1
|
Yang Y, Zhu L, Xu Y, Liang L, Liu L, Chen X, Li H, Liu H. The progress and prospects of targeting the adenosine pathway in cancer immunotherapy. Biomark Res 2025; 13:75. [PMID: 40390144 PMCID: PMC12090549 DOI: 10.1186/s40364-025-00784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 04/26/2025] [Indexed: 05/21/2025] Open
Abstract
Despite the notable success of cancer immunotherapy, its effectiveness is often limited in a significant proportion of patients, highlighting the need to explore alternative tumor immune evasion mechanisms. Adenosine, a key metabolite accumulating in hypoxic tumor regions, has emerged as a promising target in oncology. Inhibiting the adenosinergic pathway not only inhibits tumor progression but also holds potential to enhance immunotherapy outcomes. Multiple therapeutic strategies targeting this pathway are being explored, ranging from preclinical studies to clinical trials. This review examines the complex interactions between adenosine, its receptors, and the tumor microenvironment, proposing strategies to target the adenosinergic axis to boost anti-tumor immunity. It also evaluates early clinical data on pharmacological inhibitors of the adenosinergic pathway and discusses future directions for improving clinical responses.
Collapse
Affiliation(s)
- Yuying Yang
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Lin Zhu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yantao Xu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Long Liang
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Li Liu
- Molecular Biology Research Center and Center for Medical Genetics, School of Life Sciences, Central South University, Changsha, Hunan, 410078, China
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Hui Li
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| | - Hong Liu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Central South University, Changsha, Hunan, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China.
| |
Collapse
|
2
|
Chen Y, Song Y, Zhang C, Jin P, Fu Y, Wang G, Tang L, Chen J, Xu X, Huang P. Ultrasound-responsive release of CD39 inhibitor overcomes adenosine-mediated immunosuppression in triple-negative breast cancer. J Control Release 2025; 383:113819. [PMID: 40345625 DOI: 10.1016/j.jconrel.2025.113819] [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: 03/03/2025] [Revised: 04/28/2025] [Accepted: 05/04/2025] [Indexed: 05/11/2025]
Abstract
Triple-negative breast cancer (TNBC), an exceptionally aggressive subtype of breast cancer, is characterized by a poor prognosis and limited treatment options. Although immunotherapy has shown promise for the treatment of TNBC, the immunosuppressive accumulation of adenosine (ADO) in the tumor microenvironment (TME) contributes to immune evasion and tumor progression. To address this challenge, we introduce a novel ultrasound-responsive liposomal system (BFPL) designed to inhibit ADO production and enhance the effectiveness of sonoimmunotherapy. BFPL consists of lipid membranes loaded with an endoplasmic reticulum (ER)-targeting sonosensitizer (PMPS) and a reactive oxygen species (ROS)-responsive CD39 inhibitor (FPL-67156) polyplex, synthesized via the thin-film hydration method. Upon ultrasound irradiation, BFPL generates substantial ROS, inducing robust immunogenic cell death (ICD) through ER stress. Concurrently, ROS-mediated deboronation of the polyplex releases FPL-67156, which inhibits ATP degradation into ADO, thereby promoting dendritic cell maturation and activating effector T cells. Moreover, BFPL effectively triggers a potent antitumor immune response and enhances the efficacy of anti-PD-L1 immunotherapy. Thus, by modulating metabolic pathways to counteract ADO-associated barriers in ICD therapy, this innovative approach holds potential for improving immunotherapy outcomes in TNBC.
Collapse
Affiliation(s)
- Yijie Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Department of Ultrasound, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Yue Song
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Chao Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Peile Jin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Yuhan Fu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Guowei Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Lina Tang
- Department of Ultrasound, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou 350014, China
| | - Jifan Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Xiaodan Xu
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China; Research Center of Ultrasound in Medicine and Biomedical Engineering, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou 310009, China.
| |
Collapse
|
3
|
Wu X, Zhang J, Deng Z, Sun X, Zhang Y, Zhang C, Wang J, Yu X, Yang G. Bacteria-based biohybrids for remodeling adenosine-mediated immunosuppression to boost radiotherapy-triggered antitumor immune response. Biomaterials 2025; 316:123000. [PMID: 39674101 DOI: 10.1016/j.biomaterials.2024.123000] [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: 07/17/2024] [Revised: 11/18/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
Radiotherapy (RT) can trigger immunogenic cell death (ICD) in tumor cells and release adenosine triphosphate (ATP) to activate antitumor immunity. However, the formation of immunosuppressive adenosine (ADO) mediated by ectonucleotidases including CD39 and CD73, can exacerbate the immunosuppressive effects. Herein, a radiosensitizer-based metal-organic framework (MOF) composed of bismuth (Bi) and ellagic acid (EA) was synthesized in situ on the surface of Escherichia coli Nissle 1917 (EcN) to serve as a carrier for the CD39 inhibitor sodium polyoxotungstate (POM-1). This therapeutic platform, acting as a radiosensitizer, significantly enhances cytotoxicity against tumor cells while effectively inducing ICD and releasing high concentrations of ATP. Subsequently, the released POM-1 increases the levels of pro-inflammatory extracellular ATP while preventing tumor immunosuppression caused by the accumulation of ADO. Additionally, as a natural immune adjuvant, EcN further promotes the maturation of dendritic cells (DCs) and the infiltration of cytotoxic T lymphocytes (CTLs). As a result, such treatment initiates the destruction of established tumor growth and induces strong abscopal effects, leading to a significant inhibition of tumor metastases. This strategy presents a bacterial-based biohybrid system that facilitates RT-induced ICD while simultaneously limiting the degradation of ATP into ADO, thereby achieving sustained anti-tumor immunity.
Collapse
Affiliation(s)
- Xirui Wu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Junjun Zhang
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Institute of Radiotherapy & Oncology, Soochow University, Suzhou 215004, China
| | - Zheng Deng
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xianglong Sun
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Yifan Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Cai Zhang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jiadong Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Xinke Yu
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Guangbao Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China.
| |
Collapse
|
4
|
Yang J, des Rieux A, Malfanti A. Stimuli-Responsive Nanomedicines for the Treatment of Non-cancer Related Inflammatory Diseases. ACS NANO 2025; 19:15189-15219. [PMID: 40249331 PMCID: PMC12045021 DOI: 10.1021/acsnano.5c00700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Nanomedicines offer a means to overcome the limitations associated with traditional drug dosage formulations by affording drug protection, enhanced drug bioavailability, and targeted drug delivery to affected sites. Inflamed tissues possess unique microenvironmental characteristics (including excessive reactive oxygen species, low pH levels, and hypoxia) that stimuli-responsive nanoparticles can employ as triggers to support on-demand delivery, enhanced accumulation, controlled release, and activation of anti-inflammatory drugs. Stimuli-responsive nanomedicines respond to physicochemical and pathological factors associated with diseased tissues to improve the specificity of drug delivery, overcome multidrug resistance, ensure accurate diagnosis and precision therapy, and control drug release to improve efficacy and safety. Current stimuli-responsive nanoparticles react to intracellular/microenvironmental stimuli such as pH, redox, hypoxia, or specific enzymes and exogenous stimuli such as temperature, magnetic fields, light, and ultrasound via bioresponsive moieties. This review summarizes the general strategies employed to produce stimuli-responsive nanoparticles tailored for inflammatory diseases and all recent advances, reports their applications in drug delivery, and illustrates the progress made toward clinical translation.
Collapse
Affiliation(s)
- Jingjing Yang
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Anne des Rieux
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
| | - Alessio Malfanti
- UCLouvain,
Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200, Brussels, Belgium
- Department
of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| |
Collapse
|
5
|
Hsieh CH, Chuang PC, Liu YW. Beyond Adaptive Immunity: Trained Innate Immune Responses as a Novel Frontier in Hepatocellular Carcinoma Therapy. Cancers (Basel) 2025; 17:1250. [PMID: 40227782 PMCID: PMC11987826 DOI: 10.3390/cancers17071250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Revised: 03/23/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer death globally, with the majority of cases detected at advanced stages when curative options are limited. Current systemic therapies, including immune checkpoint inhibitors, demonstrate limited efficacy with durable responses in only 15-20% of patients. This poor response is largely attributed to HCC's immunosuppressive microenvironment, which blunts effective T-cell responses. By illustrating that innate immune cells can acquire memory-like characteristics through a process known as trained immunity, recent evidence has challenged the conventional belief that innate immunity is devoid of memory. This review investigates the potential of trained immunity, which is defined by the long-term functional reprogramming of innate immune cells through epigenetic, transcriptomic, and metabolic changes, to provide new therapeutic opportunities for HCC. We discuss mechanisms by which trained immunity can transform the HCC microenvironment, including enhanced inflammatory cytokine production, repolarization of tumor-associated macrophages toward anti-tumor phenotypes, increased immune cell infiltration, and improved bridging to adaptive immunity. We further evaluate emerging therapeutic strategies leveraging trained immunity principles, including BCG vaccination, β-glucan administration, cytokine-trained NK cell therapy, and innovative combination approaches. Finally, we address potential resistance mechanisms and future directions for clinical application. By integrating trained immunity into conventional immunotherapeutic regimens, we may significantly improve outcomes for HCC patients, potentially transforming advanced disease into a more manageable condition.
Collapse
Affiliation(s)
- Ching-Hua Hsieh
- Department of Plastic Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan;
| | - Pei-Chin Chuang
- Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Yueh-Wei Liu
- Department of General Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 83301, Taiwan
- Department of General Surgery, Kaohsiung Municipal Fong Shan Hospital—Under the Management of Chang Gung Medical Foundation, Kaohsiung 83091, Taiwan
| |
Collapse
|
6
|
Kurowski K, Prozmann SN, Cabrita Figueiredo AE, Heyer J, Kind F, Schröder KM, Passlick B, Werner M, Bronsert P, Schmid S. The Ectonucleotidases CD39 and CD73 and the Purinergic Receptor P2X4 Serve as Prognostic Markers in Non-Small Cell Lung Cancer. Cancers (Basel) 2025; 17:1142. [PMID: 40227655 PMCID: PMC11987875 DOI: 10.3390/cancers17071142] [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: 02/14/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Purinergic signaling, which involves extracellular ATP (eATP), its metabolites, purinergic receptors and ectonucleotidases, plays a pivotal role in the tumor microenvironment (TME), impacting tumor progression and the antineoplastic immune response. In this study, the CD39, CD73, P2X4, and P2X7 expression in NSCLC tumor cells and the surrounding stroma of 139 resected patients was examined. METHODS The study included tissue samples from 139 NSCLC patients. Tissue microarrays (TMAs) were constructed using 1.0 mm cores from annotated tumor regions. Immunohistochemical staining for CD39, CD73, P2X4, and P2X4 was performed on 2 µm sections. TMA slides were digitized and analyzed with QuPath, where staining intensity was evaluated using a semi-quantitative H-score. Statistical analysis, including survival analysis, was performed using R, to assess the impact of biomarker expression on patient outcomes. RESULTS High CD39 expression in both tumor and stromal cells was significantly associated with prolonged PFS (respectively: p = 0.0058 and p = 0.0067), particularly in adenocarcinoma (ADC) patients (respectively: p = 0.01 and p = 0.023). In the multivariable Cox model, low CD73 expression in tumor cells correlated with longer PFS (HR: 0.47; 95% CI: [0.28, 0.8], p = 0.005), while low CD73 expression in stromal cells was linked to increased progression risk (HR: 4.81; 95% CI: [1.61, 14.4], p = 0.001). Neither P2X7 nor P2X4 demonstrated a consistent effect on PFS in univariable analyses; however, multivariable analyses suggested that P2X4 might play a prognostic role in NSCLCs (HR: 0.37; 95% CI: [0.19, 0.73], p = 0.003). CONCLUSIONS These findings underscore the importance of purinergic signaling in NSCLC prognosis and highlight the role of the ectonucleotidases CD39 and CD73 as potential therapeutic targets to enhance antineoplastic immune responses.
Collapse
Affiliation(s)
- Konrad Kurowski
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Core Facility Histopathology and Digital Pathology Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Tumorbank Comprehensive Cancer Center Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
| | - Sophie Nicole Prozmann
- Department of Thoracic Surgery, University Medical Center Freiburg, 79106 Freiburg, Germany
| | | | - Jannis Heyer
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Core Facility Histopathology and Digital Pathology Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Tumorbank Comprehensive Cancer Center Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
| | - Felix Kind
- Department of Nuclear Medicine, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Karl-Moritz Schröder
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center–University of Freiburg, 79106 Freiburg, Germany
| | - Bernward Passlick
- Department of Thoracic Surgery, University Medical Center Freiburg, 79106 Freiburg, Germany
| | - Martin Werner
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Core Facility Histopathology and Digital Pathology Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Tumorbank Comprehensive Cancer Center Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
| | - Peter Bronsert
- Institute for Surgical Pathology, Faculty of Medicine, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Core Facility Histopathology and Digital Pathology Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
- Tumorbank Comprehensive Cancer Center Freiburg, Medical Center–University of Freiburg, 79106 Freiburg, Germany
| | - Severin Schmid
- Department of Thoracic Surgery, University Medical Center Freiburg, 79106 Freiburg, Germany
| |
Collapse
|
7
|
Shin K, Park M, Kim S, Lee H, Lee Y, Kim J, Park S, Kim J, Lee K, Park CW, Kim JH, Lee EJ, Mok H, Oh SM, Lee S, Oh YM, Lee W, Shim YA, Cho YG, Park J, Lee JY, Koh YJ, Kim KH, Jang MH. Novel anti-CD73-IL-2v bispecific fusion protein augments antitumor immunity by alleviating immunosuppressive adenosine pathways in CD8 + T cells. J Immunother Cancer 2025; 13:e008594. [PMID: 40081940 PMCID: PMC11906993 DOI: 10.1136/jitc-2023-008594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/26/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Adenosine accumulated in the tumor microenvironment functions as an immune-modulating factor, exerting immunosuppressive actions via adenosine A2A/A2B receptor (A2AR/A2BR) in various immune cell types. CD73, a key enzymatic regulator responsible for adenosine production, is frequently overexpressed in diverse cancers, and its overexpression is associated with reduced responsiveness to conventional anti-cancer drug treatments such as chemotherapy, radiation therapy, targeted therapy, or immunotherapy. Despite numerous therapeutic applications of IL-2 in cancer immunotherapy, the relationship between the CD73-adenosine axis and IL-2-based immunotherapy remains largely unexplored. METHODS To evaluate the effect of CD73 blockade on IL-2 signaling of CD8+ T cells, we screened novel CD73 antibodies using human single-chain variable fragment phage library and immunized Alpaca phage library. To optimize targeting to CD73-expressing cells and reinvigorate the antitumor effect of IL-2 in adenosine-rich microenvironment, we engineered a novel bifunctional GI-αCD73/IL-2v fusion protein. Functionality of GI-αCD73/IL-2v fusion protein was assessed in the in vitro cell-based assays and the in vivo tumor-bearing mouse model or cynomolgus monkey. RESULTS IL-2-induced increase in proliferation of CD8+ T cells was not observed under adenosine-rich microenvironment. We demonstrated that the functional impairment of IL-2 signaling in CD8+ T cells in these conditions can be reversed by our anti-CD73 antibody (GI-αCD73). Furthermore, GI-αCD73/IL-2v fusion protein significantly restored the impaired proliferation of CD8+ T cells and consequently enhanced tumor cell killing under adenosine-mediated immunosuppression, surpassing the combined treatment of GI-αCD73 and Fc-IL-2v. These synergistic effects were attributed to the enhanced delivery of the IL-2v component of GI-αCD73/IL-2v to IL-2Rβγ on CD73-expressing CD8+ T cells through a cis-binding mechanism. GI-αCD73/IL-2v elicited a potent antitumor effect in both the human CD73 knock-in (hCD73 KI) mouse model and the humanized mouse model. In non-human primates, GI-αCD73/IL-2v exhibited excellent tolerability while inducing robust and durable expansions of cytotoxic lymphocytes. CONCLUSIONS GI-αCD73/IL-2v bispecific protein is a novel and potent immunocytokine with significant antitumor immunity through cis-binding on CD8+ T cells.
