1
|
Dey RK, Kumari R, Patra R, Soni DK, Biswas R, Patnaik S, Ghosh D. MicroRNA-129-5p-mediated translational repression of microglial ROCK1 leads to enhanced phagocytosis. J Biol Chem 2025:110293. [PMID: 40419128 DOI: 10.1016/j.jbc.2025.110293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 05/08/2025] [Accepted: 05/22/2025] [Indexed: 05/28/2025] Open
Abstract
ROCK1 plays an important role in phagocytosis by inducing cytoskeletal rearrangement. Although the transcriptional regulation of ROCK1 is known but its post-transcriptional regulation is underexplored. We intended to find a mechanism of microglial phagocytosis through possible post-transcriptional regulation of ROCK1. The study identified miR-129-5p as a regulator of microglial phagocytosis following exposure to an environmental stressor, arsenic, combining in silico analysis, mutational analysis, in vitro experiments, and validation in Balb/c mouse model. The in silico analysis and in vitro studies with mouse primary neonatal microglia, BV2 microglia, ex vivo microglia and human microglial cell line CHME3 revealed that arsenic exposure increases microglial phagocytic activity. Arsenic exposure was also observed to increase the level of miR-129-5p and consequently decreases the level of ROCK1 protein. In vitro experiments and mutational analysis confirmed the in silico predicted binding site of miR-129-5p on the 3'UTR of ROCK1 and also confirmed the shuttling of ROCK1 mRNA into the cytoplasmic-processing body (p-body) in mouse microglia. Downstream to ROCK1, Rac1 has also been studied to pinpoint the partners in the signaling axis. The role of miR-129-5p in microglial phagocytosis was studied in vitro and validated in vivo in BALB/c mouse by stereotactically injecting anti-miR-129-5p and assessing the phagocytosis in ex vivo microglia and co-localization of Iba1 and PSD95 in brain cryosection. Finally, experiments with arsenic, anti-miR-129-5p, ROCK1 & Rac1 siRNA in various combinations confirmed the miR-129-5p→ROCK1→Rac1→Phagocytosis signaling axis. Overall, the study revealed miR-129-5p as an important regulator of microglial phagocytosis with potential implication in synaptic plasticity and neurodegenerative complications.
Collapse
Affiliation(s)
- Rajib Kumar Dey
- Immunotoxicology Laboratory, Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ranjana Kumari
- Immunotoxicology Laboratory, Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Roni Patra
- Immunotoxicology Laboratory, Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Dharmendra Kumar Soni
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Satyakam Patnaik
- Drug and Chemical Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh 226001, India
| | - Debabrata Ghosh
- Immunotoxicology Laboratory, Systems Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
2
|
Sun R, Zhao Y, Liu Y, Zhang M, Qiu Z, Ma X, Wei L, Lu W, Liu Z, Jiang J. Extracellular matrix stiffness in endometrial cancer: driving progression and modulating treatment sensitivity via the ROCK1/YAP1 axis. Cell Death Dis 2025; 16:380. [PMID: 40368918 PMCID: PMC12078694 DOI: 10.1038/s41419-025-07697-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025]
Abstract
Endometrial cancer (EC) is among the most prevalent gynecological malignancies, with advanced or recurrent cases posing significant treatment challenges due to limited responses to conventional therapies. Growing evidence highlights the critical role of extracellular matrix (ECM) stiffness in driving tumor progression by shaping the tumor microenvironment. In this study, we demonstrate that ECM stiffness is significantly higher in EC tissues compared to normal endometrium, correlating with elevated expression of ROCK1, a mechanosensitive kinase. Using atomic force microscopy (AFM), we quantified ECM stiffness, while polyacrylamide gels with varying stiffness were employed to mimic ECM conditions in vitro. Bioinformatics analyses, immunofluorescence, Western blotting, and co-immunoprecipitation experiments revealed that ROCK1 modulates the phosphorylation of YAP1, promoting its nuclear localization and transcriptional activity, thereby driving aggressive tumor behaviors, including enhanced proliferation, migration, invasion, and reduced apoptosis. Pharmacological inhibition of ROCK1 with Y-27632 mitigated these effects, suppressing tumor growth, restoring apoptosis, and inducing cell cycle arrest. Treatment with Y-27632 improved sensitivity to chemotherapy and radiotherapy, and significantly enhanced macrophage-mediated phagocytosis, thereby boosting anti-tumor immune responses. In hormone-resistant EC cells, ROCK1 inhibition restored sensitivity to progesterone therapy. Notably, in vivo experiments in a xenograft mouse model confirmed the therapeutic potential of Y-27632, as combination therapy with progesterone showed superior tumor-suppressive effects compared to monotherapy. These findings underscore the dual role of ECM stiffness and ROCK1 in driving tumor progression and influencing treatment outcomes. By elucidating the relationship between ECM stiffness, ROCK1/YAP1 signaling, and treatment sensitivity, this study highlights the potential of targeting the ROCK1/YAP1 axis as a therapeutic strategy. ROCK1 serves as both a biomarker for prognosis and a target for improving personalized treatment approaches, offering new avenues to enhance clinical outcomes for EC patients.
Collapse
Affiliation(s)
- Rui Sun
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Ying Zhao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Yao Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Mengyao Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Ziyi Qiu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaohong Ma
- Department of Obstetrics and Gynecology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Lina Wei
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Lu
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Zhiming Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.
| | - Jie Jiang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
3
|
Liang JL, Cao Y, Lv K, Xiao B, Sun J. Amplifying Ca 2+ overload by engineered biomaterials for synergistic cancer therapy. Biomaterials 2025; 316:123027. [PMID: 39700532 DOI: 10.1016/j.biomaterials.2024.123027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/28/2024] [Accepted: 12/13/2024] [Indexed: 12/21/2024]
Abstract
Ca2+ overload is one of the most widely causes of inducing apoptosis, pyroptosis, immunogenic cell death, autophagy, paraptosis, necroptosis, and calcification of tumor cells, and has become the most valuable therapeutic strategy in the field of cancer treatment. Nevertheless, several challenges remain in translating Ca2+ overload-mediated therapeutic strategies into clinical applications, such as the precise control of Ca2+ dynamics, specificity of Ca2+ homeostasis dysregulation, as well as comprehensive mechanisms of Ca2+ regulation. Given this, we comprehensively reviewed the Ca2+-driven intracellular signaling pathways and the application of Ca2+-based biomaterials (such as CaCO3-, CaP-, CaO2-, CaSi-, CaF2-, and CaH2-) in mediating cancer diagnosis, treatment, and immunotherapy. Meanwhile, the latest researches on Ca2+ overload-mediated therapeutic strategies, as well as those combined with multiple-model therapies in mediating cancer immunotherapy are further highlighted. More importantly, the critical challenges and the future prospects of the Ca2+ overload-mediated therapeutic strategies are also discussed. By consolidating recent findings and identifying future research directions, this review aimed to advance the field of oncology therapy and contribute to the development of more effective and targeted treatment modalities.
Collapse
Affiliation(s)
- Jun-Long Liang
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Yangyang Cao
- Hangzhou Ultra-theranostics Biopharmaceuticals Technology Co., Ltd., Hangzhou, 311231, China
| | - Kaiwei Lv
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Bing Xiao
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China.
| | - Jihong Sun
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China; Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University, China; Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
4
|
Zou JX, Chang MR, Kuznetsov NA, Kee JX, Babak MV, Ang WH. Metal-based immunogenic cell death inducers for cancer immunotherapy. Chem Sci 2025; 16:6160-6187. [PMID: 40160356 PMCID: PMC11949249 DOI: 10.1039/d4sc08495k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Immunogenic cell death (ICD) has attracted enormous attention over the past decade due to its unique characteristics in cancer cell death and its role in activating innate and adaptive immune responses against tumours. Many efforts have been dedicated to screening, identifying and discovering ICD inducers, resulting in the validation of several based on metal complexes. In this review, we provide a comprehensive summary of current metal-based ICD inducers, their molecular mechanisms for triggering ICD initiation and subsequent protective antitumour immune responses, along with considerations for validating ICD both in vitro and in vivo. We also aim to offer insights into the future development of metal complexes with enhanced ICD-inducing properties and their applications in potentiating antitumour immunity.
Collapse
Affiliation(s)
- Jiao Xia Zou
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Meng Rui Chang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Nikita A Kuznetsov
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Jia Xuan Kee
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
- NUS Graduate School - Integrative Science and Engineering Programme (ISEP), National University of Singapore 21 Lower Kent Ridge Rd Singapore 119077 Singapore
| |
Collapse
|
5
|
Silver FH. The Role of Connections Between Cellular and Tissue Mechanical Elements and the Importance of Applied Energy in Mechanotransduction in Cancerous Tissue. Biomolecules 2025; 15:457. [PMID: 40305177 PMCID: PMC12025281 DOI: 10.3390/biom15040457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/08/2025] [Accepted: 03/13/2025] [Indexed: 05/02/2025] Open
Abstract
In the presence of cellular mutations and impaired mechanisms of energy transmission to the attached cells and tissues, excess energy is available to upregulate some of the mechanotransduction pathways that maintain cell and tissue structure and function. The ability to transfer applied energy through integrin-mediated pathways, cell ion channels, cell membrane, cytoskeleton-nucleoskeleton connections, cell junctions, and cell-extracellular matrix attachments provides an equilibrium for energy storage, transmission, and dissipation in tissues. Disruption in energy storage, transmission, or dissipation via genetic mutations blocks mechanical communication between cells and tissues and impairs the mechanical energy equilibrium that exists between cells and tissues. This results in local structural changes through altered regulatory pathways, which produce cell clustering, collagen encapsulation, and an epithelial-mesenchymal transition (EMT), leading to increased cellular motility along newly reorganized collagen fibers (fibrosis). The goal of this review is to postulate how changes in energy transfer between cells and the extracellular matrix may alter local energy equilibrium and mechanotransduction pathways. The changes along with cellular mutations lead to cell and ECM changes reported in cancer, which is postulated to modify mechanical equilibria between cells and their ECM. This leads to uncontrolled cancer cellular proliferation and collagen remodeling.
