efficacy was also observed when combining siGSTK1-LNPs with anti-CD47 antibody. Above all, GSTK1 blockade was identified as a promising and feasible stimulus for enhancing the effectiveness of anti-CD47 antibody, introducing a novel and effective combination approach in cancer immunotherapy.
Collapse
Affiliation(s)
- Wei-Bang Yu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jia-Jie Shi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Wei-Qing Deng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jun Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macau, China; MoE Frontiers Science Center for Precision Oncology, University of Macau, Macau, China.
| |
Collapse
|
3
|
Chen M, Zhou Y, Bao K, Chen S, Song G, Wang S. Multispecific Antibodies Targeting PD-1/PD-L1 in Cancer. BioDrugs 2025; 39:427-444. [PMID: 40106158 DOI: 10.1007/s40259-025-00712-6] [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] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
The development of immune checkpoint inhibitors has revolutionized the treatment of patients with cancer. Targeting the programmed cell death protein 1 (PD-1)/programmed cell death 1 ligand 1(PD-L1) interaction using monoclonal antibodies has emerged as a prominent focus in tumor therapy with rapid advancements. However, the efficacy of anti-PD-1/PD-L1 treatment is hindered by primary or acquired resistance, limiting the effectiveness of single-drug approaches. Moreover, combining PD-1/PD-L1 with other immune drugs, targeted therapies, or chemotherapy significantly enhances response rates while exacerbating adverse reactions. Multispecific antibodies, capable of binding to different epitopes, offer improved antitumor efficacy while reducing drug-related side effects, serving as a promising therapeutic approach in cancer treatment. Several bispecific antibodies (bsAbs) targeting PD-1/PD-L1 have received regulatory approval, and many more are currently in clinical development. Additionally, tri-specific antibodies (TsAbs) and tetra-specific antibodies (TetraMabs) are under development. This review comprehensively explores the fundamental structure, preclinical principles, clinical trial progress, and challenges associated with bsAbs targeting PD-1/PD-L1.
Collapse
Affiliation(s)
- Miaomiao Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Yuli Zhou
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kaicheng Bao
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Siyu Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Guoqing Song
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China.
| | - Siliang Wang
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China.
| |
Collapse
|
4
|
Cai W, Fan T, Xiao C, Deng Z, Liu Y, Li C, He J. Neutrophils in cancer: At the crucial crossroads of anti-tumor and pro-tumor. Cancer Commun (Lond) 2025. [PMID: 40296668 DOI: 10.1002/cac2.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
Neutrophils are important components of the immune system and play a key role in defending against pathogenic infections and responding to inflammatory cues, including cancer. Their dysregulation indicates potential disease risk factors. However, their functional importance in disease progression has often been underestimated due to their short half-life, especially as there is limited information on the role of intratumoral neutrophils. Recent studies on their prominent role in cancer have led to a paradigm shift in our understanding of the functional diversity of neutrophils. These studies highlight that neutrophils have emerged as key components of the tumor microenvironment, where they can play a dual role in promoting and suppressing cancer. Moreover, several approaches to therapeutically target neutrophils have emerged, and clinical trials are investigating their efficacy. In this review, we discussed the involvement of neutrophils in cancer initiation and progression. We summarized recent advances in therapeutic strategies targeting neutrophils and, most importantly, suggested future research directions that could facilitate the manipulation of neutrophils for therapeutic purposes in cancer patients.
Collapse
Affiliation(s)
- Wenpeng Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Yixiao Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, P. R. China
| |
Collapse
|
5
|
Sevilla CM, Mijacika A, Somoza B, Osorio JC. Protocol for assessing antibody-dependent cellular phagocytosis by primary murine and human macrophages. STAR Protoc 2025; 6:103787. [PMID: 40279242 DOI: 10.1016/j.xpro.2025.103787] [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: 10/10/2024] [Revised: 02/04/2025] [Accepted: 04/04/2025] [Indexed: 04/27/2025] Open
Abstract
Macrophages play a crucial role in mediating antibody-dependent cellular phagocytosis (ADCP), a process that enhances the effectiveness of several cancer immunotherapies. In this protocol, we outline detailed steps for isolating, differentiating, and polarizing macrophages from both mouse and human sources. Additionally, we describe an optimized technique to assess ADCP using in vitro co-culture studies followed by flow cytometry analysis. This protocol offers a reliable approach to evaluate the efficiency of macrophage-mediated phagocytosis of cancer cells during treatment with antibody-based therapies. For complete details on the use and execution of this protocol, please refer to Osorio et al.1.
Collapse
Affiliation(s)
- Carlo Miguel Sevilla
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA
| | - Andrew Mijacika
- Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA
| | - Bella Somoza
- Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Juan C Osorio
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Laboratory of Molecular Genetics and Immunology, Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
6
|
Zhao Y, Duan M, Qi Y, Xia J, Hao C, Yao W. Innate immune checkpoint SIRPα/CD47 blockade ameliorates silica-induced pulmonary fibrosis by modulating macrophage immunity. Int Immunopharmacol 2025; 156:114723. [PMID: 40279943 DOI: 10.1016/j.intimp.2025.114723] [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: 03/11/2025] [Revised: 04/12/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
Silicosis is a fibrotic disease caused by prolonged inhalation of silica particles. Signal regulatory protein alpha (SIRPα) and its ligand CD47, key innate immune checkpoints mediating inhibition of phagocytosis, have been reported to regulate organ fibrosis. However, the role of SIRPα/CD47 in silicosis remains unexplored. In this study, a silicosis mouse model was constructed and revealed a significant upregulation of SIRPα and CD47 expression in lung tissue with disease progression. In addition, the expression patterns of SIRPα and CD47 in various silicosis effector cells exhibit distinct cell specificity. Using RRx-001 to block SIRPα/CD47 signaling in mice, we observed a marked reduction in lung injury, decreased collagen deposition, and improved pulmonary function. Mechanistically, blocking SIRPα/CD47 affected T cell activation, macrophage polarization and the expression of pro-inflammatory and pro-fibrotic factors. In vitro, we found that inhibiting SIRPα/CD47 countered the silica-induced suppression of macrophage phagocytosis and induced macrophage polarization towards the M1 phenotype. Additionally, levels of soluble SIRPα and CD47 in the peripheral blood of silicosis patients were significantly higher than those in healthy controls. In summary, this study demonstrates that SIRPα/CD47-mediated immunomodulatory signaling is the driving factor for the progression of silicosis, and this pathway might serve as a therapeutic target for silicosis treatment.
Collapse
Affiliation(s)
- Youliang Zhao
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Meixiu Duan
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Yuanmeng Qi
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Jiarui Xia
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China
| | - Changfu Hao
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China.
| | - Wu Yao
- Department of Occupational Health and Occupational Disease, College of Public Health, Zhengzhou University, Zhengzhou, Henan, PR China.
| |
Collapse
|
7
|
Kim M, Choi R, Kim L, Kim YC, Noh I. Cell membrane nanoparticles in cancer therapy: From basic structure to surface functionalization. J Control Release 2025:113752. [PMID: 40254140 DOI: 10.1016/j.jconrel.2025.113752] [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: 02/21/2025] [Revised: 04/06/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Cell membrane nanoparticles (CNPs) have recently garnered significant attention as effective drug-delivery vehicles. Beyond their simple function of encapsulating cargo within a lipid bilayer structure, the cell membrane is a complex entity derived from biological materials, presenting a variety of surface proteins and glycans. Notable features that enhance their effectiveness as delivery vehicles include the inhibition of protein corona formation in the plasma and the suppression of macrophage phagocytosis, both of which contribute to prolonged blood circulation. Furthermore, CNPs exhibit homotypic targeting effects toward their cells of origin, resulting in reduced side effects, and because they are not xenobiotics, the likelihood of nonspecific immune activation is also minimized. This review focuses on various applications of CNPs in cancer therapeutic studies, examining their structural evolution and surface engineering developments. We introduce studies that leverage the inherent functionality of cell membranes and recent research in functional CNPs synthesized through genetic or chemical engineering methods. Through this review, we aim to trace the progression of CNP research, explore potential directions for their use in biomedical applications, and assess the prospects for clinical trials.
Collapse
Affiliation(s)
- Munsik Kim
- Department of Medical Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea; Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea; Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291, Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Rohbin Choi
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291, Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Lian Kim
- Department of Medical Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yeu-Chun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291, Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea.
| | - Ilkoo Noh
- Department of Medical Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea; Institute of Bioscience & Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea.
| |
Collapse
|
8
|
Du W, Chen C, Liu Y, Quan H, Xu M, Liu J, Song P, Fang Z, Yue Z, Xu H, Ling Y, Duan J, He F, Wang L. A combined "eat me/don't eat me" strategy based on exosome for acute liver injury treatment. Cell Rep Med 2025; 6:102033. [PMID: 40120577 DOI: 10.1016/j.xcrm.2025.102033] [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: 07/04/2024] [Revised: 12/09/2024] [Accepted: 02/24/2025] [Indexed: 03/25/2025]
Abstract
Drug-induced liver injury (DILI) involves multifaceted pathogenesis, necessitating effective therapeutic strategies. Wnt2, secreted by liver sinusoidal endothelial cell (LSEC), activates the Wnt/β-catenin signaling pathway to promote hepatocyte proliferation after injury. To address the dual challenges of targeted delivery and phagocytosis evasion, we develop a combined "eat me/don't eat me" strategy. RLTRKRGLK (RLTR) peptide-functionalized exosomes are engineered by inserting DMPE-PEG2000-CRLTRKRGLK into the lipid membrane of exosome derived from bEnd.3 cell. Surface-displayed RLTR mediates exosomal enrichment in LSEC, while CD47 engineering reduces macrophage clearance via "don't eat me" signaling. Then, lentiviral transfection enables stable encapsulation of functional Wnt2 mRNA into ExoCD47 (designated Wnt2@ExoCD47). In both acetaminophen (APAP) and dimethylnitrosamine (DMN)-induced murine liver injury models, RLTR-Wnt2@ExoCD47 demonstrates LSEC-specific targeting and significant hepatoprotection. This engineered exosome platform provides a therapeutic strategy for DILI.
Collapse
Affiliation(s)
- Wei Du
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Chen Chen
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - YingYing Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Huiyi Quan
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Ming Xu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - JingJing Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Ping Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - ZhiQiang Fang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - ZhenSheng Yue
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Hao Xu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - YuWei Ling
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - JuanLi Duan
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Fei He
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
9
|
Jiang S, Guo F, Li L. Biological mechanisms and immunotherapy of brain metastases in non-small cell lung cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189320. [PMID: 40220878 DOI: 10.1016/j.bbcan.2025.189320] [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: 01/26/2025] [Revised: 04/07/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related mortality worldwide, with Brain Metastases serving as a significant adverse prognostic factor. The blood-brain barrier poses a substantial challenge in the treatment of brain metastases, as it restricts the penetration of many anticancer agents. Novel immunotherapy, such as immune checkpoint inhibitors (ICIs) have emerged as promising treatment for NSCLC and its associated brain metastases. This review summarizes the biological mechanism underlying NSCLC brain metastases and provides an overview of the current landscape of immunotherapy, exploring the mechanism of action and clinical applications of these advanced treatments.