Collapse
Affiliation(s)
- Kayoung Shin
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Min Park
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Seoho Kim
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Haejong Lee
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Yuseong Lee
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Jongil Kim
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Suyoun Park
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Jisoo Kim
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Kyungwha Lee
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Chong Woo Park
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Ji-Hyun Kim
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Eun-Jin Lee
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Hyuckjun Mok
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Sung-Man Oh
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Sanghee Lee
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Young Min Oh
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Wonjae Lee
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Yaein Amy Shim
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Young-Gyu Cho
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Junsik Park
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seodaemun-gu, Korea (the Republic of)
| | - Jung-Yun Lee
- Department of Obstetrics and Gynecology, Yonsei University College of Medicine, Seodaemun-gu, Korea (the Republic of)
| | - Young Jun Koh
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Kook Hwan Kim
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
| | - Myoung Ho Jang
- Research Institute, GI Innovation Inc, Songpa-gu, Korea (the Republic of)
- World Premier International Immunology Frontier Research Center, Osaka University, Suita, Japan
| |
Collapse
|
8
|
Viel S, Vivier E, Walzer T, Marçais A. Targeting metabolic dysfunction of CD8 T cells and natural killer cells in cancer. Nat Rev Drug Discov 2025; 24:190-208. [PMID: 39668206 DOI: 10.1038/s41573-024-01098-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/06/2024] [Indexed: 12/14/2024]
Abstract
The importance of metabolic pathways in regulating immune responses is now well established, and a mapping of the bioenergetic metabolism of different immune cell types is under way. CD8 T cells and natural killer (NK) cells contribute to cancer immunosurveillance through their cytotoxic functions and secretion of cytokines and chemokines, complementing each other in target recognition mechanisms. Several immunotherapies leverage these cell types by either stimulating their activity or redirecting their specificity against tumour cells. However, the anticancer activity of CD8 T cells and NK cells is rapidly diminished in the tumour microenvironment, closely linked to a decline in their metabolic capacities. Various strategies have been developed to restore cancer immunosurveillance, including targeting bioenergetic metabolism or genetic engineering. This Review provides an overview of metabolic dysfunction in CD8 T cells and NK cells within the tumour microenvironment, highlighting current therapies aiming to overcome these issues.
Collapse
Affiliation(s)
- Sébastien Viel
- Plateforme de Biothérapie et de Production de Médicaments de Thérapie Innovante, Hôpital Edouard Herriot, Hospices Civils de Lyon, Lyon, France
| | - Eric Vivier
- Innate Pharma Research Laboratories, Innate Pharma, Marseille, France
- Aix Marseille University, CNRS, INSERM, CIML, Marseille, France
- APHM, Hôpital de la Timone, Marseille, France
- Paris Saclay Cancer Cluster, Villejuif, France
- Université Paris-Saclay, Gustave Roussy, Inserm, Prédicteurs moléculaires et nouvelles cibles en oncologie, Villejuif, France
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS UMR5308 ENS de Lyon, Lyon, France
| | - Antoine Marçais
- CIRI, Centre International de Recherche en Infectiologie, (Team Lyacts), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS UMR5308 ENS de Lyon, Lyon, France.
| |
Collapse
|
9
|
Lin L, Roccuzzo G, Yakymiv Y, Marchisio S, Ortolan E, Funaro A, Senetta R, Pala V, Bagot M, de Masson A, Battistella M, Guenova E, Ribero S, Quaglino P. The CD39/CD73/Adenosine and NAD/CD38/CD203a/CD73 Axis in Cutaneous T-Cell Lymphomas. Cells 2025; 14:309. [PMID: 39996780 PMCID: PMC11854806 DOI: 10.3390/cells14040309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 02/26/2025] Open
Abstract
Cutaneous T-cell lymphoma (CTCL), characterized by malignant T-cell proliferation primarily in the skin, includes subtypes such as mycosis fungoides (MF) and Sézary syndrome (SS). The tumor microenvironment (TME) is central to their pathogenesis, with flow cytometry and histology being the gold standards for detecting malignant T cells within the TME. Alongside emerging molecular markers, particularly clonality analysis, these tools are indispensable for accurate diagnosis and treatment planning. Of note, adenosine signaling within the TME has been shown to suppress immune responses, affecting various cell types. The expression of CD39, CD73, and CD38, enzymes involved in adenosine production, can be elevated in MF and SS, contributing to immune suppression. Conversely, the expression of CD26, part of the adenosine deaminase/CD26 complex, that degrades adenosine, is often lost by circulating tumoral cells. Flow cytometry has demonstrated increased levels of CD39 and CD73 on Sézary cells, correlating with disease progression and prognosis, while CD38 shows a variable expression, with its prognostic significance remaining under investigation. Understanding these markers' roles in the complexity of TME-mediated immune evasion mechanisms might enhance diagnostic precision and offer new therapeutic targets in CTCL.
Collapse
Affiliation(s)
- Liyun Lin
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.L.); (Y.Y.); (S.M.); (E.O.); (A.F.)
| | - Gabriele Roccuzzo
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (G.R.); (V.P.); (P.Q.)
| | - Yuliya Yakymiv
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.L.); (Y.Y.); (S.M.); (E.O.); (A.F.)
| | - Sara Marchisio
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.L.); (Y.Y.); (S.M.); (E.O.); (A.F.)
| | - Erika Ortolan
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.L.); (Y.Y.); (S.M.); (E.O.); (A.F.)
| | - Ada Funaro
- Laboratory of Immunogenetics, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (L.L.); (Y.Y.); (S.M.); (E.O.); (A.F.)
| | - Rebecca Senetta
- Pathology Unit, Department of Oncology, University of Turin, 10126 Turin, Italy;
| | - Valentina Pala
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (G.R.); (V.P.); (P.Q.)
| | - Martine Bagot
- Dermatology Department, Saint-Louis Hospital, AP-HP, Université Paris Cité, Inserm U976, 75010 Paris, France; (M.B.); (A.d.M.)
| | - Adèle de Masson
- Dermatology Department, Saint-Louis Hospital, AP-HP, Université Paris Cité, Inserm U976, 75010 Paris, France; (M.B.); (A.d.M.)
| | - Maxime Battistella
- Department of Pathology, Hôpital Saint-Louis, AP-HP, Université Paris Cité, Inserm U976, 75010 Paris, France;
| | - Emmanuella Guenova
- Department of Dermatology, Lausanne University Hospital (CHUV) and Faculty of Biology and Medicine, University of Lausanne, 1007 Lausanne, Switzerland;
- University Institute and Clinic for Immunodermatology, Medical Faculty, Johannes Kepler University, 4020 Linz, Austria
| | - Simone Ribero
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (G.R.); (V.P.); (P.Q.)
| | - Pietro Quaglino
- Section of Dermatology, Department of Medical Sciences, University of Turin, 10126 Turin, Italy; (G.R.); (V.P.); (P.Q.)
| |
Collapse
|
10
|
Zhang X, Shi C, Liu Q, Zhong Y, Zhu L, Zhao Y. Combination of adenosine blockade and ferroptosis for photo-immunotherapy of triple negative breast cancer with aptamer-modified copper sulfide. J Mater Chem B 2025; 13:2504-2519. [PMID: 39834279 DOI: 10.1039/d4tb02125h] [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: 01/22/2025]
Abstract
Combination of immunotherapy and photothermal therapy (PTT) provides a promising therapeutic performance for tumors. However, it still faces negative feedback from suppressive factors such as adenosine. Herein, we developed a new nanodrug that can combine adenosine blockade and ferroptosis to promote the photoimmunotherapy of triple negative breast cancer (TNBC). The nanodrug, named CuS-PEG@Apt, was constructed via the modification of copper sulfide (CuS) nanoparticles with adenosine aptamer and PEG. CuS-PEG@Apt could be effectively enriched in the tumor site and locally generate a strong photothermal effect, directly ablating tumors and inducing immunogenic death (ICD). On the other hand, the aptamers could block the adenosine pathway to inhibit the immune suppression by adenosine, which further promoted the anti-tumor immunity. Moreover, the CuS nanoparticles could consume GSH and inhibit GPX4 to cause the ferroptosis of tumor cells. Collectively, CuS-PEG@Apt achieved potent efficacy of tumor suppression via the combination of PTT, immune activation and ferroptosis, representing an appealing platform for TNBC treatment.
Collapse
Affiliation(s)
- Xingyu Zhang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Chengyu Shi
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Qiao Liu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Yuting Zhong
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Lipeng Zhu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| | - Yuetao Zhao
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, 410012, China.
| |
Collapse
|
11
|
Wardell CM, Boardman DA, Levings MK. Harnessing the biology of regulatory T cells to treat disease. Nat Rev Drug Discov 2025; 24:93-111. [PMID: 39681737 DOI: 10.1038/s41573-024-01089-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2024] [Indexed: 12/18/2024]
Abstract
Regulatory T (Treg) cells are a suppressive subset of CD4+ T cells that maintain immune homeostasis and restrain inflammation. Three decades after their discovery, the promise of strategies to harness Treg cells for therapy has never been stronger. Multiple clinical trials seeking to enhance endogenous Treg cells or deliver them as a cell-based therapy have been performed and hint at signs of success, as well as to important limitations and unanswered questions. Strategies to deplete Treg cells in cancer are also in active clinical testing. Furthermore, multi-dimensional methods to interrogate the biology of Treg cells are leading to a refined understanding of Treg cell biology and new approaches to harness tissue-specific functions for therapy. A new generation of Treg cell clinical trials is now being fuelled by advances in nanomedicine and synthetic biology, seeking more precise ways to tailor Treg cell function. This Review will discuss recent advances in our understanding of human Treg cell biology, with a focus on mechanisms of action and strategies to assess outcomes of Treg cell-targeted therapies. It highlights results from recent clinical trials aiming to enhance or inhibit Treg cell activity in a variety of diseases, including allergy, transplantation, autoimmunity and cancer, and discusses ongoing strategies to refine these approaches.
Collapse
Affiliation(s)
- Christine M Wardell
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dominic A Boardman
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Megan K Levings
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada.
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
12
|
Kolbe C, Kauer J, Brinkmann B, Dreger P, Huber W, Müller-Tidow C, Dietrich S, Roider T. Blocking the CD39/CD73 pathway synergizes with anti-CD20 bispecific antibody in nodal B-cell lymphoma. J Immunother Cancer 2025; 13:e009245. [PMID: 39884778 PMCID: PMC11784132 DOI: 10.1136/jitc-2024-009245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 01/14/2025] [Indexed: 02/01/2025] Open
Abstract
Bispecific antibodies (BsAb) have emerged as a leading treatment modality in patients suffering from B-cell non-Hodgkin's lymphoma (B-NHL). However, treatment failure is common and may potentially be attributed to pre-existing or emerging T-cell exhaustion. CD39 catalyzes-together with CD73-the hydrolysis of immunogenic ATP into immunosuppressive adenosine and thus actively promotes an immunosuppressive micromilieu. Previously, we and others demonstrated that CD39+ T-cell subsets may have an adverse impact on the efficacy of T-cell-engaging immunotherapies. In this study, we applied an autologous ex vivo culture model of primary lymph node-derived T cells to investigate the potential of anti-CD39 or anti-CD73 blocking antibodies as T-cell enhancing combination partners of an anti-CD20 BsAb. Existing single-cell data of patient samples examined in this study were used to detect potential biomarkers predicting combination benefits. Combining anti-CD20 BsAb with anti-CD39 or anti-CD73 blocking antibodies induced synergistic effects on tumor cell killing, T-cell expansion and secretion of cytokines, including granzyme B, perforin, interleukin-10, interferon-γ, and tumor necrosis factor-α. We discovered that blockade of the CD39/CD73 pathway was particularly effective in patients with a high proportion of Programmed cell death protein 1 (PD-1)+ T-cell immunoglobulin and mucin-domain containing-3 (TIM3)+ exhausted T cells. Also, expression of CD39 in effector memory T cells indicated superior treatment benefit ex vivo. In summary, our study holds significant relevance as it introduces the combination of bispecific and anti-CD39 or anti-CD73 antibodies as a synergistic treatment approach in B-NHL, while also suggesting potential indicators to identify patients that might benefit from this treatment.