Collapse
Affiliation(s)
- Frederick H Silver
- Department of Pathology and Laboratory Medicine, Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, New Brunswick, NJ 08854, USA
| |
Collapse
|
6
|
Lin YY, Lan HY, Teng HW, Wang YP, Lin WC, Hwang WL. Colorectal cancer stem cells develop NK cell resistance via homotypic cell-in-cell structures suppressed by Stathmin1. Theranostics 2025; 15:4308-4324. [PMID: 40225568 PMCID: PMC11984389 DOI: 10.7150/thno.110379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/05/2025] [Indexed: 04/15/2025] Open
Abstract
Rationale: Advances in cancer therapies have significantly improved patient survival; however, tumors enriched in cancer stem cells (CSCs) have poor treatment responses. CSCs are a key source of tumor heterogeneity, contributing to therapeutic resistance and unfavorable patient outcomes. In the tumor microenvironment (TME), cell-in-cell (CIC) structures, where one cell engulfs another, have been identified as markers of poor prognosis. Despite their clinical relevance, the mechanisms underlying CIC formation across different tumor cell subpopulations remain largely unknown. Elucidating these processes could provide novel insights and therapeutic opportunities to address aggressive, treatment-resistant cancers. Method: Fluorescent mCherry-carrying colorectal cancer stem cells (CRCSCs) were expanded as spheroids in serum-free media and cocultured with either parental cancer cell-expressing Venus fluorescent protein or CFSE dye-stained immune cells (T cells, M1/M2 macrophages, neutrophils, and NK cells) or treated with EGFR- or PD-L1-targeting antibodies to assess the formation of CIC structures. Genes potentially crucial for the formation of CIC structures were knocked down or overexpressed, and their effects on CIC formation were evaluated. The clinical relevance of the in vitro findings was confirmed through analysis of formalin-fixed, paraffin-embedded (FFPE) human colorectal cancer (CRC) specimens. Results: CRCSCs have a strong predilection for serving as the outer cell in a CIC structure and forming homotypic CIC structures predominantly with parental CRC cells. The frequency of CIC structure formation increased when the cells were exposed to anti-PD-L1 antibody treatment. Both the outer CRCSC in a CIC structure and CRCSCs released from a homotypic CIC structure showed enhanced resistance to the cytotoxicity of NK-92MI cells. Restoration of Stathmin1 (STMN1) expression but not RAC1 knockdown in CRCSCs reduced the homotypic CIC frequency, disrupted the outer cell fate in CIC structures, and increased cell susceptibility to NK-92MI cytotoxicity. In CRC patients, CIC structures are associated with poor tumor differentiation, negative STMN1 expression, and poor prognosis. Conclusion: CSCs play a crucial role in informing CIC structures in CRC. CIC structure formation partially depends on low STMN1 expression and confers a survival advantage under NK cytotoxicity. Targeting this pathway may significantly improve immunotherapy's efficacy for CRC patients.
Collapse
Affiliation(s)
- Yen-Yu Lin
- Department of Pathology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City 24352, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Hsin-Yi Lan
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Hao-Wei Teng
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ya-Pei Wang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Wen-Chun Lin
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Wei-Lun Hwang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| |
Collapse
|
7
|
Ji X, Wang G, Pan D, Xu S, Lei X. Efficacy and safety of pembrolizumab in advanced gastric and gastroesophageal junction cancer: a systematic review and meta-analysis. BMC Gastroenterol 2025; 25:173. [PMID: 40087572 PMCID: PMC11908035 DOI: 10.1186/s12876-025-03754-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Pembrolizumab, a PD-1 inhibitor, has shown potential for treating advanced gastric and gastroesophageal junction (GEJ) cancer. This meta-analysis evaluates its efficacy and safety, alone or combined with chemotherapy, in this population. METHODS A systematic review and meta-analysis were conducted in accordance with PRISMA guidelines. Databases including PubMed, Embase, the Cochrane Central Register of Controlled Trials, and Web of Science were searched up to October 31, 2024. Twelve studies comprising 4,069 patients were included. The primary outcomes were overall survival (OS) and progression-free survival (PFS); secondary outcomes included objective response rate (ORR), adverse events (AEs), and grade ≥ 3 AEs. Effect sizes were calculated using mean differences (MDs) and odds ratios (ORs) with 95% confidence intervals (CIs). RESULTS Pembrolizumab combined with chemotherapy significantly improved OS (MD = 1.92 months; 95% CI: 0.94 to 2.91) and ORR (MD = 11.05%; 95% CI: 6.29 to 15.82) compared to chemotherapy alone. Pembrolizumab monotherapy did not show a significant effect on OS (MD = 0.24 months; 95% CI: -1.15 to 1.63) and was associated with a significant reduction in PFS (MD = -2.28 months; 95% CI: -2.85 to -1.71) compared to chemotherapy alone. For safety, pembrolizumab monotherapy significantly reduced the risk of AEs (OR = 0.68; 95% CI: 0.57 to 0.81) and grade ≥ 3 AEs (OR = 0.39; 95% CI: 0.30 to 0.51) compared to chemotherapy. Pembrolizumab combined with chemotherapy did not significantly alter the risk of AEs (OR = 1.01; 95% CI: 0.90 to 1.13) or grade ≥ 3 AEs (OR = 1.12; 95% CI: 0.99 to 1.27) compared to chemotherapy alone. CONCLUSION Pembrolizumab combined with chemotherapy improves survival and response rates with a manageable safety profile in advanced gastric and GEJ cancers. Monotherapy shows limited efficacy, highlighting the need for combination strategies and patient selection.
Collapse
Affiliation(s)
- Xiaoying Ji
- Department of Pharmacy, Yiwu Central Hospital, Yiwu, Zhejiang, 322000, China
| | - Guoping Wang
- Department of Pharmacy, Yiwu Central Hospital, Yiwu, Zhejiang, 322000, China
| | - Dandan Pan
- Department of Pharmacy, Yiwu Central Hospital, Yiwu, Zhejiang, 322000, China
| | - Shanxia Xu
- Quzhou Zhong Da Lang Yuan Nursing Home, Quzhou, Zhejiang, 324000, China
| | - Xinming Lei
- The Quzhou Afliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, Zhejiang, 324000, China.
| |
Collapse
|
8
|
Lin J, Li B, Guo X, Li G, Zhang Q, Wang W. Key Mechanisms of Oxidative Stress-Induced Ferroptosis in Heart Failure with Preserved Ejection Fraction and Potential Therapeutic Approaches. Rev Cardiovasc Med 2025; 26:26613. [PMID: 40160560 PMCID: PMC11951494 DOI: 10.31083/rcm26613] [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: 09/18/2024] [Revised: 12/02/2024] [Accepted: 12/10/2024] [Indexed: 04/02/2025] Open
Abstract
The prevalence of heart failure with preserved ejection fraction (HFpEF) is increasing annually, particularly among patients with metabolic disorders such as hypertension and diabetes. However, there is currently no treatment capable of altering the natural course of HFpEF. Recently, the interplay between oxidative stress and ferroptosis in cardiovascular diseases has drawn extensive attention; however, minimal research has been published on the mechanisms of oxidative stress and ferroptosis in HFpEF. This paper reviews the relevant mechanisms through which oxidative stress is induced and promotes ferroptosis during the development of HFpEF. The review also explores more efficacious treatment approaches for HFpEF by inhibiting oxidative stress and ferroptosis, thereby offering a theoretical foundation for verifying the feasibility of these methods for further research. As tumor-targeted therapy progresses, the survival period of tumor patients is prolonged, and cardiovascular events have gradually emerged as one of the most crucial causes of death among tumor patients. Hence, inhibiting the vascular endothelial growth factor (VEGF) pathway has become a major target in tumor treatment, significantly enhancing patient survival. Nevertheless, secondary cardiovascular complications and events, such as myocardial injury and subsequent heart failure, have severely impacted patient survival and quality of life. Therefore we have also explored the potential mechanism through which novel targeted anti-cancer drugs induce HFpEF via ferroptosis. Additionally, we reviewed the specific modes of action for preventing and treating HFpEF without influencing their anti-cancer therapeutic effect.
Collapse
Affiliation(s)
- Junling Lin
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Bingtao Li
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Xueqi Guo
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Guodong Li
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Qi Zhang
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| | - Wenjuan Wang
- Department of Cardiovascular Center, First Affiliated Hospital of Huzhou University, 313000 Huzhou, Zhejiang, China
| |
Collapse
|
9
|
Kim S, Kim YK, Kim S, Choi YS, Lee I, Joo H, Kim J, Kwon M, Park S, Jo MK, Choi Y, D'Souza T, Jung JW, Zakhem E, Lenzini S, Woo J, Choi H, Park J, Park SY, Kim GB, Nam GH, Kim IS. Dual-mode action of scalable, high-quality engineered stem cell-derived SIRPα-extracellular vesicles for treating acute liver failure. Nat Commun 2025; 16:1903. [PMID: 39988725 PMCID: PMC11847939 DOI: 10.1038/s41467-025-57133-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 02/06/2025] [Indexed: 02/25/2025] Open
Abstract
Acute liver failure (ALF) is a life-threatening condition caused by rapid hepatocyte death and impaired liver regeneration. Here we show that extracellular vesicles engineered to express Signal Regulatory Protein Alpha (SIRP-EVs), produced via a scalable 3D bioreactor process with high yield and purity, exhibit significant therapeutic potential by targeting damaged cells and promoting tissue repair. SIRP-EVs block CD47, a crucial inhibitory signal on necroptotic cells, to enhance macrophage-mediated clearance of dying hepatocytes. They also deliver regenerative cargo from mesenchymal stem cells, reprogramming macrophages to support liver regeneration. In male animal models, SIRP-EVs significantly reduce liver injury markers and improve survival, demonstrating their dual-function therapeutic efficacy. By integrating the resolution of necroptosis with regenerative macrophage reprogramming, SIRP-EVs represent a promising platform for restoring liver function. These findings support the development of EV-based in vivo macrophage reprogramming therapies for ALF and other inflammation-driven diseases, paving the way for the clinical application of engineered EV therapeutics.
Collapse
Grants
- This research was funded by National Research Foundation of Korea (NRF) funded by the Ministry of Science and ICT (Grant Number: RS-2017-NR022964).
- This research was supported by SHIFTBIO INC., Korean Fund for Regenerative Medicine funded by Ministry of Science and ICT, and Ministry of Health and Welfare (Grant Number: 23C0111L1), a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (Grant Number: RS-2023-KH136648), and a grant of the BIG3 Project, funded by the Ministry of SMEs and Startups, Republic of Korea (Grant Number: RS-2022-TI022422).
- This research was supported by a grant of the Korea Health Technology R&D Project through the Korea Health Industry Development Institute (KHIDI), funded by the Ministry of Health & Welfare, Republic of Korea (Grant Number: RS-2023-KH136648; RS-2023-KH140007).