Collapse
Affiliation(s)
- Sitong Jiang
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Fengzhu Guo
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Lin Li
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
10
|
Liao H, Chen M, Liao Z, Luo Y, Chen S, Wang L, Wang Z, Niu C. MnO 2-based nanoparticles remodeling tumor micro-environment to augment sonodynamic immunotherapy against breast cancer. Biomater Sci 2025. [PMID: 40202432 DOI: 10.1039/d5bm00189g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/10/2025]
Abstract
The tumor microenvironment (TME) is characterized by a complex array of factors, including aerobic conditions, high glutathione (GSH) levels, acidic pH, and elevated hydrogen peroxide (H2O2) content, all of which promote cancer progression and contribute to poor prognosis. Fortunately, these challenges can be addressed using MnO2-based nanomaterials. In this study, we have designed and synthesized a Curcumin/MnO2@PLGA@4T1 cell membrane (CMP@4T1m) system aimed at remodelling the TME and enhancing sonodynamic immunotherapy for breast cancer. Through the homologous targeting ability of 4T1m, CMP@4T1m efficiently accumulates at the tumor site. Upon ultrasound irradiation, curcumin (Cur) acts as a sonosensitizer, generating cytotoxic reactive oxygen species (ROS) that induce immunogenic cell death (ICD), activate T-cell responses, and repolarize protumoral M2-like macrophages to antitumoral M1-like macrophages. In the TME, which is mildly acidic and enriched with GSH and H2O2, MnO2 not only oxidizes GSH to glutathione disulfide (GSSG) but also reacts with H2O2 and H+ to produce oxygen, alleviating hypoxia and significantly enhancing the sonodynamic immunotherapy effect. Additionally, Mn2+ generated during this process converts H2O2 into cytotoxic hydroxyl radicals (˙OH). This study thus lays the foundation for advancing cancer nanomedicine, offering a novel approach that integrates TME remodelling with sonodynamic immunotherapy.
Collapse
Affiliation(s)
- Haiqin Liao
- Department of Ultrasound, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
- Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Clinical Research Center for Ultrasound and Treatment in Hunan Province, Hunan 410011, China
| | - Mingyu Chen
- Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Clinical Research Center for Ultrasound and Treatment in Hunan Province, Hunan 410011, China
| | - Zhipeng Liao
- Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Clinical Research Center for Ultrasound and Treatment in Hunan Province, Hunan 410011, China
| | - Yi Luo
- Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Clinical Research Center for Ultrasound and Treatment in Hunan Province, Hunan 410011, China
| | - Sijie Chen
- Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Clinical Research Center for Ultrasound and Treatment in Hunan Province, Hunan 410011, China
| | - Long Wang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Engineering Research Center of Biomedical Metal and Ceramic Implants, Xiangya Hospital, Central South University, Changsha, China
| | - Zhigang Wang
- Department of Ultrasound, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China.
| | - Chengcheng Niu
- Department of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Research Center of Ultrasonography, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Clinical Research Center for Ultrasound and Treatment in Hunan Province, Hunan 410011, China
| |
Collapse
|
11
|
Gong JR, Lee J, Han Y, Cho KH. DDX54 downregulation enhances anti-PD1 therapy in immune-desert lung tumors with high tumor mutational burden. Proc Natl Acad Sci U S A 2025; 122:e2412310122. [PMID: 40172969 PMCID: PMC12002276 DOI: 10.1073/pnas.2412310122] [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: 06/19/2024] [Accepted: 02/25/2025] [Indexed: 04/04/2025] Open
Abstract
High tumor mutational burden (TMB-H) is a predictive biomarker for the responsiveness of cancer to immune checkpoint inhibitor (ICI) therapy that indicates whether immune cells can sufficiently recognize cancer cells as nonself. However, about 30% of all cancers from The Cancer Genome Atlas (TCGA) are classified as immune-desert tumors lacking T cell infiltration despite TMB-H. Since the underlying mechanism of these immune-desert tumors has yet to be unraveled, there is a pressing need to transform such immune-desert tumors into immune-inflamed tumors and thereby enhance their responsiveness to anti-PD1 therapy. Here, we present a systems framework for identifying immuno-oncotargets, based on analysis of gene regulatory networks, and validating the effect of these targets in transforming immune-desert into immune-inflamed tumors. In particular, we identify DEAD-box helicases 54 (DDX54) as a master regulator of immune escape in immune-desert lung cancer with TMB-H and show that knockdown of DDX54 can increase immune cell infiltration and lead to improved sensitivity to anti-PD1 therapy.
Collapse
Affiliation(s)
- Jeong-Ryeol Gong
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
| | - Jungeun Lee
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
| | - Younghyun Han
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
| | - Kwang-Hyun Cho
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology, Daejeon34141, Republic of Korea
| |
Collapse
|
12
|
Zhang C, Wang H, Li X, Jiang Y, Sun G, Yu H. Enhancing antitumor immunity: the role of immune checkpoint inhibitors, anti-angiogenic therapy, and macrophage reprogramming. Front Oncol 2025; 15:1526407. [PMID: 40260303 PMCID: PMC12009726 DOI: 10.3389/fonc.2025.1526407] [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: 11/11/2024] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer treatment has long been hindered by the complexity of the tumor microenvironment (TME) and the mechanisms that tumors employ to evade immune detection. Recently, the combination of immune checkpoint inhibitors (ICIs) and anti-angiogenic therapies has emerged as a promising approach to improve cancer treatment outcomes. This review delves into the role of immunostimulatory molecules and ICIs in enhancing anti-tumor immunity, while also discussing the therapeutic potential of anti-angiogenic strategies in cancer. In particular, we highlight the critical role of endoplasmic reticulum (ER) stress in angiogenesis. Moreover, we explore the potential of macrophage reprogramming to bolster anti-tumor immunity, with a focus on restoring macrophage phagocytic function, modulating hypoxic tumor environments, and targeting cytokines and chemokines that shape immune responses. By examining the underlying mechanisms of combining ICIs with anti-angiogenic therapies, we also review recent clinical trials and discuss the potential of biomarkers to guide and predict treatment efficacy.
Collapse
Affiliation(s)
- Chong Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Xinying Li
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yuxin Jiang
- Department of Nephrology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hanqing Yu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
13
|
Feng Z, Wang L, Li Y, Wei Y, Zhou Y, Wang S, Zhang X, Jiang C, Liao X, Kang Y, Xiao F, Zhang W. CD47-amyloid-β-CD74 signaling triggers adaptive immunosuppression in sepsis. EMBO Rep 2025:10.1038/s44319-025-00442-4. [PMID: 40185975 DOI: 10.1038/s44319-025-00442-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/18/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025] Open
Abstract
Sepsis is defined as life-threatening organ dysfunction caused by a dysregulated host response to infection. However, how this dysregulation occurs remains to be elucidated. In this study, we use single-cell RNA sequencing (scRNA-seq) and conventional RNA-seq to analyze the immune landscape of sepsis and observe that adaptive immunity is acutely and strongly suppressed. This systemic immunosuppression occurs not only in the peripheral blood but also in all other immune compartments, including the spleen, lymph nodes, and bone marrow. Clinical data show that these adaptive immunity-related genes may have the potential to be used to distinguish patients with sepsis from those with common infections. CD47 is found to play a pivotal role in this immunosuppression by inducing the production of amyloid-β (Aβ), which interacts with CD74 on B cells, leading to B-cell suppression and subsequent adaptive immunosuppression. Blocking CD47-Aβ signaling significantly reduces organ injury and improves the survival rate of septic mice by restoring phagocytic cell functions and alleviating B-cell suppression and adaptive immunosuppression.
Collapse
Affiliation(s)
- Zhongxue Feng
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lijun Wang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Li
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yonggang Wei
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yueyue Zhou
- Frontier Medical Center, Xin Chuan Road, Zhong He Street, 610212, Chengdu, Sichuan, China
| | - Siying Wang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaoqi Zhang
- Department of Orthodontics, State Key laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Chunling Jiang
- Department of Anesthesiology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Xuelian Liao
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Kang
- Department of Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Fei Xiao
- Department of Intensive Care Unit of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Wei Zhang
- Institute of Critical Care Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
14
|
Xu X, Lu X, Chen X, Yao A, Lai W. Elevated CD47 Expression Impairs Elimination of Photoaged Fibroblasts by Macrophages and Serves as a Potential Biomarker for Photoaging. J Cosmet Dermatol 2025; 24:e70098. [PMID: 40202158 PMCID: PMC11980025 DOI: 10.1111/jocd.70098] [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] [Revised: 01/07/2025] [Accepted: 02/23/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND CD47 could negatively regulate macrophage-mediated phagocytosis and contribute to senescent cells accumulation in aging. However, it remains unknown whether CD47 is overexpressed in photoaged skin and involved in photoaging pathogenesis. AIMS To investigate the expression, clinical significance, and mechanism of CD47 in photoaging. METHODS Sun-exposed (n = 10) and sun-protected (n = 10) skin samples were collected from elderly subjects and stained for CD47, and its association with collagen and elastin content and p16 expression was subsequently analyzed. A cellular photoaging model was then established to examine CD47 expression in photoaged fibroblasts. Furthermore, the influence of photoaged fibroblasts on macrophage-mediated phagocytosis and elimination was assessed by constructing a co-culture system. SiRNA was applied to block the CD47/SIRPα axis to determine its role in this process. Finally, the activation of the CD47/SIRPα axis was evaluated in skin samples. RESULTS We showed the increased dermal CD47 expression in sun-exposed aged skin, which was closely correlated with the reduced collagen content and enhanced elastin accumulation and dermal p16 expression. Next, elevated CD47 was detected in both sun-exposed aged skin-derived fibroblasts and photoaged ones. We discovered that photoaged fibroblasts impaired the phagocytotic function of co-cultured macrophages via CD47/SIRPα axis, and blocking the CD47/SIRPα axis could improve their elimination. Moreover, the CD47/SIRPα axis was found to be activated in the sun-exposed aged skin. CONCLUSIONS The present study demonstrated for the first time that CD47 was highly expressed and involved in mediating photoaged fibroblasts accumulation, providing important evidence for CD47 as a potential biomarker and therapeutic target for photoaging.
Collapse
Affiliation(s)
- Xinya Xu
- Department of DermatologyThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Xinhua Lu
- Department of NeurosurgeryForesea Life Insurance Guangzhou General HospitalGuangzhouChina
| | - Xinling Chen
- Department of DermatologyThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Amin Yao
- Department of DermatologyThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Wei Lai
- Department of DermatologyThe Third Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| |
Collapse
|
15
|
Ahvati H, Roudi R, Sobhani N, Safari F. CD47 as a potent target in cancer immunotherapy: A review. Biochim Biophys Acta Rev Cancer 2025; 1880:189294. [PMID: 40057140 DOI: 10.1016/j.bbcan.2025.189294] [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: 02/22/2025] [Accepted: 03/02/2025] [Indexed: 03/22/2025]
Abstract
Cancer is the second-highest cause of death worldwide. Accordingly, finding new cancer treatments is of great interest to researchers. The current platforms to fight cancer such as chemotherapy, radiotherapy, and surgery are limited in efficacy, especially in the metastatic setting. In this war against cancer, the immune system is a powerful ally, but tumor cells often outsmart it through alternative pathways. Cluster of differentiation 47 (CD47), a protein that normally prevents healthy cells from being attacked by immune cells, is often overexpressed on cancer cells. This makes CD47 a prime target for immunotherapy. Blocking of CD47 has the potential to unleash the immune system's cell populations-such as myeloid cells, macrophages, and T cells-to allow the immune system to discover and destroy cancer cells more successfully. In this review, we aimed to provide the latest information and findings about the roles of CD47 in the regulation of various cellular pathways and, thus, the importance of CD47 as a potential target in cancer therapy.