Collapse
Affiliation(s)
- Clara Kolbe
- Heidelberg University Hospital Department of Hematology Oncology and Rheumatology, Heidelberg, Germany
| | - Joseph Kauer
- Heidelberg University Hospital Department of Hematology Oncology and Rheumatology, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Heidelberg, Baden-Württemberg, Germany
| | - Berit Brinkmann
- Heidelberg University Hospital Department of Hematology Oncology and Rheumatology, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Heidelberg, Baden-Württemberg, Germany
| | - Peter Dreger
- Heidelberg University Hospital Department of Hematology Oncology and Rheumatology, Heidelberg, Germany
| | - Wolfgang Huber
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Heidelberg, Baden-Württemberg, Germany
- Genome Biology Unit, EMBL, Heidelberg, Baden-Württemberg, Germany
| | - Carsten Müller-Tidow
- Heidelberg University Hospital Department of Hematology Oncology and Rheumatology, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Heidelberg, Baden-Württemberg, Germany
| | - Sascha Dietrich
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Heidelberg, Baden-Württemberg, Germany
- University Hospital of Düsseldorf Department of Haematology Oncology and Clinical Immunology, Düsseldorf, Nordrhein-Westfalen, Germany
| | - Tobias Roider
- Heidelberg University Hospital Department of Hematology Oncology and Rheumatology, Heidelberg, Germany
- Molecular Medicine Partnership Unit (MMPU), Heidelberg, Heidelberg, Baden-Württemberg, Germany
- Medical Faculty Heidelberg, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
13
|
Yang X, Zhou Y, Zhou F, Bao L, Wang Z, Li Z, Ding F, Kuang H, Liu H, Tan S, Qiu X, Jing H, Liu S, Ma D. T Cell-Derived Apoptotic Extracellular Vesicles Ameliorate Bone Loss via CD39 and CD73-Mediated ATP Hydrolysis. Int J Nanomedicine 2025; 20:1083-1100. [PMID: 39895982 PMCID: PMC11784384 DOI: 10.2147/ijn.s491222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025] Open
Abstract
Background Osteoporosis is a major public health concern characterized by decreased bone density. Among various therapeutic strategies, apoptotic extracellular vesicles (ApoEVs) have emerged as promising agents in tissue regeneration. Specifically, T cell-derived ApoEVs have shown substantial potential in facilitating bone regeneration. However, it remains unclear whether ApoEVs can promote bone mass recovery through enzymatic activity mediated by membrane surface molecules. Therefore, this study aimed to investigate whether T cell-derived ApoEVs could promote bone mass recovery in osteoporosis mice and reveal the underlying mechanisms. Methods ApoEVs were isolated through sequential centrifugation, and their proteomic profiles were identified via mass spectrometry. Western blot and immunogold staining confirmed the enrichment of CD39 and CD73 on ApoEVs. The role of CD39 and CD73 in hydrolyzing adenosine triphosphate (ATP) to adenosine was evaluated by quantifying the levels of ATP and adenosine. Inhibitors of CD39 and CD73, and an A2BR antagonist were used to explore the molecular mechanism of ApoEVs in promoting bone regeneration. Results ApoEVs significantly reduced bone loss and promote the osteogenic differentiation of BMMSCs in ovariectomy (OVX) mice. We observed increased levels of extracellular ATP and a decrease in CD39 and CD73, key enzymes in ATP-to-adenosine conversion in bone marrow of OVX mice. We found that ApoEVs are enriched with CD39 and CD73 on their membranes, which enable the hydrolysis of extracellular ATP to adenosine both in vitro and in vivo. The adenosine generated by ApoEVs inhibits the inflammatory response and promotes osteogenesis through A2BR and downstream PKA signaling. Conclusion T cell-derived ApoEVs are enriched with CD39 and CD73, enabling them to hydrolyze extracellular ATP to adenosine, thereby promoting bone regeneration via A2BR and PKA signaling pathway. Our data underscore the substantive role of T cell-derived ApoEVs to treat osteoporosis, thus providing new ideas for the development of ApoEVs-based therapies in tissue regeneration.
Collapse
Affiliation(s)
- Xiaoshan Yang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, People’s Republic of China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Yang Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Fuxing Zhou
- Department of Gynecology and Obstetrics, Xijing Hospital, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Lili Bao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Zhengyan Wang
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, 250012, People’s Republic of China
| | - Zihan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Feng Ding
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Huijuan Kuang
- Department of Orthopaedics, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- State Key Laboratory for Manufacturing System Engineering, Xi’an Jiaotong University, Xi’an, 710054, People’s Republic of China
| | - Huan Liu
- Department of Otolaryngology Head and Neck Surgery, Peking University Third Hospital, Beijing, 100871, People’s Republic of China
| | - Shenglong Tan
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, People’s Republic of China
| | - Xinyuan Qiu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, People’s Republic of China
| | - Huan Jing
- Department of Endodontics, Guangdong Provincial High-level Clinical Key Specialty, Guangdong Province Engineering Research Center of Oral Disease Diagnosis and Treatment, Peking University Shenzhen Hospital, Shenzhen, 518036, People’s Republic of China
| | - Shiyu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Key Laboratory of Stomatology, Department of Oral Biology, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, Xi’an, 710032, People’s Republic of China
| | - Dandan Ma
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, People’s Republic of China
| |
Collapse
|
14
|
Ma M, Zhang Y, Pu K, Tang W. Nanomaterial-enabled metabolic reprogramming strategies for boosting antitumor immunity. Chem Soc Rev 2025; 54:653-714. [PMID: 39620588 DOI: 10.1039/d4cs00679h] [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: 01/21/2025]
Abstract
Immunotherapy has become a crucial strategy in cancer treatment, but its effectiveness is often constrained. Most cancer immunotherapies focus on stimulating T-cell-mediated immunity by driving the cancer-immunity cycle, which includes tumor antigen release, antigen presentation, T cell activation, infiltration, and tumor cell killing. However, metabolism reprogramming in the tumor microenvironment (TME) supports the viability of cancer cells and inhibits the function of immune cells within this cycle, presenting clinical challenges. The distinct metabolic needs of tumor cells and immune cells require precise and selective metabolic interventions to maximize therapeutic outcomes while minimizing adverse effects. Recent advances in nanotherapeutics offer a promising approach to target tumor metabolism reprogramming and enhance the cancer-immunity cycle through tailored metabolic modulation. In this review, we explore cutting-edge nanomaterial strategies for modulating tumor metabolism to improve therapeutic outcomes. We review the design principles of nanoplatforms for immunometabolic modulation, key metabolic pathways and their regulation, recent advances in targeting these pathways for the cancer-immunity cycle enhancement, and future prospects for next-generation metabolic nanomodulators in cancer immunotherapy. We expect that emerging immunometabolic modulatory nanotechnology will establish a new frontier in cancer immunotherapy in the near future.
Collapse
Affiliation(s)
- Muye Ma
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
| | - Yongliang Zhang
- Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 5 Science Dr 2, Singapore, 117545, Singapore
- Immunology Programme, Life Sciences Institute, National University of Singapore, 28 Medical Dr, Singapore, 117597, Singapore
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore.
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921, Singapore
| | - Wei Tang
- Department of Diagnostic Radiology, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr, Singapore, 117597, Singapore.
- Department of Pharmacy and Pharmaceutic Sciences, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| |
Collapse
|
15
|
Iser IC, Bertoni APS, Beckenkamp LR, Consolaro MEL, Maria-Engler SS, Wink MR. Adenosinergic Signalling in Cervical Cancer Microenvironment. Expert Rev Mol Med 2025; 27:e5. [PMID: 39762204 PMCID: PMC11707834 DOI: 10.1017/erm.2024.30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 08/20/2024] [Accepted: 08/30/2024] [Indexed: 01/11/2025]
Abstract
Despite the emergence of the first human papillomavirus vaccine, the incidence of cervical cancer is still responsible for more than 350,000 deaths yearly. Over the past decade, ecto-5'-nucleotidase (CD73/5'-NT) and extracellular adenosine (ADO) signalling has been the subject of many investigations to target cancer progression. In general, the adenosinergic axis has been linked to tumourigenic effects. However, CD73 can play contradictory effects, probably dependent on the tumour type, tumour microenvironment and tumour stage, thus being in some circumstances, inversely related to tumour progression. We herein reviewed the pathophysiological function of CD73 in cervical cancer and performed in silico analysis of the main components of the adenosinergic signalling in human tissues of cervical cancer compared to non-tumour cervix tissue. Our data showed that the NT5E gene, that encoded CD73, is hypermethylated, leading to a decreased CD73 expression in cervical cancer cells compared to normal cells. Consequently, the high availability of ADO cytoplasmatic/extracellular leads to its conversion to AMP by ADK, culminating in global hypermethylation. Therefore, epigenetic modulation may reveal a new role for CD73 in cervical cancer.
Collapse
Affiliation(s)
- Isabele Cristiana Iser
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| | - Ana Paula Santin Bertoni
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| | - Liziane Raquel Beckenkamp
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| | - Marcia Edilaine Lopes Consolaro
- Department of Clinical Analysis and Biomedicine, Division of Clinical Cytology, State University of Maringá, Maringá, PR, Brazil
| | - Silvya Stuchi Maria-Engler
- Department of Clinical Chemistry and Toxicology, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Marcia Rosângela Wink
- Department of Basics Health Sciences and Laboratory of Cell Biology, Federal University of Health Sciences of Porto Alegre, Porto Alegre, RS, Brazil
| |
Collapse
|
16
|
Zhou H, Bao G, Zhao J, Zhu X. Nuclear Molecular Imaging for Evaluating T Cell Exhaustion. Mol Pharm 2025; 22:103-112. [PMID: 39586059 DOI: 10.1021/acs.molpharmaceut.4c00970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
T cells are indispensable for the therapeutic efficacy of cancer immunotherapies, including immune checkpoint blockade. However, prolonged antigen exposure also drives T cells into exhaustion, which is characterized by upregulated inhibitory molecules, impaired effector functions, reduced cytotoxicity, altered metabolism, etc. Noninvasive monitoring of T cell exhaustion allows a timely identification of cancer patients that are most likely to benefit from immunotherapies. In this Review, we briefly explain the biological cascades underlying the modulation of inhibitory molecules, present a concise update on the nuclear molecular imaging tracers of T cell exhaustion, and then discuss the potential opportunities for future development.
Collapse
Affiliation(s)
- Huimin Zhou
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Guangfa Bao
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Department of Nuclear Medicine, The First People's Hospital of Yunnan Province, Kunming, Yunnan 650032, China
| | - Jun Zhao
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Department of Anatomy, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Xiaohua Zhu
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| |
Collapse
|
17
|
Fan G, Na J, Shen Z, Lin F, Zhong L. Heterogeneity of tumor-associated neutrophils in hepatocellular carcinoma. Mol Immunol 2025; 177:1-16. [PMID: 39642781 DOI: 10.1016/j.molimm.2024.11.009] [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: 09/19/2024] [Revised: 11/08/2024] [Accepted: 11/17/2024] [Indexed: 12/09/2024]
Abstract
Neutrophils are the most abundant cell type in human blood and play a crucial role in the immune system and development of tumors. This review begins with the generation and development of neutrophils, traces their release from the bone marrow into the bloodstream, and finally discusses their role in the hepatocellular carcinoma (HCC) microenvironment. It elaborates in detail the mechanisms by which tumor-associated neutrophils (TANs) exert antitumor or protumor effects under the influence of various mediators in the tumor microenvironment. Neutrophils can exert antitumor effects through direct cytotoxic action. However, they can also accelerate the formation and progression of HCC by being recruited and infiltrated, promoting tumor angiogenesis, and maintaining an immunosuppressive microenvironment. Therefore, based on the heterogeneity and plasticity of neutrophils in tumor development, this review summarizes the current immunotherapies targeting TANs, discusses potential opportunities and challenges, and provides new insights into exploring more promising strategies for treating HCC.
Collapse
Affiliation(s)
- Guixiang Fan
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, China; Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Zhen Shen
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, China; Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China
| | - Faquan Lin
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, China.
| | - Liping Zhong
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, China; Pharmaceutical College, Guangxi Medical University, Nanning, Guangxi 530021, China.
| |
Collapse
|
18
|
Xu JG, Chen S, He Y, Zhu X, Wang Y, Ye Z, Zhou JC, Wu X, Zhang L, Ren X, Jia H, Yu H, Wei X, Feng Y, Chen X, Cui X, Pan X, Wang S, Xia S, Shang H, Pu Y, Xu W, Li H, Chen Q, Chen Z, Wang M, Yan X, Shi H, Li M, Xia Y, Bellelli R, Dong S, He J, Huang J, Cai CL, Zhu X, Zhan Y, Wan L. An antibody cocktail targeting two different CD73 epitopes enhances enzyme inhibition and tumor control. Nat Commun 2024; 15:10872. [PMID: 39738003 PMCID: PMC11685497 DOI: 10.1038/s41467-024-55207-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
CD73, an ectoenzyme responsible for adenosine production, is often elevated in immuno-suppressive tumor environments. Inhibition of CD73 activity holds great promise as a therapeutic strategy for CD73-expressing cancers. In this study, we have developed a therapeutic anti-human CD73 antibody cocktail, HB0045. HB0045 is a 1:1 mixture of two humanized monoclonal IgG1 antibodies (mAbs), HB0038 and HB0039. The cocktail not only harnesses the advantages of its parental mAbs in enzyme inhibition but also shows a significantly greater capability of promoting T cell proliferation in vitro. Structural analyses show that HB0045 effectively locks the CD73 dimer in a "partially open" non-active conformation through a double lock mechanism. In various animal models of syngeneic and xenograft tumors, HB0045 inhibits tumor growth more potently than the single mAbs. Collectively, our findings provide functional and structural insights into the mechanism of a CD73-targeting antibody cocktail.
