1
|
Xu S, Tang Y, Tong J. Dexmedetomidine alleviates the pro-tumor activity of perioperative stress in tumor-bearing mice: an alternative approach of psycho-physiological intervention. World J Surg Oncol 2025; 23:103. [PMID: 40156037 PMCID: PMC11951540 DOI: 10.1186/s12957-025-03665-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/19/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND The immediate perioperative period (IPP) usually is highly stressful and has significant effects on the postoperative recurrence/metastasis of tumors. Effective methods for limiting the impact of the IPP on postoperative recurrence/metastasis of tumors remain scarce. We aimed to determine the effects of dexmedetomidine (DEX) treatment during the IPP on postoperative recurrence/metastasis of tumors and the stress response. MATERIALS AND METHODS The clinical perioperative setting was mimicked via tumor resection and perioperative restraint stress in tumor-bearing mice with or without DEX during the IPP. The stress response was assessed using stress hormone and interleukin (IL)-6 levels in peripheral blood. Tumor cell growth was measured via in vivo bioluminescent imaging, cell viability assay, wound-healing assay, and Western blotting. RESULTS In tumor-bearing mice, DEX during the IPP limited the growth of implanted tumor cells and stress response in a dose-dependent manner. The serum from mice without DEX promoted cultured tumor cell growth, which was alleviated by beta-adrenergic receptor blocker propranolol or IL-6 antibody. Relative to the serum from mice without DEX, the serum from mice with DEX had lower stress hormone and IL-6 levels, as well as weaker effects on tumor growth promotion. Dexmedetomidine supplementation during culture had no significant effects on tumor cells. CONCLUSIONS Dexmedetomidine alleviates the pro-tumor activity of perioperative stress in abdominal tumors.
Collapse
Affiliation(s)
- Shanqing Xu
- Department of Anaesthesiology, Third Xiangya Hospital, Central South University, Changsha, China
- Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, China
| | - Yongzhong Tang
- Department of Anaesthesiology, Third Xiangya Hospital, Central South University, Changsha, China
- Center of Clinical Pharmacology, Central South University, Changsha, China
| | - Jianbin Tong
- Department of Anaesthesiology, Third Xiangya Hospital, Central South University, Changsha, China.
- Hunan Province Key Laboratory of Brain Homeostasis, Third Xiangya Hospital, Central South University, Changsha, China.
- Department of Anesthesiology, Third Xiangya Hospital, 138 Tongzipo Road, Yuelu District, Changsha, Hunan, 410013, P.R. China.
| |
Collapse
|
2
|
Chi X, Wang SH, Gao J, Su J, Du YZ, Xu XL. Carrier-Free Nanocombo-Sensitized Photoimmunotherapy via Activation of α2-Adrenergic Receptors. ACS APPLIED MATERIALS & INTERFACES 2025; 17:16437-16452. [PMID: 40040324 PMCID: PMC11931486 DOI: 10.1021/acsami.4c18052] [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: 10/19/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 03/06/2025]
Abstract
Photodynamic therapy (PDT)-based photoimmunotherapy has attracted increasing attention in the field of cancer immunotherapy. Nonetheless, monotherapy alone proves insufficient in eliciting robust and enduring tumor immunogenicity within the "cold" microenvironment of triple-negative breast cancer. Therefore, it is imperative to integrate phototherapy and immunostimulation strategies to achieve synergistic effects. Here, we developed a carrier-free nanocombo comprising a photosensitizer (chlorin e6, Ce6) and an α2-adrenergic receptor (α2-AR) agonist (guanfacine, GFC) to enhance photoimmunotherapy through α2-AR activation. Ce6 and GFC possessed the ability to self-assemble into spherical nanoparticles, with the resulting Ce6-GFC (CeG) exhibiting exceptional drug loading efficiency (approaching 100%) and long-lasting colloidal stability, along with effective in vivo tumor-targeting capabilities. Following near-infrared laser irradiation, CeG-mediated phototherapy instigated a rapid generation of reactive oxygen species, leading to membrane disruption and the release of tumor-associated antigens, thereby facilitating dendritic cell maturation. Furthermore, α2-AR agonists served to repolarize M2 tumor-associated macrophages toward the M1 phenotype via adenylyl cyclase-mediated activation of α2-AR, thereby promoting the recruitment and activation of cytotoxic T lymphocytes. As a result, the carrier-free nanocombo significantly enhanced the efficacy of photoimmunotherapy in combatting poorly immunogenic breast tumors in female mice. Our findings showcase a "killing two birds with one stone" approach that boosts tumor immunogenicity, mitigates tumor immunosuppression, and advances the field of photoimmunotherapy.
Collapse
Affiliation(s)
- XiaoKai Chi
- Shulan International
Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
- College of
Pharmacy, Jiamusi University, Jiamusi 154007, PR China
- Institute
of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Si-Hui Wang
- Shulan International
Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - JingJing Gao
- Shulan International
Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| | - Jin Su
- College of
Pharmacy, Jiamusi University, Jiamusi 154007, PR China
| | - Yong-Zhong Du
- Institute
of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Xiao-Ling Xu
- Shulan International
Medical College, Zhejiang Shuren University, Hangzhou 310015, PR China
| |
Collapse
|
3
|
Dong J, Su T, Wu J, Xiang Y, Song M, He C, Shao L, Yang Y, Chen S. Drug functional remapping: a new promise for tumor immunotherapy. Front Oncol 2025; 15:1519355. [PMID: 40161377 PMCID: PMC11949826 DOI: 10.3389/fonc.2025.1519355] [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: 10/29/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
The research and development of new anti-cancer drugs face challenges such as high costs, lengthy development cycles, and limited data on side effects. In contrast, the clinical safety and side effects of traditional drugs have been well established through long-term use. The development or repurposing of traditional drugs with potential applications in cancer treatment offers an economical, feasible, and promising strategy for new drug development. This article reviews the novel applications of traditional drugs in tumor immunotherapy, discussing how they can enhance tumor treatment efficacy through functional repositioning, while also reducing development time and costs. Recent advancements in cancer immunotherapy have revolutionized treatment options, but resistance to ICIs remains a significant challenge. Drug repurposing has emerged as a promising strategy to identify novel agents that can enhance the efficacy of immunotherapies by overcoming ICI resistance. A study suggests that drug repositioning has the potential to modulate immune cell activity or alter the tumor microenvironment, thereby circumventing the resistance mechanisms associated with immune checkpoint blockade. This approach provides a rapid and cost-effective pathway for identifying therapeutic candidates that can be quickly transitioned into clinical trials. To improve the effectiveness of tumor immunotherapy, it is crucial to explore systematic methods for identifying repurposed drug candidates. Methods such as high-throughput screening, computational drug repositioning, and bioinformatic analysis have been employed to efficiently identify potential candidates for cancer treatment. Furthermore, leveraging databases related to immunotherapy and drug repurposing can provide valuable resources for drug discovery and facilitate the identification of promising compounds. It focuses on the latest advancements in the use of antidiabetic drugs, antihypertensive agents, weight-loss medications, antifungal agents, and antiviral drugs in tumor immunotherapy, examining their mechanisms of action, clinical application prospects, and associated challenges. In this context, our aim is to explore these strategies and highlight their potential for expanding the therapeutic options available for cancer immunotherapy, providing valuable references for cancer research and treatment.
Collapse
Affiliation(s)
- Jiayi Dong
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ting Su
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiexiong Wu
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yu Xiang
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Minghan Song
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
| | - Canfeng He
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
| | - Lijuan Shao
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yubin Yang
- Traditional Chinese Medicine Department, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| | - Size Chen
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Provincial Engineering Research Center for Precision Medicine in Esophageal Cancer, Guangdong Pharmaceutical University, Guangzhou, China
- Key Laboratory of Monitoring Adverse Reactions Associated with Chimeric Antigen Receptor T-Cell Therapy, Guangdong Higher Education Institutions, Guangdong Pharmaceutical University, Guangzhou, China
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
4
|
Yu N, Xu Y, Sun Q, Ge Y, Guo Y, Chen M, Shan H, Zheng M, Chen Z, Zhao S, Chen X. Size-specific clonidine-loaded liposomes: Advancing melanoma microenvironment suppression with safety and precision. J Control Release 2025; 379:120-134. [PMID: 39756687 DOI: 10.1016/j.jconrel.2025.01.001] [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: 09/02/2024] [Revised: 11/26/2024] [Accepted: 01/01/2025] [Indexed: 01/07/2025]
Abstract
The immunosuppressive tumor microenvironment (TME) plays a crucial role in the progression and treatment resistance of melanoma. Modulating the TME is thus a key strategy for enhancing therapeutic outcomes. Previousstudies have identified clonidine (CLD), an α2-adrenergic receptor agonist, as a promising agent that enhances T lymphocyte infiltration and reduces myeloid-derived suppressor cells within the TME, thereby promoting antitumor immune responses. In this study, we discovered that CLD reshaped the melanoma immune microenvironment, facilitating T-cell activation and exerting antitumor effects. However, the high doses of CLD required for effective TME modulation pose significant toxicity concerns, limiting its clinical applicability. To address this, we employed the controllable cavitation-on-a-chip (CCC) platform to formulate CLD-loaded liposomes and optimize their size. This approach aimed to enhance the precision and efficacy of drug delivery while reducing systemic side effects. Our results demonstrated that size-specific CLD liposomes, particularly those at 50 nm, significantly improved tumor growth inhibition and immune cell infiltration within the TME. Moreover, these optimized liposomes mitigate adverse effects associated with high-dose CLD treatment. This study indicates the potential of CCC-optimized CLD liposomes as a safer and more effective melanoma therapy, highlighting the critical interplay between liposome size control and therapeutic outcomes in cancer treatment.
Collapse
Affiliation(s)
- Nianzhou Yu
- 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, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 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, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qi Sun
- Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China; School of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Yi Ge
- 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, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yeye Guo
- 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, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Maike 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, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Han Shan
- Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China; School of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Mingde Zheng
- Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China; School of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Zeyu Chen
- Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China; School of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China.
| | - Shuang Zhao
- 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, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, 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, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
5
|
Bezu L, Forget P, Hollmann MW, Parat MO, Piegeler T. Potential influence of different peri-operative analgesic regimens on tumour biology and outcome after oncologic surgery: A narrative review. Eur J Anaesthesiol 2025; 42:233-243. [PMID: 39743967 DOI: 10.1097/eja.0000000000002118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The management of peri-operative pain is one of the pillars of anaesthesia and is of particular importance in patients undergoing surgery for solid malignant tumours. Amongst several options, the most commonly employed analgesic regimens involve opioids, NSAIDs and regional anaesthesia techniques with different local anaesthetics. In recent years, several research reports have tried to establish a connection between peri-operative anaesthesia care and outcome after cancer surgery. Experimental studies have indicated that certain pain management substances may influence cancer progression, mainly by modifying the tumour's response to surgical stress and peri-operative inflammation. However, these promising in-vitro and in-vivo data have yet to be confirmed by randomised clinical trials. The reason for this might lie with the nature of tumour biology itself, and in the diversity of patient and tumour phenotypes. In a translational approach, future research should therefore concentrate on patient and tumour-related factors or biomarkers, which might either influence the tumour and its microenvironment or predict potential responses to interventions, including the choice of the analgesic. This might not only be relevant for the daily practice of clinical anaesthesia, but would also be of great importance for patients undergoing cancer surgery, who might be able to receive an individualised anaesthetic regimen based on their phenotypic profile.