Collapse
Affiliation(s)
- Hiva Ahvati
- School of Biology, College of Science, University of Tehran, Tehran, Iran
| | - Raheleh Roudi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA.
| | - Navid Sobhani
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fatemeh Safari
- Department of Biology, Faculty of Science, University of Guilan, Rasht, Iran
| |
Collapse
|
16
|
Tarannum M, Ding X, Barisa M, Hu S, Anderson J, Romee R, Zhang J. Engineering innate immune cells for cancer immunotherapy. Nat Biotechnol 2025; 43:516-533. [PMID: 40229380 DOI: 10.1038/s41587-025-02629-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/05/2025] [Indexed: 04/16/2025]
Abstract
Innate immune cells, including natural killer cells, macrophages and γδ T cells, are gaining prominence as promising candidates for cancer immunotherapy. Unlike conventional T cells, these cells possess attributes such as inherent antitumor activity, rapid immune responses, favorable safety profiles and the ability to target diverse malignancies without requiring prior antigen sensitization. In this Review, we examine the engineering strategies used to enhance their anticancer potential. We discuss challenges associated with each cell type and summarize insights from preclinical and clinical work. We propose strategies to address existing barriers, providing a perspective on the advancement of innate immune engineering as a powerful modality in anticancer treatment.
Collapse
Affiliation(s)
- Mubin Tarannum
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Xizhong Ding
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow Transplantation Center of the First Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
| | - Marta Barisa
- Cancer Section, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Sabrina Hu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John Anderson
- Cancer Section, Developmental Biology and Cancer Department, UCL Great Ormond Street Institute of Child Health, University College London, London, UK.
| | - Rizwan Romee
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA.
| | - Jin Zhang
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences, and Bone Marrow Transplantation Center of the First Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
17
|
Chen Y, Kou Y, Ni Y, Yang H, Xu C, Fan H, Liu H. Microglia efferocytosis: an emerging mechanism for the resolution of neuroinflammation in Alzheimer's disease. J Neuroinflammation 2025; 22:96. [PMID: 40159486 PMCID: PMC11955113 DOI: 10.1186/s12974-025-03428-0] [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: 02/04/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by significant neuroinflammatory responses. Microglia, the immune cells of the central nervous system, play a crucial role in the pathophysiology of AD. Recent studies have indicated that microglial efferocytosis is an important mechanism for clearing apoptotic cells and cellular debris, facilitating the resolution of neuroinflammation. This review summarizes the biological characteristics of microglia and the mechanisms underlying microglial efferocytosis, including the factors and signaling pathways that regulate efferocytosis, the interactions between microglia and other cells that influence this process, and the role of neuroinflammation in AD. Furthermore, we explore the role of microglial efferocytosis in AD from three perspectives: its impact on the clearance of amyloid plaques, its regulation of neuroinflammation, and its effects on neuroprotection. Finally, we summarize the current research status on enhancing microglial efferocytosis to alleviate neuroinflammation and improve AD, as well as the future challenges of this approach as a therapeutic strategy for AD.
Collapse
Affiliation(s)
- Yongping Chen
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, Heilongjiang Province, P. R. China
| | - Yuhong Kou
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, Heilongjiang Province, P. R. China
| | - Yang Ni
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
| | - Haotian Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, Heilongjiang Province, P. R. China
| | - Cailin Xu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, Heilongjiang Province, P. R. China.
| | - Huanqi Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
18
|
Yang J, Xing F, Hu F, Hou M, Dong H, Cheng J, Li W, Yan R, Xu J, Xu K, Pan L. Super-resolution microscopy unveils the nanoscale organization and self-limiting clustering of CD47 in human erythrocytes. J Mol Cell Biol 2025; 16:mjae041. [PMID: 39367479 PMCID: PMC11992563 DOI: 10.1093/jmcb/mjae041] [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: 06/03/2023] [Revised: 11/16/2023] [Accepted: 10/03/2024] [Indexed: 10/06/2024] Open
Abstract
The transmembrane protein CD47, an innate immune checkpoint protein, plays a pivotal role in preventing healthy erythrocytes from immune clearance. Our study utilized stochastic optical reconstruction microscopy (STORM) and single-molecule analysis to investigate the distribution of CD47 on the human erythrocyte membrane. Contrary to previous findings in mouse erythrocytes, we discovered that CD47 exists in randomly distributed monomers rather than in clusters across the human erythrocyte membrane. Using secondary antibody-induced crosslinking, we found that CD47 aggregates into stable clusters within minutes. By comparing these STORM results with those of the fully mobile protein CD59 and the cytoskeleton-bound membrane protein glycophorin C under similar conditions, as well as devising two-color STORM co-labeling and co-clustering experiments, we further quantitatively revealed an intermediate, self-limiting clustering behavior of CD47, elucidating its fractional (∼14%) attachment to the cytoskeleton. Moreover, we report reductions in both the amount of CD47 and its clustering capability in aged erythrocytes, providing new insight into erythrocyte senescence. Together, the combination of STORM and secondary antibody-based crosslinking unveils the unique self-limiting clustering behavior of CD47 due to its fractional cytoskeleton attachment.
Collapse
Affiliation(s)
- Jianyu Yang
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Fulin Xing
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Fen Hu
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Mengdi Hou
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Hao Dong
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Jiayu Cheng
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
| | - Wan Li
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | - Rui Yan
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | - Jingjun Xu
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518083, China
| | - Ke Xu
- Department of Chemistry, University of California, Berkeley, CA 94720, USA
| | - Leiting Pan
- The Key Laboratory of Weak-Light Nonlinear Photonics of Education Ministry, School of Physics and TEDA Institute of Applied Physics, Nankai University, Tianjin 300071, China
- Shenzhen Research Institute of Nankai University, Shenzhen 518083, China
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin 300071, China
| |
Collapse
|
19
|
Zhao T, Luo Y, Sun Y, Wei Z. Characterizing macrophage diversity in colorectal malignancies through single-cell genomics. Front Immunol 2025; 16:1526668. [PMID: 40191203 PMCID: PMC11968368 DOI: 10.3389/fimmu.2025.1526668] [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: 11/12/2024] [Accepted: 03/10/2025] [Indexed: 04/09/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors of the digestive tract, with increasing incidence and mortality rates, posing a significant burden on human health. Its progression relies on various mechanisms, among which the tumor microenvironment and tumor-associated macrophages (TAMs) have garnered increasing attention. Macrophage infiltration in various solid tumors is associated with poor prognosis and is linked to chemotherapy resistance in many cancers. These significant biological behaviors depend on the heterogeneity of macrophages. Tumor-promoting TAMs comprise subpopulations characterized by distinct markers and unique transcriptional profiles, rendering them potential targets for anticancer therapies through either depletion or reprogramming from a pro-tumoral to an anti-tumoral state. Single-cell RNA sequencing technology has significantly enhanced our research resolution, breaking the traditional simplistic definitions of macrophage subtypes and deepening our understanding of the diversity within TAMs. However, a unified elucidation of the nomenclature and molecular characteristics associated with this diversity remains lacking. In this review, we assess the application of conventional macrophage polarization subtypes in colorectal malignancies and explore several unique subtypes defined from a single-cell omics perspective in recent years, categorizing them based on their potential functions.
Collapse
Affiliation(s)
- Tingshuo Zhao
- First Clinical Medical College, Shanxi Medical University, Tai Yuan, China
| | - Yinyi Luo
- First Clinical Medical College, Shanxi Medical University, Tai Yuan, China
| | - Yuanjie Sun
- First Clinical Medical College, Shanxi Medical University, Tai Yuan, China
| | - Zhigang Wei
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Shanxi Medical University, Tai Yuan, China
| |
Collapse
|
20
|
Zhang B, Zou Y, Tang Q, Yuan Z, Jiang K, Zhang Z, Chen S, Wu Q, Zhou X, Zhang X. SIRPα modulates microglial efferocytosis and neuroinflammation following experimental subarachnoid hemorrhage via the SHP1/STAT6 axis. J Neuroinflammation 2025; 22:88. [PMID: 40108663 PMCID: PMC11924727 DOI: 10.1186/s12974-025-03414-6] [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: 12/04/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Subarachnoid hemorrhage induces extensive neuronal cell death, leading to the release of damage-associated molecular patterns (DAMPs). These DAMPs, along with hemoglobin and cell corpses, trigger localized inflammation. Signal regulatory protein alpha (SIRPα) plays a crucial role in efferocytosis by acting as a "don't eat-me" signal, modulating inflammation and tissue homeostasis. However, the precise function and regulatory mechanisms of SIRPα in efferocytosis remain unclear. Proteomic analysis of cerebrospinal fluid (CSF) reveals that SIRPα levels are significantly elevated in the CSF of SAH patients and correlate with clinical outcomes. In vivo and in vitro studies show that microglial knockdown of SIRPα promotes efferocytosis and attenuates neuroinflammation following SAH. SIRPα inhibits efferocytosis by recruiting and phosphorylating SHP1 and SHP2 through phosphorylation of four tyrosine residues in its cytoplasmic domain, with SHP1 playing a particularly critical role. Mutation of these tyrosine residues to non-phosphorylatable alanine residues enhances efferocytosis and reduces neuroinflammation in vitro. RNA-seq analysis suggests that this mutation upregulates the expression of "eat-me" signals, MerTK and CD36, and identifies STAT6 as a key transcription factor involved in this process. In conclusion, SIRPα plays a central role in regulating microglia efferocytosis and neuroinflammation after SAH via the SHP1/STAT6 axis. Targeting this pathway may provide a promising therapeutic approach for SAH.
Collapse
Affiliation(s)
- Bingtao Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yan Zou
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qikai Tang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zixuan Yuan
- Department of Neurosurgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Kun Jiang
- Department of Neurosurgery, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Zhaoxiang Zhang
- Department of Neurosurgery, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Shujuan Chen
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qi Wu
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaoming Zhou
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Xin Zhang
- Department of Neurosurgery, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
21
|
Wang Q, He J, Lei T, Li X, Yue S, Liu C, Hu Q. New insights into cancer immune checkpoints landscape from single-cell RNA sequencing. Biochim Biophys Acta Rev Cancer 2025; 1880:189298. [PMID: 40088992 DOI: 10.1016/j.bbcan.2025.189298] [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: 01/09/2025] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Immune checkpoint blockade (ICB) therapy represents a pivotal advancement in tumor immunotherapy by restoring the cytotoxic lymphocytes' anti-tumor activity through the modulation of immune checkpoint functions. Nevertheless, many patients experience suboptimal therapeutic outcomes, likely due to the immunosuppressive tumor microenvironment, drug resistance, and other factors. Single-cell RNA sequencing has assisted to precisely investigate the immune infiltration patterns before and after ICB treatment, enabling a high-resolution depiction of previously unrecognized functional interaction among immune checkpoints. This review addresses the heterogeneity between tumor microenvironments that respond to or resist ICB therapy, highlighting critical factors underlying the variation in immunotherapy efficacy and elucidating treatment failure. Furthermore, a comprehensive examination is provided of how specific ICBs modulate immune and tumor cells to achieve anti-tumor effects and generate treatment resistance, alongside a summary of emerging immune checkpoints identified as promising targets for cancer immunotherapy through single-cell RNA sequencing applications.
Collapse
Affiliation(s)
- Qian Wang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiahui He
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tianyu Lei
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaohui Li
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China
| | - Shengqin Yue
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Chao Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing 100034, China.