Collapse
MESH Headings
- 5'-Nucleotidase/immunology
- 5'-Nucleotidase/antagonists & inhibitors
- Animals
- Humans
- Mice
- GPI-Linked Proteins/immunology
- GPI-Linked Proteins/antagonists & inhibitors
- GPI-Linked Proteins/metabolism
- Epitopes/immunology
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Cell Proliferation/drug effects
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/chemistry
- Neoplasms/immunology
- Neoplasms/drug therapy
- Female
- T-Lymphocytes/immunology
- T-Lymphocytes/drug effects
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/chemistry
- Mice, Inbred BALB C
- Combined Antibody Therapeutics
Collapse
Affiliation(s)
- Jin-Gen Xu
- Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Scienes, Guangzhou, China
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Shi Chen
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yang He
- Institute of Molecular and Cell Biology, Singapore, Singapore
| | - Xi Zhu
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yanting Wang
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Zhifeng Ye
- Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Scienes, Guangzhou, China
| | - Jin Chuan Zhou
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Xuanhui Wu
- Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Scienes, Guangzhou, China
| | - Lei Zhang
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Xiaochen Ren
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Huifeng Jia
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Haijia Yu
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Xiaoyue Wei
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yujie Feng
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Xiaofang Chen
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Xiaopei Cui
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Xianfei Pan
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Shaojie Wang
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Simin Xia
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Hongjie Shang
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yueqing Pu
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Wei Xu
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Haidong Li
- College of Biology and Pharmacy, Yulin Normal University, Yulin, China
| | - Qian Chen
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Zeyu Chen
- Biortus Biosciences Co. Ltd, Jiangyin, China
| | - Manfu Wang
- Biortus Biosciences Co. Ltd, Jiangyin, China
| | | | - Hui Shi
- Biortus Biosciences Co. Ltd, Jiangyin, China
| | - Mingwei Li
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China
| | - Yisui Xia
- Medical School, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University, Shenzhen, China
| | - Roberto Bellelli
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Charterhouse Square, Barbican, London, UK
| | - Shunli Dong
- Huzhou Key Laboratory of Molecular Medicine, Huzhou Central Hospital, Huzhou, China
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou, China
| | - Jun He
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Jun Huang
- The MOE Key Laboratory of Biosystems Homeostasis & Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Chen-Leng Cai
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Xiangyang Zhu
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China.
| | - Yifan Zhan
- Drug Discovery, Shanghai Huaota Biopharmaceutical Co. Ltd., Shanghai, China.
| | - Li Wan
- Key Laboratory of Immune Response and Immunotherapy, Guangzhou Institutes of Biomedicine and Health (GIBH), Chinese Academy of Scienes, Guangzhou, China.
- Institute of Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China.
| |
Collapse
|
19
|
Baek S, Cui K. Targeting CD200 in Breast Cancer: Opportunities and Challenges in Immunotherapeutic Strategies. Int J Mol Sci 2024; 26:115. [PMID: 39795972 PMCID: PMC11719565 DOI: 10.3390/ijms26010115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
One of the key factors that contribute to tumor progression and resistance is the immunosuppressive microenvironment of the tumor. CD200 is a recently identified cell surface glycoprotein recognized as an important molecule in breast cancer for its versatile modulation of the immune response via its receptor, CD200R. The interaction between CD200 and CD200R suppresses the immune activities against tumor cells and allows them to be undetected and, in doing so, to escape from the destructive capability of the immune cells. Here, we review recent advances and future trends in CD200-targeted therapies for cancer treatments. We also discuss molecular pathways that include variable expressions across different cancer types and their importance in treatment options.
Collapse
Affiliation(s)
| | - Kui Cui
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
20
|
Liu D, Zhao J, Li L, Wang J, Wang C, Wu Y, Huang Y, Xing D, Chen W. CD73: agent development potential and its application in diabetes and atherosclerosis. Front Immunol 2024; 15:1515875. [PMID: 39735551 PMCID: PMC11672340 DOI: 10.3389/fimmu.2024.1515875] [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: 10/23/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
CD73, an important metabolic and immune escape-promoting gene, catalyzes the hydrolysis of adenosine monophosphate (AMP) to adenosine (ADO). AMP has anti-inflammatory and vascular relaxant properties, while ADO has a strong immunosuppressive effect, suggesting that CD73 has pro-inflammatory and immune escape effects. However, CD73 also decreased proinflammatory reaction, suggesting that CD73 has a positive side to the body. Indeed, CD73 plays a protective role in diabetes, while with age, CD73 changes from anti-atherosclerosis to pro-atherosclerosis. The upregulation of CD73 with agents, including AGT-5, Aire-overexpressing DCs, Aspirin, BAFFR-Fc, CD4+ peptide, ICAs, IL-2 therapies, SAgAs, sCD73, stem cells, RAD51 inhibitor, TLR9 inhibitor, and VD, decreased diabetes and atherosclerosis development. However, the downregulation of CD73 with agents, including benzothiadiazine derivatives and CD73 siRNA, reduced atherosclerosis. Notably, many CD73 agents were investigated in clinical trials. However, no agents were used to treat diabetes and atherosclerosis. Most agents were CD73 inhibitors. Only FP-1201, a CD73 agonist, was investigated in clinical trials but its further development was discontinued. In addition, many lncRNAs, circRNAs, and genes are located at the same chromosomal location as CD73. In particular, circNT5E promoted CD73 expression. circNT5E may be a promising target for agent development. This mini-review focuses on the current state of knowledge of CD73 in diabetes, atherosclerosis, and its potential role in agent development.
Collapse
Affiliation(s)
- Dan Liu
- Guangdong Provincial People’s Hospital, Zhuhai Hospital (Jinwan Central Hospital of Zhuhai), Zhuhai, Guangdong, China
| | - Jingjing Zhao
- Sleep Medicine Center, Huai’an No.3 People’s Hospital, Huaian Second Clinical College of Xuzhou Medical University, Huaian, China
| | - Ling Li
- Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Jie Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Chao Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Yudong Wu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, China
| | - Yucun Huang
- Guangdong Provincial People’s Hospital, Zhuhai Hospital (Jinwan Central Hospital of Zhuhai), Zhuhai, Guangdong, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Wujun Chen
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong, China
| |
Collapse
|
21
|
Rajkumari S, Singh J, Agrawal U, Agrawal S. Myeloid-derived suppressor cells in cancer: Current knowledge and future perspectives. Int Immunopharmacol 2024; 142:112949. [PMID: 39236460 DOI: 10.1016/j.intimp.2024.112949] [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: 05/30/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024]
Abstract
MDSCs (myeloid-derived suppressor cells) are crucial for immune system evasion in cancer. They accumulate in peripheral blood and tumor microenvironment, suppressing immune cells like T-cells, natural killer cells and dendritic cells. They promote tumor angiogenesis and metastasis by secreting cytokines and growth factors and contribute to a tumor-promoting environment. The accumulation of MDSCs in cancer patients has been linked to poor prognosis and resistance to various cancer therapies. Targeting MDSCs and their immunosuppressive mechanisms may improve treatment outcomes and enhance immune surveillance by developing drugs that inhibit MDSC function, by preventing their accumulation and by disrupting the tumor-promoting environment. This review presents a detailed overview of the MDSC research in cancer with regulation of their development and function. The relevance of MDSC as a prognostic and predictive biomarker in different types of cancers, along with recent advancements on the therapeutic approaches to target MDSCs are discussed in detail.
Collapse
Affiliation(s)
- Sunanda Rajkumari
- ICMR National Institute of Medical Statistics, Ansari Nagar, New Delhi 110029, India
| | - Jaspreet Singh
- ICMR National Institute of Pathology, Safdarjung Hospital Campus, Ansari Nagar, New Delhi 110029, India
| | - Usha Agrawal
- Asian Institute of Public Health University (AIPH) University, 1001 Haridamada, Jatani, Near IIT Bhubaneswar, Bhubaneswar 751002, India
| | - Sandeep Agrawal
- Discovery Research Division, Indian Council of Medical Research, Ansari Nagar, New Delhi 110029, India.
| |
Collapse
|
22
|
Zhao Y, Sun J, Xu XL, Su J, Du YZ. The potential of nanosystems in disrupting adenosine signaling pathways for tumor immunotherapy. Expert Opin Drug Deliv 2024; 21:1755-1770. [PMID: 39434697 DOI: 10.1080/17425247.2024.2417687] [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: 08/02/2024] [Revised: 09/04/2024] [Accepted: 10/14/2024] [Indexed: 10/23/2024]
Abstract
INTRODUCTION Adenosine (ADO) is a naturally occurring nucleoside primarily synthesized through the hydrolysis of extracellular adenosine triphosphate. Within the tumor microenvironment, ADO levels substantially increase, resulting in suppressed immune responses. AREAS COVERED Nanosystems offer a promising approach for precise drug delivery to tumor lesions. In this review, we provide an overview of the current research progress in the development of nanosystems that modulate adenosine signaling for tumor immunotherapy. These nanosystems are designed to target adenosine-hydrolyzing proteins, increase adenosine decomposition, and antagonize adenosine receptors. EXPERT OPINION Based on the literature review, adenosine has great potential in tumor immunotherapy, and nano-drug delivery system has great application prospects in targeted cancer therapy in the near future due to its superior characteristics.
Collapse
Affiliation(s)
- Yutong Zhao
- College of Pharmacy, Jiamusi University, Jiamusi, PR China
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| | - Jingqi Sun
- College of Pharmacy, Jiamusi University, Jiamusi, PR China
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Xiao-Ling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, PR China
| | - Jin Su
- College of Pharmacy, Jiamusi University, Jiamusi, PR China
| | - Yong-Zhong Du
- College of Pharmacy, Jiamusi University, Jiamusi, PR China
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, PR China
| |
Collapse
|
23
|
Zhang Y, Wang W, Liu Q, Jiang J, Zhao P, Huang C, Li Y, Fu Y. CD19 +CD73 + B cells infiltration indicates poor prognosis and unfavorable responses to immunotherapy in gastric cancer. Int Immunopharmacol 2024; 141:113002. [PMID: 39213870 DOI: 10.1016/j.intimp.2024.113002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVES Cluster of Differentiation 73 (CD73) is expressed on immune cells and plays a significant role in tumor inhibition by suppressing antitumor immunity. The objectives of this study were to explore the expression and functional mechanisms of CD73 on B cells in patients with gastric cancer (GC). METHODS The prognostic significance of CD19+CD73+ B cells was evaluated in 390 GC patients through dual immunohistochemistry staining. Flow cytometry was employed to analyze the phenotype of the CD19 subpopulation using fresh tumor and non-tumor tissue samples from 8 GC patients. A bioinformatics analysis of CD19+CD73+ B cells was also performed within the scRNA-seq cohort, and the CD19+ B cell subtype was assessed using multiple immunofluorescence staining. RESULTS The infiltration of CD19+CD73+ B cells was observed to be elevated in gastric cancer (GC) tissue compared to normal tissues. A strong correlation was observed between high CD19+CD73+ B cell infiltration, poor overall survival, and diminished responsiveness to neoadjuvant immunotherapy in GC. These cells emerged as a novel subset of regulatory B cells (Bregs) linked to adenosine metabolism and the exhaustion of CD8+ T cells. The CD19+CD73+ B cells also correlated with the production of immunosuppressive cytokines IL-10 and TGFB1. Further analysis indicated an association between CD19+CD73+ B cells and advanced-stage GC. CONCLUSIONS The presence of CD19+CD73+ B cells in GC may serve as a prognostic indicator for clinical outcomes and a predictive marker for poor responsiveness to neoadjuvant immunotherapy. The correlation between the presence of CD19+CD73+ B cells and CD8+ T cell exhaustion, along with immunosuppression, highlights the tumor-promoting function of these cells.
Collapse
Affiliation(s)
- Yawei Zhang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wendong Wang
- Department of Breast Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qi Liu
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jianwu Jiang
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Zhao
- Department of Surgery, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Changjun Huang
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yingying Li
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Fu
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
24
|
Chen S, Zhu H, Jounaidi Y. Comprehensive snapshots of natural killer cells functions, signaling, molecular mechanisms and clinical utilization. Signal Transduct Target Ther 2024; 9:302. [PMID: 39511139 PMCID: PMC11544004 DOI: 10.1038/s41392-024-02005-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 11/15/2024] Open
Abstract
Natural killer (NK) cells, initially identified for their rapid virus-infected and leukemia cell killing and tumor destruction, are pivotal in immunity. They exhibit multifaceted roles in cancer, viral infections, autoimmunity, pregnancy, wound healing, and more. Derived from a common lymphoid progenitor, they lack CD3, B-cell, or T-cell receptors but wield high cytotoxicity via perforin and granzymes. NK cells orchestrate immune responses, secreting inflammatory IFNγ or immunosuppressive TGFβ and IL-10. CD56dim and CD56bright NK cells execute cytotoxicity, while CD56bright cells also regulate immunity. However, beyond the CD56 dichotomy, detailed phenotypic diversity reveals many functional subsets that may not be optimal for cancer immunotherapy. In this review, we provide comprehensive and detailed snapshots of NK cells' functions and states of activation and inhibitions in cancer, autoimmunity, angiogenesis, wound healing, pregnancy and fertility, aging, and senescence mediated by complex signaling and ligand-receptor interactions, including the impact of the environment. As the use of engineered NK cells for cancer immunotherapy accelerates, often in the footsteps of T-cell-derived engineering, we examine the interactions of NK cells with other immune effectors and relevant signaling and the limitations in the tumor microenvironment, intending to understand how to enhance their cytolytic activities specifically for cancer immunotherapy.
Collapse
Affiliation(s)
- Sumei Chen
- Department of Radiation Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China.
| | - Haitao Zhu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Youssef Jounaidi
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
25
|
Qu T, Sun Y, Zhao J, Liu N, Yang J, Lyu D, Huang W, Zhan W, Li T, Yao Z, Yan R, Zhang H, Hong H, Shi L, Meng X, Yin B. Scoulerine: A natural isoquinoline alkaloid targeting SLC6A3 to treat RCC. Biomed Pharmacother 2024; 180:117524. [PMID: 39395255 DOI: 10.1016/j.biopha.2024.117524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024] Open
Abstract
Scoulerine, an isoquinoline alkaloid derived from the corydalis plant, exhibits diverse therapeutic properties against tumors, Alzheimer's disease, and inflammation. This research delves into the pharmacological impact and underlying mechanism of scoulerine on renal cell carcinoma (RCC). Our findings suggest that Scoulerine displays promise as a potential therapeutic agent for RCC, demonstrating notable inhibitory effects in both in vivo and in vitro models. In addition, scoulerine inhibited the viability of 769-P and 786-O cell lines in a time-dependent and dose-dependent manner, and promoted the level of apoptosis associated with B-cell lymphoma-2 associated X protein (Bax). Moreover, the administration of scoulerine resulted in a significant suppression of the mitogen activated protein kinase (MAPK) signaling pathway. Subsequently, utilizing bioinformatics and spatial transcriptomic databases, we identified solute carrier family 6 Member 3 (SLC6A3) as the most promising target of scoulerine. Through experimental validation, we confirmed the functional and therapeutic relevance of SLC6A3 in scoulerine-mediated treatment of RCC. The results of our study indicate a significant affinity between scoulerine and SLC6A3, with competitive inhibition of this interaction leading to a reduction in the inhibitory impact of scoulerine on RCC cell viability. In conclusion, our findings suggest that scoulerine may induce apoptosis in RCC by targeting SLC6A3 and inhibiting the activation of the MAPK signaling pathway, thereby positioning it as a promising natural compound for potential future RCC treatment.