Collapse
Affiliation(s)
- Lucillia Bezu
- From the Département d'Anesthésie, Chirurgie et Interventionnel (LB), U1138 Metabolism, Cancer and Immunity, Gustave Roussy, Villejuif, France (LB), Department of Anesthesiology, Perioperative and Pain Medicine, School of Medicine, Stanford University, Stanford, California, USA (LB), Aberdeen Centre for Arthritis and Musculoskeletal Health (Epidemiology Group), Institute of Applied Health Sciences, School of Medicine, Medical Sciences and Nutrition (PF), Anaesthesia department, NHS Grampian, Aberdeen, UK (PF), IMAGINE UR UM 103, Montpellier University, Anesthesia Critical Care, Emergency and Pain Medicine Division, Nîmes University Hospital, Nîmes, France (PF), Pain and Opioids after Surgery (PANDOS) European Society of Anaesthesiology and Intensive Care (ID ESAIC_RG_PAND) Research Group, Brussels, Belgium (PF), Department of Anaesthesiology, Amsterdam UMC, Amsterdam, The Netherlands (MWH), School of Pharmacy, The University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba Qld, Australia (M-OP), Department of Anaesthesiology and Intensive Care, University of Leipzig Medical Center, Leipzig, Germany (TP), EuroPeriscope, ESAIC Onco-Anaesthesiology Research Group, Brussels, Belgium (TP, LB, PF, MWH)
| | | | | | | | | |
Collapse
|
6
|
Fan NW, Yu M, Wang S, Blanco T, Luznik Z, Chauhan SK, Viswanath V, Gil D, Held K, Chen Y, Dana R. Activation of α2B/2C adrenergic receptor ameliorates ocular surface inflammation through enhancing regulatory T cell function. Mucosal Immunol 2025; 18:176-187. [PMID: 39522611 PMCID: PMC11869812 DOI: 10.1016/j.mucimm.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
There is an unmet need for effectively treating dry eye disease (DED), a T cell-mediated chronic, inflammatory ocular surface disorder. Given the potential of nonneuronal adrenergic system in modulating T cell response, we herein investigated the therapeutic efficacy and the underlying mechanisms of a specific alpha 2 adrenergic receptor agonist (AGN-762, selective for α2B/2C receptor subtypes) in a mouse model of DED. Experimental DED was treated with the AGN-762 by oral gavage, either at disease induction or after disease establishment, and showed sustained amelioration, along with reduced expression of DED-pathogenic cytokines in ocular surface tissues, decreased corneal MHC-II+CD11b+ cells and lymphoid Th17 cells, and higher function of regulatory T cells (Treg). In vitro culture of DED-derived effector T helper cells (Teff) with AGN-762 failed to suppress Th17 response, while culture of DED-Treg with AGN-762 led to enhanced suppressive function of Treg and their IL-10 production. Adoptive transfer of AGN-762-pretreated DED-Treg in syngeneic B6.Rag1-/- mice effectively suppressed DED Teff-mediated disease and Th17 response, and the effect was abolished by the neutralization of IL-10. In conclusion, our findings demonstrate that α2B/2C adrenergic receptor agonism effectively ameliorates persistent corneal epitheliopathy in DED by enhancing IL-10 production from Treg and thus restoring their immunoregulatory function.
Collapse
Affiliation(s)
- Nai-Wen Fan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, Taipei Veterans General Hospital, Taiwan; School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Man Yu
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA; Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Shudan Wang
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Tomas Blanco
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Zala Luznik
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA
| | - Veena Viswanath
- (Former) Development Sciences, AbbVie Inc., Irvine, CA 92612, USA
| | - Daniel Gil
- (Former) Biological Research, Allergan plc, Irvine, CA 92612, USA
| | - Katherine Held
- Ophthalmology Discovery Research, AbbVie Inc., Irvine, CA 92612, USA
| | - Yihe Chen
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA.
| | - Reza Dana
- Schepens Eye Research Institute of Massachusetts Eye and Ear, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
7
|
Lee YK, Xiao C, Zhou X, Wang L, McReynolds MG, Wu Z, Purisic E, Kim H, Li X, Pang ZP, Dai J, Peng J, Yang N, Yue Z. Bipolar and schizophrenia risk gene AKAP11 encodes an autophagy receptor coupling the regulation of PKA kinase network homeostasis to synaptic transmission. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.12.30.630813. [PMID: 39803523 PMCID: PMC11722322 DOI: 10.1101/2024.12.30.630813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Human genomic studies have identified protein-truncating variants in AKAP11 associated with both bipolar disorder and schizophrenia, implicating a shared disease mechanism driven by loss-of-function. AKAP11, a protein kinase A (PKA) adaptor, plays a key role in degrading the PKA-RI complex through selective autophagy. However, the neuronal functions of AKAP11 and the impact of its loss-of-function remains largely uncharacterized. Through multi-omics approaches, cell biology, and electrophysiology analysis in mouse models and human induced neurons, we delineated a central role of AKAP11 in coupling PKA kinase network regulation to synaptic transmission. Loss of AKAP11 disrupted PKA activity and impaired cellular functions that significantly overlap with pathways associated with the psychiatric disease. Moreover, we identified interactions between AKAP11, the PKA-RI adaptor SPHKAP, and the ER-resident autophagy-related proteins VAPA/B, which co-adapt and mediate PKA-RI degradation. Notably, AKAP11 deficiency impaired neurotransmission and decreased presynaptic protein levels in neurons, providing key insights into the mechanism underlying AKAP11-associated psychiatric diseases.
Collapse
|
8
|
Wang YH, Yang X, Liu CC, Wang X, Yu KD. Unraveling the peripheral nervous System's role in tumor: A Double-edged Sword. Cancer Lett 2025; 611:217451. [PMID: 39793755 DOI: 10.1016/j.canlet.2025.217451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 01/01/2025] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
The peripheral nervous system (PNS) includes all nerves outside the brain and spinal cord, comprising various cells like neurons and glial cells, such as schwann and satellite cells. The PNS is increasingly recognized for its bidirectional interactions with tumors, exhibiting both pro- and anti-tumor effects. Our review delves into the complex mechanisms underlying these interactions, highlighting recent findings that challenge the conventional understanding of PNS's role in tumorigenesis. We emphasize the contradictory results in the literature and propose novel perspectives on how these discrepancies can be resolved. By focusing on the PNS's influence on tumor initiation, progression, and microenvironment remodeling, we provide a comprehensive analysis that goes beyond the structural description of the PNS. Our review suggests that a deeper comprehension of the PNS-tumor crosstalk is pivotal for developing targeted anticancer strategies. We conclude by emphasizing the need for future research to unravel the intricate dynamics of the PNS in cancer, which may lead to innovative diagnostic tools and therapeutic approaches.
Collapse
Affiliation(s)
- Yan-Hao Wang
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China
| | - Xuan Yang
- Department of General Surgery, Shanxi Provincial People's Hospital, Taiyuan, 030000, PR China
| | - Cui-Cui Liu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China
| | - Xin Wang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Ke-Da Yu
- Department of Breast Surgery, Fudan University Shanghai Cancer Center and Cancer Institute, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China; Key Laboratory of Breast Cancer in Shanghai, Shanghai, 200032, PR China.
| |
Collapse
|
9
|
Stagg J, Gutkind JS. Targeting G Protein-Coupled Receptors in Immuno-Oncological Therapies. Annu Rev Pharmacol Toxicol 2025; 65:315-331. [PMID: 39270681 DOI: 10.1146/annurev-pharmtox-061724-080852] [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: 09/15/2024]
Abstract
The advent of cancer immunotherapy based on PD-1 and CTLA-4 immune checkpoint blockade (ICB) has revolutionized cancer treatment. However, many cancers do not respond to ICB, highlighting the urgent need for additional approaches to achieve durable cancer remission. The large family of G protein-coupled receptors (GPCRs) is the target of more than 30% of all approved drugs, but GPCRs have been underexploited in cancer immunotherapy. In this review, we discuss the central role of GPCRs in immune cell migration and function and describe how single-cell transcriptomic studies are illuminating the complexity of the human tumor immune GPCRome. These receptors include multiple GPCRs expressed in CD8 T cells that are activated by inflammatory mediators, protons, neurotransmitters, and metabolites that accumulate in the tumor microenvironment, thereby promoting T cell dysfunction. We also discuss new opportunities to target GPCRs as a multimodal approach to enhance the response to ICB for a myriad of human malignancies.
Collapse
Affiliation(s)
- John Stagg
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada;
| | - J Silvio Gutkind
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California, USA;
| |
Collapse
|
10
|
Sheng Y, Qiao C, Zhang Z, Shi X, Yang L, Xi R, Yu J, Liu W, Zhang G, Wang F. Calcium Channel Blocker Lacidipine Promotes Antitumor Immunity by Reprogramming Tryptophan Metabolism. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409310. [PMID: 39585774 PMCID: PMC11744582 DOI: 10.1002/advs.202409310] [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: 08/07/2024] [Revised: 10/14/2024] [Indexed: 11/27/2024]
Abstract
Dysfunction of calcium channels is involved in the development and progression of some cancers. However, it remains unclear the role of calcium channel inhibitors in tumor immunomodulation. Here, calcium channel blocker lacidipine is identified to potently inhibit the enzymatic activity and expression of indoleamine 2,3-dioxygenase 1 (IDO1), a rate-limiting enzyme in tryptophan metabolism. Lacidipine activates effector T cells and incapacitates regulatory T cells (Tregs) to augment the anti-tumor effect of chemotherapeutic agents in breast cancer by converting immunologically "cold" into "hot" tumors. Mechanistically, lacidipine targets calcium channels (CaV1.2/1.3) to inhibit Pyk2-JAK1-calmodulin complex-mediated IDO1 transcription suppression, which suppresses the kynurenine pathway and maintains the total nicotinamide adenine dinucleotide (NAD) pool by regulating NAD biosynthesis. These results reveal a new function of calcium channels in IDO1-mediated tryptophan metabolism in tumor immunity and warrant further development of lacidipine for the metabolic immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Yuwen Sheng
- Center for Natural Products ResearchChengdu Institute of BiologyChinese Academy of SciencesChengdu610041China
| | - Chong Qiao
- Center for Natural Products ResearchChengdu Institute of BiologyChinese Academy of SciencesChengdu610041China
| | - Zhonghui Zhang
- School Of Pharmaceutical SciencesSun Yat‐Sen UniversityGuangzhou511400China
| | - Xiaoke Shi
- Center for Natural Products ResearchChengdu Institute of BiologyChinese Academy of SciencesChengdu610041China
- University of Chinese Academy of SciencesBeijing100049China
| | - Linhan Yang
- Center for Natural Products ResearchChengdu Institute of BiologyChinese Academy of SciencesChengdu610041China
- University of Chinese Academy of SciencesBeijing100049China
| | - Ruiying Xi
- Center for Natural Products ResearchChengdu Institute of BiologyChinese Academy of SciencesChengdu610041China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jialing Yu
- Center for Natural Products ResearchChengdu Institute of BiologyChinese Academy of SciencesChengdu610041China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wanli Liu
- State Key Laboratory of Membrane BiologySchool of Life SciencesInstitute for ImmunologyBeijing Advanced Innovation Center for Structural BiologyBeijing Key Lab for Immunological Research on Chronic DiseasesBeijing100084China
| | - Guolin Zhang
- Center for Natural Products ResearchChengdu Institute of BiologyChinese Academy of SciencesChengdu610041China
| | - Fei Wang
- Center for Natural Products ResearchChengdu Institute of BiologyChinese Academy of SciencesChengdu610041China
| |
Collapse
|
11
|
Liu Y, Liu F, Zeng Y, Lin L, Yu H, Zhang S, Yang W. Hydrogel systems for spatiotemporal controlled delivery of immunomodulators: engineering the tumor immune microenvironment for enhanced cancer immunotherapy. Front Cell Dev Biol 2024; 12:1514595. [PMID: 39735340 PMCID: PMC11681625 DOI: 10.3389/fcell.2024.1514595] [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: 10/21/2024] [Accepted: 11/28/2024] [Indexed: 12/31/2024] Open
Abstract
Tumor immunotherapy, modulating innate and adaptive immunity, has become an important therapeutic strategy. However, the tumor immune microenvironment's (TIME) complexity and heterogeneity challenge tumor immunotherapy. Hydrogel is a hydrophilic three-dimensional (3D) mesh structure with good biocompatibility and drug release control, which is widely used in drug delivery, agriculture, industry, etc. Hydrogels loaded with immune cells, cytokines, immune checkpoint inhibitors, and anti-tumor drugs can achieve targeted delivery and ultimately activate the immune response in the TIME. In this review, we will summarize the components of the TIME and their immune effects, the emerging immunomodulatory agents, the characteristics and functions of hydrogels, and how hydrogels regulate innate and adaptive immune cells in the TIME.