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China; Renmin Hospital of Wuhan Economic and Technological Development Zone (Hannan), Wuhan 430090, China.
| |
Collapse
|
22
|
Zhou X, He J, Song H, Zhao W, Li R, Han W, Li Q. Regulation of macrophage efferocytosis by the CLCF1/NF-κB pathway improves neurological and cognitive impairment following CO poisoning. Brain Behav Immun 2025; 127:126-146. [PMID: 40081779 DOI: 10.1016/j.bbi.2025.03.003] [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: 09/02/2024] [Revised: 02/19/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025] Open
Abstract
Severe carbon monoxide (CO) poisoning can induce structural and functional damage to the nervous system, resulting in persistent cognitive impairments. Properly terminating inflammation caused by neuronal damage is essential for tissue repair. Macrophages clear cell corpses and fragments through efferocytosis and produce cytokines to coordinate the immune response, thus promoting neuronal repair and regeneration. However, within the microenvironment of the CO-affected nervous system, macrophage efferocytosis is disrupted. Our study found that macrophages regulate efferocytosis by releasing Cardiotrophin-like cytokine factor 1 (CLCF1), which modulates the NF-κB pathway in both macrophages and microglia, thereby controlling inflammation and promoting nervous system repair. Furthermore, efferocytosis regulates the secretion of cytokines such as TNF-α, IL-1β, and IL-10, promoting M2 polarization of macrophages, which aids in neuronal repair and regeneration. Regulating macrophage CLCF1 expression also leads to improvements in the memory, learning, and motor abilities of rats poisoned with CO.
Collapse
Affiliation(s)
- Xudong Zhou
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518060, PR China; The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, PR China
| | - Jingjing He
- The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, PR China
| | - Huiping Song
- Department of Traditional Chinese Medicine II, Rehabilitation University Qingdao Central Hospital, Qingdao, Shandong 266042, PR China
| | - Weiwei Zhao
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, PR China
| | - Rui Li
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518060, PR China
| | - Wei Han
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518060, PR China
| | - Qin Li
- Emergency Department, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong 518060, PR China; The First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250355, PR China.
| |
Collapse
|
23
|
Cao X, Wan S, Wu B, Liu Z, Xu L, Ding Y, Huang H. Antitumor Research Based on Drug Delivery Carriers: Reversing the Polarization of Tumor-Associated Macrophages. Mol Pharm 2025; 22:1174-1197. [PMID: 39868820 DOI: 10.1021/acs.molpharmaceut.4c01277] [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] [Indexed: 01/28/2025]
Abstract
The development of malignant tumors is a complex process that involves the tumor microenvironment (TME). An immunosuppressive TME presents significant challenges to current cancer therapies, serving as a key mechanism through which tumor cells evade immune detection and play a crucial role in tumor progression and metastasis. This impedes the optimal effectiveness of immunotherapeutic approaches, including cytokines, immune checkpoint inhibitors, and cancer vaccines. Tumor-associated macrophages (TAMs), a major component of tumor-infiltrating immune cells, exhibit dual functionalities: M1-like TAMs suppress tumorigenesis, while M2-like TAMs promote tumor growth and metastasis. Consequently, the development of various nanocarriers aimed at polarizing M2-like TAMs to M1-like phenotypes through distinct mechanisms has emerged as a promising therapeutic strategy to inhibit tumor immune escape and enhance antitumor responses. This Review covers the origin and types of TAMs, common pathways regulating macrophage polarization, the role of TAMs in tumor progression, and therapeutic strategies targeting TAMs, aiming to provide a comprehensive understanding and guidance for future research and clinical applications.
Collapse
Affiliation(s)
- Xinyu Cao
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Shen Wan
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Bingyu Wu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Zhikuan Liu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Lixing Xu
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| | - Yu Ding
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, China
| | - Haiqin Huang
- Department of Pharmaceutics, School of Pharmacy, Nantong University, Nantong 226001, China
| |
Collapse
|
24
|
Zhang J, Wang F, Sun Z, Ye J, Chu H. Multidimensional applications of prussian blue-based nanoparticles in cancer immunotherapy. J Nanobiotechnology 2025; 23:161. [PMID: 40033359 DOI: 10.1186/s12951-025-03236-x] [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: 10/25/2024] [Accepted: 02/16/2025] [Indexed: 03/05/2025] Open
Abstract
Immunotherapy holds notable progress in the treatment of cancer. However, the clinical therapeutic effect remains a significant challenge due to immune-related side effects, poor immunogenicity, and immunosuppressive microenvironment. Nanoparticles have emerged as a revolutionary tool to surmount these obstacles and amplify the potency of immunotherapeutic agents. Prussian blue nanoparticles (PBNPs) exhibit multi-dimensional immune function in cancer immunotherapy, including acting as a nanocarrier to deliver immunotherapeutic agents, as a photothermal agent to improve the efficacy of immunotherapy through photothermal therapy, as a nanozyme to regulate tumor microenvironment, and as an iron donor to induce immune events related to ferroptosis and tumor-associated macrophages polarization. This review focuses on the advances and applications of PBNPs in cancer immunotherapy. First, the biomedical functions of PBNPs are introduced. Then, based on the immune function of PBNPs, we systematically reviewed the multidimensional application of PBNPs in cancer immunotherapy. Finally, the challenges and future developments of PBNPs-based cancer immunotherapy are highlighted.
Collapse
Affiliation(s)
- Jiayi Zhang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Fang Wang
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Zhaogang Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Hongqian Chu
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing, 101149, China.
- Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| |
Collapse
|
25
|
Huang Y, Gao Y, Lin Z, Miao H. Involvement of the ubiquitin-proteasome system in the regulation of the tumor microenvironment and progression. Genes Dis 2025; 12:101240. [PMID: 39759114 PMCID: PMC11697063 DOI: 10.1016/j.gendis.2024.101240] [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: 06/29/2023] [Revised: 10/11/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2025] Open
Abstract
The tumor microenvironment is a complex environment comprising tumor cells, non-tumor cells, and other critical non-cellular components. Some studies about tumor microenvironment have recently achieved remarkable progress in tumor treatment. As a substantial part of post-translational protein modification, ubiquitination is a crucial player in maintaining protein stability in cell signaling, cell growth, and a series of cellular life activities, which are also essential for regulating tumor cells or other non-tumor cells in the tumor microenvironment. This review focuses on the role and function of ubiquitination and deubiquitination modification in the tumor microenvironment while discussing the prospect of developing inhibitors targeting ubiquity-related enzymes, thereby providing ideas for future research in cancer therapy.
Collapse
Affiliation(s)
- Yulan Huang
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Yuan Gao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
| | - Zhenghong Lin
- School of Life Sciences, Chongqing University, Chongqing 401331, China
| | - Hongming Miao
- Department of Pathophysiology, College of High Altitude Military Medicine, Army Medical University, Chongqing 400038, China
- School of Life Sciences, Chongqing University, Chongqing 401331, China
- Jinfeng Laboratory, Chongqing 401329, China
| |
Collapse
|
26
|
Jeppesen DK, Sanchez ZC, Kelley NM, Hayes JB, Ambroise J, Koory EN, Krystofiak E, Taneja N, Zhang Q, Dungan MM, Perkins OL, Tyska MJ, Knapik EW, Dean KM, Doran AC, Coffey RJ, Burnette DT. Blebbisomes are large, organelle-rich extracellular vesicles with cell-like properties. Nat Cell Biol 2025; 27:438-448. [PMID: 39984653 PMCID: PMC11906356 DOI: 10.1038/s41556-025-01621-0] [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: 09/18/2023] [Accepted: 01/14/2025] [Indexed: 02/23/2025]
Abstract
Cells secrete a large variety of extracellular vesicles (EVs) to engage in cell-to-cell and cell-to-environment intercellular communication. EVs are functionally involved in many physiological and pathological processes by interacting with cells that facilitate transfer of proteins, lipids and genetic information. However, our knowledge of EVs is incomplete. Here we show that cells actively release exceptionally large (up to 20 µm) membrane-enclosed vesicles that exhibit active blebbing behavior, and we, therefore, have termed them blebbisomes. Blebbisomes contain an array of cellular organelles that include functional mitochondria and multivesicular endosomes, yet lack a definable nucleus. We show that blebbisomes can both secrete and internalize exosomes and microvesicles. Blebbisomes are released from normal and cancer cells, can be observed by direct imaging of cancer cells in vivo and are present in normal bone marrow. We demonstrate that cancer-derived blebbisomes contain a plethora of inhibitory immune checkpoint proteins, including PD-L1, PD-L2, B7-H3, VISTA, PVR and HLA-E. These data identify a very large, organelle-containing functional EV that act as cell-autonomous mobile communication centres capable of integrating and responding to signals in the extracellular environment.
Collapse
Affiliation(s)
- Dennis K Jeppesen
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Zachary C Sanchez
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Noah M Kelley
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - James B Hayes
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Jessica Ambroise
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Emma N Koory
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Evan Krystofiak
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Nilay Taneja
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Qin Zhang
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew M Dungan
- Department of Molecular Pathology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Olivia L Perkins
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Matthew J Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Ela W Knapik
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Kevin M Dean
- Lydia Hill Department of Bioinformatics, University of Texas Southwestern, Dallas, TX, USA
| | - Amanda C Doran
- Department of Molecular Pathology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA
| | - Dylan T Burnette
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine Basic Sciences, Nashville, TN, USA.
| |
Collapse
|
27
|
Huang HY, Zheng XN, Tian L. Vascular-Associated Mononuclear Phagocytes: First-Line Soldiers Ambushing Metastasis. Bioessays 2025; 47:e202400261. [PMID: 39988942 DOI: 10.1002/bies.202400261] [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: 11/05/2024] [Revised: 12/10/2024] [Accepted: 12/12/2024] [Indexed: 02/25/2025]
Abstract
Mononuclear phagocytes (MPs), which consist of dendritic cells, monocytes, and macrophages, are distributed throughout the body and actively eliminate invading microorganisms and abnormal cells. Depending on the local microenvironment, MPs manifest considerably various lifespans and phenotypes to maintain tissue homeostasis. Vascular-associated mononuclear phagocytes (VaMPs) are the special subsets of MPs that are localized either within the lumen side or on the apical surface of vessels, acting as the critical sentinels to recognize and defend against disseminated tumor cells. In this review, we introduce three major types of VaMPs, patrolling monocytes, Kupffer cells, and perivascular macrophages, and discuss their emerging roles in immunosurveillance during incipient metastasis. We also explore the roles of lineage-determining transcription factors and cell surface receptors that endow VaMPs with potent anti-tumor activity. Finally, we highlight the molecular and cellular mechanisms that drive the phenotypic plasticity of VaMPs and summarize combinatory strategies for targeting VaMPs in overt metastasis.
Collapse
Affiliation(s)
- Han-Ying Huang
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Xin-Nan Zheng
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| | - Lin Tian
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, P. R. China
| |
Collapse
|
28
|
Li Y, Meng Z, Fan C, Rong H, Xi Y, Liao Q. Identification and multi-omics analysis of essential coding and long non-coding genes in colorectal cancer. Biochem Biophys Rep 2025; 41:101938. [PMID: 40034256 PMCID: PMC11874739 DOI: 10.1016/j.bbrep.2025.101938] [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: 12/05/2024] [Revised: 01/19/2025] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Essential genes are indispensable for the survival of cancer cell. CRISPR/Cas9-based pooled genetic screens have distinguished the essential genes and their functions in distinct cellular processes. Nevertheless, the landscape of essential genes at the single cell levels and the effect on the tumor microenvironment (TME) remains limited. Here, we identified 396 essential protein-coding genes (ESPs) by integration of 8 genome-wide CRISPR loss-of-function screen datasets of colorectal cancer (CRC) cell lines and single-cell RNA sequencing (scRNA-seq) data of CRC tissues. Then, 29 essential long non-coding genes (ESLs) were predicted using Hypergeometric Test (HT) and Personalized PageRank (PPR) algorithms based on ESPs and co-expressed network constructed from scRNA-seq. CRISPR/Cas9 knockout experiment verified the effect of several ESPs and ESLs on the survival of CRC cell line. Furthermore, multi-omics features of ESPs and ESLs were illustrated by examining their expression patterns and transcription factor (TF) regulatory network at the single cell level, as well as DNA mutation and DNA methylation events at bulk level. Finally, through integrating multiple intracellular regulatory networks with cell-cell communication network (CCN), we elucidated that CD47 and MIF are regulated by multiple CRC essential genes, and the anti-cancer drugs sunitinib can interfere the expression of them potentially. Our findings provide a comprehensive asset of CRC ESPs and ESLs, sheding light on the mining of potential therapy targets for CRC.