Collapse
Affiliation(s)
| | | | | | | | - Inkyu Lee
- SHIFTBIO INC, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Hyemin Joo
- SHIFTBIO INC, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | | | - Minjeong Kwon
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Seryoung Park
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Min Kyoung Jo
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | - Jiwan Woo
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hongyoon Choi
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
- Portrai, Inc, Seoul, Republic of Korea
| | | | - Seung-Yoon Park
- Department of Biochemistry, School of Medicine, Dongguk University, Gyeongju, Republic of Korea
| | | | - Gi-Hoon Nam
- SHIFTBIO INC, Seoul, Republic of Korea.
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.
| |
Collapse
|
10
|
Kim SA, Kim S, Hong Y, Choi Y, Lee Y, Kwon M, Park SY, Jeong C, Nam GH, Han RT, Kim IS. Immunogenic clearance combined with PD-1 blockade elicits antitumor effect by promoting the recruitment and expansion of the effector memory-like CD8 +T cell. Transl Oncol 2025; 51:102209. [PMID: 39608213 PMCID: PMC11635775 DOI: 10.1016/j.tranon.2024.102209] [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: 07/21/2024] [Revised: 09/30/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
Immune checkpoint inhibition shows promise for cancer treatment, but only a minority of patients respond. Combination strategies have been explored to overcome this resistance. Combining immunogenic clearance using immunogenic cell death inducers with a rho kinase inhibitor enhances phagocytosis of immunogenically dying cancer cells by antigen-presenting cells, stimulating tumor-specific immune responses by activating CD8+T cells via dendritic cell-mediated priming. This approach increases the responsiveness of immune checkpoint blockade (ICB)-resistant cancer to ICB. However, the precise mechanisms remain unclear. This study elucidates cellular mechanisms of immunogenic clearance enhancing ICB response. Using single-cell RNA sequencing, we observed an increase in effector memory-like CD8+T cells within the tumor microenvironment with combined treatment. We propose this cell cluster may originate from proliferating CD8+T cells elevated by immunogenic clearance. Notably, abundant effector memory-like CD8+T cells in ICB-responsive patients suggest their antitumor effect. Thus, increasing this cell population through enhanced T cell priming may improve the response of ICB-resistant tumors.
Collapse
Affiliation(s)
- Seong A Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Seohyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Department of Research and Development, ShiftBio, Seoul 02751, Republic of Korea
| | - Yeonsun Hong
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Yoonjeong Choi
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Department of Research and Development, ShiftBio, Seoul 02751, Republic of Korea
| | - Yeji Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Minsu Kwon
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea
| | - Seung-Yoon Park
- Department of Biochemistry, School of Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Cherlhyun Jeong
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Department of Biological Chemistry, Korea University of Science and Technology, KIST Campus, 02841, Republic of Korea
| | - Gi-Hoon Nam
- Department of Research and Development, ShiftBio, Seoul 02751, Republic of Korea; Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul 02841, Republic of Korea.
| | - Rafael T Han
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea; Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.
| |
Collapse
|
11
|
Dong Y, Lu M, Yin Y, Wang C, Dai N. Tumor Biomechanics-Inspired Future Medicine. Cancers (Basel) 2024; 16:4107. [PMID: 39682291 DOI: 10.3390/cancers16234107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Malignant tumors pose a significant global health challenge, severely threatening human health. Statistics from the World Health Organization indicate that, in 2022, there were nearly 20 million new cancer cases and 9.7 million cancer-related deaths. Therefore, it is urgently necessary to study the pathogenesis of cancer and explore effective diagnostic and treatment strategies. In recent years, research has highlighted the importance of mechanical cues in tumors, which have become a new hallmark of cancer and a key factor in regulating tumor behavior. This suggests that studying the mechanical properties of tumors may open potential new avenues for understanding the pathogenesis, diagnosis, and therapeutic intervention of cancer. This review summarizes the mechanical characteristics of tumors and the development of tumor diagnostics and treatments targeting specific mechanical factors. Finally, we propose new ideas and insights for the application of mechanomedicine in cancer diagnosis and treatment in the future.
Collapse
Affiliation(s)
- Yuqing Dong
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Mengnan Lu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Yuting Yin
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Cong Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| | - Ningman Dai
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, China
| |
Collapse
|
12
|
Mao C, Fang J, Zou S, Huang Y, Chen X, Ding X, Fang Z, Zhang N, Lou Y, Chen Z, Ding W, Ma Z. Discovery of the First-in-Class Dual-Target ROCK/HDAC Inhibitor with Potent Antitumor Efficacy in Vivo That Trigger Antitumor Immunity. J Med Chem 2024; 67:20619-20638. [PMID: 39523548 DOI: 10.1021/acs.jmedchem.4c02183] [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: 11/16/2024]
Abstract
Triple-negative breast cancer (TNBC) represents a highly aggressive and heterogeneous malignancy. Currently, multitarget drug approaches present a promising therapeutic approach for TNBC. Utilizing a combinatorial chemistry strategy to construct a virtual screening database, dual ROCK/HDAC-targeting benzothiophene compounds were identified. Notably, compound 10h effectively inhibits ROCK1/2 and HDAC1/2/3/6/8 while demonstrating potent antiproliferative activity against breast cancer cells. In an orthotopic mouse model of breast cancer, 10h significantly suppressed tumor growth without apparent toxicity. Importantly, 10h induced immunogenic cell death (ICD), promoted dendritic cells (DCs) maturation, and activated T cells, thereby initiating antitumor immunity. In conclusion, compound 10h is a novel dual-target ROCK/HDAC inhibitor that represents a promising treatment strategy for TNBC.
Collapse
Affiliation(s)
- Churu Mao
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Jiebin Fang
- Hainan Institute of Zhejiang University, Sanya 572025, China
| | - Shijie Zou
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Yun Huang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Xiaoming Chen
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Xia Ding
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Zhangyun Fang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Ningjing Zhang
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
| | - Yijie Lou
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Zhe Chen
- Key Laboratory of Digestive Pathophysiology of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Wanjing Ding
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
- Hainan Institute of Zhejiang University, Sanya 572025, China
| | - Zhongjun Ma
- Institute of Marine Biology and Pharmacology, Ocean College, Zhejiang University, Zhoushan 316021, China
- Hainan Institute of Zhejiang University, Sanya 572025, China
| |
Collapse
|
13
|
Zhao H, Luo K, Liu M, Cai Y, Liu S, Li S, Zhao Y, Zhang H. Immune regulation and prognostic prediction model establishment and validation of PSMB6 in lung adenocarcinoma. Front Genet 2024; 15:1458047. [PMID: 39507618 PMCID: PMC11538069 DOI: 10.3389/fgene.2024.1458047] [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: 07/01/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024] Open
Abstract
Lung cancer is one of the most common malignant tumors, and patients are often diagnosed at an advanced stage, posing a substantial risk to human health, so it is crucial to establish a model to forecast the prognosis of patients with lung cancer. Recent research has indicated that proteasome 20S subunit 6 (PSMB6) may be closely associated with anti-apoptotic pathways, and proliferation transduction signals in tumor cells of different tumors. However, the precise role of PSMB6 in the immunoregulatory processes within lung adenocarcinoma (LUAD) is yet to be elucidated. By analyzing the TCGA database, we discovered a positive correlation between the expression of PSMB6 and tumor growth trends, and lung adenocarcinoma patients with elevated PSMB6 expression levels had a worse prognosis. Our findings suggest a close correlation between PSMB6 expression levels, immune cell infiltration and immune checkpoint gene expression, which suggests that PSMB6 may become a new independent prognostic indicator. In addition, we developed a prognostic model of PSMB6-regulated immune infiltration-associated genes by analyzing the link between PSMB6 and the immune microenvironment. This model can not only predict the prognosis of lung adenocarcinoma but also forecasts the patient's reaction to immunotherapy. The validity of this research outcome has been confirmed by the GSE31210 and IMvigor210 cohorts. Analysis of the Kaplan-Meier Plotter database indicates that individuals with elevated levels of PSMB6 expression exhibit a poorer prognosis. Additionally, in vitro experiments demonstrated that knockdown of PSMB6 inhibits the proliferation, migration, and invasion of lung adenocarcinoma cells while promoting their apoptosis. Overall, our findings suggest that PSMB6 could remarkably influence the management and treatment of lung adenocarcinoma, opening new avenues for targeted immunotherapeutic strategies.
Collapse
Affiliation(s)
- Haiyang Zhao
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- Department of Thoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Innovation Centre for Science and Technology, Nanchong, China
| | - Kexin Luo
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- Department of Thoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Innovation Centre for Science and Technology, Nanchong, China
| | - Meihan Liu
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- North Sichuan Medical College, Innovation Centre for Science and Technology, Nanchong, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yuanze Cai
- North Sichuan Medical College, Nanchong, China
| | - Siman Liu
- North Sichuan Medical College, Nanchong, China
| | - Shijuan Li
- Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Yongsheng Zhao
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- Department of Thoracic Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hongpan Zhang
- Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
- North Sichuan Medical College, Nanchong, China
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Therapeutic Proteins Key Laboratory of Sichuan Province, Nanchong, China
| |
Collapse
|
14
|
Lu M, Xu Z, Xu F, Yin C, Guo H, Cheng B. Mechanical network motifs as targets for mechanomedicine. Drug Discov Today 2024; 29:104145. [PMID: 39182599 DOI: 10.1016/j.drudis.2024.104145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/26/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
The identification and analysis of network motifs has been widely used in the functional analysis of signaling components, disease discovery and other fields. The positive feedback loop (PFL) is a simple but important network motif. The formation of a PFL is regulated by mechanical cues such as substrate stiffness, fiber stretching and cell compression in the cell microenvironment. Here, we propose a new term, 'mechanical PFL', and analyze the mechanisms of mechanical PFLs at molecular, subcellular and cellular scales. More and more therapies are being targeted against mechanosignaling pathways at the experimental and preclinical stages, and exploring mechanical PFLs as potential mechanomedicine targets could be a new direction for disease treatment.