Collapse
Affiliation(s)
- Yanting Liu
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Fang Liu
- Department of Neurosurgery, Department of Urology, Medical Research Center, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
- College of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Yan Zeng
- West China School of Basic Medical Sciences and Forensic Medicine, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Liangbin Lin
- Department of Neurosurgery, Department of Urology, Medical Research Center, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
- Obesity and Metabolism Medicine-Engineering Integration Laboratory, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
- The Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Hui Yu
- Department of Neurosurgery, Department of Urology, Medical Research Center, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Sunfu Zhang
- Department of Neurosurgery, Department of Urology, Medical Research Center, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| | - Wenyong Yang
- Department of Oncology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
- Department of Neurosurgery, Department of Urology, Medical Research Center, The Second Chengdu Hospital Affiliated to Chongqing Medical University, The Affiliated Hospital of Southwest Jiaotong University, The Third People’s Hospital of Chengdu, Chengdu, China
| |
Collapse
|
12
|
Yang P, Chen X, Yu F, Wang L, Li M, Bai Z, Xu H. CAR T cells secreting NGF-neutralizing scFv enhance efficacy in clear cell renal cell carcinoma by relieving immunosuppression through immunosympathectomy. J Immunother Cancer 2024; 12:e009910. [PMID: 39653553 PMCID: PMC11629019 DOI: 10.1136/jitc-2024-009910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cells have demonstrated remarkable breakthroughs in treating hematologic malignancies, yet their efficacy in solid tumors is limited by the immunosuppressive microenvironment. Sympathetic nerves significantly contribute to this immunosuppressive milieu in solid tumors. However, the impact of tumor sympathetic denervation on enhancing CAR T-cell antitumor efficacy remains unclear. METHODS We screened for sympathetic gene sets in various types of cancers and investigated the association of sympathetic nerves with immunosuppression in renal clear cell carcinoma. Using antibodies to block the nerve growth factor (NGF) pathway, we explored sympathetic nerve distribution in tumor tissues and tumor progression. Additionally, we engineered CAR T cells to secrete NGF single chain fragment variable (scFv) to achieve tumor immunosympathectomy and assessed their antitumor efficacy. Bulk RNA sequencing and single-cell RNA sequencing analyses were conducted to evaluate changes in immune cell phenotypes within the tumor microenvironment. RESULTS Blocking the NGF pathway with antibodies effectively reduced sympathetic nerve distribution in tumor tissues and delayed tumor progression. CAR T cells engineered to secrete NGF scFv achieved a similar tumor immunosympathectomy and exhibited enhanced tumor suppression. RNA sequencing analyses revealed that this augmented effect was primarily due to the inhibition of the terminal exhaustion phenotype in tumor-infiltrating CD8 T cells and the prevention of macrophage polarization from M1 to M2. This approach maintained a stronger antitumor immune state at the tumor site. Additionally, splenic T cells also exhibited a more potent immune effector phenotype following the infusion of NGF scFv-secreting CAR T cells. CONCLUSIONS Our results suggest that immunosympathectomy is a novel approach to weaken tumor microenvironment immunosuppression and synergistically enhance CAR T-cell efficacy against solid tumors.
Collapse
Affiliation(s)
- Peiwei Yang
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province, China Pharmaceutical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
| | - Xi Chen
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province, China Pharmaceutical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
| | - Fan Yu
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province, China Pharmaceutical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
| | - Lan Wang
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, Beijing, China
| | - Meng Li
- NHC Key Laboratory of Research on Quality and Standardization of Biotech Products, NMPA Key Laboratory for Quality Research and Evaluation of Biological Products, State Key Laboratory of Drug Regulatory Science, National Institutes for Food and Drug Control, Beijing, China
| | - Zongke Bai
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province, China Pharmaceutical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
| | - Hanmei Xu
- The Engineering Research Center of Synthetic Polypeptide Drug Discovery and Evaluation, Jiangsu Province, China Pharmaceutical University, Nanjing, Jiangsu Province, China
- State Key Laboratory of Natural Medicines, Ministry of Education, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
13
|
Wei X, Qiu J, Lai R, Wei T, Lin Z, Huang S, Jiang Y, Kuang Z, Zeng H, Gong Y, Xie X, Yang J, Zhang Y, Zhang S, Zou Z, Gao X, Bai X. A human organoid drug screen identifies α2-adrenergic receptor signaling as a therapeutic target for cartilage regeneration. Cell Stem Cell 2024; 31:1813-1830.e8. [PMID: 39353427 DOI: 10.1016/j.stem.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 07/09/2024] [Accepted: 09/02/2024] [Indexed: 10/04/2024]
Abstract
Directed differentiation of stem cells toward chondrogenesis in vitro and in situ to regenerate cartilage suffers from off-target differentiation and hypertrophic tendency. Here, we generated a cartilaginous organoid system from human expanded pluripotent stem cells (hEPSCs) carrying a COL2A1mCherry and COL10A1eGFP double reporter, enabling real-time monitoring of chondrogenesis and hypertrophy. After screening 2,040 FDA-approved drugs, we found that α-adrenergic receptor (α-AR) antagonists, especially phentolamine, stimulated chondrogenesis but repressed hypertrophy, while α2-AR agonists reduced chondrogenesis and induced hypertrophy. Phentolamine prevented cartilage degeneration in hEPSC cartilaginous organoid and human cartilage explant models and stimulated microfracture-activated endogenous skeletal stem cells toward hyaline-like cartilage regeneration without fibrotic degeneration in situ. Mechanistically, α2-AR signaling induced hypertrophic degeneration via cyclic guanosine monophosphate (cGMP)-dependent secretory leukocyte protease inhibitor (SLPI) production. SLPI-deleted cartilaginous organoid was degeneration resistant, facilitating large cartilage defect healing. Ultimately, targeting α2-AR/SLPI was a promising and clinically feasible strategy to regenerate cartilage via promoting chondrogenesis and repressing hypertrophy.
Collapse
Affiliation(s)
- Xiaocui Wei
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Histology and Embryology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jingyang Qiu
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Ruijun Lai
- Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China
| | - Tiantian Wei
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhijie Lin
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shijiang Huang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yuanjun Jiang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhanpeng Kuang
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Hao Zeng
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan Gong
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaoling Xie
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jun Yang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yue Zhang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sheng Zhang
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhipeng Zou
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xuefei Gao
- Department of Physiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, Guangdong, China.
| | - Xiaochun Bai
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; Academy of Orthopedics, Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital, Southern Medical University, Guangzhou 510630, China.
| |
Collapse
|
14
|
Luo Z, Wu W, Sun Q, Wang J. Accurate and transferable drug-target interaction prediction with DrugLAMP. Bioinformatics 2024; 40:btae693. [PMID: 39570605 PMCID: PMC11629708 DOI: 10.1093/bioinformatics/btae693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 10/29/2024] [Accepted: 11/14/2024] [Indexed: 11/22/2024] Open
Abstract
MOTIVATION Accurate prediction of drug-target interactions (DTIs), especially for novel targets or drugs, is crucial for accelerating drug discovery. Recent advances in pretrained language models (PLMs) and multi-modal learning present new opportunities to enhance DTI prediction by leveraging vast unlabeled molecular data and integrating complementary information from multiple modalities. RESULTS We introduce DrugLAMP (PLM-assisted multi-modal prediction), a PLM-based multi-modal framework for accurate and transferable DTI prediction. DrugLAMP integrates molecular graph and protein sequence features extracted by PLMs and traditional feature extractors. We introduce two novel multi-modal fusion modules: (i) pocket-guided co-attention (PGCA), which uses protein pocket information to guide the attention mechanism on drug features, and (ii) paired multi-modal attention (PMMA), which enables effective cross-modal interactions between drug and protein features. These modules work together to enhance the model's ability to capture complex drug-protein interactions. Moreover, the contrastive compound-protein pre-training (2C2P) module enhances the model's generalization to real-world scenarios by aligning features across modalities and conditions. Comprehensive experiments demonstrate DrugLAMP's state-of-the-art performance on both standard benchmarks and challenging settings simulating real-world drug discovery, where test drugs/targets are unseen during training. Visualizations of attention maps and application to predict cryptic pockets and drug side effects further showcase DrugLAMP's strong interpretability and generalizability. Ablation studies confirm the contributions of the proposed modules. AVAILABILITY AND IMPLEMENTATION Source code and datasets are freely available at https://github.com/Lzcstan/DrugLAMP. All data originate from public sources.
Collapse
Affiliation(s)
- Zhengchao Luo
- Department of Big Data and Biomedical AI, College of Future Technology, Peking University, Beijing 100871, China
| | - Wei Wu
- Department of Big Data and Biomedical AI, College of Future Technology, Peking University, Beijing 100871, China
| | - Qichen Sun
- School of Mathematical Sciences, Peking University, Beijing 100871, China
| | - Jinzhuo Wang
- Department of Big Data and Biomedical AI, College of Future Technology, Peking University, Beijing 100871, China
| |
Collapse
|
15
|
Moreno P, Ohara Y, Craig AJ, Liu H, Yang S, Dorsey TH, Zhang L, Panigrahi G, Cawley H, Azizian A, Gaedcke J, Ghadimi M, Hanna N, Hussain SP. ADRA2A promotes the classical/progenitor subtype and reduces disease aggressiveness of pancreatic cancer. Carcinogenesis 2024; 45:845-856. [PMID: 39136088 PMCID: PMC11584292 DOI: 10.1093/carcin/bgae056] [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: 03/14/2024] [Revised: 07/09/2024] [Accepted: 08/12/2024] [Indexed: 08/21/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) manifests diverse molecular subtypes, including the classical/progenitor and basal-like/squamous subtypes, with the latter known for its aggressiveness. We employed integrative transcriptome and metabolome analyses to identify potential genes contributing to the molecular subtype differentiation and its metabolic features. Our comprehensive analysis revealed that adrenoceptor alpha 2A (ADRA2A) was downregulated in the basal-like/squamous subtype, suggesting its potential role as a candidate suppressor of this subtype. Reduced ADRA2A expression was significantly associated with a high frequency of lymph node metastasis, higher pathological grade, advanced disease stage, and decreased survival among PDAC patients. In vitro experiments demonstrated that ADRA2A transgene expression and ADRA2A agonist inhibited PDAC cell invasion. Additionally, ADRA2A-high condition downregulated the basal-like/squamous gene expression signature, while upregulating the classical/progenitor gene expression signature in our PDAC patient cohort and PDAC cell lines. Metabolome analysis conducted on the PDAC cohort and cell lines revealed that elevated ADRA2A levels were associated with suppressed amino acid and carnitine/acylcarnitine metabolism, which are characteristic metabolic profiles of the classical/progenitor subtype. Collectively, our findings suggest that heightened ADRA2A expression induces transcriptome and metabolome characteristics indicative of classical/progenitor subtype with decreased disease aggressiveness in PDAC patients. These observations introduce ADRA2A as a candidate for diagnostic and therapeutic targeting in PDAC.