Collapse
Affiliation(s)
- Yanguo Li
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, Zhejiang, China
| | - Zixing Meng
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Chengjiang Fan
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Hao Rong
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Yang Xi
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Qi Liao
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
29
|
Du F, Wang G, Dai Q, Huang J, Li J, Liu C, Du K, Tian H, Deng Q, Xie L, Zhao X, Zhang Q, Yang L, Li Y, Wu Z, Zhang Z. Targeting novel regulated cell death: disulfidptosis in cancer immunotherapy with immune checkpoint inhibitors. Biomark Res 2025; 13:35. [PMID: 40012016 DOI: 10.1186/s40364-025-00748-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The battle against cancer has evolved over centuries, from the early stages of surgical resection to contemporary treatments including chemotherapy, radiation, targeted therapies, and immunotherapies. Despite significant advances in cancer treatment over recent decades, these therapies remain limited by various challenges. Immune checkpoint inhibitors (ICIs), a cornerstone of tumor immunotherapy, have emerged as one of the most promising advancements in cancer treatment. Although ICIs, such as CTLA-4 and PD-1/PD-L1 inhibitors, have demonstrated clinical efficacy, their therapeutic impact remains suboptimal due to patient-specific variability and tumor immune resistance. Cell death is a fundamental process for maintaining tissue homeostasis and function. Recent research highlights that the combination of induced regulatory cell death (RCD) and ICIs can substantially enhance anti-tumor responses across multiple cancer types. In cells exhibiting high levels of recombinant solute carrier family 7 member 11 (SLC7A11) protein, glucose deprivation triggers a programmed cell death (PCD) pathway characterized by disulfide bond formation and REDOX (reduction-oxidation) reactions, termed "disulfidptosis." Studies suggest that disulfidptosis plays a critical role in the therapeutic efficacy of SLC7A11high cancers. Therefore, to investigate the potential synergy between disulfidptosis and ICIs, this study will explore the mechanisms of both processes in tumor progression, with the goal of enhancing the anti-tumor immune response of ICIs by targeting the intracellular disulfidptosis pathway.
Collapse
Affiliation(s)
- Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Guojun Wang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qian Dai
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Jiang Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Junxin Li
- Department of pharmacy, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Congxing Liu
- Department of Pharmacy, Chengfei Hospital, Chengdu, 610000, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pediatrics, Luzhou Maternal and Child Health Hospital, Luzhou Second People's Hospital, Luzhou, 646000, Sichuan, China
| | - Hua Tian
- School of Nursing, Chongqing College of Humanities, Science & Technology, Chongqing, 401520, China
| | - Qiwei Deng
- Heruida Pharmaceutical Co.,ltd, Haikou, Hainan, 570100, China
| | - Longxiang Xie
- The TCM Hospital of Longquanyi District, Chengdu, 610100, Sichuan, China
| | - Xin Zhao
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qimin Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Lan Yang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhigui Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhuo Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
30
|
Yang Q, Shu Y, Chen Y, Qi Z, Hu S, Zhang Y, Qin Y, Xu X, Hu J, Huang A, Cheng P. Expression of SIRPα-Fc by oncolytic virus enhances antitumor efficacy through tumor microenvironment reprogramming. Front Immunol 2025; 16:1513555. [PMID: 40070841 PMCID: PMC11893986 DOI: 10.3389/fimmu.2025.1513555] [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: 10/18/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025] Open
Abstract
Oncolytic viruses (OVs) selectively replicate within tumors, directly killing cancer cells and promoting a systemic immune response by releasing tumor antigens. These features make OVs a promising approach in tumor immunotherapy, offering targeted treatment with fewer side effects. Despite these advantages, OVs are primarily administered via intratumoral injection, limiting their effectiveness for advanced, systemic cancers. Among OVs, oncolytic adenoviruses (oAdVs) are the most widely studied due to their well-understood gene regulation, safety, and stability. In this study, a modified oAdV vector, pDC316-oAd-SA, was engineered to express the SIRPα-mIgG1Fc gene, designed to remodel tumor-associated macrophages (TAMs) and enhance anti-tumor immunity. This vector, along with a control virus (Ad-ON), was evaluated both in vitro and in vivo. The modified oAd-SA significantly improved macrophage phagocytosis and showed superior tumor regression in murine models. Additionally, while both oAdVs increased T cell infiltration in the tumor microenvironment, oAd-SA specifically enhanced T cell immune function. The study also revealed that oAdVs modulate TAMs differently across tumor types, with oAd-SA therapy particularly increasing TAM phagocytosis and promoting an anti-tumor response.
Collapse
Affiliation(s)
- Qingzhe Yang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yongheng Shu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yanwei Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhongbing Qi
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shichuan Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yao Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Qin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xianglin Xu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jianchuan Hu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Anliang Huang
- Department of Pathology, Chengdu Fifth People’s Hospital, Chengdu, China
| | - Ping Cheng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
31
|
Salminen A. Cooperation between inhibitory immune checkpoints of senescent cells with immunosuppressive network to promote immunosenescence and the aging process. Ageing Res Rev 2025; 106:102694. [PMID: 39984130 DOI: 10.1016/j.arr.2025.102694] [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: 11/22/2024] [Revised: 12/30/2024] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
The accumulation of senescent cells within tissues promotes the aging process by remodelling the functions of the immune system. For many years, it has been known that senescent cells secrete pro-inflammatory cytokines and chemokines, a phenotype called the senescence-associated secretory phenotype (SASP). Chemokines and colony-stimulating factors stimulate myelopoiesis and recruit myeloid cells into aging tissues. Interestingly, recent studies have demonstrated that senescent cells are not only secretory but they also express an increased level of ligand proteins for many inhibitory immune checkpoint receptors. These ligands represent "don't eat me" markers in senescent cells and moreover, they are able to induce an exhaustion of many immune cells, such as surveying natural killer (NK) cells, cytotoxic CD8+ T cells, and macrophages. The programmed cell death protein-1 (PD-1) and its ligand PD-L1 represent the best known inhibitory immune checkpoint pathway. Importantly, the activation of inhibitory checkpoint receptors, e.g., in chronic inflammatory states, can also induce certain immune cells to differentiate toward their immunosuppressive phenotype. This can be observed in myeloid derived suppressor cells (MDSC), tissue regulatory T cells (Treg), and M2 macrophages. Conversely, these immunosuppressive cells stimulate in senescent cells the expression of many ligand proteins for inhibitory checkpoint receptors. Paradoxically, senescent cells not only promote the pro-inflammatory state but they maintain it at a low-grade level by expressing ligands for inhibitory immune checkpoint receptors. Thus, the cooperation between senescent cells and immunosuppressive cells enhances the senescence state of immune cells, i.e., immune senescence/exhaustion, and cellular senescence within tissues via bystander effects.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, Kuopio FI-70211, Finland.
| |
Collapse
|
32
|
Ding J, Zhao X, Long S, Sun W, Du J, Fan J, Peng X. A Dual Stimuli-Responsive Nanoimmunomodulator for Antitumor Synergy of Macrophages and T Cells. ACS NANO 2025; 19:6468-6478. [PMID: 39919169 DOI: 10.1021/acsnano.4c17285] [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: 02/09/2025]
Abstract
Only a minority of patients benefit from current T-cell-focused adaptive immunotherapies, underscoring the need to engage innate immune cells, particularly macrophages, for multilayered tumor control. However, high-efficacy therapeutics capable of orchestrating multiple immune cells remain scarce. Herein, a dual stimuli-responsive nanoimmunomodulator (6EPP@si) that caters specifically to the tumor microenvironment (TME) is presented for the antitumor synergy of macrophages and T cells. Using the functional polymer-based carrier, we co-deliver the endoplasmic reticulum (ER)-localized photosensitizer 6E and small interfering RNA targeting CD47 (siCD47) into breast tumors. Within the acidic and high-glutathione TME, 6EPP@si undergoes self-lysosome escape and nanocleavage for precise, on-demand drug release. Consequently, siCD47 released into the cytoplasm enables potent CD47 silencing, while the ER-targeted photosensitizer 6E induces immunogenic cell death through reactive oxygen species-based ER stress, triggering the release of damage-associated molecular patterns, including calreticulin surface translocation. 6EPP@si enhances macrophage phagocytosis by modulating both antiphagocytic and prophagocytic signals and also promotes antigen presentation to activate T cells. In orthotopic breast tumor and spontaneous lung metastatic tumor models, this combined approach demonstrates robust antitumor effects and effective antimetastatic immunity, offering a meaningful strategy to simultaneously activate multiple immune cells for enhancing cancer immunotherapy.
Collapse
Affiliation(s)
- Junying Ding
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Xueze Zhao
- Department of Chemistry, The University of Hong Kong, Hong Kong SAR 999077, China
| | - Saran Long
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Jianjun Du
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Jiangli Fan
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- Ningbo Institute of Dalian University of Technology, Ningbo 315016, China
| | - Xiaojun Peng
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian 116024, China
- State Key Laboratory of Fine Chemicals, College of Materials Science and Engineering, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
33
|
Li L, Zhang J, Zhang Y, Zhao R, Yang F, Yan Y, Wang Q, Xie M. Biofilm-modified Prussian blue improves memory function in late-stage Alzheimer's disease mice with triple therapy. Int J Pharm 2025; 670:125112. [PMID: 39732217 DOI: 10.1016/j.ijpharm.2024.125112] [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: 08/29/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that is significantly characterized by cognitive and memory impairments, which worsen significantly with age. In the late stages of AD, metal ion disorders and an imbalance of reactive oxygen species (ROS) levels occur in the brain microenvironment, which causes abnormal aggregation of β-amyloid (Aβ), leading to a significant worsening of the AD symptoms. Therefore, we designed a composite nanomaterial of macrophage membranes-encapsulated Prussian blue nanoparticles (PB NPs/MM). Prussian blue nanoparticles (PB NPs) are capable of chelating Cu2+ and reducing ROS. Macrophage membranes (MM) have advantages over liposomal and erythrocyte membrane carriers, including inflammatory targeting capabilities and more effective immune evasion. Concurrently, the excellent photothermal ability of PB NPs can briefly open the blood-brain barrier (BBB) under near-infrared laser irradiation, which improves the transport efficiency of PB NPs/MM across the BBB and ablates Aβ deposition, thus achieving optimal therapeutic efficacy. In vitro experiments demonstrated that PB NPs/MM is a multifunctional nanosystem, which can effectively inhibit Cu2+-induced Aβ monomers aggregation, photothermally depolymerize Aβ fibrils, and attenuate oxidative stress through the combined treatment of chelating metals, photothermal therapy and scavenging ROS. In behavioral experiments, it also significantly improved the cognitive and learning deficits in late-stage APP/PS1 mice, thereby providing new ideas for the treatment of late-stage AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Lianxin Li
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Jiayang Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Yuewen Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Ruixin Zhao
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Fengmei Yang
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Yujiao Yan
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China
| | - Qi Wang
- Department of Neurology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, PR China.
| | - Meng Xie
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu 212013, PR China.
| |
Collapse
|
34
|
Felix Oghenemaro E, Uthirapathy S, Nathiya D, Kaur P, Ravi Kumar M, Verma A. Role of glutaminyl-peptide cyclo-transferase-like protein (QPCTL) in cancer: From molecular mechanisms to immunotherapy. Gene 2025; 937:149153. [PMID: 39653089 DOI: 10.1016/j.gene.2024.149153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Glutaminyl-peptide cyclotransferase-like protein (QPCTL) is a newly discovered enzyme that has sparked interest owing to its possible role in cancer genesis and progression. Initially discovered as a post-translational modification regulator of protein maturation, QPCTL has emerged as a key participant in cancer biology. Recent research has linked QPCTL to numerous essential cancer-related processes, including cell proliferation, migration, invasion, and apoptosis. Furthermore, QPCTL expression changes have been seen in a variety of cancer types, underlining its potential as a diagnostic and prognostic marker. The molecular mechanisms behind QPCTL's participation in cancer will be examined in this review. We investigate its involvement in the control of signaling pathways and the modification of cellular activities that are important in cancer. We also examine the clinical importance of QPCTL, including as its relationship with tumor development, metastasis, and response to treatment. We also discuss the possible therapeutic implications of targeting QPCTL in cancer therapy. QPCTL is a prospective target for the development of innovative anticancer treatments due to its participation in several cancer-associated pathways.