Collapse
Affiliation(s)
- Tianrui Qu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Yu Sun
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Jingying Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Nanqi Liu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China
| | - Jianli Yang
- Department of Laboratory Animals, China Medical University, Shenyang, Liaoning 110122, China
| | - Dantong Lyu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China
| | - Wenjie Huang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China
| | - Weizhen Zhan
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China
| | - Tao Li
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China
| | - Zichuan Yao
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China
| | - Rongbo Yan
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110000, China
| | - Haiyan Zhang
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Hong Hong
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Liye Shi
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Xin Meng
- Department of Biochemistry and Molecular Biology, School of Life Sciences, China Medical University, Shenyang, Liaoning 110122, China.
| | - Bo Yin
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, China.
| |
Collapse
|
26
|
Iqbal J, Bano S, Khan IA, Sévigny J, Huang Q. Ectonucleotidase inhibitors: an updated patent review (2017-2023). Expert Opin Ther Pat 2024; 34:1167-1176. [PMID: 39460640 DOI: 10.1080/13543776.2024.2423023] [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: 04/23/2024] [Revised: 09/09/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
INTRODUCTION The main enzymes that hydrolyzes nucleotides at the cell surface are nucleoside triphosphate diphosphohydrolases (NTPDases), ecto-nucleotide pyrophosphatases/phosphodiesterases (ENPPs), alkaline phosphatases (APs) and ecto-5'- nucleotidase (e5'NT, CD73) and by regulating the concentration of nucleotides at the cell surface, these enzymes have the potential to affect various conditions such as fibrosis, cancer metastasis, pruritus, inflammation, and autoimmune diseases. Thus, they represent a prospective therapeutic target. AREA COVERED A number of molecules, including nucleoside/nucleotide and non-nucleoside analogues, and bicyclic compounds, have shown strong potential as ectonucleotidase inhibitors. This review covers the chemistry and clinical uses of ectonucleotidase inhibitors patented between 2017 and 2023. EXPERT OPINION By binding to their specific P1 and P2 receptors at the cell surface, nucleosides and nucleotides regulate a number of pathophysiological events such as inflammation, fibrosis, cancer, and autoimmune diseases. Interestingly, these nucleotides can be hydrolyzed to nucleosides by several cell surface enzymes called ectonucleotidases. The development of small molecules that modulate ectonucleotidase activity is, therefore, of therapeutic value. This review provides valuable insights into recent advancements, including combination therapy and enhanced selectivity, which are poised to shape the future of ectonucleotidase inhibition through a comprehensive analysis of patents.
Collapse
Affiliation(s)
- Jamshed Iqbal
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Pakistan
| | - Sehrish Bano
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Pakistan
| | - Imtiaz Ali Khan
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Pakistan
| | - Jean Sévigny
- Département de microbiologie-infectiologie et d'immunologie, Faculté de Médecine, Université Laval, Quebec city, QC, Canada
- Centre de recherche du CHU de Québec - Université Laval, Quebec city, QC, Canada
| | - Qing Huang
- Institute of Intelligent Machines, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| |
Collapse
|
27
|
Qi Y, Zhang L, Liu Y, Li Y, Liu Y, Zhang Z. Targeted modulation of myeloid-derived suppressor cells in the tumor microenvironment: Implications for cancer therapy. Biomed Pharmacother 2024; 180:117590. [PMID: 39423752 DOI: 10.1016/j.biopha.2024.117590] [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: 08/05/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) represent a heterogeneous population of immature myeloid cells originating from the bone marrow, known for their potent immunosuppressive functions that contribute to tumor immune evasion and progression. This paper provides a comprehensive analysis of the multifaceted interactions between MDSCs and tumors, exploring their distinct phenotypes and immunosuppressive mechanisms. Key roles of MDSCs in tumor biology are discussed, including their involvement in the formation of the pre-metastatic niche, facilitation of angiogenesis, enhancement of vascular permeability, suppression of tumor cell apoptosis, and promotion of resistance to cancer therapies. Additionally, the review highlights recent advances in the development of MDSC-targeting therapies, with a focus on their potential to enhance anti-tumor immunity. The therapeutic potential of Traditional Chinese Medicine (TCM) in modulating MDSC quantity and function is also explored, suggesting a novel approach to cancer treatment by integrating traditional and modern therapeutic strategies.
Collapse
Affiliation(s)
- Yafeng Qi
- Clinical School of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Liying Zhang
- School of Integrative Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Yeyuan Liu
- Clinical School of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Yangyang Li
- Clinical School of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Yongqi Liu
- School of Basic Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Zhiming Zhang
- Department of Oncology, Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, Gansu 730000, China.
| |
Collapse
|
28
|
Wu M, Mao L, Zhai X, Liu J, Wang J, Li L, Duan J, Wang J, Lin S, Li J, Yu S. Microenvironmental alkalization promotes the therapeutic effects of MSLN-CAR-T cells. J Immunother Cancer 2024; 12:e009510. [PMID: 39433427 PMCID: PMC11499857 DOI: 10.1136/jitc-2024-009510] [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] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by high invasion, prone metastasis, frequent recurrence and poor prognosis. Unfortunately, the curative effects of current clinical therapies, including surgery, radiotherapy, chemotherapy and immunotherapy, are still limited in patients with TNBC. In this study, we showed that the heterogeneous expression at the protein level and subcellular location of mesothelin (MSLN), a potential target for chimeric antigen receptor-T (CAR-T) cell therapy in TNBC, which is caused by acidification of the tumor microenvironment, may be the main obstacle to therapeutic efficacy. Alkalization culture or sodium bicarbonate administration significantly promoted the membrane expression of MSLN and enhanced the killing efficiency of MSLN-CAR-T cells both in vitro and in vivo, and the same results were also obtained in other cancers with high MSLN expression, such as pancreatic and ovarian cancers. Moreover, mechanistic exploration revealed that the attenuation of autophagy-lysosome function caused by microenvironmental alkalization inhibited the degradation of MSLN. Hence, alkalization of the microenvironment improves the consistency and high expression of the target antigen MSLN and constitutes a routine method for treating diverse solid cancers via MSLN-CAR-T cells.
Collapse
Affiliation(s)
- Min Wu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Jin-feng Laboratory, Chongqing, Chongqing, China
| | - Ling Mao
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Xuejia Zhai
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Deaprtment of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Jie Liu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Junhan Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Langhong Li
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Jiangjie Duan
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Jin-feng Laboratory, Chongqing, Chongqing, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Jin-feng Laboratory, Chongqing, Chongqing, China
| | - Shuang Lin
- International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Jianjun Li
- Deaprtment of Oncology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
| | - Shicang Yu
- Department of Stem Cell and Regenerative Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, Chongqing, China
- Jin-feng Laboratory, Chongqing, Chongqing, China
| |
Collapse
|
29
|
Anderson AE, Parashar K, Jin K, Clor J, Stagnaro CE, Vani U, Singh J, Chen A, Guan Y, Talukdar P, Sathishkumar P, Juat DJ, Singh H, Kushwaha R, Zhao X, Kaplan A, Seitz L, Walters MJ, Fernandez-Salas E, Walker NP, Bowman CE. Characterization of AB598, a CD39 Enzymatic Inhibitory Antibody for the Treatment of Solid Tumors. Mol Cancer Ther 2024; 23:1471-1482. [PMID: 38797955 PMCID: PMC11443198 DOI: 10.1158/1535-7163.mct-23-0865] [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: 12/06/2023] [Revised: 01/31/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
AB598 is a CD39 inhibitory antibody being pursued for the treatment of solid tumors in combination with chemotherapy and immunotherapy. CD39 metabolizes extracellular adenosine triphosphate (eATP), an alarmin capable of promoting antitumor immune responses, into adenosine, an immuno-inhibitory metabolite. By inhibiting CD39, the consumption of eATP is reduced, resulting in a proinflammatory milieu in which eATP can activate myeloid cells to promote antitumor immunity. The preclinical characterization of AB598 provides a mechanistic rationale for combining AB598 with chemotherapy in the clinic. Chemotherapy can induce ATP release from tumor cells and, when preserved by AB598, both chemotherapy-induced eATP and exogenously added ATP promote the function of monocyte-derived dendritic cells via P2Y11 signaling. Inhibition of CD39 in the presence of ATP can promote inflammasome activation in in vitro-derived macrophages, an effect mediated by P2X7. In a MOLP8 murine xenograft model, AB598 results in full inhibition of intratumoral CD39 enzymatic activity, an increase in intratumoral ATP, a decrease of extracellular CD39 on tumor cells, and ultimately, control of tumor growth. In cynomolgus monkeys, systemic dosing of AB598 results in effective enzymatic inhibition in tissues, full peripheral and tissue target engagement, and a reduction in cell surface CD39 both in tissues and in the periphery. Taken together, these data support a promising therapeutic strategy of harnessing the eATP generated by standard-of-care chemotherapies to prime the tumor microenvironment for a productive antitumor immune response.
Collapse
Affiliation(s)
| | | | - Ke Jin
- Arcus Biosciences, Inc., Hayward, California.
| | - Julie Clor
- Arcus Biosciences, Inc., Hayward, California.
| | | | - Urvi Vani
- Arcus Biosciences, Inc., Hayward, California.
| | | | - Ada Chen
- Arcus Biosciences, Inc., Hayward, California.
| | - Yihong Guan
- Arcus Biosciences, Inc., Hayward, California.
| | | | | | | | - Hema Singh
- Arcus Biosciences, Inc., Hayward, California.
| | | | | | | | - Lisa Seitz
- Arcus Biosciences, Inc., Hayward, California.
| | | | | | | | | |
Collapse
|
30
|
Sposito M, Eccher S, Scaglione I, Avancini A, Rossi A, Pilotto S, Belluomini L. The frontier of neoadjuvant therapy in non-small cell lung cancer beyond PD-(L)1 agents. Expert Opin Biol Ther 2024; 24:1025-1037. [PMID: 39311630 DOI: 10.1080/14712598.2024.2408292] [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: 06/18/2024] [Accepted: 09/20/2024] [Indexed: 09/26/2024]
Abstract
INTRODUCTION While surgical resection is the cornerstone of treatment for resectable lung cancer, neoadjuvant/adjuvant chemotherapy has shown limited improvement in survival rates over the past decades. With the success of immune checkpoint inhibitors (ICIs) in advanced NSCLC, there is growing interest in their application in earlier stages of the disease. Recent approvals for neoadjuvant/adjuvant ICIs in stage II-IIIA NSCLC highlight this shift in treatment paradigms. AREAS COVERED In this review, we aim to explore available data regarding alternative agents beyond the PD-(L)1 inhibitors, such as monoclonal antibodies against CTLA4, LAG3, TIGIT, antiangiogenic drugs, and novel therapies (antibody drug conjugates, bispecific antibodies) in neoadjuvant/perioperative regimens. EXPERT OPINION Novel agents and combinations (with or without ICI or/and chemotherapy), guided by molecular profiling and immune phenotyping, showed promise in improving surgical and survival outcomes. Crucial is, also in early setting, to identifying biomarkers predictive of treatment efficacy in order to personalize neoadjuvant/perioperative treatment strategies.
Collapse
Affiliation(s)
- Marco Sposito
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and Verona University Hospital Trust, Verona, Italy
| | - Serena Eccher
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and Verona University Hospital Trust, Verona, Italy
| | - Ilaria Scaglione
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and Verona University Hospital Trust, Verona, Italy
| | - Alice Avancini
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and Verona University Hospital Trust, Verona, Italy
| | - Antonio Rossi
- Oncology Centre of Excellence, Therapeutic Science & Strategy Unit, Milan, Italy
| | - Sara Pilotto
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and Verona University Hospital Trust, Verona, Italy
| | - Lorenzo Belluomini
- Section of Innovation Biomedicine - Oncology Area, Department of Engineering for Innovation Medicine (DIMI), University of Verona and Verona University Hospital Trust, Verona, Italy
| |
Collapse
|
31
|
Sun Y, Yinwang E, Wang S, Wang Z, Wang F, Xue Y, Zhang W, Zhao S, Mou H, Chen S, Jin L, Li B, Ye Z. Phenotypic and spatial heterogeneity of CD8 + tumour infiltrating lymphocytes. Mol Cancer 2024; 23:193. [PMID: 39251981 PMCID: PMC11382426 DOI: 10.1186/s12943-024-02104-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/30/2024] [Indexed: 09/11/2024] Open
Abstract
CD8+ T cells are the workhorses executing adaptive anti-tumour response, and targets of various cancer immunotherapies. Latest advances have unearthed the sheer heterogeneity of CD8+ tumour infiltrating lymphocytes, and made it increasingly clear that the bulk of the endogenous and therapeutically induced tumour-suppressive momentum hinges on a particular selection of CD8+ T cells with advantageous attributes, namely the memory and stem-like exhausted subsets. A scrutiny of the contemporary perception of CD8+ T cells in cancer and the subgroups of interest along with the factors arbitrating their infiltration contextures, presented herein, may serve as the groundwork for future endeavours to probe further into the regulatory networks underlying their differentiation and migration, and optimise T cell-based immunotherapies accordingly.
Collapse
Affiliation(s)
- Yikan Sun
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Eloy Yinwang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Shengdong Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Zenan Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Fangqian Wang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Yucheng Xue
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Wenkan Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Shenzhi Zhao
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Haochen Mou
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Shixin Chen
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Lingxiao Jin
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China
| | - Binghao Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China.
| | - Zhaoming Ye
- Department of Orthopedic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, Zhejiang, China.
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, Zhejiang, China.