Collapse
Affiliation(s)
- Paloma Moreno
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Yuuki Ohara
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Amanda J Craig
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Huaitian Liu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Shouhui Yang
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Tiffany H Dorsey
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Lin Zhang
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Gatikrushna Panigrahi
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Helen Cawley
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Azadeh Azizian
- Städtisches Klinikum Karlsruhe, Moltkestraße 90, 76133 Karlsruhe, Germany
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Jochen Gaedcke
- Städtisches Klinikum Karlsruhe, Moltkestraße 90, 76133 Karlsruhe, Germany
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Michael Ghadimi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Nader Hanna
- Division of General and Oncologic Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, United States
- Division of Surgical Oncology, Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - S Perwez Hussain
- Pancreatic Cancer Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| |
Collapse
|
16
|
Degavre C, Lepez A, Ibanez S, François C, Głowacka K, Guilbaud C, Laloux-Morris F, Esfahani H, Brusa D, Bouzin C, Feron O. In situ endoscopic photodynamic therapy combined with immature DC vaccination induces a robust T cell response against peritoneal carcinomatosis. J Immunother Cancer 2024; 12:e009752. [PMID: 39500528 PMCID: PMC11552574 DOI: 10.1136/jitc-2024-009752] [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: 10/20/2024] [Indexed: 11/13/2024] Open
Abstract
BACKGROUND Immunogenic cell death (ICD) and ferroptosis have recently emerged as key factors in the anticancer immune response. Among the treatments able to induce ICD and the associated release of danger signals is photodynamic therapy (PDT). Ferroptosis for its part results from lipid peroxidation and is induced by CD8+ T cells to kill nearby cancer cells on IFN-γ production. We aimed to combine the two concepts, that is, to evaluate whether the strong pro-oxidant effects of PDT may promote ferroptosis and antigen release and to develop a procedure for in situ PDT to prepare the soil for highly endocytotic immature dendritic cell (iDC) adoptive transfer. This approach was implemented for managing peritoneal carcinomatosis, a lesion often associated with poor outcomes. METHODS We used three-dimensional (3D) heterotypic spheroids made of cancer cells, exposed them to a white light-activated OR141 photosensitizer (PS), and subsequently complexified them by adding iDC and naive lymphocytes. We next used a model of mouse peritoneal carcinomatosis and administered PDT using laparoscopy to locally induce photoactivation using the endoscope light. The immune response following adoptive transfer of iDC was tracked both in vivo and ex vivo using isolated immune cells from in situ vaccinated mice. RESULTS Cancer cells undergoing PDT-induced cell death significantly increased ICD markers and the infiltration of iDCs in spheroids, relying on ferroptosis. These actions induced the sequential activation of CD8+ and CD4+ T cells as revealed by a significant spheroid 3D structure deterioration and, remarkably, were not recapitulated by conventional ferroptosis inducer RSL3. Using LED light from an endoscope for in situ photoactivation of PS enabled us to apply the vaccination modality in mice with peritoneal tumors. Consecutive intraperitoneal injection of iDCs resulted in delayed tumor growth, increased survival rates, and prevented tumor relapse on rechallenge. CD8+ T cell response was supported by depletion experiments, nodal detection of early activated T cells, and ex vivo T cell-induced cytotoxicity toward spheroids. CONCLUSIONS The combination of in situ PDT locally delivered by an endoscope light and iDC administration induces a durable memory immune response against peritoneal carcinomatosis thereby opening new perspectives for the treatment of a life-threatening condition.
Collapse
Affiliation(s)
- Charline Degavre
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Anouk Lepez
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Sebastien Ibanez
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Clémence François
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Katarzyna Głowacka
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Céline Guilbaud
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Florine Laloux-Morris
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
| | - Hrag Esfahani
- IPHY Platform, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Davide Brusa
- CytoFlux-Flow Cytometry and Cell Sorting Platform, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Caroline Bouzin
- Imaging Platform 2IP, Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Olivier Feron
- Institut de Recherche Expérimentale et Clinique (IREC), Pole of Pharmacology and Therapeutics (FATH), Cancer Translational Research laboratory, UCLouvain, Brussels, Belgium
- Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), WEL Research Institute, Wavre, Belgium
| |
Collapse
|
17
|
Kizil B, De Virgiliis F, Scheiermann C. Neural control of tumor immunity. FEBS J 2024; 291:4670-4679. [PMID: 39304984 DOI: 10.1111/febs.17280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/02/2024] [Accepted: 09/09/2024] [Indexed: 11/02/2024]
Abstract
Communication between the nervous system and the immune system has evolved to optimally respond to potentially dangerous stimuli both from within and outside the body. Tumors pose a severe threat to an organism and current therapies are insufficient for tumor regression in the majority of cases. Studies show that tumors are innervated by peripheral nerves from the sensory, parasympathetic and sympathetic nervous systems. Interactions between cancer cells, nerves and immune cells regulate overall tumor progression. Clinical studies have indicated the potential of targeting the peripheral nervous system for promoting anti-tumor immune responses. This view point provides an opinion on the current evidence and therapeutic potential of manipulating neuro-immune communications in cancer.
Collapse
Affiliation(s)
- Burak Kizil
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - Francesco De Virgiliis
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
| | - Christoph Scheiermann
- Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Switzerland
- Geneva Center for Inflammation Research (GCIR), Geneva, Switzerland
- Translational Research Centre in Onco-Hematology (CRTOH), Geneva, Switzerland
- Institute of Genetics and Genomics of Geneva (iGE3), Switzerland
- Biomedical Center, Institute for Cardiovascular Physiology and Pathophysiology, Walter Brendel-Center for Experimental Medicine, Faculty of Medicine, Ludwig-Maximilians-Universität Munich, Planegg-Martinsried, Germany
| |
Collapse
|
18
|
Wang C, Zhang M, Li S, Gong M, Luo MY, Zhang MC, Zou JH, Shen N, Xu L, Lei HM, Bi L, Zhu L, Wang Z, Chen HZ, Zhou L, Shen Y. A phosphoglycerate mutase 1 allosteric inhibitor restrains TAM-mediated colon cancer progression. Acta Pharm Sin B 2024; 14:4819-4831. [PMID: 39664444 PMCID: PMC11628787 DOI: 10.1016/j.apsb.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/27/2024] [Accepted: 07/26/2024] [Indexed: 12/13/2024] Open
Abstract
Colorectal cancer (CRC) is a prevalent malignant tumor often leading to liver metastasis and mortality. Despite some success with PD-1/PD-L1 immunotherapy, the response rate for colon cancer patients remains relatively low. This is closely related to the immunosuppressive tumor microenvironment mediated by tumor-associated macrophages (TAMs). Our previous work identified that a phosphoglycerate mutase 1 (PGAM1) allosteric inhibitor, HKB99, exerts a range of anti-tumor activities in lung cancer. Here, we found that upregulation of PGAM1 correlates with increased levels of M2-like tumor-associated macrophages (TAMs) in human colon cancer samples, particularly in liver metastatic tissues. HKB99 suppressed tumor growth and metastasis in cell culture and syngeneic tumor models. M2-polarization, induced by colon cancer cell co-culture, was reversed by HKB99. Conversely, the increased migration of colon cancer cells by M2-TAMs was remarkably restrained by HKB99. Notably, a decrease in TAM infiltration was required for the HKB99-mediated anti-tumor effect, along with an increase in CD8+ T cell infiltration. Moreover, HKB99 improved the efficacy of anti-PD-1 treatment in syngeneic tumors. Overall, this study highlights HKB99's inhibitory activity in TAM-mediated colon cancer progression. Targeting PGAM1 could lead to novel therapeutic strategies and enhance the effectiveness of existing immunotherapies for colon cancer.
Collapse
Affiliation(s)
- Cheng Wang
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai 200025, China
| | - Minghao Zhang
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai 200025, China
| | - Shunyao Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Miaomiao Gong
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai 200025, China
| | - Ming-yu Luo
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai 200025, China
| | - Mo-cong Zhang
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai 200025, China
| | - Jing-Hua Zou
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai 200025, China
| | - Ningxiang Shen
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai 200025, China
| | - Lu Xu
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai 200025, China
| | - Hui-min Lei
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai 200025, China
| | - Ling Bi
- Department of Medical Oncology & Cancer Institute of Integrative Medicine, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai 200025, China
| | - Zhengting Wang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Hong-zhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Lu Zhou
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ying Shen
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
- Collaborative Innovation Center for Clinical and Translational Science by Chinese Ministry of Education & Shanghai, Shanghai 200025, China
| |
Collapse
|
19
|
Qi Q, Pang J, Chen Y, Tang Y, Wang H, Gul S, Sun Y, Tang W, Sheng M. Targeted Drug Screening Leveraging Senescence-Induced T-Cell Exhaustion Signatures in Hepatocellular Carcinoma. Int J Mol Sci 2024; 25:11232. [PMID: 39457014 PMCID: PMC11508728 DOI: 10.3390/ijms252011232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most prevalent cancer and a leading cause of cancer-related mortality globally, with most patients diagnosed at advanced stages and facing limited early treatment options. This study aimed to identify characteristic genes associated with T-cell exhaustion due to senescence in hepatocellular carcinoma patients, elucidating the interplay between senescence and T-cell exhaustion. We constructed prognostic models based on five signature genes (ENO1, STMN1, PRDX1, RAN, and RANBP1) linked to T-cell exhaustion, utilizing elastic net regression. The findings indicate that increased expression of ENO1 in T cells may contribute to T-cell exhaustion and Treg infiltration in hepatocellular carcinoma. Furthermore, molecular docking was employed to screen small molecule compounds that target the anti-tumor effects of these exhaustion-related genes. This study provides crucial insights into the diagnosis and treatment of hepatocellular carcinoma, establishing a strong foundation for the development of predictive biomarkers and therapeutic targets for affected patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Wenru Tang
- Laboratory of Molecular Genetics of Aging & Tumor, Medicine School, Kunming University of Science and Technology, Kunming 650500, China; (Q.Q.); (J.P.); (Y.C.); (Y.T.); (H.W.); (S.G.); (Y.S.)
| | - Miaomiao Sheng
- Laboratory of Molecular Genetics of Aging & Tumor, Medicine School, Kunming University of Science and Technology, Kunming 650500, China; (Q.Q.); (J.P.); (Y.C.); (Y.T.); (H.W.); (S.G.); (Y.S.)
| |
Collapse
|
20
|
Ren C, Zeng Y, Qiu L, Luo D, Wang J, Chen X, Yan Y. Study on the effects of intraoperative administration of dexmedetomidine on the prognosis and survival outcomes of patients with colorectal cancer. Heliyon 2024; 10:e38241. [PMID: 39381090 PMCID: PMC11456851 DOI: 10.1016/j.heliyon.2024.e38241] [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: 06/04/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024] Open
Abstract
Background The perioperative period of tumor surgery commonly utilizes dexmedetomidine as an adjuvant analgesic for anesthesia. Nevertheless, there is a paucity of research investigating its influence on the prognosis of colorectal cancer (CRC). This article primarily aims to examine the correlation between the intraoperative administration of dexmedetomidine and recurrence-free survival (RFS) and overall survival (OS) of colorectal cancer patients, as well as its prognostic implications on survival. Methods According to the exclusion criteria, 76 patients undergoing laparoscopic radical resection of CRC under general anesthesia were enrolled at Huizhou Central People's Hospital in 2014. Kaplan-Meier method was used for univariate survival analysis of clinical prognostic factors, RFS, and OS in patients with CRC Cox regression analysis was used for multivariate survival analysis. Results A total of 76 patients with CRC were enrolled in this study. Among them, 36 patients were treated with dexmedetomidine (group D), and 40 patients were not treated with dexmedetomidine (group C) during the operation. Survival analysis showed that the RFS and OS of patients in group D were significantly higher than those in group C (P = 0.046 and P = 0.021, respectively). Multivariate regression analysis demonstrated that the intraoperative administration of dexmedetomidine independently predicted a protective effect on OS (P = 0.025). Conclusions The intraoperative application of dexmedetomidine as an adjuvant analgesic has a protective effect on the prognosis and survival of patients with CRC and can improve the overall survival rate. Additionally, it influences the recurrence status of patients to a certain extent. These results suggest that dexmedetomidine significantly benefits on the long-term prognosis of patients with CRC.