Collapse
Affiliation(s)
- Enwa Felix Oghenemaro
- Delta State University, Department of Pharmaceutical Microbiology, Faculty of Pharmacy, PMB 1, Abraka, Delta State, Nigeria
| | - Subasini Uthirapathy
- Faculty of Pharmacy, Pharmacology Department, Tishk International University, Erbil, Kurdistan Region, Iraq.
| | - Deepak Nathiya
- Department of Pharmacy Practice, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India.
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - M Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Ashish Verma
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| |
Collapse
|
35
|
Sun Y, Wu X, Li J, Verma CS, Yu J, Miserez A. Peptide-Based Complex Coacervates Stabilized by Cation-π Interactions for Cell Engineering. J Am Chem Soc 2025; 147:4284-4295. [PMID: 39864072 DOI: 10.1021/jacs.4c14469] [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: 01/28/2025]
Abstract
Complex coacervation is a form of liquid-liquid phase separation, whereby two types of macromolecules, usually bearing opposite net charges, self-assemble into dense microdroplets driven by weak molecular interactions. Peptide-based coacervates have recently emerged as promising carriers to deliver large macromolecules (nucleic acids, proteins and complex thereof) inside cells. Thus, it is essential to understand their assembly/disassembly mechanisms at the molecular level in order to tune the thermodynamics of coacervates formation and the kinetics of cargo release upon entering the cell. In this study, we designed histidine-rich peptides consisting of modular sequences in which we systematically incorporate cationic, anionic, or aromatic residues at specific positions along the sequence in order to modulate intermolecular interactions and the resulting coacervation stability. We show that cation-π interactions between arginine and aromatic side chains are particularly efficient in stabilizing complex coacervates, and these interactions can be disrupted in the protein-rich intracellular environment, triggering the disassembly of complex coacervates followed by cargo release. With the additional grafting of a disulfide-based self-immolative side chain, these complex coacervates exhibited enhanced stability and could deliver proteins, mRNA, and CRISPR/Cas9 genome editing tools with tunable release kinetics into cells. This capability extends to challenging cell types, such as macrophages. Our study highlights the critical role of cation-π interactions in the design of peptide-based coacervates, expanding the biomedical and biotechnology potential of this emerging intracellular delivery platform.
Collapse
Affiliation(s)
- Yue Sun
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Xi Wu
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Jianguo Li
- Agency for Science, Technology and Research (A*STAR), Bioinformatics Institute, 30 Biopolis Street, Matrix, Singapore 138671, Singapore
- Singapore Eye Research Institute, Singapore 169856, Singapore
| | - Chandra Shekhar Verma
- Agency for Science, Technology and Research (A*STAR), Bioinformatics Institute, 30 Biopolis Street, Matrix, Singapore 138671, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore 117558, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Jing Yu
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, Singapore 636921, Singapore
| | - Ali Miserez
- Center for Sustainable Materials (SusMat), School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| |
Collapse
|
36
|
Zhong W, Zhang G, Yue K, Song Y, Zhao Z. MMP2 enzyme-responsive extracellular vesicles as dual-targeted carriers to promote the phagocytosis of macrophages. Colloids Surf B Biointerfaces 2025; 246:114365. [PMID: 39531838 DOI: 10.1016/j.colsurfb.2024.114365] [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: 08/08/2024] [Revised: 11/03/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Combination therapy using inhibition of tumor cell escape and alteration of the tumor microenvironment offers a new strategy for cancer treatment. This study aimed to develop an extracellular vesicle (EV) carrier that regulates tumor cells and the tumor microenvironment to achieve efficient tumor immunotherapy. The ligand modified on carriers targets the immune checkpoint CD47 protein, blocking tumor cell escape. This ligand is cleaved by the MMP2 enzyme and assembles into nanofibers, extending the retention time in the tumor. The carriers target the CD206 protein, enabling efficient uptake by M2 macrophages. Carriers with a high density of ligands (anti-CD206) exhibit strong receptorligand interactions with tumor cells. Due to their high rigidity, these EVs have difficulty deforming during the transmembrane process, reducing resistance and resulting in low uptake efficiency by M2 cells. The optimal uptake efficiency by M2 macrophages is achieved when the mass ratio of ligand to EVs is 1:25. Crocin loaded in EVs facilitates the polarization of M2 macrophages into M1 cells, which can phagocytize tumor cells. This study reveals a potential strategy for using extracellular vesicles in tumor treatment.
Collapse
Affiliation(s)
- Weishen Zhong
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Genpei Zhang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Kai Yue
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing 100083, China; Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong 528399, China.
| | - Yongmei Song
- State Key Laboratory of Molecular Oncology, National Cancer Center, China; National Clinical Research Center for Cancer, China; Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zitong Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center, China; National Clinical Research Center for Cancer, China; Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
37
|
Wang L, Hu Z, Zhang W, Wang Z, Cao M, Cao X. Promoting macrophage phagocytosis of cancer cells for effective cancer immunotherapy. Biochem Pharmacol 2025; 232:116712. [PMID: 39675588 DOI: 10.1016/j.bcp.2024.116712] [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: 10/02/2024] [Revised: 11/26/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
Cancer therapy has been revolutionized by immunotherapeutic agents exploiting adaptive antitumor immunity in the past two decades. However, the overall response rate of these immunotherapies is limited, and patients also develop resistance upon treatment, promoting a rapidly growing exploration of anti-tumor innate immunity for effective cancer therapy. Among these, macrophage immunotherapy through harnessing macrophage phagocytosis has been thrust into the spotlight due to its potential for simultaneously inducing cancer cell killing effect and mobilizing adaptive antitumor responses. Here in this review, we summarize the current macrophage immunotherapy such as therapeutic antibodies, phagocytosis checkpoint blockades, and CAR-macrophages with a particular emphasis on the resistant mechanisms limiting their therapeutic effects. Moreover, we further survey the efforts being placed to seek synergistic mechanisms and combination strategies for promoting macrophage phagocytosis which might stand as next-generation cancer immunotherapy.
Collapse
Affiliation(s)
- Lei Wang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Ziyi Hu
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Wencan Zhang
- Shanghai Key Laboratory of Veterinary Biotechnology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhixin Wang
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China
| | - Ming Cao
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xu Cao
- Shanghai Frontiers Science Center for Drug Target Identification and Delivery, and the Engineering Research Center of Cell and Therapeutic Antibody of the Ministry of Education, School of Pharmaceutical Sciences, National Key Laboratory of Innovative Immunotherapy, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
38
|
Liu Z, Li Y, Ren Y, Chen J, Weng S, Zhou Z, Luo P, Chen Q, Xu H, Ba Y, Zuo A, Liu S, Zhang Y, Pan T, Han X. Efferocytosis: The Janus-Faced Gatekeeper of Aging and Tumor Fate. Aging Cell 2025; 24:e14467. [PMID: 39748782 PMCID: PMC11822654 DOI: 10.1111/acel.14467] [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: 07/20/2024] [Revised: 10/30/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
From embryogenesis to aging, billions of cells perish daily in mammals. The multistep process by which phagocytes engulf these deceased cells without eliciting an inflammatory response is called efferocytosis. Despite significant insights into the fundamental mechanisms of efferocytosis, its implications in disorders such as aging and cancer remain elusive. Upon summarizing and analyzing existing studies on efferocytosis, it becomes evident that efferocytosis is our friend in resolving inflammation, yet it transforms into our foe by facilitating tumor development and metastasis. This review illuminates recent discoveries regarding the emerging mechanisms of efferocytosis in clearing apoptotic cells, explores its connections with aging, examines its influence on tumor development and metastasis, and identifies the regulatory factors of efferocytosis within the tumor microenvironment. A comprehensive understanding of these efferocytosis facets offers insights into crucial physiological and pathophysiological processes, paving the way for innovative therapeutic approaches to combat aging and cancer.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
- Institute of Basic Medical SciencesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Yan Li
- Medical School of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuqing Ren
- Department of Respiratory and Critical Care MedicineThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Jingqi Chen
- Medical School of Zhengzhou UniversityZhengzhouHenanChina
| | - Siyuan Weng
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Zhaokai Zhou
- Department of UrologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Peng Luo
- The Department of Oncology, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Quan Chen
- Department of Neurosurgery, Xiangya HospitalCentral South UniversityChangshaHunanChina
| | - Hui Xu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuhao Ba
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Anning Zuo
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Shutong Liu
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Yuyuan Zhang
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
| | - Teng Pan
- Longgang District Maternity & Child Healthcare Hospital of Shenzhen City (Longgang Maternity and Child Institute of Shantou University Medical College)ShenzhenGuangdongChina
| | - Xinwei Han
- Department of Interventional RadiologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Institute of Zhengzhou UniversityZhengzhouHenanChina
- Interventional Treatment and Clinical Research Center of Henan ProvinceZhengzhouHenanChina
| |
Collapse
|
39
|
Yu L, Sun Y, Xie L, Tan X, Wang P, Xu S. Targeting QPCTL: An Emerging Therapeutic Opportunity. J Med Chem 2025; 68:929-943. [PMID: 39746038 DOI: 10.1021/acs.jmedchem.4c02247] [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: 01/04/2025]
Abstract
Glutaminyl cyclases, including glutaminyl-peptide cyclotransferase (QPCT) and glutaminyl-peptide cyclotransferase-like protein (QPCTL), primarily catalyze the cyclization of N-terminal glutamine or glutamate to pyroglutamate (pGlu). QPCTL, in particular, modifies the N-terminus of CD47, thereby regulating its interaction with signal-regulatory protein alpha (SIRPα) and modulating phagocytosis of tumor cells by immune cells. Additionally, QPCTL cyclizes the N-termini of CCL2, CCL7, and CX3CL1, influencing the tumor microenvironment and inflammatory responses in cancer and other disorders. Consequently, QPCTL is considered a valuable therapeutic target for several human diseases. However, the development of QPCTL inhibitors remains in its early stages. This perspective summarizes the structural features, catalytic mechanisms, and biological functions of QPCTL, along with its recent advances in small-molecule inhibitors. It provides valuable insights into the development of novel QPCTL inhibitors.