Collapse
Affiliation(s)
- Mengnan Lu
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710054, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Zhao Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; The Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; The Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China
| | - Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710054, PR China.
| | - Hui Guo
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; Department of Medical Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710054, PR China.
| | - Bo Cheng
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, PR China; The Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, PR China.
| |
Collapse
|
15
|
Feroz W, Park BS, Siripurapu M, Ntim N, Kilroy MK, Sheikh AMA, Mishra R, Garrett JT. Non-Muscle Myosin II A: Friend or Foe in Cancer? Int J Mol Sci 2024; 25:9435. [PMID: 39273383 PMCID: PMC11395477 DOI: 10.3390/ijms25179435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Non-muscle myosin IIA (NM IIA) is a motor protein that belongs to the myosin II family. The myosin heavy chain 9 (MYH9) gene encodes the heavy chain of NM IIA. NM IIA is a hexamer and contains three pairs of peptides, which include the dimer of heavy chains, essential light chains, and regulatory light chains. NM IIA is a part of the actomyosin complex that generates mechanical force and tension to carry out essential cellular functions, including adhesion, cytokinesis, migration, and the maintenance of cell shape and polarity. These functions are regulated via light and heavy chain phosphorylation at different amino acid residues. Apart from physiological functions, NM IIA is also linked to the development of cancer and genetic and neurological disorders. MYH9 gene mutations result in the development of several autosomal dominant disorders, such as May-Hegglin anomaly (MHA) and Epstein syndrome (EPS). Multiple studies have reported NM IIA as a tumor suppressor in melanoma and head and neck squamous cell carcinoma; however, studies also indicate that NM IIA is a critical player in promoting tumorigenesis, chemoradiotherapy resistance, and stemness. The ROCK-NM IIA pathway regulates cellular movement and shape via the control of cytoskeletal dynamics. In addition, the ROCK-NM IIA pathway is dysregulated in various solid tumors and leukemia. Currently, there are very few compounds targeting NM IIA, and most of these compounds are still being studied in preclinical models. This review provides comprehensive evidence highlighting the dual role of NM IIA in multiple cancer types and summarizes the signaling networks involved in tumorigenesis. Furthermore, we also discuss the role of NM IIA as a potential therapeutic target with a focus on the ROCK-NM IIA pathway.
Collapse
Affiliation(s)
- Wasim Feroz
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Briley SoYoung Park
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
- Cancer Research Scholars Program, College of Allied Health Sciences, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Meghna Siripurapu
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Nicole Ntim
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Mary Kate Kilroy
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | | | - Rosalin Mishra
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| | - Joan T. Garrett
- Department of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, Cincinnati, OH 45229, USA; (W.F.); (B.S.P.); (M.S.); (N.N.); (M.K.K.); (R.M.)
| |
Collapse
|
16
|
Wei M, Wang X, Mo Y, Kong C, Zhang M, Qiu G, Tang Z, Chen J, Wu F. Combined Effects of Anti-PD-L1 and Nanosonodynamic Therapy on HCC Immune Activation in Mice: An Investigation. Int J Nanomedicine 2024; 19:7215-7236. [PMID: 39050875 PMCID: PMC11268760 DOI: 10.2147/ijn.s427144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 05/29/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction Current therapeutic strategies, including immune checkpoint blockade (ICB), exhibit limited efficacy in treating hepatocellular carcinoma (HCC). Nanoparticles, particularly those that can accumulate specifically within tumors and be activated by sonodynamic therapy (SDT), can induce immunogenic cell death (ICD); however, ICD alone has not achieved satisfactory therapeutic effectiveness. This study investigates whether combining ICB with ICD induced by nanoparticle-mediated SDT could enhance anti-tumor immunity and inhibit HCC growth. Methods We developed an iron-based micelle nanodelivery system encapsulating the Near-Infrared Dye IR-780, which was surface-modified with a cyclic tripeptide composed of arginine-glycine-aspartic acid (cRGD). This led to the synthesis of targeted IR780@FOM-cRGD nanoparticles. These nanoparticles were specifically engineered to kill tumor cells under sonication, activate immunogenic cell death (ICD), and be used in conjunction with immune checkpoint blockade (ICB) for the treatment of hepatocellular carcinoma (HCC). Results The synthesized IR780@FOM-cRGD nanoparticles had an average diameter of 28.23±1.750 nm and a Zeta potential of -23.95±1.926. Confocal microscopy demonstrated that IR780@FOM-cRGD could target HCC cells while minimizing toxicity to healthy cells. Upon sonodynamic activation, these nanoparticles consumed significant amounts of oxygen and generated substantial reactive oxygen species (ROS), effectively killing tumor cells and inhibiting the proliferation, invasion, and migration of H22 cells. Hemolysis assays confirmed the in vivo safety of the nanoparticles, and in vivo fluorescence imaging revealed significant accumulation in tumor tissues. Mouse model experiments showed that combining ICB(which induced by Anti-PD-L1) with ICD (which induced by IR780@FOM-cRGD), could effectively activated anti-tumor immunity and suppressed tumor growth. Discussion This study highlights the potential of IR780@FOM-cRGD nanoparticles to facilitate tumor eradication and immune activation when used in conjunction with Anti-PD-L1 therapy. This combination represents a non-invasive, efficient, and targeted approach for the treatment of hepatocellular carcinoma (HCC). By integrating sonodynamic therapy with immunotherapy, this strategy promises to substantially improve the effectiveness of traditional treatments in combating HCC, offering new avenues for clinical application and therapeutic innovation.
Collapse
Affiliation(s)
- Meng Wei
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
| | - Xiaobo Wang
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
| | - Yunhai Mo
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
| | - Cunqing Kong
- Medical Imaging Center, Affiliated Taihe Hospital, Hubei University of Medicine, Hubei, 442000, People’s Republic of China
| | - Mengqi Zhang
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
- Department of Interventional Therapy, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
| | - Guanhua Qiu
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
- Department of Ultrasound, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
| | - Zhihong Tang
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
| | - Jie Chen
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
| | - Feixiang Wu
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, 530021, People’s Republic of China
- Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Ministry of Education/Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, 530021, People’s Republic of China
| |
Collapse
|
17
|
Choi Y, Kim SA, Jung H, Kim E, Kim YK, Kim S, Kim J, Lee Y, Jo MK, Woo J, Cho Y, Lee D, Choi H, Jeong C, Nam GH, Kwon M, Kim IS. Novel insights into paclitaxel's role on tumor-associated macrophages in enhancing PD-1 blockade in breast cancer treatment. J Immunother Cancer 2024; 12:e008864. [PMID: 39009452 PMCID: PMC11253755 DOI: 10.1136/jitc-2024-008864] [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: 06/27/2024] [Indexed: 07/17/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) poses unique challenges due to its complex nature and the need for more effective treatments. Recent studies showed encouraging outcomes from combining paclitaxel (PTX) with programmed cell death protein-1 (PD-1) blockade in treating TNBC, although the exact mechanisms behind the improved results are unclear. METHODS We employed an integrated approach, analyzing spatial transcriptomics and single-cell RNA sequencing data from TNBC patients to understand why the combination of PTX and PD-1 blockade showed better response in TNBC patients. We focused on toll-like receptor 4 (TLR4), a receptor of PTX, and its role in modulating the cross-presentation signaling pathways in tumor-associated macrophages (TAMs) within the tumor microenvironment. Leveraging insights obtained from patient-derived data, we conducted in vitro experiments using immunosuppressive bone marrow-derived macrophages (iBMDMs) to validate if PTX could augment the cross-presentation and phagocytosis activities. Subsequently, we extended our study to an in vivo murine model of TNBC to ascertain the effects of PTX on the cross-presentation capabilities of TAMs and its downstream impact on CD8+ T cell-mediated immune responses. RESULTS Data analysis from TNBC patients revealed that the activation of TLR4 and cross-presentation signaling pathways are crucial for the antitumor efficacy of PTX. In vitro studies showed that PTX treatment enhances the cross-presentation ability of iBMDMs. In vivo experiments demonstrated that PTX activates TLR4-dependent cross-presentation in TAMs, improving CD8+ T cell-mediated antitumor responses. The efficacy of PTX in promoting antitumor immunity was elicited when combined with PD-1 blockade, suggesting a complementary interaction. CONCLUSIONS This study reveals how PTX boosts the effectiveness of PD-1 inhibitors in treating TNBC. We found that PTX activates TLR4 signaling in TAMs. This activation enhances their ability to present antigens, thereby boosting CD8+ T cell antitumor responses. These findings not only shed light on PTX's immunomodulatory role in TNBC but also underscore the potential of targeting TAMs' antigen presentation capabilities in immunotherapy approaches.
Collapse
Affiliation(s)
- Yoonjeong Choi
- SHIFTBIO INC, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
| | - Seong A Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Hanul Jung
- SHIFTBIO INC, Seoul, Republic of Korea
- Department of Otolaryngology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eunhae Kim
- SHIFTBIO INC, Seoul, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | | | | | | | - Yeji Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Min Kyoung Jo
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Jiwan Woo
- Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Yakdol Cho
- Research Animal Resource Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | | | - Hongyoon Choi
- Portrai Inc, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Cherlhyun Jeong
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Division of Biomedical Science and Technology, KIST School, University of Science and Technology, Seoul, Republic of Korea
| | - Gi-Hoon Nam
- SHIFTBIO INC, Seoul, Republic of Korea
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Minsu Kwon
- Department of Otolaryngology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea
- Chemical and Biological Integrative Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| |
Collapse
|
18
|
Elzoghby AO, Samir O, Emam HE, Soliman A, Abdelgalil RM, Elmorshedy YM, Elkhodairy KA, Nasr ML. Engineering nanomedicines for immunogenic eradication of cancer cells: Recent trends and synergistic approaches. Acta Pharm Sin B 2024; 14:2475-2504. [PMID: 38828160 PMCID: PMC11143780 DOI: 10.1016/j.apsb.2024.03.022] [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: 08/01/2023] [Revised: 02/07/2024] [Accepted: 03/09/2024] [Indexed: 06/05/2024] Open
Abstract
Resistance to cancer immunotherapy is mainly attributed to poor tumor immunogenicity as well as the immunosuppressive tumor microenvironment (TME) leading to failure of immune response. Numerous therapeutic strategies including chemotherapy, radiotherapy, photodynamic, photothermal, magnetic, chemodynamic, sonodynamic and oncolytic therapy, have been developed to induce immunogenic cell death (ICD) of cancer cells and thereby elicit immunogenicity and boost the antitumor immune response. However, many challenges hamper the clinical application of ICD inducers resulting in modest immunogenic response. Here, we outline the current state of using nanomedicines for boosting ICD of cancer cells. Moreover, synergistic approaches used in combination with ICD inducing nanomedicines for remodeling the TME via targeting immune checkpoints, phagocytosis, macrophage polarization, tumor hypoxia, autophagy and stromal modulation to enhance immunogenicity of dying cancer cells were analyzed. We further highlight the emerging trends of using nanomaterials for triggering amplified ICD-mediated antitumor immune responses. Endoplasmic reticulum localized ICD, focused ultrasound hyperthermia, cell membrane camouflaged nanomedicines, amplified reactive oxygen species (ROS) generation, metallo-immunotherapy, ion modulators and engineered bacteria are among the most innovative approaches. Various challenges, merits and demerits of ICD inducer nanomedicines were also discussed with shedding light on the future role of this technology in improving the outcomes of cancer immunotherapy.