- Department of Orthopedics, Musculoskeletal Tumor Center, The Second Affiliated Hospital of Zhejiang, University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
32
|
Fathi M, Zarei A, Moghimi A, Jalali P, Salehi Z, Gholamin S, Jadidi-Niaragh F. Combined cancer immunotherapy based on targeting adenosine pathway and PD-1/PDL-1 axis. Expert Opin Ther Targets 2024; 28:757-777. [PMID: 39305018 DOI: 10.1080/14728222.2024.2405090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Accepted: 09/12/2024] [Indexed: 10/02/2024]
Abstract
INTRODUCTION Cancer immunotherapy has revolutionized the field of oncology, offering new hope to patients with advanced malignancies. Tumor-induced immunosuppression limits the effectiveness of current immunotherapeutic strategies, such as PD-1/PDL-1 checkpoint inhibitors. Adenosine, a purine nucleoside molecule, is crucial to this immunosuppression because it stops T cells from activating and helps regulatory T cells grow. Targeting the adenosine pathway and blocking PD-1/PDL-1 is a potential way to boost the immune system's response to tumors. AREAS COVERED This review discusses the current understanding of the adenosine pathway in tumor immunology and the preclinical and clinical data supporting the combination of adenosine pathway inhibitors with PD-1/PDL-1 blockade. We also discuss the challenges and future directions for developing combination immunotherapy targeting the adenosine pathway and the PD-1/PDL-1 axis for cancer treatment. EXPERT OPINION The fact that the adenosine signaling pathway controls many immune system processes suggests that it has a wide range of therapeutic uses. Within the next five years, there will be tremendous progress in this area, and the standard of care for treating malignant tumors will have switched from point-to-point therapy to the integration of immunological networks comprised of multiple signaling pathways, like the adenosine axis.
Collapse
Affiliation(s)
- Mehrdad Fathi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Asieh Zarei
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ata Moghimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Sharareh Gholamin
- City of Hope Beckman Research Institute and Medical Center, Duarte, CA, USA
- City of Hope Department of Radiation Oncology, Duarte, CA, USA
| | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
33
|
Shukla S, Dalai P, Agrawal-Rajput R. Metabolic crosstalk: Extracellular ATP and the tumor microenvironment in cancer progression and therapy. Cell Signal 2024; 121:111281. [PMID: 38945420 DOI: 10.1016/j.cellsig.2024.111281] [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: 04/28/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Adenosine 5'-triphosphate (ATP) is a vital element in energy information. It plays a critical role in transmitting signals inside the body, which is necessary for controlling the life activities of all cells, including tumor cells [1]. Its significance extends from intracellular signaling pathways to tumor regression. Purinergic signaling, a form of extracellular paracrine signaling, relies on purine nucleotides. Extracellular ectonucleotidases convert these purine nucleotides to their respective di and mono-phosphate nucleoside forms, contributing significantly to immune biology, cancer biology, and inflammation studies. ATP functions as a mighty damage-linked molecular pattern when released outside the cell, accumulating in inflammatory areas. In the tumor microenvironment (TME), purinergic receptors such as ATP-gated ion channels P2X1-5 and G protein-coupled receptors (GPCR) (P2Y) interact with ATP and other nucleotides, influencing diverse immune cell activities. CD39 and CD73-mediated extracellular ATP degradation contributes to immunosuppression by diminishing ATP-dependent activation and generating adenosine (ADO), potentially hindering antitumor immunity and promoting tumor development. Unraveling the complexities of extracellular ATP (e-ATP) and ADO effects on the TME poses challenges in identifying optimal treatment targets, yet ongoing investigations aim to devise strategies combating e-ATP/ADO-induced immunosuppression, ultimately enhancing anti-tumor immunity. This review explores e-ATP metabolism, its purinergic signaling, and therapeutic strategies targeting associated receptors and enzymes.
Collapse
Affiliation(s)
- Sourav Shukla
- Immunology Lab, Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar 382426, Gujarat, India
| | - Parameswar Dalai
- Immunology Lab, Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar 382426, Gujarat, India
| | - Reena Agrawal-Rajput
- Immunology Lab, Biotechnology and Bioengineering, Indian Institute of Advanced Research, Gandhinagar 382426, Gujarat, India.
| |
Collapse
|
34
|
Huang T, Ren X, Tang X, Wang Y, Ji R, Guo Q, Ma Q, Zheng Y, Hu Z, Zhou Y. Current perspectives and trends of CD39-CD73-eAdo/A2aR research in tumor microenvironment: a bibliometric analysis. Front Immunol 2024; 15:1427380. [PMID: 39188712 PMCID: PMC11345151 DOI: 10.3389/fimmu.2024.1427380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/17/2024] [Indexed: 08/28/2024] Open
Abstract
Background and objective Extracellular adenosine (eAdo) bridges tumor metabolism and immune regulation. CD39-CD73-eAdo/A2aR axis regulates tumor microenvironment (TME) and immunotherapy response. In the era of immunotherapy, exploring the impact of the CD39-CD73-eAdo/A2aR axis on TME and developing targeted therapeutic drugs to enhance the efficacy of immunotherapy are the current research hotspots. This study summarizes and explores the research trends and hotspots of the adenosine axis in the field of TME to provide ideas for further in-depth research. Methods Literature information was obtained from the Web of Science core collection database. The VOS viewer and the bibliometric tool based on R were used to quantify and identify cooperation information and individual influence by analyzing the detailed information of the global annual publication volume, country/region and institution distribution, article authors and co-cited authors, and journal distribution of these articles. At the same time, the distribution of author keywords and the co-occurrence of author keywords, highly cited articles, and highly co-cited references of CD39-CD73-eAdo/A2aR in the field of TME were analyzed to determine research hotspots and trends. Result 1,721 articles published in the past ten years were included in this study. Through bibliometric analysis, we found that (1) 69 countries and regions explored the effect of the CD39-CD73-eAdo/A2aR on TME, and the research was generally on the rise. Researchers in the United States dominated research in this area, with the highest total citation rate. China had the most significant number of publications. (2) Harvard University has published the most articles in this field. (3) 12,065 authors contributed to the publication of papers in this field, of which 23 published at least eight papers. STAGG J had significant academic influence, with 24 published articles and 2,776 citations. Co-cited authors can be clustered into three categories. Stagg J, Allard B, Ohta A, and Antonioli, L occupied a central position in the network. (4) 579 scholarly journals have published articles in this field. The journal FRONTIERS IN IMMUNOLOGY published the most significant number of papers, with 97 articles and a total of 2,317 citations, and the number of publications increased year by year. (5) "The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets" was the most frequently local cited article (163 times). The "A2A adenosine receptor protects tumors from antitumor T cells" was the most co-cited reference (224 times). (6) Through the analysis of author keywords, we found that the relationship between adenosine and immunotherapy was a core concept for many researchers in this field. Breast cancer, melanoma, colorectal cancer, ovarian cancer, glioblastoma, pancreatic cancer, hepatocellular carcinoma, and lung cancer were the most frequent cancer types in adenosine-related tumor studies. Immunotherapy, immunosuppression, immune checkpoint, and immune checkpoint inhibitors were the hot keywords in the research, reflecting the importance of the adenosine metabolic pathway in tumor immunotherapy. The keywords such as Immunogenic cell death, T cells, Sting, regulatory T cells, innate immunity, and immune infiltration demonstrated the pathways by which adenosine affected the TME. The famous author keywords in recent years have been immunotherapy, immunogenic cell death, inflammation, lung cancer, and gastric cancer. Conclusion The effect of CD39-CD73-eAdo/A2aR on the infiltration and function of various immune cells in TME, tumor immunotherapy response, and patient prognosis has attracted the attention of researchers from many countries/regions. American scholars still dominate the research in this field, but Chinese scholars produce the most research results. The journal FRONTIERS IN IMMUNOLOGY has published the wealthiest research in the field. Stagg J was a highly influential researcher in this field. Further exploration of targeted inhibition of CD39-CD73-eAdo/A2aR alone or in combination with other immunotherapy, radiotherapy, and chemotherapy in treating various cancer types and developing effective clinical therapeutic drugs are continuous research hotspots in this field.
Collapse
Affiliation(s)
- Tian Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiangqing Ren
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaolong Tang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- The Second Department of Gastrointestinal Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Rui Ji
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Qinghong Guo
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Qian Ma
- The First Department of Geriatrics, Xianyang First People’s Hospital, Xianyang, China
| | - Ya Zheng
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zenan Hu
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yongning Zhou
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
35
|
Xu S, Ma Y, Jiang X, Wang Q, Ma W. CD39 transforming cancer therapy by modulating tumor microenvironment. Cancer Lett 2024; 597:217072. [PMID: 38885807 DOI: 10.1016/j.canlet.2024.217072] [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: 05/03/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/20/2024]
Abstract
CD39 is a pivotal enzyme in cancer, regulating immune response and tumor progression via extracellular ATP and adenosine in the tumor microenvironment (TME). Beyond its established immunoregulatory function, CD39 influences cancer cell angiogenesis and metabolism, opening new frontiers for therapeutic interventions. Current research faces gaps in understanding CD39's full impact across cancer types, with ongoing debates about its potential beyond modulating immune evasion. This review distills CD39's multifaceted roles, examining its dual actions and implications for cancer prognosis and treatment. We analyze the latest therapeutic strategies, highlighting the need for an integrated approach that combines molecular insights with TME dynamics to innovate cancer care. This synthesis underscores CD39's integral role, charting a course for precision oncology that seeks to unravel controversies and harness CD39's therapeutic promise for improved cancer outcomes.
Collapse
Affiliation(s)
- Suling Xu
- Department of Dermatology, The First Affiliated Hospital of Ningbo University School of Medicine, Ningbo, Zhejiang, 315020, China.
| | - Yuhan Ma
- Department of Dermatology, The First Affiliated Hospital of Ningbo University School of Medicine, Ningbo, Zhejiang, 315020, China.
| | - Xinyu Jiang
- Department of Dermatology, The First Affiliated Hospital of Ningbo University School of Medicine, Ningbo, Zhejiang, 315020, China.
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
| | - Wenxue Ma
- Department of Medicine, Sanford Stem Cell Institute, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
36
|
Botticelli A, Cirillo A, d'Amati G, Di Gioia C, Corsi A, Della Rocca C, Santini D, Carletti R, Pisano A, Polimeni A, De Vincentiis M, Valentini V, di Cristofano C, Romeo U, Cerbelli E, Messineo D, De Felice F, Leopizzi M, Cerbelli B. The role of CD73 in predicting the response to immunotherapy in head and neck cancer patients. Pathol Res Pract 2024; 260:155415. [PMID: 38996615 DOI: 10.1016/j.prp.2024.155415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 05/15/2024] [Accepted: 06/16/2024] [Indexed: 07/14/2024]
Abstract
Immunotherapy has a crucial role in the treatment of recurrent/metastatic head and neck squamous cell carcinoma (R/M HNSCC). However, only a small percentage of patients achieve long-term benefit in terms of overall response and survival. It was shown that HNSCC has an immunosuppressive microenvironment due to high levels of regulatory T cells and immunosuppressive molecules, such as LAG3 and CD73. The aim of our study was to investigate if the expression of CD73 by neoplastic and immune cells could affect the efficacy of anti-PD-1 immunotherapy. We reviewed data from 50 patients with R/M HNSCC receiving first line immunotherapy with or without chemotherapy based on a combined positive score (CPS). CD73 expression by cancer and immune cells was evaluated on pre-treatment and the percentage of stained cells was recorded. We analysed the association between CD73 expression on neoplastic and immune cells and early progression (EP), defined as progression occurring within 3 months. In 88 % of patients the primary tumour site was in the oral cavity or larynx. All patients received pembrolizumab associated in 40 % of cases to chemotherapy. CD73 was positive in 82 % and 96 % of cases on neoplastic and immune cells, respectively. The median value of CD73 was 32 % for neoplastic cells and 10 % for the immune ones. We observed a significant association between the CD73 expression on neoplastic cells over the median value and EP disease. We didn't record a correlation between the expression of CD73 on immune cells and early progression. Our findings suggest that higher expression of CD73 on neoplastic cells could predict resistance to immunotherapy in patients with CPS positive R/M HNSCC. The addition of this biomarker to routine evaluation of CPS could help to select the patients primary resistant to anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Andrea Botticelli
- Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome 00161, Italy
| | - Alessio Cirillo
- Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome 00161, Italy; Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena, 324, Rome 00161, Italy
| | - Giulia d'Amati
- Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome 00161, Italy
| | - Cira Di Gioia
- Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome 00161, Italy
| | - Alessandro Corsi
- Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome 00161, Italy
| | - Carlo Della Rocca
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University, Roma 00185, Italy
| | - Daniele Santini
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University, Roma 00185, Italy
| | - Raffaella Carletti
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Annalinda Pisano
- Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome 00161, Italy
| | - Antonella Polimeni
- Odontostomatological and Maxillo-Facial Science, 'Sapienza' University of Rome, Rome 00185, Italy
| | - Marco De Vincentiis
- Odontostomatological and Maxillo-Facial Science, 'Sapienza' University of Rome, Rome 00185, Italy
| | - Valentino Valentini
- Odontostomatological and Maxillo-Facial Science, 'Sapienza' University of Rome, Rome 00185, Italy
| | - Claudio di Cristofano
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University, Roma 00185, Italy
| | - Umberto Romeo
- Odontostomatological and Maxillo-Facial Science, 'Sapienza' University of Rome, Rome 00185, Italy
| | - Edoardo Cerbelli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena, 324, Rome 00161, Italy
| | - Daniela Messineo
- Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome 00161, Italy
| | - Francesca De Felice
- Department of Radiological, Oncological and Pathological Science, Policlinico Umberto I, "Sapienza" University of Rome, Rome 00161, Italy
| | - Martina Leopizzi
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University, Roma 00185, Italy
| | - Bruna Cerbelli
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University, Roma 00185, Italy.
| |
Collapse
|
37
|
Giraulo C, Orlando L, Morretta E, Voli A, Plaitano P, Cicala C, Potaptschuk E, Müller CE, Tosco A, Monti MC, Morello S. High levels of soluble CD73 unveil resistance to BRAF inhibitors in melanoma cells. Biomed Pharmacother 2024; 177:117033. [PMID: 38941889 DOI: 10.1016/j.biopha.2024.117033] [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: 05/15/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024] Open
Abstract
Melanoma cells express high levels of CD73 that produce extracellular immunosuppressive adenosine. Changes in the CD73 expression occur in response to tumor environmental factors, contributing to tumor phenotype plasticity and therapeutic resistance. Previously, we have observed that CD73 expression can be up-regulated on the surface of melanoma cells in response to nutritional stress. Here, we explore the mechanism by which melanoma cells release soluble CD73 under low nutrient availability and whether this might be affected by agents targeting the proto-oncogene B-Raf (BRAF). We found that starved melanoma cells can release high levels of CD73, able to convert AMP into adenosine, and this activity is abrogated by selective CD73 inhibitors, APCP or PSB-12489. The release of CD73 from melanoma cells is mediated by the matrix metalloproteinase MMP-9. Indeed, MMP-9 inhibitors significantly reduce the levels of CD73 released from the cells, while its surface levels increase. Of relevance, melanoma cells, harboring an activating BRAF mutation, upon treatment with dabrafenib or vemurafenib, show a strong reduction of CD73 cell expression and reduced levels of CD73 released into the extracellular space. Conversely, melanoma cells resistant to dabrafenib show high expression of membrane-bound CD73 and soluble CD73 released into the culture medium. In summary, our data indicate that CD73 is released from melanoma cells. The expression of CD73 is associated with response to BRAF inhibitors. Melanoma cells developing resistance to dabrafenib show increased expression of CD73, including soluble CD73 released from cells, suggesting that CD73 is involved in acquiring resistance to treatment.