Collapse
Affiliation(s)
- Chu Ren
- Shantou University Medical College, Shantou, 515041, Guangdong Province, China
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou 516001, Guangdong Province, China
| | - Ying Zeng
- Department of Pathophysiology, Guangdong Medical University, Dongguan 523808, Guangdong Province, China
| | - Liuji Qiu
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou 516001, Guangdong Province, China
| | - Dexing Luo
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou 516001, Guangdong Province, China
| | - Junfang Wang
- Central Laboratory, Medical College of Jiaying University, Meizhou, 514031, Guangdong Province, China
| | - Xin Chen
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou 516001, Guangdong Province, China
| | - Yan Yan
- Shantou University Medical College, Shantou, 515041, Guangdong Province, China
- Department of Anesthesiology, Huizhou Central People's Hospital, Huizhou 516001, Guangdong Province, China
| |
Collapse
|
21
|
Yaniv D, Mattson B, Talbot S, Gleber-Netto FO, Amit M. Targeting the peripheral neural-tumour microenvironment for cancer therapy. Nat Rev Drug Discov 2024; 23:780-796. [PMID: 39242781 DOI: 10.1038/s41573-024-01017-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2024] [Indexed: 09/09/2024]
Abstract
As the field of cancer neuroscience expands, the strategic targeting of interactions between neurons, cancer cells and other elements in the tumour microenvironment represents a potential paradigm shift in cancer treatment, comparable to the advent of our current understanding of tumour immunology. Cancer cells actively release growth factors that stimulate tumour neo-neurogenesis, and accumulating evidence indicates that tumour neo-innervation propels tumour progression, inhibits tumour-related pro-inflammatory cytokines, promotes neovascularization, facilitates metastasis and regulates immune exhaustion and evasion. In this Review, we give an up-to-date overview of the dynamics of the tumour microenvironment with an emphasis on tumour innervation by the peripheral nervous system, as well as current preclinical and clinical evidence of the benefits of targeting the nervous system in cancer, laying a scientific foundation for further clinical trials. Combining empirical data with a biomarker-driven approach to identify and hone neuronal targets implicated in cancer and its spread can pave the way for swift clinical integration.
Collapse
Affiliation(s)
- Dan Yaniv
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brandi Mattson
- The Neurodegeneration Consortium, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sebastien Talbot
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Frederico O Gleber-Netto
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Moran Amit
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- UTHealth Houston Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
22
|
Xie CK, Liao CY, Lin HY, Wu YD, Lu FC, Huang XX, Wang ZW, Li G, Lin CF, Hu JF, Chen YH, Li QW, Chen LQ, Chen HX, Chen S. Sulindac (K-80003) with nab-paclitaxel and gemcitabine overcomes drug-resistant pancreatic cancer. Mol Cancer 2024; 23:215. [PMID: 39350121 PMCID: PMC11441089 DOI: 10.1186/s12943-024-02128-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
The Nab-paclitaxel combined with gemcitabine (AG) regimen is the main chemotherapy regimen for pancreatic cancer, but drug resistance often occurs. Currently, the ability to promote sensitization in drug-resistant cases is an important clinical issue, and the strategy of repurposing conventional drugs is a promising strategy. This study aimed to identify a classic drug that targets chemotherapy resistance's core signaling pathways and combine it with the AG regimen to enhance chemosensitivity. We also aimed to find reliable predictive biomarkers of drug combination sensitivity. Using RNA sequencing, we found that abnormal PI3K/Akt pathway activation plays a central role in mediating resistance to the AG regimen. Subsequently, through internal and external verification of randomly selected AG-resistant patient-derived organoid (PDO) and PDO xenograft models, we discovered for the first time that the classic anti-inflammatory drug sulindac K-80003, an inhibitor of the PI3K/Akt pathway that we focused on, promoted sensitization in half (14/28) of AG-resistant pancreatic ductal adenocarcinoma cases. Through RNA-sequencing, multiplex immunofluorescent staining, and immunohistochemistry experiments, we identified cFAM124A as a novel biomarker through which sulindac K-80003 promotes AG sensitization. Its role as a sensitization marker is explained via the following mechanism: cFAM124A enhances both the mRNA expression of cathepsin L and the activity of the cathepsin L enzyme. This dual effect stimulates the cleavage of RXRα, leading to large amounts of truncated RXRα, which serves as a direct target of K-80003. Consequently, this process results in the pathological activation of the PI3K/Akt pathway. In summary, our study provides a new treatment strategy and novel biological target for patients with drug-resistant pancreatic cancer.
Collapse
Affiliation(s)
- Cheng-Ke Xie
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Cheng-Yu Liao
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350001, China
- Fuzhou University, Fuzhou, 350001, China
| | - Hong-Yi Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Yong-Ding Wu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Feng-Chun Lu
- Department of General Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Xiao-Xiao Huang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350001, China
- Fuzhou University, Fuzhou, 350001, China
| | - Zu-Wei Wang
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350001, China
- Fuzhou University, Fuzhou, 350001, China
| | - Ge Li
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Cai-Feng Lin
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350001, China
- Fuzhou University, Fuzhou, 350001, China
| | - Jian-Fei Hu
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Yin-Hao Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Qiao-Wei Li
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China
- Fujian Provincial Center for Geriatrics, Fuzhou, 350001, China
- Fujian Key Laboratory of Geriatrics, Fuzhou, 350001, China
| | - Li-Qun Chen
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350108, China.
- Institute of Applied Genomics, Fuzhou University, Fuzhou, 350108, China.
| | - Hui-Xing Chen
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
- Fujian Medical University Cancer Center, Fuzhou, 350001, China.
| | - Shi Chen
- Shengli Clinical Medical College of Fujian Medical University, Fuzhou, 350001, China.
- Department of Hepatobiliary Pancreatic Surgery, Fuzhou University Affiliated Provincial Hospital, Fujian Provincial Hospital, Fuzhou, 350001, China.
- Fuzhou University, Fuzhou, 350001, China.
- Fujian Provincial Center for Geriatrics, Fuzhou, 350001, China.
- Fujian Key Laboratory of Geriatrics, Fuzhou, 350001, China.
| |
Collapse
|
23
|
Manoleras AV, Sloan EK, Chang A. The sympathetic nervous system shapes the tumor microenvironment to impair chemotherapy response. Front Oncol 2024; 14:1460493. [PMID: 39381049 PMCID: PMC11458372 DOI: 10.3389/fonc.2024.1460493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 08/30/2024] [Indexed: 10/10/2024] Open
Abstract
The tumor microenvironment influences cancer progression and response to treatments, which ultimately impacts the survival of patients with cancer. The sympathetic nervous system (SNS) is a core component of solid tumors that arise in the body. In addition to influencing cancer progression, a role for the SNS in the effectiveness of cancer treatments is beginning to emerge. This review explores evidence that the SNS impairs chemotherapy efficacy. We review findings of studies that evaluated the impact of neural ablation on chemotherapy outcomes and discuss plausible mechanisms for the impact of neural signaling on chemotherapy efficacy. We then discuss implications for clinical practice, including opportunities to block neural signaling to improve response to chemotherapy.
Collapse
Affiliation(s)
| | | | - Aeson Chang
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| |
Collapse
|
24
|
Viswanath V, Mistry S, Cabrera-Ghayouri S, Leang R, Frail D, Donello J, Gil D. Sustained Alleviation of Autoimmunity by Activating α2B-adrenergic Receptors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:435-441. [PMID: 38940628 DOI: 10.4049/jimmunol.2300893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 06/10/2024] [Indexed: 06/29/2024]
Abstract
Catecholamines binding to α- and β-adrenergic receptors on immune cells have recently been shown to play an important role in regulating immune responses. Although α2-adrenergic receptors are known to modulate the immune response in different ways, the therapeutic exploration of their utility has been limited by the lack of agonists selective for the three α2-adrenergic subtypes. We report in this study the identification of the agonist AGN-762, which activates α2B- and α2C-adrenergic subtypes, but not the α2A subtype. We show that AGN-762 reduced clinical disease in an experimental autoimmune encephalitis model of autoimmune disease via direct or indirect effects on T regulatory cells. The activity of AGN-762 was abrogated by depletion of T regulatory cells, which express the α2B-adrenergic receptor. Furthermore, a drug-induced shift to an anti-inflammatory phenotype was demonstrated in immune cells in the spleen of drug-treated experimental autoimmune encephalitis mice. AGN-762 does not display sedative and cardiovascular side effects associated with α2A subtype agonists. Immune modulation by selective α2-adrenergic agonists represents a novel, to our knowledge, approach for treating autoimmune disease.
Collapse
MESH Headings
- Animals
- Mice
- Receptors, Adrenergic, alpha-2/metabolism
- Receptors, Adrenergic, alpha-2/immunology
- Autoimmunity/immunology
- Adrenergic alpha-2 Receptor Agonists/pharmacology
- Adrenergic alpha-2 Receptor Agonists/therapeutic use
- Mice, Inbred C57BL
- T-Lymphocytes, Regulatory/immunology
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Humans
- Female
- Disease Models, Animal
Collapse
Affiliation(s)
| | - Shruti Mistry
- Ophthalmology Discovery Research, AbbVie Inc., Irvine, CA
| | | | - Ronika Leang
- Ophthalmology Discovery Research, AbbVie Inc., Irvine, CA
| | - Don Frail
- Alceptor Therapeutics, Newport Beach, CA
| | | | - Daniel Gil
- Alceptor Therapeutics, Newport Beach, CA
| |
Collapse
|
25
|
Dong J, Che J, Wu Y, Deng Y, Jiang X, He Z, Zhang J. Dexmedetomidine promotes colorectal cancer progression via Piwil2 signaling. Cell Oncol (Dordr) 2024; 47:1459-1474. [PMID: 38592610 DOI: 10.1007/s13402-024-00944-8] [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: 03/27/2024] [Indexed: 04/10/2024] Open
Abstract
PURPOSE α2-adrenoceptor agonist dexmedetomidine (DEX) has been reported to promote tumorigenesis. Stem-cell protein Piwil2 is associated with cancer progression. Whether Piwil2 plays a role in tumor-promoting effects of DEX is unknown. METHODS We examined the expression of Piwil2 in human colorectal cancer (CRC) cell lines with/without DEX treatment. We also studied the roles of Piwil2 in proliferation, invasion, migration, as well as expressions of epithelial-mesenchymal transition (EMT)-related proteins in DEX-treated in vitro and in vivo CRC models. And the experiments with genetic and pharmacological treatments were conducted to investigate the underlying molecular mechanism. RESULTS RNA-sequencing (RNA-seq) analysis found Piwil2 is one of most upregulated genes upon DEX treatment in CRC cells. Furthermore, Piwil2 protein levels significantly increased in DEX-treated CRC cancer cells, which promoted proliferation, invasion, and migration in both CRC cell lines and human tumor xenografts model. Mechanistically, DEX increased nuclear factor E2-related factor 2 (Nrf2) expression, which enhanced Piwil2 transcription via binding to its promoter. Furthermore, in vitro experiments with Piwil2 knockdown or Siah2 inhibition indicated that DEX promoted EMT process and tumorigenesis through Siah2/PHD3/HIF1α pathway. The experiments with another α2-adrenoceptor agonist Brimonidine and antagonists yohimbine and atipamezole also suggested the role of Piwil2 signaling in tumor-promoting effects via an α2 adrenoceptor-dependent manner. CONCLUSION DEX promotes CRC progression may via activating α2 adrenoceptor-dependent Nrf2/Piwil2/Siah2 pathway and thus EMT process. Our work provides a novel insight into the mechanism underlying tumor-promoting effects of α2-adrenoceptor agonists.