Collapse
Affiliation(s)
- Ahmed O. Elzoghby
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Omar Samir
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Hagar E. Emam
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Ahmed Soliman
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| | - Riham M. Abdelgalil
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Yomna M. Elmorshedy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Kadria A. Elkhodairy
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University, Alexandria 21521, Egypt
| | - Mahmoud L. Nasr
- Division of Engineering in Medicine and Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston 02115, MA, USA
| |
Collapse
|
19
|
Shi B, Lou X, Ma F, Nie Y, Chen H, Huang Y, Zhang W, Wang T. Perylene-Mediated Cytoskeletal Dysfunction Remodels Cancer-Associated Fibroblasts to Augment Antitumor Immunotherapy. Adv Healthc Mater 2024; 13:e2303837. [PMID: 38183408 DOI: 10.1002/adhm.202303837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/18/2023] [Indexed: 01/08/2024]
Abstract
Targeted reprogramming of cancer-associated fibroblasts (CAFs) is one of the most essential cancer therapies. However, how to reprogram active CAFs toward deactivated state still remains immense challenge. To tackle this challenge, herein, one perylene N, N'-bis(2-((dimethylammonium)ethylene)-2-(methoxylethyl))-1, 6, 7, 12-tetrachloroperylene-3, 4, 9, 10-tetracarboxylic diimide (PDIC-OC) is prepared, which can trigger endogenous reactive oxygen species (ROS) burst to result in cytoskeletal dysfunction and cell apoptosis so that suppress transforming growth factor β (TGF-β) production. As a result, PDIC-OC can reprogram the activated CAFs and relieve immunosuppressive tumor microenvironment by efficient polarization of M2-typed macrophages into M1-typed ones, downregulation of alpha-smooth muscle actin (α-SMA), alleviation of hypoxic state to promote infiltration of cytotoxic T lymphocytes, and ultimately realizes outstanding antitumor performance on B16F10 tumor-xenografted and lung-metastatic mouse model even at low concentration of 1 mg kg-1 body weight. This work thus presents a novel strategy that cytoskeleton dysfunction and cell apoptosis cooperatively suppress the secretion of TGF-β to reprogram CAFs and meanwhile clarifies intrinsic mechanism for perylene-triggered chemo-immunotherapy against hypoxic tumors.
Collapse
Affiliation(s)
- Bing Shi
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Zhengzhou, Henan, 475001, P. R. China
| | - Xue Lou
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Zhengzhou, Henan, 475001, P. R. China
| | - Feiyan Ma
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Zhengzhou, Henan, 475001, P. R. China
| | - Yanling Nie
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Zhengzhou, Henan, 475001, P. R. China
| | - Haoxing Chen
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Zhengzhou, Henan, 475001, P. R. China
| | - Yongwei Huang
- Laboratory for NanoMedical Photonics, School of Basic Medical Science, Henan University, Zhengzhou, Henan, 475001, P. R. China
| | - Wei Zhang
- Life and Health Intelligent Research Institute, Tianjin Key Laboratory of Life and Health Detection, Tianjin University of Technology, Tianjin, 300387, P. R. China
| | - Tie Wang
- Life and Health Intelligent Research Institute, Tianjin Key Laboratory of Life and Health Detection, Tianjin University of Technology, Tianjin, 300387, P. R. China
| |
Collapse
|
20
|
Yang Q, Zhuo Z, Qiu X, Luo R, Guo K, Wu H, Jiang R, Li J, Lian Q, Chen P, Sha W, Chen H. Adverse clinical outcomes and immunosuppressive microenvironment of RHO-GTPase activation pattern in hepatocellular carcinoma. J Transl Med 2024; 22:122. [PMID: 38297333 PMCID: PMC10832138 DOI: 10.1186/s12967-024-04926-0] [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: 08/09/2023] [Accepted: 01/23/2024] [Indexed: 02/02/2024] Open
Abstract
BACKGROUND Emerging evidence suggests that Rho GTPases play a crucial role in tumorigenesis and metastasis, but their involvement in the tumor microenvironment (TME) and prognosis of hepatocellular carcinoma (HCC) is not well understood. METHODS We aim to develop a tumor prognosis prediction system called the Rho GTPases-related gene score (RGPRG score) using Rho GTPase signaling genes and further bioinformatic analyses. RESULTS Our work found that HCC patients with a high RGPRG score had significantly worse survival and increased immunosuppressive cell fractions compared to those with a low RGPRG score. Single-cell cohort analysis revealed an immune-active TME in patients with a low RGPRG score, with strengthened communication from T/NK cells to other cells through MIF signaling networks. Targeting these alterations in TME, the patients with high RGPRG score have worse immunotherapeutic outcomes and decreased survival time in the immunotherapy cohort. Moreover, the RGPRG score was found to be correlated with survival in 27 other cancers. In vitro experiments confirmed that knockdown of the key Rho GTPase-signaling biomarker SFN significantly inhibited HCC cell proliferation, invasion, and migration. CONCLUSIONS This study provides new insight into the TME features and clinical use of Rho GTPase gene pattern at the bulk-seq and single-cell level, which may contribute to guiding personalized treatment and improving clinical outcome in HCC.
Collapse
Affiliation(s)
- Qi Yang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zewei Zhuo
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Xinqi Qiu
- Cancer Prevention Center, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Ruibang Luo
- Department of Computer Science, The University of Hong Kong, Hong Kong, 999077, SAR, China
| | - Kehang Guo
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
- Department of Critical Care Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Huihuan Wu
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Rui Jiang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
| | - Jingwei Li
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Qizhou Lian
- Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518118, Guangdong, China.
- Cord Blood Bank, Guangzhou Institute of Eugenics and Perinatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 511436, Guangdong, China.
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, 999077, SAR, China.
| | - Pengfei Chen
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Weihong Sha
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
21
|
Xu X, Yao L. Recent advances in the development of Rho kinase inhibitors (2015-2021). Med Res Rev 2024; 44:406-421. [PMID: 37265266 DOI: 10.1002/med.21980] [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: 12/23/2021] [Revised: 01/27/2023] [Accepted: 05/23/2023] [Indexed: 06/03/2023]
Abstract
Rho-associated coiled-coil kinases (ROCKs) are key downstream effectors of small GTPases. ROCK plays a central role in diverse cellular events with accumulating evidence supporting the concept that ROCK is important in tumor development and progression. Numerous ROCK inhibitors have been investigated for their therapeutic potential in the treatment of cancers. In this article, we review recent research progress on ROCK inhibitors, especially those with potential for the treatment of cancers, reported in the literature from 2015 to 2021. Most ROCK inhibitors show potent in vitro and in vivo antitumor activities and have potential in the treatment of cancers.
Collapse
Affiliation(s)
- Xiangrong Xu
- Yantai University Hospital, Yantai University, Yantai, China
| | - Lei Yao
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| |
Collapse
|
22
|
Zhou J, Guo H, Liu L, Jin Z, Zhang W, Tang T. Identification of immune-related hub genes and construction of an immune-related gene prognostic index for low-grade glioma. J Cell Mol Med 2023; 27:3851-3863. [PMID: 37775993 PMCID: PMC10718158 DOI: 10.1111/jcmm.17960] [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: 02/19/2023] [Revised: 08/14/2023] [Accepted: 09/08/2023] [Indexed: 10/01/2023] Open
Abstract
Low-grade glioma (LGG) poses significant management challenges and has a dismal prognosis. While immunotherapy has shown significant promise in cancer treatment, its progress in glioma has confronted with challenges. In our study, we aimed to develop an immune-related gene prognostic index (IRGPI) which could be used to evaluate the response and efficacy of LGG patients with immunotherapy. We included a total of 529 LGG samples from TCGA database and 1152 normal brain tissue samples from the GTEx database. Immune-related differentially expressed genes (DEGs) were screened. Then, we used weighted gene co-expression network analysis (WGCNA) to identify immune-related hub genes in LGG patients and performed Cox regression analysis to construct an IRGPI. The median IRGPI was used as the cut-off value to categorize LGG patients into IRGPI-high and low subgroups, and the molecular and immune mechanism in IRGPI-defined subgroups were analysed. Finally, we explored the relationship between IRGPI-defined subgroups and immunotherapy related indicators in patients after immunotherapy. Three genes (RHOA, NFKBIA and CCL3) were selected to construct the IRGPI. In a survival analysis using TCGA cohort as a training set, patients in the IRGPI-low subgroup had a better OS than those in IRGPI-high subgroup, consistent with the results in CGGA cohort. The comprehensive results showed that IRGPI-low subgroup had a more abundant activated immune cell population and lower TIDE score, higher MSI, higher TMB score, lower T cell dysfunction score, more likely benefit from ICIs therapy. IRGPI is a promising biomarker in the field of LGG ICIs therapy to distinguish the prognosis, the molecular and immunological characteristics of patients.
Collapse
Affiliation(s)
- Jing Zhou
- Department of Oncology, Shanxi Province Academy of Traditional Chinese MedicineShanxi Province Hospital of Traditional Chinese MedicineTaiyuanChina
| | - Hao Guo
- Department of AnesthesiologyShanxi Provincial People's HospitalTaiyuanChina
| | - Likun Liu
- Department of Oncology, Shanxi Province Academy of Traditional Chinese MedicineShanxi Province Hospital of Traditional Chinese MedicineTaiyuanChina
| | - Zengcai Jin
- Department of Oncology, Shanxi Province Academy of Traditional Chinese MedicineShanxi Province Hospital of Traditional Chinese MedicineTaiyuanChina
| | - Wencui Zhang
- Department of Oncology, Shanxi Province Academy of Traditional Chinese MedicineShanxi Province Hospital of Traditional Chinese MedicineTaiyuanChina
| | - Tao Tang
- Department of Integrated Traditional Chinese and Western Medicine, Institute of Integrative MedicineXiangya Hospital, Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| |
Collapse
|
23
|
Qin Y, Zhang H, Li Y, Xie T, Yan S, Wang J, Qu J, Ouyang F, Lv S, Guo Z, Wei H, Yu CY. Promotion of ICD via Nanotechnology. Macromol Biosci 2023; 23:e2300093. [PMID: 37114599 DOI: 10.1002/mabi.202300093] [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: 03/07/2023] [Revised: 04/17/2023] [Indexed: 04/29/2023]
Abstract
Immunotherapy represents the most promising treatment strategy for cancer, but suffers from compromised therapeutic efficiency due to low immune activity of tumor cells and an immunosuppressive microenvironment, which significantly hampers the clinical translations of this treatment strategy. To promote immunotherapy with desired therapeutic efficiency, immunogenic cell death (ICD), a particular type of death capable of reshaping body's antitumor immune activity, has drawn considerable attention due to the potential to stimulate a potent immune response. Still, the potential of ICD effect remains unsatisfactory because of the intricate tumor microenvironment and multiple drawbacks of the used inducing agents. ICD has been thoroughly reviewed so far with a general classification of ICD as a kind of immunotherapy strategy and repeated discussion of the related mechanism. However, there are no published reviews, to the authors' knowledge, providing a systematic summarization on the enhancement of ICD via nanotechnology. For this purpose, this review first discusses the four stages of ICD according to the development mechanisms, followed by a comprehensive description on the use of nanotechnology to enhance ICD in the corresponding four stages. The challenges of ICD inducers and possible solutions are finally summarized for future ICD-based enhanced immunotherapy.