Collapse
Affiliation(s)
- Lei Yu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yaoliang Sun
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Longyan Xie
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Xiao Tan
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Shilin Xu
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
40
|
Tang D, He S, Yang Y, Zeng Y, Xiong M, Ding D, Wei W, Lyu Y, Zhang XB, Tan W. Microenvironment-confined kinetic elucidation and implementation of a DNA nano-phage with a shielded internal computing layer. Nat Commun 2025; 16:923. [PMID: 39843440 PMCID: PMC11754784 DOI: 10.1038/s41467-025-56219-9] [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: 06/30/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
Multiple receptor analysis-based DNA molecular computation has been developed to mitigate the off-target effect caused by nonspecific expression of cell membrane receptors. However, it is quite difficult to involve nanobodies into molecular computation with programmed recognition order because of the "always-on" response mode and the inconvenient molecular programming. Here we propose a spatial segregation-based molecular computing strategy with a shielded internal computing layer termed DNA nano-phage (DNP) to program nanobody into DNA molecular computation and build a series of kinetic models to elucidate the mechanism of microenvironment-confinement. We explain the contradiction between fast molecular diffusion and effective DNA computation using a "diffusion trap" theory and comprehensively overcome the kinetic bottleneck of DNP by determining the rate-limiting step. We predict and verify that identifying trace amount of target cells in complex cell mixtures is an intrinsic merit of microenvironment-confined DNA computation. Finally, we show that DNP can efficiently work in complex human blood samples by shielding the interference of erythrocytes and enhance phagocytosis of macrophages toward target cells by blocking CD47-SIRPα pathway.
Collapse
Affiliation(s)
- Decui Tang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, China
| | - Shuoyao He
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, China
| | - Yani Yang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, China
| | - Yuqi Zeng
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, China
| | - Mengyi Xiong
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, China
| | - Ding Ding
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yifan Lyu
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, China.
- Furong Laboratory, Changsha, Hunan, China.
| | - Xiao-Bing Zhang
- Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Aptamer Engineering Center of Hunan Province, Hunan University, Changsha, Hunan, China.
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| |
Collapse
|
41
|
Laddha K, Sobhia ME. Optimizing antibody stability and efficacy in CD47- SIRPα inhibition via computational approaches. Mol Divers 2025:10.1007/s11030-024-11037-x. [PMID: 39832086 DOI: 10.1007/s11030-024-11037-x] [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: 09/21/2024] [Accepted: 10/28/2024] [Indexed: 01/22/2025]
Abstract
CD47, a cell surface protein, serves as a "don't eat me" signal that prevents immune cells from engulfing healthy cells upon its interaction with SIRPα. Cancer cells exploit this mechanism by overexpressing CD47 to evade immune destruction. Blocking the interaction between CD47 and its receptor, SIRPα, is a promising therapeutic strategy. Targeting the interactions between these surface proteins with small molecules is quite challenging, and on the other hand, antibodies offer potential. However, the interactions between antigen (CD47) and antibody (B6H12.2) play a crucial role in this scenario, and increasing the affinity by mutating the interacting residues might impact the inclination and effectiveness of the antibody towards antigen. Thus, this study focuses on designing antibodies with increased affinity and stability towards the antigen compared to the wild-type. Residual scanning calculations were performed to mutate the interacting as well as the hydrophobic residues of the antibody and affinity was assessed. Computational approaches, including antigen-antibody docking studies and molecular dynamics simulations, were employed to evaluate the affinity, stability and therapeutic potential of these modified antibodies.
Collapse
Affiliation(s)
- Kapil Laddha
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, 160062, India
| | - M Elizabeth Sobhia
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Education and Research, S.A.S Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
42
|
Kong D, Chen Y, Yin Y, Liu Z, Yang F, Li X, Shen D, Zhang J. PD-L1 monoclonal antibody alleviated MI injury of left ventricular function via modulating CD47/SHP2/SIRPα/SYK/FcγR signalings in tumor associated macrophages. Sci Rep 2025; 15:2303. [PMID: 39824849 PMCID: PMC11748645 DOI: 10.1038/s41598-024-85065-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: 04/09/2024] [Accepted: 12/30/2024] [Indexed: 01/20/2025] Open
Abstract
To investigate how PD-L1 monoclonal antibodies (mAbs) affect the left ventricular function in mice with myocardial infarction (MI) and through what mechanisms they exert their effects. In vivo experiments were conducted using 27 female BALB/c mice, which were divided equally into 3 groups. Cardiac function was assessed by ultrasound. Heart tissue and breast cancer tumor samples were isolated, and the content of cGAMP was measured using LC-MS/MS. The extent of myocardial infarction was evaluated by Masson staining. In vitro experiments involved dividing macrophages, treated with different inducers, into 8 groups. Protein expression levels in each group were analyzed by Western blotting, and the macrophages were transplanted into experimental mice for observation. In the in vivo experiments, ultrasound examination showed that PD-L1 mAb improved cardiac function in mice with breast cancer and MI. Both cGAMP content measurement and Masson staining results indicated that PD-L1 mAb had a therapeutic effect on mice with breast cancer and MI, improving the infarct condition and slowing tumor progression. In vitro Western blotting analysis revealed that PD-L1 mAb can modulate the CD47/SHP2/SIRPα/SYK/FcγR signaling pathway, thereby affecting breast cancer. Treatment with a STING inhibitor significantly reduced the cGAMP effect, leading to improved left ventricular function in mice with MI. PD-L1 monoclonal antibodies improve left ventricular function in mice with myocardial infarction by modulating the CD47/SHP2/SIRPα/SYK/FcγR signaling pathway in tumor-associated macrophages and inhibiting the expression of cGAMP.
Collapse
Affiliation(s)
- Deyou Kong
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yongzhen Chen
- Department of Function, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Yajuan Yin
- Department of Cardiology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhikun Liu
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fang Yang
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Xiaohong Li
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Dongxing Shen
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| | - Jun Zhang
- Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China.
| |
Collapse
|
43
|
Zhang Z, Zhao L, Huang T, Chen Z, Zhao Y, Liang J, Ao X, Jia X, Kang L, Kong L, Jing Q, Hu J, Gu L, Pan F, Hu Z, He L, Zhou M, Chen J, Guo Z. A self-activated and protective module enhances the preclinical performance of allogeneic anti-CD70 CAR-T cells. Front Immunol 2025; 15:1531294. [PMID: 39906740 PMCID: PMC11792090 DOI: 10.3389/fimmu.2024.1531294] [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: 11/20/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Introduction Allogeneic chimeric antigen receptor T (CAR-T) therapy, also known as universal CAR-T (UCAR-T) therapy, offers broad applicability, high production efficiency, and reduced costs, enabling quicker access for patients. However, clinical application remains limited by challenges such as immune rejection, and issues with potency and durability. Methods We first screened a safe and effective anti-CD70 scFv to construct anti-CD70 CAR-T cells. Anti-CD70 UCAR-T cells were then generated by knocking out TRAC, B2M, and HLA-DRA. To address the limitations of UCAR-T therapy, we developed an 'all-in-one' self-activated and protective (SAP) module, integrated into the CAR scaffold. The SAP module consists of the CD47 extracellular domain, a mutant interleukin 7 receptor alpha (IL7Rα) transmembrane domain, and the IL7Rα intracellular domain, designed to protect UCAR-T cells from host immune attacks and enhance their survival. Results SAP UCAR-T cells demonstrated significantly reduced immune rejection from the innate immune system, as evidenced by enhanced survival and functionality both in vitro and in vivo. The modified UCAR-T cells exhibited improved persistence, with no observed safety concerns. Furthermore, SAP UCAR-T cells maintained process stability during scale-up production, indicating the potential for large-scale manufacturing. Discussion Our findings highlight the SAP module as a promising strategy for the preclinical development of anti-CD70 UCAR-T, paving the way for an 'off-the-shelf' cell therapy product.
Collapse
Affiliation(s)
- Zhao Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lianfeng Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tinghui Huang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhengliang Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
- Department of Research and Development, Nanjing Calmhome Cell & Gene Engineering Institute Co., Ltd., Nanjing, China
| | - Yaoyao Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
- Department of Research and Development, Nanjing Calmhome Cell & Gene Engineering Institute Co., Ltd., Nanjing, China
| | - Junqing Liang
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xudong Ao
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xiaoqiong Jia
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Lei Kang
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Linghui Kong
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Qi Jing
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Jianhua Hu
- Center of Biotherapy, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Lili Gu
- Department of Research and Development, Nanjing Calmhome Cell & Gene Engineering Institute Co., Ltd., Nanjing, China
| | - Feiyan Pan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lingfeng He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Muya Zhou
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jiannan Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
44
|
Chen Y, Chen X, Zhang Y, Wang M, Yang M, Wang R, Yan X, Shao S, Xin H, Hu Q, Wei W, Ping Y. Macrophage-specific in vivo RNA editing promotes phagocytosis and antitumor immunity in mice. Sci Transl Med 2025; 17:eadl5800. [PMID: 39813319 DOI: 10.1126/scitranslmed.adl5800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 07/09/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
Macrophages play a central role in antitumor immunity, making them an attractive target for gene therapy strategies. However, macrophages are difficult to transfect because of nucleic acid sensors that can trigger the degradation of foreign plasmid DNA. Here, we developed a macrophage-specific editing (MAGE) system by which compact plasmid DNA encoding a CasRx editor can be delivered to macrophages by a poly(β-amino ester) (PBAE) carrier to bypass the DNA sensor and enable RNA editing in vitro and in vivo. We identified a four-arm branched PBAE with 1-(2-aminoethyl)-4-methylpiperazine end-capping (PBAE29) that enables highly efficient macrophage transfection. PBAE29-mediated transfection of cultured macrophages stimulated less inflammatory cytokine production and inflammasome activation compared with traditional lipofectamine or electroporation-mediated plasmid delivery. Transfection efficiency was further improved by delivering CasRx by minicircle plasmid. The MAGE system incorporated a layer of carboxylated-mannan coating to target macrophage mannose receptors and a macrophage-specific promoter for enhanced selectivity. The delivery of CasRx with guide RNA targeting the transcripts for sialic acid-binding immunoglobulin similar to lectin 10 and signal regulatory protein alpha expression resulted in effective protein knockdown, improving macrophage phagocytosis. The MAGE system also showed efficacy in targeting macrophages in vivo, stimulating antitumor immune responses and reducing tumor volume in murine tumor models, including patient-derived pancreatic adenocarcinoma xenografts in humanized mice. In sum, the MAGE system presents a promising platform for in vivo macrophage-specific delivery of RNA editing tools that can be applied as a cancer therapy.
Collapse
Affiliation(s)
- Yuxuan Chen
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Xiaohong Chen
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, MOE Joint International Research Laboratory of Pancreatic Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yao Zhang
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Meng Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, MOE Joint International Research Laboratory of Pancreatic Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Minqi Yang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, MOE Joint International Research Laboratory of Pancreatic Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Ruiji Wang
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Xiaojie Yan
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| | - Shiyi Shao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, MOE Joint International Research Laboratory of Pancreatic Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Huhu Xin
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Qida Hu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, MOE Joint International Research Laboratory of Pancreatic Diseases, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou 311121, China
| |
Collapse
|
45
|
Yang Y, Li J, Zhang J, Wu H, Yang Y, Guo H, Zhang D, Ge C, Zhou B, Ma L, Dong W. PPAB001, a novel bispecific antibody against CD47 and CD24, enhances anti-PD-L1 efficacy in triple-negative breast cancer via reprogramming tumor-associated macrophages towards M1 phenotype. Int Immunopharmacol 2025; 144:113740. [PMID: 39622130 DOI: 10.1016/j.intimp.2024.113740] [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: 09/08/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/15/2024]
Abstract
Triple-negative breast cancer (TNBC) is a biologically aggressive tumor with a strong association with a high recurrence rate and poor prognosis. Although anti-PD-L1 antibody, Tecentriq has been approved by FDA for treating TNBC, the overall response rate (ORR) is still generally less than 20 %. PPAB001 is a novel bispecific antibody simultaneously targeting CD47 and CD24. In the present study, we firstly evaluated the activity of PPAB001 on promoting the phagocytosis of TNBC cell lines. And the efficacy by combination of PPAB001 and Tecentriq was also assessed in TNBC 4T-1 mouse model. Moreover, the expression profiling of macrophage-associated genes and surface markers were evaluated upon the combinatorial treatment by flow cytometry, Western blot and IHC analysis. Cell signaling involved in M1 macrophage polarization was further identified via the analysis of RNA-seq, Western blot and immunofluorescnece. Our results demonstrated that PPAB001 effectively promotes phagocytosis of macrophages against human TNBC cell lines and significantly delayed TNBC tumor growth, especially when combined with Tecentriq treatment which may be attributed to the mechanism that simultaneous blockade of CD47 and CD24 signaling maximized the polarization toward M1 phenotype polarization. Especially, RNA-seq analysis and Western blotting further revealed that CXCL9/10-CXCR3 axis was markedly up-regulated and JAK/STAT1 pathway was activated upon treatment with PPAB001 plus Tecentriq. Overall, our results underscore that simultaneous blockade of CD47 and CD24 is a potential therapeutic option to improve the efficacy of anti-PD-L1 therapy mainly by resetting tumor-associated macrophages toward M1 phenotype.