Collapse
Affiliation(s)
- Jing Dong
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Ji Che
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yuanyuan Wu
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Yixu Deng
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Xuliang Jiang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Zhiyong He
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China
| | - Jun Zhang
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China.
- Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong'An Road, Xuhui District, Shanghai, 200032, P. R. China.
| |
Collapse
|
26
|
Xu X, Lu Y, Cao L, Miao Y, Li Y, Cui Y, Han T. Tumor-intrinsic P2RY6 drives immunosuppression by enhancing PGE 2 production. Cell Rep 2024; 43:114469. [PMID: 38996067 DOI: 10.1016/j.celrep.2024.114469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 05/21/2024] [Accepted: 06/24/2024] [Indexed: 07/14/2024] Open
Abstract
Despite the success of anti-programmed cell death-1 (anti-PD-1) immunotherapy, many cancer patients remain unresponsive, and reliable predictive biomarkers are lacking. Here, we show that aberrant expression of the pyrimidinergic receptor P2RY6 is frequent in human cancers and causes immune evasion. In mouse syngeneic and human xenograft tumor models, ectopic expression of P2RY6 shapes an immunosuppressive tumor microenvironment (TME) to enhance tumor growth and resistance to immunotherapy, whereas deletion of P2RY6 from tumors with high P2RY6 expression inflames the TME to inhibit tumor growth. As a G protein-coupled receptor, P2RY6 activates Gq/phospholipase C-β signaling and stimulates the synthesis of prostaglandin E2, which is a key mediator of immunosuppression in the TME. In contrast to the essential role of P2RY6 in tumors, global deletion of P2ry6 from mice does not compromise viability. Our study thus nominates P2RY6 as a precision immunotherapy target for patients with high tumor-intrinsic P2RY6 expression.
Collapse
Affiliation(s)
- Xilong Xu
- College of Life Sciences, Beijing Normal University, Beijing 100875, China; National Institute of Biological Sciences, Beijing 102206, China
| | - Yi Lu
- National Institute of Biological Sciences, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China
| | - Longzhi Cao
- National Institute of Biological Sciences, Beijing 102206, China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yang Miao
- National Institute of Biological Sciences, Beijing 102206, China; PTN Joint Graduate Program, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yamei Li
- National Institute of Biological Sciences, Beijing 102206, China; Graduate School of Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yue Cui
- College of Life Sciences, Beijing Normal University, Beijing 100875, China; National Institute of Biological Sciences, Beijing 102206, China
| | - Ting Han
- College of Life Sciences, Beijing Normal University, Beijing 100875, China; National Institute of Biological Sciences, Beijing 102206, China; Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 102206, China.
| |
Collapse
|
27
|
Wang L, Ge J, Han H, Jia Y, Qin Y. Crosstalk between the nervous system and tumor microenvironment: Functional aspects and potential therapeutic strategies. Cancer Lett 2024; 594:216986. [PMID: 38797233 DOI: 10.1016/j.canlet.2024.216986] [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: 03/15/2024] [Revised: 05/19/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Recent advancements in understanding the tumor microenvironment (TME) have highlighted the critical role of the nervous system in cancer progression. This review comprehensively examines how the nervous system influences various aspects of tumorigenesis, including growth, motility, immune response, angiogenesis, and the behavior of cancer-associated fibroblasts (CAFs). We delineate the neurodevelopmental mechanisms associated with cancer, such as the secretion of neurotrophins and exosomes by cancer cells. Furthermore, we explore the emerging therapeutic strategy of targeting nerves associated with tumors. Evidence supporting this approach includes studies demonstrating direct tumor growth inhibition, enhanced efficacy of immunotherapy when combined with nervous system-modulating drugs, and the suppression of tumor blood vessel formation through nerve targeting. Finally, we discuss the current challenges in this field and emphasize the need for further exploration within cancer neuroscience.
Collapse
Affiliation(s)
- Lei Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Zhengzhou, 450052, PR China
| | - Jingjing Ge
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Sun Yat-Sen University Cancer Center, No. 651, Dongfeng Road East, Guangzhou, 510060, PR China
| | - Huiqiong Han
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Zhengzhou, 450052, PR China
| | - Yongxu Jia
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Zhengzhou, 450052, PR China
| | - Yanru Qin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, No.1, Jianshe East Road, Zhengzhou, 450052, PR China.
| |
Collapse
|
28
|
Yang Y, Sun L, Liu X, Liu W, Zhang Z, Zhou X, Zhao X, Zheng R, Zhang Y, Guo W, Wang X, Li X, Pang J, Li F, Tao Y, Shi D, Shen W, Wang L, Zang J, Li S. Neurotransmitters: Impressive regulators of tumor progression. Biomed Pharmacother 2024; 176:116844. [PMID: 38823279 DOI: 10.1016/j.biopha.2024.116844] [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: 03/20/2024] [Revised: 05/22/2024] [Accepted: 05/26/2024] [Indexed: 06/03/2024] Open
Abstract
In contemporary times, tumors have emerged as the primary cause of mortality in the global population. Ongoing research has shed light on the significance of neurotransmitters in the regulation of tumors. It has been established that neurotransmitters play a pivotal role in tumor cell angiogenesis by triggering the transformation of stromal cells into tumor cells, modulating receptors on tumor stem cells, and even inducing immunosuppression. These actions ultimately foster the proliferation and metastasis of tumor cells. Several major neurotransmitters have been found to exert modulatory effects on tumor cells, including the ability to restrict emergency hematopoiesis and bind to receptors on the postsynaptic membrane, thereby inhibiting malignant progression. The abnormal secretion of neurotransmitters is closely associated with tumor progression, suggesting that focusing on neurotransmitters may yield unexpected breakthroughs in tumor therapy. This article presents an analysis and outlook on the potential of targeting neurotransmitters in tumor therapy.
Collapse
Affiliation(s)
- Yumei Yang
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Lei Sun
- Department of Critical Care Medicine, The First Hospital of Harbin, No 151, Diduan Street, Daoli District, Harbin, China
| | - Xuerou Liu
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Wei Liu
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Zhen Zhang
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Xingqi Zhou
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Xinli Zhao
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Ruijie Zheng
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Yongjun Zhang
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Wanqing Guo
- School of Pharmacy, Bengbu Medical University, Bengbu, China
| | - Xiaoli Wang
- College of Pharmacy, Anhui University of Traditional Chinese Medicine, China
| | - Xian Li
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Jinlong Pang
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Feng Li
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Yu Tao
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China
| | - Dongmin Shi
- Department of Day Surgery Ward, The First Hospital of Harbin, No 151, Diduan Street, Daoli District, Harbin, China
| | - Wenyi Shen
- Department of Respiratory and Critical Care Medicine, Lianshui County People's Hospital, Jiangsu, China
| | - Liping Wang
- Department of Day Surgery Ward, The First Hospital of Harbin, No 151, Diduan Street, Daoli District, Harbin, China
| | - Jialan Zang
- Department of Day Surgery Ward, The First Hospital of Harbin, No 151, Diduan Street, Daoli District, Harbin, China.
| | - Shanshan Li
- School of Pharmacy, Bengbu Medical University, Bengbu, China; Anhui Province Engineering Technology Research Center of Biochemical Pharmaceutical, Bengbu, China.
| |
Collapse
|
29
|
Mandal SK, Yadav P, Sheth RA. The Neuroimmune Axis and Its Therapeutic Potential for Primary Liver Cancer. Int J Mol Sci 2024; 25:6237. [PMID: 38892423 PMCID: PMC11172507 DOI: 10.3390/ijms25116237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
The autonomic nervous system plays an integral role in motion and sensation as well as the physiologic function of visceral organs. The nervous system additionally plays a key role in primary liver diseases. Until recently, however, the impact of nerves on cancer development, progression, and metastasis has been unappreciated. This review highlights recent advances in understanding neuroanatomical networks within solid organs and their mechanistic influence on organ function, specifically in the liver and liver cancer. We discuss the interaction between the autonomic nervous system, including sympathetic and parasympathetic nerves, and the liver. We also examine how sympathetic innervation affects metabolic functions and diseases like nonalcoholic fatty liver disease (NAFLD). We also delve into the neurobiology of the liver, the interplay between cancer and nerves, and the neural regulation of the immune response. We emphasize the influence of the neuroimmune axis in cancer progression and the potential of targeted interventions like neurolysis to improve cancer treatment outcomes, especially for hepatocellular carcinoma (HCC).
Collapse
Affiliation(s)
| | | | - Rahul A. Sheth
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1471, Houston, TX 77030-4009, USA; (S.K.M.); (P.Y.)
| |
Collapse
|
30
|
Shu LZ, Ding YD, Zhang JY, He RS, Xiao L, Pan BX, Deng H. Interactions between MDSCs and the Autonomic Nervous System: Opportunities and Challenges in Cancer Neuroscience. Cancer Immunol Res 2024; 12:652-662. [PMID: 38568775 DOI: 10.1158/2326-6066.cir-23-0976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/11/2024] [Accepted: 03/19/2024] [Indexed: 04/05/2024]
Abstract
Myeloid-derived suppressor cells (MDSC) are a population of heterogeneous immune cells that are involved in precancerous conditions and neoplasms. The autonomic nervous system (ANS), which is composed of the sympathetic nervous system and the parasympathetic nervous system, is an important component of the tumor microenvironment that responds to changes in the internal and external environment mainly through adrenergic and cholinergic signaling. An abnormal increase of autonomic nerve density has been associated with cancer progression. As we discuss in this review, growing evidence indicates that sympathetic and parasympathetic signals directly affect the expansion, mobilization, and redistribution of MDSCs. Dysregulated autonomic signaling recruits MDSCs to form an immunosuppressive microenvironment in chronically inflamed tissues, resulting in abnormal proliferation and differentiation of adult stem cells. The two components of the ANS may also be responsible for the seemingly contradictory behaviors of MDSCs. Elucidating the underlying mechanisms has the potential to provide more insights into the complex roles of MDSCs in tumor development and lay the foundation for the development of novel MDSC-targeted anticancer strategies.