Collapse
Affiliation(s)
- Yang Qin
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Haitao Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Yunxian Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Ting Xie
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Shuang Yan
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jiaqi Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Jun Qu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Feijun Ouyang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Shaoyang Lv
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Zifen Guo
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, 421001, China
| |
Collapse
|
24
|
Kim GB, Kim S, Hwang YH, Kim S, Lee I, Kim SA, Goo J, Yang Y, Jeong C, Nam GH, Kim IS. Harnessing Oncolytic Extracellular Vesicles for Tumor Cell-Preferential Cytoplasmic Delivery of Misfolded Proteins for Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2300527. [PMID: 37226374 DOI: 10.1002/smll.202300527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 05/12/2023] [Indexed: 05/26/2023]
Abstract
In this study, extracellular vesicles (EVs) are reimagined as more than just a cellular waste disposal system and are repurposed for cancer immunotherapy. Potent oncolytic EVs (bRSVF-EVs) loaded with misfolded proteins (MPs) are engineered, which are typically considered cellular debris. By impairing lysosomal function using bafilomycin A1 and expressing the respiratory syncytial virus F protein, a viral fusogen, MPs are successfully loaded into the EVs expressing RSVF. bRSVF-EVs preferentially transplant a xenogeneic antigen onto cancer cell membranes in a nucleolin-dependent manner, triggering an innate immune response. Furthermore, bRSVF-EV-mediated direct delivery of MPs into the cancer cell cytoplasm initiates endoplasmic reticulum stress and immunogenic cell death (ICD). This mechanism of action leads to substantial antitumor immune responses in murine tumor models. Importantly, when combined with PD-1 blockade, bRSVF-EV treatment elicits robust antitumor immunity, resulting in prolonged survival and complete remission in some cases. Overall, the findings demonstrate that utilizing tumor-targeting oncolytic EVs for direct cytoplasmic delivery of MPs to induce ICD in cancer cells represents a promising approach for enhancing durable antitumor immunity.
Collapse
Affiliation(s)
- Gi Beom Kim
- SHIFTBIO.INC, Seoul, 02751, Republic of Korea
| | | | - Yeong Ha Hwang
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Seohyun Kim
- SHIFTBIO.INC, Seoul, 02751, Republic of Korea
| | - Inkyu Lee
- SHIFTBIO.INC, Seoul, 02751, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Seong A Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jiyoung Goo
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyunghee University, Seoul, 02447, Republic of Korea
| | - Yoosoo Yang
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Cherlhyun Jeong
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyunghee University, Seoul, 02447, Republic of Korea
| | - Gi-Hoon Nam
- SHIFTBIO.INC, Seoul, 02751, Republic of Korea
- Department of Biochemistry and Molecular Biology, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - In-San Kim
- SHIFTBIO.INC, Seoul, 02751, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Chemical and Biological Integrative Research Center, Biomedical Research Division, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| |
Collapse
|
25
|
Wang Y, Wang W, Gu R, Chen J, Chen Q, Lin T, Wu J, Hu Y, Yuan A. In Situ Vaccination with Mitochondria-Targeting Immunogenic Death Inducer Elicits CD8 + T Cell-Dependent Antitumor Immunity to Boost Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300286. [PMID: 37127892 PMCID: PMC10369267 DOI: 10.1002/advs.202300286] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/28/2023] [Indexed: 05/03/2023]
Abstract
In situ vaccination can elicit systemic antitumor immunity to potentiate immune checkpoint blockade (ICB) in poorly immunogenic tumors. Herein, an immunogenic cell death (ICD) inducer for in situ vaccination, which is based on a mitochondria-targeting modification of fenofibric acid (FFa), a lipid-lowering drug with potential inhibitory efficacy of respiratory complex I is developed. Mitochondria-targeting FFa (Mito-FFa) inhibits complex I efficiently and increases mitochondrial ROS (mtROS) generation, which further triggers endoplasmic reticulum (ER) stress with unprecedented calreticulin (CRT) exposure on tumor cellular membranes. Moreover, the generated mtROS also oxidizes mitochondrial DNA (mtDNA) and promotes it leakage into the cytoplasm for cGAS-STING-dependent type I interferon (IFN-I) secretion. The synchronous CRT exposure and IFN-I secretion successively improve the uptake of tumor antigens, maturation of dendritic cells (DCs) and cross-priming of CD8+ T cells. In a poorly immunogenic 4T1 tumor model, a single intratumoral (i.t.) Mito-FFa injection turns immune-cold tumors into hot ones and elicits systemic tumor-specific CD8+ T cells responses against primary and metastatic tumors. Furthermore, the synergistic effect with PD-L1 blockade and good bio-safety of i.t. Mito-FFa administration suggest the great translational potential of Mito-FFa in tumor immunotherapy.
Collapse
Affiliation(s)
- Yuxiang Wang
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
| | - Weiran Wang
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
| | - Rong Gu
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
| | - Jing Chen
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
| | - Qian Chen
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
| | - Tingsheng Lin
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
- Department of UrologyNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNanjing210093P. R. China
| | - Jinhui Wu
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
- Jiangsu Key Laboratory for Nano TechnologyNanjing UniversityNanjing210093P. R. China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
- Jiangsu Key Laboratory for Nano TechnologyNanjing UniversityNanjing210093P. R. China
| | - Ahu Yuan
- State Key Laboratory of Pharmaceutical BiotechnologyMedical School and School of Life ScienceNanjing UniversityNanjing210093P. R. China
- Jiangsu Key Laboratory for Nano TechnologyNanjing UniversityNanjing210093P. R. China
| |
Collapse
|
26
|
Zhu X, Li S. Nanomaterials in tumor immunotherapy: new strategies and challenges. Mol Cancer 2023; 22:94. [PMID: 37312116 PMCID: PMC10262535 DOI: 10.1186/s12943-023-01797-9] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 05/31/2023] [Indexed: 06/15/2023] Open
Abstract
Tumor immunotherapy exerts its anti-tumor effects by stimulating and enhancing immune responses of the body. It has become another important modality of anti-tumor therapy with significant clinical efficacy and advantages compared to chemotherapy, radiotherapy and targeted therapy. Although various kinds of tumor immunotherapeutic drugs have emerged, the challenges faced in the delivery of these drugs, such as poor tumor permeability and low tumor cell uptake rate, had prevented their widespread application. Recently, nanomaterials had emerged as a means for treatment of different diseases due to their targeting properties, biocompatibility and functionalities. Moreover, nanomaterials possess various characteristics that overcome the defects of traditional tumor immunotherapy, such as large drug loading capacity, precise tumor targeting and easy modification, thus leading to their wide application in tumor immunotherapy. There are two main classes of novel nanoparticles mentioned in this review: organic (polymeric nanomaterials, liposomes and lipid nanoparticles) and inorganic (non-metallic nanomaterials and metallic nanomaterials). Besides, the fabrication method for nanoparticles, Nanoemulsions, was also introduced. In summary, this review article mainly discussed the research progress of tumor immunotherapy based on nanomaterials in the past few years and offers a theoretical basis for exploring novel tumor immunotherapy strategies in the future.
Collapse
Affiliation(s)
- Xudong Zhu
- Department of General Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China
| | - Shenglong Li
- Second Ward of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, People's Republic of China.
| |
Collapse
|
27
|
Zhou Z, Zhang H, Tao Y, Jie H, Zhao J, Zang J, Li H, Wang Y, Wang T, Zhao H, Li Y, Guo C, Zhu F, Mao H, Zhang L, Liu F, Wang Q. CX3CR1 hi macrophages sustain metabolic adaptation by relieving adipose-derived stem cell senescence in visceral adipose tissue. Cell Rep 2023; 42:112424. [PMID: 37086405 DOI: 10.1016/j.celrep.2023.112424] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 10/20/2022] [Accepted: 04/06/2023] [Indexed: 04/23/2023] Open
Abstract
Adipose-derived stem cells (ASCs) drive healthy visceral adipose tissue (VAT) expansion via adipocyte hyperplasia. Obesity induces ASC senescence that causes VAT dysfunction and metabolic disorders. It is challenging to restrain this process by biological intervention, as mechanisms of controlling VAT ASC senescence remain unclear. We demonstrate that a population of CX3CR1hi macrophages is maintained in mouse VAT during short-term energy surplus, which sustains ASCs by restraining their senescence, driving adaptive VAT expansion and metabolic health. Long-term overnutrition induces diminishment of CX3CR1hi macrophages in mouse VAT accompanied by ASC senescence and exhaustion, while transferring CX3CR1hi macrophages restores ASC reservoir and triggers VAT beiging to alleviate the metabolic maladaptation. Mechanistically, visceral ASCs attract macrophages via MCP-1 and shape their CX3CR1hi phenotype via exosomes; these macrophages relieve ASC senescence by promoting the arginase1-eIF5A hypusination axis. These findings identify VAT CX3CR1hi macrophages as ASC supporters and unravel their therapeutic potential for metabolic maladaptation to obesity.