Collapse
Affiliation(s)
- Caterina Giraulo
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy; PhD Program in Drug Discovery and Development, University of Salerno, Fisciano, SA, Italy
| | - Lavinia Orlando
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy; PhD Program in Drug Discovery and Development, University of Salerno, Fisciano, SA, Italy
| | - Elva Morretta
- Department of Pharmacy, University of Naples "Federico II", Napoli, NA, Italy
| | - Antonia Voli
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy; PhD Program in Drug Discovery and Development, University of Salerno, Fisciano, SA, Italy
| | - Paola Plaitano
- Department of Pharmacy, University of Naples "Federico II", Napoli, NA, Italy
| | - Carla Cicala
- Department of Pharmacy, University of Naples "Federico II", Napoli, NA, Italy
| | - Eugen Potaptschuk
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Christa E Müller
- PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn, Germany
| | - Alessandra Tosco
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy
| | - Maria Chiara Monti
- Department of Pharmacy, University of Naples "Federico II", Napoli, NA, Italy
| | - Silvana Morello
- Department of Pharmacy, University of Salerno, Fisciano, SA, Italy.
| |
Collapse
|
38
|
Ou J, Kang Y, Medlegeh, Fu K, Zhang Y, Yang W. An analysis of the vaginal microbiota and cervicovaginal metabolomics in cervical lesions and cervical carcinoma. Heliyon 2024; 10:e33383. [PMID: 39040371 PMCID: PMC11260971 DOI: 10.1016/j.heliyon.2024.e33383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 05/16/2024] [Accepted: 06/20/2024] [Indexed: 07/24/2024] Open
Abstract
Background To explore the role of vaginal microbiota and metabolomics in the progression of cervical dysplasia. Methods The patient group consists of female patients with low-grade, high-grade cervical dysplasia, and cervical cancer. Normal cervix samples from health volunteers were used as controls. The metabolic fingerprints of cervicovaginal lavage were analyzed using liquid chromatography-mass spectrometry, while the vaginal microbiota was examined through 16S rRNA sequencing. Bioinformatic analysis was adopted to investigate the interplay between hosts and microbes. The vaginal metabolic and microbiota profiles of 90 female patients with cervical dysplasia and 10 controls were analyzed to discover the biological characteristics underlying the progression of cervical cancer. Results We found that Valyl-Glutamate, N, N'-Diacetylbenzidine, and Oxidized glutathione, which were involved in oxidative stress response, were discriminators to distinguish the normal cervix, invasive cervical carcinomas, and CIN3 from others. Cervical carcinoma was characterized by a large variety of vaginal microbes (dominated by non-Lactobacillus communities) compared to the control. These microbes affected amino acid and nucleotide metabolism, producing metabolites with cervical carcinoma and genital inflammation compared to the control group. Conclusions This study revealed that cervicovaginal metabolic profiles were determined by cervical cancer, vaginal microbiota, and their interplays. ROS metabolism can be used to discriminate normal cervix, CIN3, and invasive cervical carcinoma.
Collapse
Affiliation(s)
- Jie Ou
- Department of Gyneacology, Xiangya Hospital Central South University, Changsha, Hunan, 410008, China
| | - Yanan Kang
- Department of Gyneacology, Xiangya Hospital Central South University, Changsha, Hunan, 410008, China
| | - Medlegeh
- Department of Gyneacology, Xiangya Hospital Central South University, Changsha, Hunan, 410008, China
| | - Kun Fu
- Department of Gyneacology, Xiangya Hospital Central South University, Changsha, Hunan, 410008, China
| | - Yu Zhang
- Department of Gyneacology, Xiangya Hospital Central South University, Changsha, Hunan, 410008, China
| | - Wenqing Yang
- Department of Gyneacology, Xiangya Hospital Central South University, Changsha, Hunan, 410008, China
| |
Collapse
|
39
|
Rossari F, Foti S, Camera S, Persano M, Casadei-Gardini A, Rimini M. Treatment options for advanced hepatocellular carcinoma: the potential of biologics. Expert Opin Biol Ther 2024; 24:455-470. [PMID: 38913107 DOI: 10.1080/14712598.2024.2363234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 05/30/2024] [Indexed: 06/25/2024]
Abstract
INTRODUCTION Advanced hepatocellular carcinoma (HCC) represents a significant global health burden, whose treatment has been recently revolutionized by the advent of biologic treatments. Despite that, innovative therapeutic regimens and approaches, especially immune-based, remain to be explored aiming at extending the therapeutic benefits to a wider population of patients. AREAS COVERED This review comprehensively discusses the evolving landscape of biological treatment modalities for advanced HCC, including immune checkpoint inhibitors, antiangiogenic monoclonal antibodies, tumor-targeting monoclonal antibodies either naked or drug-conjugated, therapeutic vaccines, oncolytic viruses, adoptive cell therapies, and cytokine-based therapies. Key clinical trials and preclinical studies are examined, highlighting the actual or potential impact of these interventions in reshaping treatment paradigms for HCC. EXPERT OPINION Tailored and rational combination strategies, leveraging the synergistic effects of different modalities, represent a promising approach to maximize treatment efficacy in advanced HCC, which should aim at conversion endpoints to increase the fraction of patients eligible for curative approaches. The identification of predictive biomarkers holds the key to optimizing patient selection and improving therapeutic outcomes.
Collapse
Affiliation(s)
- Federico Rossari
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Silvia Foti
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Silvia Camera
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Mara Persano
- Medical Oncology, University and University Hospital of Cagliari, Cagliari, Italy
| | - Andrea Casadei-Gardini
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| | - Margherita Rimini
- Department of Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute Hospital, Milan, Italy
| |
Collapse
|
40
|
Coban B, Wang Z, Liao CY, Beslmüller K, Timmermans MA, Martens JW, Hundscheid JH, Slutter B, Zweemer AJ, Neubert E, Danen EH. GRHL2 suppression of NT5E/CD73 in breast cancer cells modulates CD73-mediated adenosine production and T cell recruitment. iScience 2024; 27:109738. [PMID: 38706844 PMCID: PMC11068632 DOI: 10.1016/j.isci.2024.109738] [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: 10/10/2023] [Revised: 03/03/2024] [Accepted: 04/10/2024] [Indexed: 05/07/2024] Open
Abstract
Tumor tissues often contain high extracellular adenosine, promoting an immunosuppressed environment linked to mesenchymal transition and immune evasion. Here, we show that loss of the epithelial transcription factor, GRHL2, triggers NT5E/CD73 ecto-enzyme expression, augmenting the conversion of AMP to adenosine. GRHL2 binds an intronic NT5E sequence and is negatively correlated with NT5E/CD73 in breast cancer cell lines and patients. Remarkably, the increased adenosine levels triggered by GRHL2 depletion in MCF-7 breast cancer cells do not suppress but mildly increase CD8 T cell recruitment, a response mimicked by a stable adenosine analog but prevented by CD73 inhibition. Indeed, NT5E expression shows a positive rather than negative association with CD8 T cell infiltration in breast cancer patients. These findings reveal a GRHL2-regulated immune modulation mechanism in breast cancers and show that extracellular adenosine, besides its established role as a suppressor of T cell-mediated cytotoxicity, is associated with enhanced T cell recruitment.
Collapse
Affiliation(s)
- Bircan Coban
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | - Zi Wang
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
- Department of clinical laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Chen-yi Liao
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | - Klara Beslmüller
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | - Mieke A.M. Timmermans
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - John W.M. Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, Rotterdam, the Netherlands
| | | | - Bram Slutter
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | - Annelien J.M. Zweemer
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | - Elsa Neubert
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| | - Erik H.J. Danen
- Leiden Academic Center for Drug Research, Leiden University, Leiden, the Netherlands
| |
Collapse
|
41
|
Ryan AT, Kim M, Lim K. Immune Cell Migration to Cancer. Cells 2024; 13:844. [PMID: 38786066 PMCID: PMC11120175 DOI: 10.3390/cells13100844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 04/27/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Immune cell migration is required for the development of an effective and robust immune response. This elegant process is regulated by both cellular and environmental factors, with variables such as immune cell state, anatomical location, and disease state that govern differences in migration patterns. In all cases, a major factor is the expression of cell surface receptors and their cognate ligands. Rapid adaptation to environmental conditions partly depends on intrinsic cellular immune factors that affect a cell's ability to adjust to new environment. In this review, we discuss both myeloid and lymphoid cells and outline key determinants that govern immune cell migration, including molecules required for immune cell adhesion, modes of migration, chemotaxis, and specific chemokine signaling. Furthermore, we summarize tumor-specific elements that contribute to immune cell trafficking to cancer, while also exploring microenvironment factors that can alter these cellular dynamics within the tumor in both a pro and antitumor fashion. Specifically, we highlight the importance of the secretome in these later aspects. This review considers a myriad of factors that impact immune cell trajectory in cancer. We aim to highlight the immunotherapeutic targets that can be harnessed to achieve controlled immune trafficking to and within tumors.
Collapse
Affiliation(s)
- Allison T. Ryan
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Minsoo Kim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| | - Kihong Lim
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA; (A.T.R.); (M.K.)
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester, Rochester, NY 14642, USA
| |
Collapse
|
42
|
Witt M, Oliveira-Ferrer L, Koch-Nolte F, Menzel S, Hell L, Sturmheit T, Seubert E, Weimer P, Ding Y, Qi M, Schmalfeldt B, Bokemeyer C, Fiedler W, Wellbrock J, Brauneck F. Expression of CD39 is associated with T cell exhaustion in ovarian cancer and its blockade reverts T cell dysfunction. Oncoimmunology 2024; 13:2346359. [PMID: 38737794 PMCID: PMC11087076 DOI: 10.1080/2162402x.2024.2346359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/12/2024] [Accepted: 04/18/2024] [Indexed: 05/14/2024] Open
Abstract
Immune exhaustion is a hallmark of ovarian cancer. Using multiparametric flow cytometry, the study aimed to analyze protein expression of novel immunological targets on CD3+ T cells isolated from the peripheral blood (n = 20), malignant ascites (n = 16), and tumor tissue (n = 6) of patients with ovarian cancer (OVCA). The study revealed an increased proportion of effector memory CD8+ T cells in OVCA tissue and malignant ascites. An OVCA-characteristic PD-1high CD8+ T cell population was detected, which differed from PD-1lowCD8+ T cells by increased co-expression of TIGIT, CD39, and HLA-DR. In addition, these OVCA-characteristic CD8+ T cells showed reduced expression of the transcription factor TCF-1, which may also indicate reduced effector function and memory formation. On the contrary, the transcription factor TOX, which significantly regulates terminal T cell-exhaustion, was found more frequently in these cells. Further protein and gene analysis showed that CD39 and CD73 were also expressed on OVCA tumor cells isolated from solid tumors (n = 14) and malignant ascites (n = 9). In the latter compartment, CD39 and CD73 were also associated with the expression of the "don't eat me" molecule CD24 on tumor cells. Additionally, ascites-derived CD24+EpCAM+ tumor cells showed a higher frequency of CD39+ or CD73+ cells. Furthermore, CD39 expression was associated with unfavorable clinical parameters. Expression of CD39 on T cells was upregulated through CD3/CD28 stimulation and its blockade by a newly developed nanobody construct resulted in increased proliferation (eFluor), activation (CD25 and CD134), and production of cytotoxic cytokines (IFN-γ, TNF-α, and granzyme-B) of CD8+ T cells.
Collapse
Affiliation(s)
- Marius Witt
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, Hamburg, Germany
| | | | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stephan Menzel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Core Facility Nanobodies, University of Bonn, Bonn, Germany
| | | | | | - Elisa Seubert
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, Hamburg, Germany
| | - Pauline Weimer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, Hamburg, Germany
| | - Yi Ding
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Minyue Qi
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Barbara Schmalfeldt
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, Hamburg, Germany
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, Hamburg, Germany
| | - Franziska Brauneck
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
43
|
Arora C, Matic M, Bisceglia L, Di Chiaro P, De Oliveira Rosa N, Carli F, Clubb L, Nemati Fard LA, Kargas G, Diaferia GR, Vukotic R, Licata L, Wu G, Natoli G, Gutkind JS, Raimondi F. The landscape of cancer-rewired GPCR signaling axes. CELL GENOMICS 2024; 4:100557. [PMID: 38723607 PMCID: PMC11099383 DOI: 10.1016/j.xgen.2024.100557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 02/17/2024] [Accepted: 04/10/2024] [Indexed: 05/15/2024]
Abstract
We explored the dysregulation of G-protein-coupled receptor (GPCR) ligand systems in cancer transcriptomics datasets to uncover new therapeutics opportunities in oncology. We derived an interaction network of receptors with ligands and their biosynthetic enzymes. Multiple GPCRs are differentially regulated together with their upstream partners across cancer subtypes and are associated to specific transcriptional programs and to patient survival patterns. The expression of both receptor-ligand (or enzymes) partners improved patient stratification, suggesting a synergistic role for the activation of GPCR networks in modulating cancer phenotypes. Remarkably, we identified many such axes across several cancer molecular subtypes, including many involving receptor-biosynthetic enzymes for neurotransmitters. We found that GPCRs from these actionable axes, including, e.g., muscarinic, adenosine, 5-hydroxytryptamine, and chemokine receptors, are the targets of multiple drugs displaying anti-growth effects in large-scale, cancer cell drug screens, which we further validated. We have made the results generated in this study freely available through a webapp (gpcrcanceraxes.bioinfolab.sns.it).