Collapse
Affiliation(s)
- Yun Yang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
| | - Jianqin Li
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Jing Zhang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - He Wu
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Yan Yang
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Huaizu Guo
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, Shanghai, China
| | - Dapeng Zhang
- State Key Laboratory of Macromolecular Drugs and Large-scale Manufacturing, Shanghai, China
| | - Chunpo Ge
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Bei Zhou
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Ling Ma
- School of Nursing and Health, Zhengzhou University, Zhengzhou, China.
| | - Weihua Dong
- International Joint Research Laboratory for Recombinant Pharmaceutical Protein Expression System of Henan, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
46
|
Liu Y, Tan H, Dai J, Lin J, Zhao K, Hu H, Zhong C. Targeting macrophages in cancer immunotherapy: Frontiers and challenges. J Adv Res 2025:S2090-1232(24)00622-2. [PMID: 39778768 DOI: 10.1016/j.jare.2024.12.043] [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: 10/11/2024] [Accepted: 12/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Cancer immunotherapy has emerged as a groundbreaking approach in cancer treatment, primarily realized through the manipulation of immune cells, notably T cell adoption and immune checkpoint blockade. Nevertheless, the manipulation of T cells encounters formidable hurdles. Macrophages, serving as the pivotal link between innate and adaptive immunity, play crucial roles in phagocytosis, cytokine secretion, and antigen presentation. Consequently, macrophage-targeted therapies have garnered significant attention. AIM OF REVIEW We aim to provide the most cutting-edge insights and future perspectives for macrophage-targeted therapies, fostering the development of novel and effective cancer treatments. KEY SCIENTIFIC CONCEPTS OF REVIEW To date, the forefront strategies for macrophage targeting encompass: altering their plasticity, harnessing CAR-macrophages, and targeting phagocytosis checkpoints. Macrophages are characterized by their remarkable diversity and plasticity, offering a unique therapeutic target. In this context, we critically analyze the innovative strategies aimed at transforming macrophages from their M2 (tumor-promoting) to M1 (tumor-suppressing) phenotype. Furthermore, we delve into the design principles, developmental progress, and advantages of CAR-macrophages. Additionally, we illuminate the challenges encountered in targeting phagocytosis checkpoints on macrophages and propose potential strategies to overcome these obstacles.
Collapse
Affiliation(s)
- Yu'e Liu
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China; Department of Pediatric Hematology-Oncology, Boston Children's Hospital, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| | - Huabing Tan
- Department of Infectious Diseases, Hepatology Institute, Renmin Hospital, Hubei University of Medicine, Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, Hubei Province 442000, China; General internal medicine, Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, 430048, China
| | - Jingyuan Dai
- School of Computer Science and Information Systems, Northwest Missouri State University, Maryville, MO 64468, USA
| | - Jianghua Lin
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China
| | - Kaijun Zhao
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China.
| | - Haibo Hu
- Department of Cardiothoracic Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China.
| | - Chunlong Zhong
- Department of Neurosurgery, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai 200120, China.
| |
Collapse
|
47
|
Ma C, Li Y, Li M, Lv C, Tian Y. Targeting immune checkpoints on myeloid cells: current status and future directions. Cancer Immunol Immunother 2025; 74:40. [PMID: 39751898 PMCID: PMC11699031 DOI: 10.1007/s00262-024-03856-6] [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: 09/11/2024] [Accepted: 10/07/2024] [Indexed: 01/04/2025]
Abstract
Myeloid cells accumulate extensively in most tumors and play a critical role in immunosuppression of the tumor microenvironment (TME). Like T cells, myeloid cells also express immune checkpoint molecules, which induce the immunosuppressive phenotype of these cells. In this review, we summarize the tumor-promoting function and immune checkpoint expression of four types of myeloid cells: macrophages, neutrophils, dendritic cells, and myeloid-derived suppressor cells, which are the main components of the TME. By summarizing the research status of myeloid checkpoints, we propose that blocking immune checkpoints on myeloid cells might be an effective strategy to reverse the immunosuppressive status of the TME. Moreover, combining nanotechnology, cellular therapy, and bispecific antibodies to achieve precise targeting of myeloid immune checkpoints can help to avoid the adverse effects of systemic administration, ultimately achieving a balance between efficacy and safety in cancer therapy.
Collapse
Affiliation(s)
- Chuhan Ma
- Department of General Surgery, Shengjing Hospital of China Medical University, ShenyangLiaoning Province, 110004, China
| | - Yang Li
- Department of General Surgery, Shengjing Hospital of China Medical University, ShenyangLiaoning Province, 110004, China
| | - Min Li
- Department of Mammary Gland, Dalian Women and Children's Medical Center (Group), DalianLiaoning Province, 116000, China
| | - Chao Lv
- Department of General Surgery, Shengjing Hospital of China Medical University, ShenyangLiaoning Province, 110004, China.
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, ShenyangLiaoning Province, 110004, China.
| |
Collapse
|
48
|
Salminen A. The role of inhibitory immune checkpoint receptors in the pathogenesis of Alzheimer's disease. J Mol Med (Berl) 2025; 103:1-19. [PMID: 39601807 PMCID: PMC11739239 DOI: 10.1007/s00109-024-02504-x] [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: 06/24/2024] [Revised: 10/16/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
There is mounting evidence that microglial cells have a key role in the pathogenesis of Alzheimer's disease (AD). In AD pathology, microglial cells not only are unable to remove β-amyloid (Aβ) plaques and invading pathogens but also are involved in synaptic pruning, chronic neuroinflammation, and neuronal degeneration. Microglial cells possess many different inhibitory immune checkpoint receptors, such as PD-1, LILRB2-4, Siglecs, and SIRPα receptors, which can be targeted by diverse cell membrane-bound and soluble ligand proteins to suppress the functions of microglia. Interestingly, in the brains of AD patients there are elevated levels of many of the inhibitory ligands acting via these inhibitory checkpoint receptors. For instance, Aβ oligomers, ApoE4, and fibronectin are able to stimulate the LILRB2-4 receptors. Increased deposition of sialoglycans, e.g., gangliosides, inhibits microglial function via Siglec receptors. AD pathology augments the accumulation of senescent cells, which are known to possess a high level of PD-L1 proteins, and thus, they can evade immune surveillance. A decrease in the expression of SIRPα receptor in microglia and its ligand CD47 in neurons enhances the phagocytic pruning of synapses in AD brains. Moreover, cerebral neurons contain inhibitory checkpoint receptors which can inhibit axonal growth, reduce synaptic plasticity, and impair learning and memory. It seems that inappropriate inhibitory immune checkpoint signaling impairs the functions of microglia and neurons thus promoting AD pathogenesis. KEY MESSAGES: Microglial cells have a major role in the pathogenesis of AD. A decline in immune activity of microglia promotes AD pathology. Microglial cells and neurons contain diverse inhibitory immune checkpoint receptors. The level of ligands for inhibitory checkpoint receptors is increased in AD pathology. Impaired signaling of inhibitory immune checkpoint receptors promotes AD pathology.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
49
|
Priya, Kumar A, Kumar D. Molecular heterogeneity and MYC dysregulation in triple-negative breast cancer: genomic advances and therapeutic implications. 3 Biotech 2025; 15:33. [PMID: 39777154 PMCID: PMC11700964 DOI: 10.1007/s13205-024-04195-0] [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: 08/25/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by a diverse range of molecular features that have been extensively studied. MYC plays a critical role in regulating metabolism, differentiation, proliferation, cell growth, and apoptosis. Dysregulation of MYC is associated with poor prognosis and contributes to the development and progression of breast cancer. A particularly intriguing aspect of TNBC is its association with tumors in BRCA1 mutation carriers, especially in younger women. MYC may also contribute to resistance to adjuvant treatments. For TNBC, targeting MYC-regulated pathways in combination with inhibitors of other carcinogenic pathways offers a promising therapeutic approach. Several signaling pathways regulate TNBC, and targeting these pathways could lead to effective therapeutic strategies for breast cancer. Advances in genomic tools, such as CRISPR-Cas9, next-generation sequencing, and whole-exome sequencing, are revolutionizing breast cancer diagnoses. These technologies have significantly enhanced our understanding of MYC oncogenesis, particularly through CRISPR-Cas9 and NGS. Targeting MYC and its partner MAX could provide valuable insights into TNBC. Moreover, the therapeutic potential of targeting MYC-driven signaling mechanisms and their interactions with other oncogenic pathways, including PI3K/AKT/mTOR and Wnt/β-catenin, is increasingly recognized. Next-generation sequencing and CRISPR-Cas9 represent significant breakthroughs in genomic tools that open new opportunities to explore MYC's role in TNBC and facilitate the development of personalized treatment plans. This review discusses the future clinical applications of personalized treatment strategies for patients with TNBC.
Collapse
Affiliation(s)
- Priya
- School of Health Sciences and Technology (SoHST), UPES, Dehradun, Uttarakhand 248007 India
| | - Arun Kumar
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar 801505 India
| | - Dhruv Kumar
- School of Health Sciences and Technology (SoHST), UPES, Dehradun, Uttarakhand 248007 India
| |
Collapse
|
50
|
Che Z, Wang W, Zhang L, Lin Z. Therapeutic strategies targeting CD47-SIRPα signaling pathway in gastrointestinal cancers treatment. J Pharm Anal 2025; 15:101099. [PMID: 39881799 PMCID: PMC11772969 DOI: 10.1016/j.jpha.2024.101099] [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: 03/28/2024] [Revised: 09/04/2024] [Accepted: 09/07/2024] [Indexed: 01/31/2025] Open
Abstract
Gastrointestinal (GI) cancers are prevalent globally, with leading incidence and mortality rates among malignant tumors. Despite notable advancements in surgical resection, radiotherapy, and chemotherapy, the overall survival rates remain low. Hence, it is imperative to explore alternative approaches that enhance patient outcomes. Cluster of differentiation 47 (CD47), serving as an early diagnostic marker, is predominantly overexpressed in GI cancers and associated with poor prognosis. Targeting the CD47-signal regulatory protein alpha (SIRPα) signaling pathway may provide a novel strategy for GI cancers treatment. This study summarizes current knowledge of the structure and function of CD47 and SIRPα, their roles in signaling pathways, the prognostic significance of CD47, therapeutic strategies targeting the CD47-SIRPα signaling pathway in GI cancer, and highlights key issues for future investigations.
Collapse
Affiliation(s)
- Zhengping Che
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| | - Wei Wang
- Department of Cancer Center, Chongqing University Three Gorges Hospital, School of Medicine, Chongqing University, Chongqing, 404000, China
| | - Lin Zhang
- Department of Gastroenterology, Chongqing University Jiangjin Hospital, Chongqing, 402260, China
| | - Zhenghong Lin
- School of Life Sciences, Chongqing University, Chongqing, 401331, China
| |
Collapse
|