Collapse
Affiliation(s)
- Zixin Zhou
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Huiying Zhang
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yan Tao
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Haipeng Jie
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jingyuan Zhao
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Jinhao Zang
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Huijie Li
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yalin Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Tianci Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Hui Zhao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Yuan Li
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China; Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Chun Guo
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Faliang Zhu
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Haiting Mao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Lining Zhang
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Fengming Liu
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Qun Wang
- Key Laboratory of Infection and Immunity of Shandong Province, Shandong Provincial Clinical Research Center for Immune Diseases and Gout, Department of Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China.
| |
Collapse
|
28
|
Barcelo J, Samain R, Sanz-Moreno V. Preclinical to clinical utility of ROCK inhibitors in cancer. Trends Cancer 2023; 9:250-263. [PMID: 36599733 DOI: 10.1016/j.trecan.2022.12.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/25/2022] [Accepted: 12/02/2022] [Indexed: 01/03/2023]
Abstract
ROCK belongs to the AGC family of Ser/Thr protein kinases that are involved in many cellular processes. ROCK-driven actomyosin contractility regulates cytoskeletal dynamics underpinning cell migration, proliferation, and survival in many cancer types. ROCK1/2 play key protumorigenic roles in several subtypes and stages of cancer development. Therefore, successfully targeting ROCK and its downstream effectors presents an interesting avenue for cancer treatment. Because local use of ROCK inhibitors will reduce the side effects of systemic administration, we propose different therapeutic strategies and latest-generation ROCK inhibitors for use in the clinic.
Collapse
Affiliation(s)
- Jaume Barcelo
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Remi Samain
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | | |
Collapse
|
29
|
Mishra AK, Banday S, Bharadwaj R, Ali A, Rashid R, Kulshreshtha A, Malonia SK. Macrophages as a Potential Immunotherapeutic Target in Solid Cancers. Vaccines (Basel) 2022; 11:55. [PMID: 36679900 PMCID: PMC9863216 DOI: 10.3390/vaccines11010055] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/17/2022] [Accepted: 12/18/2022] [Indexed: 12/28/2022] Open
Abstract
The revolution in cancer immunotherapy over the last few decades has resulted in a paradigm shift in the clinical care of cancer. Most of the cancer immunotherapeutic regimens approved so far have relied on modulating the adaptive immune system. In recent years, strategies and approaches targeting the components of innate immunity have become widely recognized for their efficacy in targeting solid cancers. Macrophages are effector cells of the innate immune system, which can play a crucial role in the generation of anti-tumor immunity through their ability to phagocytose cancer cells and present tumor antigens to the cells of adaptive immunity. However, the macrophages that are recruited to the tumor microenvironment predominantly play pro-tumorigenic roles. Several strategies targeting pro-tumorigenic functions and harnessing the anti-tumorigenic properties of macrophages have shown promising results in preclinical studies, and a few of them have also advanced to clinical trials. In this review, we present a comprehensive overview of the pathobiology of TAMs and their role in the progression of solid malignancies. We discuss various mechanisms through which TAMs promote tumor progression, such as inflammation, genomic instability, tumor growth, cancer stem cell formation, angiogenesis, EMT and metastasis, tissue remodeling, and immunosuppression, etc. In addition, we also discuss potential therapeutic strategies for targeting TAMs and explore how macrophages can be used as a tool for next-generation immunotherapy for the treatment of solid malignancies.
Collapse
Affiliation(s)
- Alok K. Mishra
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Shahid Banday
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Ravi Bharadwaj
- Department of Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Amjad Ali
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Romana Rashid
- Department of Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Ankur Kulshreshtha
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Sunil K. Malonia
- Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
30
|
Chen Z, Yue Z, Yang K, Li S. Nanomaterials: small particles show huge possibilities for cancer immunotherapy. J Nanobiotechnology 2022; 20:484. [DOI: 10.1186/s12951-022-01692-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 10/27/2022] [Indexed: 11/17/2022] Open
Abstract
AbstractWith the economy's globalization and the population's aging, cancer has become the leading cause of death in most countries. While imposing a considerable burden on society, the high morbidity and mortality rates have continuously prompted researchers to develop new oncology treatment options. Anti-tumor regimens have evolved from early single surgical treatment to combined (or not) chemoradiotherapy and then to the current stage of tumor immunotherapy. Tumor immunotherapy has undoubtedly pulled some patients back from the death. However, this strategy of activating or boosting the body's immune system hardly benefits most patients. It is limited by low bioavailability, low response rate and severe side effects. Thankfully, the rapid development of nanotechnology has broken through the bottleneck problem of anti-tumor immunotherapy. Multifunctional nanomaterials can not only kill tumors by combining anti-tumor drugs but also can be designed to enhance the body's immunity and thus achieve a multi-treatment effect. It is worth noting that the variety of nanomaterials, their modifiability, and the diversity of combinations allow them to shine in antitumor immunotherapy. In this paper, several nanobiotics commonly used in tumor immunotherapy at this stage are discussed, and they activate or enhance the body's immunity with their unique advantages. In conclusion, we reviewed recent advances in tumor immunotherapy based on nanomaterials, such as biological cell membrane modification, self-assembly, mesoporous, metal and hydrogels, to explore new directions and strategies for tumor immunotherapy.
Collapse
|
31
|
Advantage of extracellular vesicles in hindering the CD47 signal for cancer immunotherapy. J Control Release 2022; 351:727-738. [DOI: 10.1016/j.jconrel.2022.09.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 11/20/2022]
|
32
|
Guo HL, Xie XY, Xu M. Application of nanomaterials in combined thermal ablation and immunotherapy for liver tumors. Shijie Huaren Xiaohua Zazhi 2022; 30:829-837. [DOI: 10.11569/wcjd.v30.i19.829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Thermal ablation is one of the important treatments for liver tumors, but the postoperative recurrence rate is high. Thermal ablation has been reported to trigger the release of tumor-associated antigens, which in turn initiates antitumor immune response. However, this anti-tumor immune effect cannot effectively suppress tumor recurrence due to the obstacles of antigen presentation, the formation of tumor-suppressive immune microenvironment, and the hypoxic and hypovascular tumor microenvironment. Therefore, using immunotherapy to enhance the antitumor immune effect after thermal ablation is a potential strategy to improve the prognosis of tumor patients. However, free immune drugs have the disadvantages of poor targeting and short half-life. Nanomaterials have the advantages of strong modifiability, controllable drug ratio, and excellent targeting. Based on the characteristics of the tumor immune microenvironment after thermal ablation, scholars have designed nano-immunopharmaceuticals that can increase the tumor permeability of immune drugs, stimulate antigen presentation, and reshape the tumor immune microenvironment. This review focuses on the role of nanomaterials in tumor ablation combined with immunotherapy for liver tumors.
Collapse
Affiliation(s)
- Huan-Ling Guo
- Department of Medical Ultrasonics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province. China
| | - Xiao-Yan Xie
- Department of Medical Ultrasonics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province. China
| | - Ming Xu
- Department of Medical Ultrasonics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, Guangdong Province. China
| |
Collapse
|
33
|
Li X, Gao Y, Liu X, Hu X, Li Y, Sun J, Wang P, Wu H, Kim H, Ramalingam M, Xie S, Wang R. Ultrasound and laser-promoted dual-gas nano-generator for combined photothermal and immune tumor therapy. Front Bioeng Biotechnol 2022; 10:1005520. [PMID: 36177188 PMCID: PMC9513372 DOI: 10.3389/fbioe.2022.1005520] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/19/2022] [Indexed: 11/28/2022] Open
Abstract
The combination of photothermal therapy (PTT) and immune tumor therapy has emerged as a promising avenue for cancer treatment. However, the insufficient immune response caused by inefficient immunogenic cell death (ICD) inducers and thermal resistance, immunosuppression, and immune escape resulting from the hypoxic microenvironment of solid tumors severely limit its efficacy. Herein, we report an ultrasound and laser-promoted dual-gas nano-generator (calcium carbonate-polydopamine-manganese oxide nanoparticles, CPM NPs) for enhanced photothermal/immune tumor therapy through reprogramming tumor hypoxic microenvironment. In this system, CPM NPs undergo reactive decomposition in a moderately acidic tumor, resulting in the generation of calcium, manganese ions, carbon dioxide (CO2), and oxygen (O2). Calcium and manganese ions act as adjuvants that trigger an immune response. The cancer cell membrane rupture caused by sudden burst of bubbles (CO2 and O2) under ultrasound stimulation and the photothermal properties of PDA also contributed to the ICD effect. The generation of O2 alleviates tumor hypoxia and thus reduces hypoxia-induced heat resistance and immunosuppressive effects, thereby improving the therapeutic efficacy of combination PTT and immune therapy. The present study provides a novel approach for the fabrication of a safe and effective tumor treatment platform for future clinical applications.
Collapse
Affiliation(s)
- XinYu Li
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai, China
- Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai, China
| | - Yong Gao
- Binzhou Medical University Hospital, Binzhou, China
| | - XinZheng Liu
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai, China
- Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai, China
| | - XiaoQian Hu
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai, China
| | - YunMeng Li
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai, China
| | - JunXi Sun
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai, China
| | - PingYu Wang
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai, China
| | - Hongkai Wu
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong, China
| | - HaeWon Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Korea
- Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Korea
| | - Murugan Ramalingam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, Korea
- Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, Korea
- School of Basic Medical Sciences, Chengdu University, Chengdu, China
- *Correspondence: Murugan Ramalingam, ; ShuYang Xie, ; RanRan Wang,
| | - ShuYang Xie
- Key Laboratory of Tumor Molecular Biology, Binzhou Medical University, Yantai, China
- *Correspondence: Murugan Ramalingam, ; ShuYang Xie, ; RanRan Wang,
| | - RanRan Wang
- Institute of Rehabilitation Medicine, School of Rehabilitation Medicine, Binzhou Medical University, Yantai, China
- Shandong Laboratory of Advanced Materials and Green Manufacturing, Yantai, China
- *Correspondence: Murugan Ramalingam, ; ShuYang Xie, ; RanRan Wang,
| |
Collapse
|
34
|
Zhu C, Fang Z, Peng L, Gao F, Peng W, Song F. Curcumin Suppresses the Progression of Colorectal Cancer by Improving Immunogenic Cell Death Caused by Irinotecan. Chemotherapy 2022; 67:211-222. [DOI: 10.1159/000518121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/27/2021] [Indexed: 11/19/2022]
Abstract
<b><i>Background:</i></b> Irinotecan (IRI) is a common chemotherapeutic drug for colorectal cancer; however, the mechanism underlying its immunomodulatory effect remains unclear. Curcumin (CUR), an adjuvant drug with anti-inflammatory and antitumor effects, has been studied extensively, although its synergistic antitumor effect remains unclear. <b><i>Methods:</i></b> The effects of CUR and IRI on oxidative stress and their antitumor effects were detected by flow cytometry. Endoplasmic reticulum stress-related proteins including CHOP and BiP, and immunogenic cell death (ICD) proteins including calreticulin (CALR) and high mobility group box 1 (HMGB1), were detected by Western blotting. IFN-γ and TNF-α levels in the serum of mice were detected by ELISA. <b><i>Results:</i></b> IRI in combination with CUR had synergistic antitumor effects in CT-26 colon carcinoma cells. Combination treatment with IRI and CUR was more effective than IRI or CUR alone. IRI and CUR combination treatment significantly upregulated ICD-related proteins including CALR and HMGB1 and had a greater antitumor effect than IRI or CUR single treatment in vivo. CUR may synergistically improve the antitumor effect of IRI by promoting the ICD effect. <b><i>Conclusion:</i></b> Combination therapy with IRI and CUR may be an option for first-line chemotherapy in some patients with advanced colorectal cancer.