Collapse
Affiliation(s)
- Chakit Arora
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Marin Matic
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Luisa Bisceglia
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Pierluigi Di Chiaro
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Natalia De Oliveira Rosa
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Francesco Carli
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Lauren Clubb
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lorenzo Amir Nemati Fard
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Giorgos Kargas
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy
| | - Giuseppe R Diaferia
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - Ranka Vukotic
- Azienda Ospedaliero-Universitaria Pisana, Via Roma, 67, 56126 Pisa, Italy
| | - Luana Licata
- Department of Biology, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Guanming Wu
- Division of Bioinformatics and Computational Biology, Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, USA
| | - Gioacchino Natoli
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milano, Italy
| | - J Silvio Gutkind
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Francesco Raimondi
- Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy; Laboratorio di Biologia Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126 Pisa, Italy.
| |
Collapse
|
44
|
Wang L, Zhang J, Zhang W, Zheng M, Guo H, Pan X, Li W, Yang B, Ding L. The inhibitory effect of adenosine on tumor adaptive immunity and intervention strategies. Acta Pharm Sin B 2024; 14:1951-1964. [PMID: 38799637 PMCID: PMC11119508 DOI: 10.1016/j.apsb.2023.12.004] [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: 07/05/2023] [Revised: 10/02/2023] [Accepted: 11/14/2023] [Indexed: 05/29/2024] Open
Abstract
Adenosine (Ado) is significantly elevated in the tumor microenvironment (TME) compared to normal tissues. It binds to adenosine receptors (AdoRs), suppressing tumor antigen presentation and immune cell activation, thereby inhibiting tumor adaptive immunity. Ado downregulates major histocompatibility complex II (MHC II) and co-stimulatory factors on dendritic cells (DCs) and macrophages, inhibiting antigen presentation. It suppresses anti-tumor cytokine secretion and T cell activation by disrupting T cell receptor (TCR) binding and signal transduction. Ado also inhibits chemokine secretion and KCa3.1 channel activity, impeding effector T cell trafficking and infiltration into the tumor site. Furthermore, Ado diminishes T cell cytotoxicity against tumor cells by promoting immune-suppressive cytokine secretion, upregulating immune checkpoint proteins, and enhancing immune-suppressive cell activity. Reducing Ado production in the TME can significantly enhance anti-tumor immune responses and improve the efficacy of other immunotherapies. Preclinical and clinical development of inhibitors targeting Ado generation or AdoRs is underway. Therefore, this article will summarize and analyze the inhibitory effects and molecular mechanisms of Ado on tumor adaptive immunity, as well as provide an overview of the latest advancements in targeting Ado pathways in anti-tumor immune responses.
Collapse
Affiliation(s)
- Longsheng Wang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jie Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wenxin Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingming Zheng
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hongjie Guo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaohui Pan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wen Li
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China
| | - Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
| |
Collapse
|
45
|
Yang H, Zhang Z, Zhao K, Zhang Y, Yin X, Zhu G, Wang Z, Yan X, Li X, He T, Wang K. Targeting the adenosine signaling pathway in macrophages for cancer immunotherapy. Hum Immunol 2024; 85:110774. [PMID: 38521664 DOI: 10.1016/j.humimm.2024.110774] [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: 10/18/2023] [Revised: 02/14/2024] [Accepted: 03/04/2024] [Indexed: 03/25/2024]
Abstract
One of the ways in which macrophages support tumorigenic growth is by producing adenosine, which acts to dampen antitumor immune responses and is generated by both tumor and immune cells in the tumor microenvironment (TME). Two cell surface expressed molecules, CD73 and CD39, boost catalytic adenosine triphosphate, leading to further increased adenosine synthesis, under hypoxic circumstances in the TME. There are four receptors (A1, A2A, A2B, and A3) expressed on macrophages that allow adenosine to perform its immunomodulatory effect. Researchers have shown that adenosine signaling is a key factor in tumor progression and an attractive therapeutic target for treating cancer. Several antagonistic adenosine-targeting biological therapies that decrease the suppressive action of tumor-associated macrophages have been produced and explored to transform this result from basic research into a therapeutic advantage. Here, we'll review the newest findings from studies of pharmacological compounds that target adenosine receptors, and their potential therapeutic value based on blocking the suppressive action of macrophages in tumors.
Collapse
Affiliation(s)
- Han Yang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Zongliang Zhang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Kai Zhao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Yulian Zhang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Xinbao Yin
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Guanqun Zhu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Zhenlin Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Xuechuan Yan
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Xueyu Li
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China
| | - Tianzhen He
- Nantong University, Institute of Special Environmental Medicine, Nantong, China.
| | - Ke Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shangdong, China.
| |
Collapse
|
46
|
Ray A, Du T, Wan X, Song Y, Pillai SC, Musa MA, Fang T, Moore J, Blank B, Du X, Chen X, Warne R, Sutimantanapi D, Lui F, Zavorotinskaya T, Colas C, Friedman L, Junttila MR, Chauhan D, Anderson KC. A novel small molecule inhibitor of CD73 triggers immune-mediated multiple myeloma cell death. Blood Cancer J 2024; 14:58. [PMID: 38594241 PMCID: PMC11004003 DOI: 10.1038/s41408-024-01019-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 04/11/2024] Open
Abstract
CD73 is the key ectoenzyme involved in the generation of AMP-derived adenosine, which contributes to immunosuppression in the MM BM milieu. Blocking CD73 activity with a potent, selective, orally bioavailable CD73 inhibitor ORIC-533 decreases adenosine generation, overcomes immune suppression, and restores immune cell-mediated MM cell lysis. Based on these preclinical studies, a multi-center clinical trial of ORIC-533 has been initiated in patients with relapsed refractory MM (NCT05227144).
Collapse
Affiliation(s)
- Arghya Ray
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Ting Du
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Xueping Wan
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Yan Song
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Sindhu C Pillai
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Md Abu Musa
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Teng Fang
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jared Moore
- ORIC Pharmaceuticals, Inc., South San Francisco, CA, USA
| | - Brian Blank
- ORIC Pharmaceuticals, Inc., South San Francisco, CA, USA
| | - Xiaohui Du
- ORIC Pharmaceuticals, Inc., South San Francisco, CA, USA
| | - Xi Chen
- ORIC Pharmaceuticals, Inc., South San Francisco, CA, USA
| | - Robert Warne
- ORIC Pharmaceuticals, Inc., South San Francisco, CA, USA
| | | | - Fang Lui
- ORIC Pharmaceuticals, Inc., South San Francisco, CA, USA
| | | | | | - Lori Friedman
- ORIC Pharmaceuticals, Inc., South San Francisco, CA, USA
| | | | - Dharminder Chauhan
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | - Kenneth C Anderson
- The LeBow Institute for Myeloma Therapeutics and Jerome Lipper Myeloma Center, Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
47
|
Battesti G, Thonnart N, Bozonnat A, Ram‐Wolff C, de Masson A, Bensussan A, Bagot M, Marie‐Cardine A, Battistella M. CD39 is expressed by a wide range of cutaneous T-cell lymphomas. SKIN HEALTH AND DISEASE 2024; 4:e334. [PMID: 38577051 PMCID: PMC10988668 DOI: 10.1002/ski2.334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/06/2024]
Abstract
CD39, an ectoenzyme in the immunosuppressive CD39/CD73/adenosine pathway, known to promote solid tumour outgrowth and spreading, was investigated in both skin and blood compartments of cutaneous T cell lymphomas. CD39 was overexpressed by peripheral blood T-cells in Sezary syndrome and mycosis fungoides, and in skin-infiltrating lymphocytes of Sezary syndrome, mycosis fungoides, subcutaneous panniculitis-like T-cell lymphoma and primary cutaneous CD30-positive lymphoproliferation. Our study emphasizes the interest in using CD39/CD73/adenosine pathway blocking agents for cutaneous T cell lymphomas treatment.
Collapse
Affiliation(s)
- Gilles Battesti
- Team 1Human Immunology, Pathophysiology, ImmunotherapyINSERM U976ParisFrance
- Université Paris CitéIRSLParisFrance
| | - Nicolas Thonnart
- Team 1Human Immunology, Pathophysiology, ImmunotherapyINSERM U976ParisFrance
- Université Paris CitéIRSLParisFrance
| | - Alizée Bozonnat
- Team 1Human Immunology, Pathophysiology, ImmunotherapyINSERM U976ParisFrance
- Université Paris CitéIRSLParisFrance
| | - Caroline Ram‐Wolff
- Team 1Human Immunology, Pathophysiology, ImmunotherapyINSERM U976ParisFrance
- Université Paris CitéIRSLParisFrance
- Department of DermatologySaint‐Louis HospitalParisFrance
| | - Adèle de Masson
- Team 1Human Immunology, Pathophysiology, ImmunotherapyINSERM U976ParisFrance
- Université Paris CitéIRSLParisFrance
- Department of DermatologySaint‐Louis HospitalParisFrance
| | - Armand Bensussan
- Team 1Human Immunology, Pathophysiology, ImmunotherapyINSERM U976ParisFrance
- Université Paris CitéIRSLParisFrance
| | - Martine Bagot
- Team 1Human Immunology, Pathophysiology, ImmunotherapyINSERM U976ParisFrance
- Université Paris CitéIRSLParisFrance
- Department of DermatologySaint‐Louis HospitalParisFrance
| | - Anne Marie‐Cardine
- Team 1Human Immunology, Pathophysiology, ImmunotherapyINSERM U976ParisFrance
- Université Paris CitéIRSLParisFrance
| | - Maxime Battistella
- Team 1Human Immunology, Pathophysiology, ImmunotherapyINSERM U976ParisFrance
- Université Paris CitéIRSLParisFrance
- Department of PathologySaint‐Louis HospitalParisFrance
| |
Collapse
|
48
|
Okada Y, Suzuki H, Tanaka T, Kaneko MK, Kato Y. Epitope Mapping of an Anti-Mouse CD39 Monoclonal Antibody Using PA Scanning and RIEDL Scanning. Monoclon Antib Immunodiagn Immunother 2024; 43:44-52. [PMID: 38507671 DOI: 10.1089/mab.2023.0029] [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: 03/22/2024] Open
Abstract
A cell-surface ectonucleotidase CD39 mediates the conversion of extracellular adenosine triphosphate into immunosuppressive adenosine with another ectonucleotidase CD73. The elevated adenosine in the tumor microenvironment attenuates antitumor immunity, which promotes tumor cell immunologic escape and progression. Anti-CD39 monoclonal antibodies (mAbs), which suppress the enzymatic activity, can be applied to antitumor therapy. Therefore, an understanding of the relationship between the inhibitory activity and epitope of mAbs is important. We previously established an anti-mouse CD39 (anti-mCD39) mAb, C39Mab-1 using the Cell-Based Immunization and Screening method. In this study, we determined the critical epitope of C39Mab-1 using flow cytometry. We performed the PA tag (12 amino acids [aa])-substituted analysis (named PA scanning) and RIEDL tag (5 aa)-substituted analysis (named RIEDL scanning) to determine the critical epitope of C39Mab-1 using flow cytometry. By the combination of PA scanning and RIEDL scanning, we identified the conformational epitope, spanning three segments of 275-279, 282-291, and 306-323 aa of mCD39. These analyses would contribute to the identification of the conformational epitope of membrane proteins.
Collapse
Affiliation(s)
- Yuki Okada
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiroyuki Suzuki
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
49
|
Menzel S, Duan Y, Hambach J, Albrecht B, Wendt-Cousin D, Winzer R, Tolosa E, Rissiek A, Guse AH, Haag F, Magnus T, Koch-Nolte F, Rissiek B. Generation and characterization of antagonistic anti-human CD39 nanobodies. Front Immunol 2024; 15:1328306. [PMID: 38590528 PMCID: PMC11000232 DOI: 10.3389/fimmu.2024.1328306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
CD39 is the major enzyme controlling the levels of extracellular adenosine triphosphate (ATP) via the stepwise hydrolysis of ATP to adenosine diphosphate (ADP) and adenosine monophosphate (AMP). As extracellular ATP is a strong promoter of inflammation, monoclonal antibodies (mAbs) blocking CD39 are utilized therapeutically in the field of immune-oncology. Though anti-CD39 mAbs are highly specific for their target, they lack deep penetration into the dense tissue of solid tumors, due to their large size. To overcome this limitation, we generated and characterized nanobodies that targeted and blocked human CD39. From cDNA-immunized alpacas we selected 16 clones from seven nanobody families that bind to two distinct epitopes of human CD39. Among these, clone SB24 inhibited the enzymatic activity of CD39. Of note, SB24 blocked ATP degradation by both soluble and cell surface CD39 as a 15kD monomeric nanobody. Dimerization via fusion to an immunoglobulin Fc portion further increased the blocking potency of SB24 on CD39-transfected HEK cells. Finally, we confirmed the CD39 blocking properties of SB24 on human PBMCs. In summary, SB24 provides a new small biological antagonist of human CD39 with potential application in cancer therapy.
Collapse
Affiliation(s)
- Stephan Menzel
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg- Eppendorf, Hamburg, Germany
- Core Facility Nanobodies, University of Bonn, Bonn, Germany
| | - Yinghui Duan
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Julia Hambach
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Birte Albrecht
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dorte Wendt-Cousin
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Riekje Winzer
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eva Tolosa
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anne Rissiek
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Cytometry und Cell Sorting Core Unit, Dept. of Stem Cell Transplantation, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Andreas H. Guse
- Department of Biochemistry and Molecular Cell Biology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Koch-Nolte
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
50
|
Wang H, Wei Y, Wang N. Purinergic pathways and their clinical use in the treatment of acute myeloid leukemia. Purinergic Signal 2024:10.1007/s11302-024-09997-8. [PMID: 38446337 DOI: 10.1007/s11302-024-09997-8] [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: 12/05/2023] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Despite the use of various therapies such as hematopoietic stem cell transplantation and chimeric antigen receptor T cell therapy (CAR-T), the prognosis of patients with acute myeloid leukemia (AML) is still generally poor. However, immunotherapy is currently a hot topic in the treatment of hematological tumors. Extracellular adenosine triphosphate (ATP) can be converted to adenosine diphosphate (ADP) via CD39, and ADP can be converted to adenosine via CD73, which can bind to P1 and P2 receptors to exert immunomodulatory effects. Research on the mechanism of the purinergic signaling pathway can provide a new direction for the treatment of AML, and inhibitors of this signaling pathway have been discovered by several researchers and gradually applied in the clinic. In this paper, the mechanism of the purinergic signaling pathway and its clinical application are described, revealing a new target for the treatment of AML and subsequent improvement in patient prognosis.
Collapse
Affiliation(s)
- Huijuan Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yujie Wei
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Na Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|