Collapse
Affiliation(s)
- Lin-Zhen Shu
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Rehabiliation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Tumor Immunology Institute, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Yi-Dan Ding
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Rehabiliation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Tumor Immunology Institute, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Jin-Yao Zhang
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Rui-Shan He
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Rehabiliation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Tumor Immunology Institute, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Li Xiao
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Rehabiliation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Tumor Immunology Institute, Nanchang University, Nanchang, Jiangxi, China
- Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Huan Deng
- The Fourth Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Rehabiliation Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- Tumor Immunology Institute, Nanchang University, Nanchang, Jiangxi, China
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
31
|
Zhang H, Yang Y, Cao Y, Guan J. Effects of chronic stress on cancer development and the therapeutic prospects of adrenergic signaling regulation. Biomed Pharmacother 2024; 175:116609. [PMID: 38678960 DOI: 10.1016/j.biopha.2024.116609] [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: 01/19/2024] [Revised: 04/14/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024] Open
Abstract
Long-term chronic stress is an important factor in the poor prognosis of cancer patients. Chronic stress reduces the tissue infiltration of immune cells in the tumor microenvironment (TME) by continuously activating the adrenergic signaling, inhibits antitumor immune response and tumor cell apoptosis while also inducing epithelial-mesenchymal transition (EMT) and tumor angiogenesis, promoting tumor invasion and metastasis. This review first summarizes how adrenergic signaling activates intracellular signaling by binding different adrenergic receptor (AR) heterodimers. Then, we focused on reviewing adrenergic signaling to regulate multiple functions of immune cells, including cell differentiation, migration, and cytokine secretion. In addition, the article discusses the mechanisms by which adrenergic signaling exerts pro-tumorigenic effects by acting directly on the tumor itself. It also highlights the use of adrenergic receptor modulators in cancer therapy, with particular emphasis on their potential role in immunotherapy. Finally, the article reviews the beneficial effects of stress intervention measures on cancer treatment. We think that enhancing the body's antitumor response by adjusting adrenergic signaling can enhance the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Oncology, The Eighth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100091, China; Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| | - Yuwei Yang
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Yan Cao
- College of Pulmonary & Critical Care Medicine, Chinese PLA General Hospital, Beijing Key Laboratory of OTIR, Beijing, 100091, China.
| | - Jingzhi Guan
- Department of Oncology, The Fifth Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing 100071, China.
| |
Collapse
|
32
|
Li Y, Gong B, Guo Y, Liu W, Chen C, Jiang S, Pan F, Song J, Yang L, Zhou G. Non-small cell lung cancer and immune checkpoint inhibitor therapy: does non-alcoholic fatty liver disease have an effect? BMC Cancer 2024; 24:535. [PMID: 38671373 PMCID: PMC11055283 DOI: 10.1186/s12885-024-12295-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Immunotherapy based on the application of immune checkpoint inhibitors (ICIs) is one of the standard treatments for advanced non-small cell lung cancer (NSCLC). Non-alcoholic fatty liver Disease (NAFLD) has demonstrated predictive value for response to immunotherapy in non-lung cancer types. Our study investigated the effect of NAFLD on the efficacy of real-life use of ICIs for patients with stage III / IV NSCLC. METHODS The clinical and imaging data of patients with stage III / IV NSCLC who were first admitted to Union Hospital, Tongji Medical College, Huazhong University of Science and Technology from March 2020 to July 2022 were retrospectively collected to ensure that they underwent at least one CT scan before treatment. A total of 479 patients were divided into the NAFLD group (Liver/Spleen density ratio ≤ 1) and the non-NAFLD group (Liver/Spleen density ratio > 1) by measuring the baseline liver and spleen CT value. The overall survival (OS), progression-free survival (PFS), objective response rate (ORR) and disease control rate (DCR) of the patients were obtained. RESULTS A total of 118 patients with NAFLD and 361 patients without NAFLD were included in the study. Patients with NAFLD tended to have higher BMI and higher total bilirubin compared to patients without NAFLD. The median duration of follow-up in the study was 22 m (IQR, 17-29 m). Both of 2 groups had a higher DCR (94% vs. 92%, p = 0.199) and ORR (38.1% vs. 44.9%, p = 0.452) respectively. There was no difference in efficacy between the two groups. In univariate analysis, NAFLD had no significant effect on PFS (p = 0.785) and OS (p = 0.851). Surprisingly, the presence of hypertension was observed to be associated with a higher OS (HR 1.471 95%CI 1.018-2.127, p = 0.040). Besides, based on multivariate analysis, lactic dehydrogenase was associated with PFS (HR 1.001 95%CI 1.000,1.002, p = 0.037) and OS (HR 1.002, 95%CI 1.001-1.003, p < 0.001). CONCLUSIONS Among patients with NSCLC, NAFLD did not result in changes in survival or disease progression after immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Yi Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Bingxin Gong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Weiwei Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chao Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Shanshan Jiang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Feng Pan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jiyu Song
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Guofeng Zhou
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
33
|
Miao SN, Chai MQ, Liu XY, Wei CY, Zhang CC, Sun NN, Fei QZ, Peng LL, Qiu H. Exercise accelerates recruitment of CD8 + T cell to promotes anti-tumor immunity in lung cancer via epinephrine. BMC Cancer 2024; 24:474. [PMID: 38622609 PMCID: PMC11021002 DOI: 10.1186/s12885-024-12224-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 04/02/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND AND PURPOSE In recent years, there has been extensive research on the role of exercise as an adjunctive therapy for cancer. However, the potential mechanisms underlying the anti-tumor therapy of exercise in lung cancer remain to be fully elucidated. As such, our study aims to confirm whether exercise-induced elevation of epinephrine can accelerate CD8+ T cell recruitment through modulation of chemokines and thus ultimately inhibit tumor progression. METHOD C57BL/6 mice were subcutaneously inoculated with Lewis lung cancer cells (LLCs) to establish a subcutaneous tumor model. The tumor mice were randomly divided into different groups to performed a moderate-intensity exercise program on a treadmill for 5 consecutive days a week, 45 min a day. The blood samples and tumor tissues were collected after exercise for IHC, RT-qPCR, ELISA and Western blot. In addition, another group of mice received daily epinephrine treatment for two weeks (0.05 mg/mL, 200 µL i.p.) (EPI, n = 8) to replicate the effects of exercise on tumors in vivo. Lewis lung cancer cells were treated with different concentrations of epinephrine (0, 5, 10, 20 µM) to detect the effect of epinephrine on chemokine levels via ELISA and RT-qPCR. RESULTS This study reveals that both pre- and post-cancer exercise effectively impede the tumor progression. Exercise led to an increase in EPI levels and the infiltration of CD8+ T cell into the lung tumor. Exercise-induced elevation of EPI is involved in the regulation of Ccl5 and Cxcl10 levels further leading to enhanced CD8+ T cell infiltration and ultimately inhibiting tumor progression. CONCLUSION Exercise training enhance the anti-tumor immunity of lung cancer individuals. These findings will provide valuable insights for the future application of exercise therapy in clinical practice.
Collapse
Affiliation(s)
- Sai-Nan Miao
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Meng-Qi Chai
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Xiang-Yu Liu
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Cheng-Yu Wei
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Cun-Cun Zhang
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Ning-Ning Sun
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Qing-Ze Fei
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Lin-Lin Peng
- School of Nursing, Anhui Medical University, 230032, Hefei, China
| | - Huan Qiu
- School of Nursing, Anhui Medical University, 230032, Hefei, China.
| |
Collapse
|
34
|
Zhu LL, Wang YH, Zhou Q. Tizanidine: Advances in Pharmacology & Therapeutics and Drug Formulations. J Pain Res 2024; 17:1257-1271. [PMID: 38529017 PMCID: PMC10962466 DOI: 10.2147/jpr.s461032] [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: 01/23/2024] [Accepted: 03/16/2024] [Indexed: 03/27/2024] Open
Abstract
Background Skeletal muscle relaxants (SMRs) are widely used in treating musculoskeletal conditions. All SMRs, with the exception of baclofen and tizanidine, are on the list of 2023 American Geriatrics Society Beers Criteria® for potentially inappropriate medication use in older adults. In our geriatric practice, off-label use of tizanidine as preemptive analgesia drove us to find recent advances in its pharmacology and therapeutics. An update review of tizanidine was thus presented, aiming to bring the latest knowledge to clinicians and promote further research and practical exploration. Methods Relevant literature up to December 2023 was identified through searches of PubMed, Web of Science, and Embase. Results Tizanidine, a centrally acting alpha-2 adrenoceptor agonist with both antispastic and antispasmodic activity, shows efficacy in the common indications for all SMRs. From the perspective of drug safety, tizanidine has lower incidences of adverse events (injury, delirium, encephalopathy, falls, and opioid overdose) compared to baclofen, no association with risk of Alzheimer's disease as with orphenadrine, no risk of serotonin syndrome like metaxalone when comedicated with serotonergic drugs, no significant pharmacokinetic changes in CYP2C19 poor metabolizers unlike diazepam and carisoprodol, and no physically addictive or abuse properties like carisoprodol and diazepam. From the perspective of new and potential therapeutic uses, tizanidine has additional benefits (eg, gastroprotection that can improve patient tolerance to nonsteroidal anti-inflammatory agents, anti-neuropathic pain, a key component of multimodal analgesia strategy to reduce early postoperative pain, and anti-tumor effects). New delivery systems of tizanidine are developing to improve the pharmacokinetics of oral products, including buccal patches, transdermal delivery systems, nasal spray, and in situ rectal gel. Conclusion Tizanidine is an SMR with unique features and may be an optimal initial choice for older adults. There would be more scientific studies, wider therapeutic applications, and new drug formulations in the future.
Collapse
Affiliation(s)
- Ling-Ling Zhu
- VIP Geriatric Ward, Division of Nursing, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Yan-Hong Wang
- Department of Pharmacy, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People’s Republic of China
| | - Quan Zhou
- Department of Pharmacy, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, People’s Republic of China
| |
Collapse
|
35
|
Sprooten J, Vanmeerbeek I, Datsi A, Govaerts J, Naulaerts S, Laureano RS, Borràs DM, Calvet A, Malviya V, Kuballa M, Felsberg J, Sabel MC, Rapp M, Knobbe-Thomsen C, Liu P, Zhao L, Kepp O, Boon L, Tejpar S, Borst J, Kroemer G, Schlenner S, De Vleeschouwer S, Sorg RV, Garg AD. Lymph node and tumor-associated PD-L1 + macrophages antagonize dendritic cell vaccines by suppressing CD8 + T cells. Cell Rep Med 2024; 5:101377. [PMID: 38232703 PMCID: PMC10829875 DOI: 10.1016/j.xcrm.2023.101377] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 08/23/2023] [Accepted: 12/18/2023] [Indexed: 01/19/2024]
Abstract
Current immunotherapies provide limited benefits against T cell-depleted tumors, calling for therapeutic innovation. Using multi-omics integration of cancer patient data, we predict a type I interferon (IFN) responseHIGH state of dendritic cell (DC) vaccines, with efficacious clinical impact. However, preclinical DC vaccines recapitulating this state by combining immunogenic cancer cell death with induction of type I IFN responses fail to regress mouse tumors lacking T cell infiltrates. Here, in lymph nodes (LNs), instead of activating CD4+/CD8+ T cells, DCs stimulate immunosuppressive programmed death-ligand 1-positive (PD-L1+) LN-associated macrophages (LAMs). Moreover, DC vaccines also stimulate PD-L1+ tumor-associated macrophages (TAMs). This creates two anatomically distinct niches of PD-L1+ macrophages that suppress CD8+ T cells. Accordingly, a combination of PD-L1 blockade with DC vaccines achieves significant tumor regression by depleting PD-L1+ macrophages, suppressing myeloid inflammation, and de-inhibiting effector/stem-like memory T cells. Importantly, clinical DC vaccines also potentiate T cell-suppressive PD-L1+ TAMs in glioblastoma patients. We propose that a multimodal immunotherapy and vaccination regimen is mandatory to overcome T cell-depleted tumors.
Collapse
Affiliation(s)
- Jenny Sprooten
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Isaure Vanmeerbeek
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Angeliki Datsi
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Jannes Govaerts
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Stefan Naulaerts
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Raquel S Laureano
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Daniel M Borràs
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Anna Calvet
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Vanshika Malviya
- Department of Microbiology, Immunology and Transplantation, KU Leuven-University of Leuven, Leuven, Belgium
| | - Marc Kuballa
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Jörg Felsberg
- Department of Neurosurgery, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Michael C Sabel
- Department of Neurosurgery, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Marion Rapp
- Department of Neurosurgery, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Christiane Knobbe-Thomsen
- Department of Neurosurgery, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Peng Liu
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Liwei Zhao
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | - Oliver Kepp
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France
| | | | - Sabine Tejpar
- Laboratory for Molecular Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Jannie Borst
- Department of Immunology and Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Center, Université Paris Saclay, Villejuif, France; Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France; Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Susan Schlenner
- Department of Microbiology, Immunology and Transplantation, KU Leuven-University of Leuven, Leuven, Belgium
| | - Steven De Vleeschouwer
- Department of Neurosurgery, University Hospitals Leuven, Leuven, Belgium; Laboratory of Experimental Neurosurgery and Neuroanatomy, Department of Neurosciences, KU Leuven, Leuven, Belgium; Leuven Brain Institute (LBI), Leuven, Belgium
| | - Rüdiger V Sorg
- Institute for Transplantation Diagnostics and Cell Therapeutics, Medical Faculty, Heinrich Heine University Hospital, Düsseldorf, Germany
| | - Abhishek D Garg
- Laboratory of Cell Stress & Immunity, Department of Cellular & Molecular Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
36
|
Chen W, Liao Y, Sun P, Tu J, Zou Y, Fang J, Chen Z, Li H, Chen J, Peng Y, Wen L, Xie X. Construction of an ER stress-related prognostic signature for predicting prognosis and screening the effective anti-tumor drug in osteosarcoma. J Transl Med 2024; 22:66. [PMID: 38229155 PMCID: PMC10792867 DOI: 10.1186/s12967-023-04794-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 12/09/2023] [Indexed: 01/18/2024] Open
Abstract
BACKGROUND Osteosarcoma is the most common malignant primary bone tumor in infants and adolescents. The lack of understanding of the molecular mechanisms underlying osteosarcoma progression and metastasis has contributed to a plateau in the development of current therapies. Endoplasmic reticulum (ER) stress has emerged as a significant contributor to the malignant progression of tumors, but its potential regulatory mechanisms in osteosarcoma progression remain unknown. METHODS In this study, we collected RNA sequencing and clinical data of osteosarcoma from The TCGA, GSE21257, and GSE33382 cohorts. Differentially expressed analysis and the least absolute shrinkage and selection operator regression analysis were conducted to identify prognostic genes and construct an ER stress-related prognostic signature (ERSRPS). Survival analysis and time dependent ROC analysis were performed to evaluate the predictive performance of the constructed prognostic signature. The "ESTIMATE" package and ssGSEA algorithm were utilized to evaluate the differences in immune cells infiltration between the groups. Cell-based assays, including CCK-8, colony formation, and transwell assays and co-culture system were performed to assess the effects of the target gene and small molecular drug in osteosarcoma. Animal models were employed to assess the anti-osteosarcoma effects of small molecular drug. RESULTS Five genes (BLC2, MAGEA3, MAP3K5, STC2, TXNDC12) were identified to construct an ERSRPS. The ER stress-related gene Stanniocalcin 2 (STC2) was identified as a risk gene in this signature. Additionally, STC2 knockdown significantly inhibited osteosarcoma cell proliferation, migration, and invasion. Furthermore, the ER stress-related gene STC2 was found to downregulate the expression of MHC-I molecules in osteosarcoma cells, and mediate immune responses through influencing the infiltration and modulating the function of CD8+ T cells. Patients categorized by risk scores showed distinct immune status, and immunotherapy response. ISOX was subsequently identified and validated as an effective anti-osteosarcoma drug through a combination of CMap database screening and in vitro and in vivo experiments. CONCLUSION The ERSRPS may guide personalized treatment decisions for osteosarcoma, and ISOX holds promise for repurposing in osteosarcoma treatment.
Collapse
Affiliation(s)
- Weidong Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yan Liao
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Pengxiao Sun
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Renal Failure Research, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jian Tu
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yutong Zou
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Ji Fang
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Ziyun Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Hongbo Li
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Junkai Chen
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yuzhong Peng
- Macau University of Science and Technology, Macau, 999078, China
| | - Lili Wen
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
| | - Xianbiao Xie
- Department of Musculoskeletal Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China.
| |
Collapse
|
37
|
Amato R, Lucchesi M, Marracci S, Filippi L, Dal Monte M. β-Adrenoceptors in Cancer: Old Players and New Perspectives. Handb Exp Pharmacol 2024; 285:665-688. [PMID: 37982890 DOI: 10.1007/164_2023_701] [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: 11/21/2023]
Abstract
Distress, or negative stress, is known to considerably increase the incidence of several diseases, including cancer. There is indeed evidence from pre-clinical models that distress causes a catecholaminergic overdrive that, mainly through the activation of β-adrenoceptors (β-ARs), results in cancer cell growth and cancer progression. In addition, clinical studies have evidenced a role of negative stress in cancer progression. Moreover, plenty of data demonstrates that β-blockers have positive effects in reducing the pro-tumorigenic activity of catecholamines, correlating with better outcomes in some type of cancers as evidenced by several clinical trials. Among β-ARs, β2-AR seems to be the main β-AR subtype involved in tumor development and progression. However, there are data indicating that also β1-AR and β3-AR may be involved in certain tumors. In this chapter, we will review current knowledge on the role of the three β-AR isoforms in carcinogenesis as well as in cancer growth and progression, with particular emphasis on recent studies that are opening new avenues in the use of β-ARs as therapeutic targets in treating tumors.
Collapse
MESH Headings
- Humans
- Neoplasms/metabolism
- Neoplasms/drug therapy
- Neoplasms/pathology
- Animals
- Receptors, Adrenergic, beta-3/metabolism
- Adrenergic beta-Antagonists/therapeutic use
- Adrenergic beta-Antagonists/pharmacology
- Receptors, Adrenergic, beta-2/metabolism
- Receptors, Adrenergic, beta-2/drug effects
- Receptors, Adrenergic, beta/metabolism
- Receptors, Adrenergic, beta/physiology
- Receptors, Adrenergic, beta-1/metabolism
- Signal Transduction
- Disease Progression
Collapse
Affiliation(s)
- Rosario Amato
- Department of Biology, University of Pisa, Pisa, Italy
| | | | | | - Luca Filippi
- Department of Clinical and Experimental Medicine, Neonatology and Neonatal Intensive Care Unit, University of Pisa, Pisa, Italy
| | | |
Collapse
|
38
|
Winkler F. Neuroscience and oncology: state-of-the-art and new perspectives. Curr Opin Neurol 2023; 36:544-548. [PMID: 37973023 DOI: 10.1097/wco.0000000000001207] [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: 11/19/2023]
Abstract
PURPOSE OF REVIEW Emerging discoveries suggest that both the central (CNS) and peripheral (PNS) nervous system are an important driver of cancer initiation, promotion, dissemination, and therapy resistance, not only in the brain but also in multiple cancer types throughout the body. This article highlights the most recent developments in this emerging field of research over the last year and provides a roadmap for the future, emphasizing its translational potential. RECENT FINDINGS Excitatory synapses between neurons and cancer cells that drive growth and invasion have been detected and characterized. In addition, a plethora of paracrine, mostly tumor-promoting neuro-cancer interactions are reported, and a neuro-immuno-cancer axis emerges. Cancer cell-intrinsic neural properties, and cancer (therapy) effects on the nervous system that cause morbidity in patients and can establish harmful feedback loops receive increasing attention. Despite the relative novelty of these findings, therapies that inhibit key mechanisms of this neuro-cancer crosstalk are developed, and already tested in clinical trials, largely by repurposing of approved drugs. SUMMARY Neuro-cancer interactions are manyfold, have multiple clinical implications, and can lead to novel neuroscience-instructed cancer therapies and improved therapies of neurological dysfunctions and cancer pain. The development of biomarkers and identification of most promising therapeutic targets is crucial.
Collapse
Affiliation(s)
- Frank Winkler
- Neurology Clinic and National Center for Tumor Diseases, University Hospital Heidelberg
- Clinical Cooperation Unit Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
39
|
Li J, Che M, Zhang B, Zhao K, Wan C, Yang K. The association between the neuroendocrine system and the tumor immune microenvironment: Emerging directions for cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2023; 1878:189007. [PMID: 37907132 DOI: 10.1016/j.bbcan.2023.189007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 09/13/2023] [Accepted: 10/16/2023] [Indexed: 11/02/2023]
Abstract
This review summarizes emerging evidence that the neuroendocrine system is involved in the regulation of the tumor immune microenvironment (TIME) to influence cancer progression. The basis of the interaction between the neuroendocrine system and cancer is usually achieved by the infiltration of nerve fibers into the tumor tissue, which is called neurogenesis; the migration of cancer cells toward nerve fibers, which is called perineural invasion (PNI), and the neurotransmitters. In addition to the traditional role of neurotransmitters in neural communications, neurotransmitters are increasingly recognized as mediators of crosstalk between the nervous system, cancer cells, and the immune system. Recent studies have revealed that not only nerve fibers but also cancer cells and immune cells within the TIME can secrete neurotransmitters, exerting influence on both neurons and themselves. Furthermore, immune cells infiltrating the tumor environment have been found to express a wide array of neurotransmitter receptors. Hence, targeting these neurotransmitter receptors may promote the activity of immune cells in the tumor microenvironment and exert anti-tumor immunity. Herein, we discuss the crosstalk between the neuroendocrine system and tumor-infiltrating immune cells, which may provide feasible cancer immunotherapy options.
Collapse
Affiliation(s)
- Jie Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Mengjie Che
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bin Zhang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kewei Zhao
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chao Wan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Key Laboratory of Precision Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
40
|
Manolios N, Papaemmanouil J, Adams DJ. The role of ion channels in T cell function and disease. Front Immunol 2023; 14:1238171. [PMID: 37705981 PMCID: PMC10497217 DOI: 10.3389/fimmu.2023.1238171] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 07/21/2023] [Indexed: 09/15/2023] Open
Abstract
T lymphocytes (T cells) are an important sub-group of cells in our immune system responsible for cell-mediated adaptive responses and maintaining immune homeostasis. Abnormalities in T cell function, lead the way to the persistence of infection, impaired immunosurveillance, lack of suppression of cancer growth, and autoimmune diseases. Ion channels play a critical role in the regulation of T cell signaling and cellular function and are often overlooked and understudied. Little is known about the ion "channelome" and the interaction of ion channels in immune cells. This review aims to summarize the published data on the impact of ion channels on T cell function and disease. The importance of ion channels in health and disease plus the fact they are easily accessible by virtue of being expressed on the surface of plasma membranes makes them excellent drug targets.
Collapse
Affiliation(s)
- Nicholas Manolios
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
- Department of Rheumatology, Westmead Hospital, Sydney, NSW, Australia
| | - John Papaemmanouil
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - David J. Adams
- Illawarra Health and Medical Research Institute (IHMRI), Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
41
|
Baek AE. cAMPing up cancer immunotherapy. Sci Signal 2023; 16:eadj2198. [PMID: 37339183 DOI: 10.1126/scisignal.adj2198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023]
Abstract
Adrenergic receptor stimulation improves cancer immunotherapy in a range of immune-competent tumor models.
Collapse
Affiliation(s)
- Amy E Baek
- Science Signaling, AAAS, Washington, DC 20005, USA.
| |
Collapse
|