Collapse
|
35
|
Dang Q, Liu Z, Liu Y, Wang W, Yuan W, Sun Z, Liu L, Wang C. LncRNA profiles from Notch signaling: Implications for clinical management and tumor microenvironment of colorectal cancer. Front Immunol 2022; 13:953405. [PMID: 35958606 PMCID: PMC9359081 DOI: 10.3389/fimmu.2022.953405] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/28/2022] [Indexed: 12/29/2022] Open
Abstract
The interplay between long non-coding RNAs (lncRNAs) and the Notch pathway involves a variety of malignancies. However, Notch-derived lncRNAs and their latent clinical significance remain elusive in colorectal cancer (CRC). In this study, we introduced a framework that could screen Notch-derived lncRNAs (named “NLncer”) and ultimately identified 24 NLncers. To further explore the clinical significance of these NLncers, we performed LASSO and Cox regression in TCGA-CRC cohort (n = 584) and then retained six lncRNAs tightly associated with prognosis. The final model (termed “NLncS”) was subsequently tested in GSE38832 (n = 122), GSE39582 (n = 573), and an in-house clinical cohort (n = 115). Ultimately, our NLncS model could serve as an independent risk factor and afford a robust performance for assessing the prognosis of CRC patients. Additionally, patients with high NLncS risk scores were characterized by upregulation of immune pathways, strong immunogenicity, abundant CD8 + T-cell infiltration, and potentially higher response rates to CTLA4 blockers, which turned out to be suitable for immunotherapy. Aiming at globally observing the characteristics of high-risk patients, somatic mutation and methylation modification analysis provide us with evidence at the genomic and transcriptomic levels. To facilitate the clinical transformability, we mined deeply into the sensitive compounds targeting high-risk individuals and identified dasatinib as a candidate agent for patients with a high Notch risk score. In conclusion, our NLncS model is a promising biomarker for optimizing the clinical management of CRC patients.
Collapse
Affiliation(s)
- Qin Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Liu
- Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China
| | - Wenkang Wang
- Department of Breast Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Chengzeng Wang, ; Lin Liu, ; Zhenqiang Sun,
| | - Lin Liu
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Chengzeng Wang, ; Lin Liu, ; Zhenqiang Sun,
| | - Chengzeng Wang
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Chengzeng Wang, ; Lin Liu, ; Zhenqiang Sun,
| |
Collapse
|
36
|
Rho kinase inhibition ameliorates vascular remodeling and blood pressure elevations in a rat model of apatinib-induced hypertension. J Hypertens 2022; 40:675-684. [PMID: 34862331 PMCID: PMC8901036 DOI: 10.1097/hjh.0000000000003060] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES Hypertension is one of the major adverse effects of tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factors. However, the mechanism underlying TKIs-induced hypertension remains unclear. Here, we explored the role of the RhoA/Rho kinase (ROCK) signaling pathway in elevation of blood pressure (BP) induced by apatinib, a selective TKI approved in China for treatment of advanced or metastatic gastric cancer. A nonspecific ROCK inhibitor, Y27632, was then combined with apatinib and its efficacy in alleviating apatinib-induced hypertension was evaluated. METHODS Normotensive female Wistar-Kyoto rats were exposed to two different doses of apatinib, or apatinib combined with Y27632, or vehicle for 2 weeks. BP was monitored by a tail-cuff plethysmography system. The mRNA levels and protein expression in the RhoA/ROCK pathway were determined, and vascular remodeling assessed. RESULTS Administration of either a high or low dose of apatinib was associated with a rapid rise in BP, reaching a plateau after 12 days. Apatinib treatment mediated upregulation of RhoA and ROCK II in the mid-aorta, more significant in the high-dose group. However, ROCK I expression showed no statistically significant differences. Furthermore, the mRNA level of GRAF3 decreased dose-dependently. Apatinib administration was also associated with decreased levels of MLCP, and elevated endothelin-1 (ET-1) and collagen I, which were accompanied with increased mid-aortic media. However, treatment with Y27632 attenuated the above changes. CONCLUSION These findings suggest that activation of the RhoA/ROCK signaling pathway could be the underlying mechanism of apatinib-induced hypertension, while ROCK inhibitor have potential therapeutic value.
Collapse
|
37
|
Leveraging cellular mechano-responsiveness for cancer therapy. Trends Mol Med 2021; 28:155-169. [PMID: 34973934 DOI: 10.1016/j.molmed.2021.11.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 12/21/2022]
Abstract
Cells sense the biophysical properties of the tumor microenvironment (TME) and adopt these signals in their development, progression, and metastatic dissemination. Recent work highlights the mechano-responsiveness of cells in tumors and the underlying mechanisms. Furthermore, approaches to mechano-modulating diverse types of cell have emerged aiming to inhibit tumor growth and metastasis. These include targeting mechanosensitive machineries in cancer cells to induce apoptosis, intervening matrix stiffening incurred by cancer-associated fibroblasts (CAFs) in both primary and metastatic tumor sites, and modulating matrix mechanics to improve immune cell therapeutic efficacy. This review is envisaged to help scientists and clinicians in cancer research to advance understanding of the cellular mechano-responsiveness in TME, and to harness these concepts for cancer mechanotherapies.
Collapse
|
38
|
Chen M, Tan Y, Hu J, Jiang Y, Wang Z, Liu Z, Chen Q. Injectable Immunotherapeutic Thermogel for Enhanced Immunotherapy Post Tumor Radiofrequency Ablation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2104773. [PMID: 34729889 DOI: 10.1002/smll.202104773] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/03/2021] [Indexed: 06/13/2023]
Abstract
Tumor radiofrequency ablation (RFA) is a local and minimally invasive application using high temperature to induce coagulative necrosis of tumor, which has been commonly used in clinic. Although the tumor fragments generated by RFA can activate the host's immune system, it may be insufficient to inhibit cancer recurrence due to many factors such as the inefficient antigen presentation by dendritic cells (DCs). In this research, a convenient local administration strategy by blocking rho-associated kinases (ROCK) is applied to amplify the immune responses triggered by RFA via promoting the phagocytosis capacity of DCs. Briefly, ROCK inhibitor, Y27632, is successfully dispersed in the amphiphilic copolymer poly(D,L-lactide-co-glycolide)-b-poly(ethyleneglycol)-b-poly(D,L-lactideco-glycolide) (PLGA-PEG-PLGA) solution, which is sol at room temperature and forms hydrogel quickly at body temperature, obviously prolonging the retention of Y27632 after injection. Interestingly, in the melanoma tumor model, the generated tumor fragments after RFA treatment are swallowed by DCs and undergo reinforced antigen presentation process with the help of gradual released Y27632, further effectively activating T cell mediated anti-tumor immune responses and significantly improving the therapeutic efficiency of RFA. Overall, such strategy remarkably prolongs the survival of mice after RFA treatment, showing great potential for clinical translation as an improvement strategy for RFA.
Collapse
Affiliation(s)
- Muchao Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Yanjun Tan
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Jiaying Hu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Yanping Jiang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Zixian Wang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| | - Qian Chen
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu, 215123, P. R. China
| |
Collapse
|
39
|
Kwon M, Nam GH, Jung H, Kim SA, Kim S, Choi Y, Lee YS, Cho HJ, Kim IS. Statin in combination with cisplatin makes favorable tumor-immune microenvironment for immunotherapy of head and neck squamous cell carcinoma. Cancer Lett 2021; 522:198-210. [PMID: 34571082 DOI: 10.1016/j.canlet.2021.09.029] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/29/2021] [Accepted: 09/19/2021] [Indexed: 12/23/2022]
Abstract
The purpose of this study was to determine whether statins can enhance anticancer effects in head and neck squamous cell carcinoma (HNSCC) when used with cisplatin and act as immunogenic cell death (ICD) inducers that can be used in cancer immunotherapy. Statins alone showed both in vitro and in vivo inhibitory effects against HNSCC, and synergistic antitumor effects were observed when combined with cisplatin in a syngeneic murine HNSCC model. Statins increased calreticulin exposure and endoplasmic reticulum stress-related signals in HNSCC cells. In addition, it was confirmed that statins could activate antigen-presenting cells and tumor-specific CD8+ T cells with an increase in their numbers in the tumor tissues and draining lymph nodes, with this effect showing significant improvement following the combination therapy with cisplatin. Moreover, in triple combination with both cisplatin and anti-programmed cell death 1 receptor (anti-PD-1) antibody, statins dramatically induced further tumor eradication and improved the survival of tumor-bearing mice. Taken together, these results demonstrate that statins, administered in combination with anti-PD-1 antibody, could enhance the anticancer effect of cisplatin and potentiate the efficacy of immunotherapy for HNSCC and present a rationale for repurposing statins as an adjuvant immunotherapeutic option for HNSCC.
Collapse
Affiliation(s)
- Minsu Kwon
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Anam Hospital, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
| | - Gi-Hoon Nam
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02456, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute Science and Technology (KIST), Seoul, 02456, Republic of Korea
| | - Hanul Jung
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Anam Hospital, Korea University College of Medicine, Seoul, 02841, Republic of Korea
| | - Seong A Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02456, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute Science and Technology (KIST), Seoul, 02456, Republic of Korea
| | - Seohyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02456, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute Science and Technology (KIST), Seoul, 02456, Republic of Korea
| | - Yeonju Choi
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Yoon Se Lee
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - Hyo Jung Cho
- Department of Gastroenterology, Ajou University Hospital, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02456, Republic of Korea; Center for Theragnosis, Biomedical Research Institute, Korea Institute Science and Technology (KIST), Seoul, 02456, Republic of Korea.
| |
Collapse
|
40
|
Kim S, Kim SA, Han J, Kim IS. Rho-Kinase as a Target for Cancer Therapy and Its Immunotherapeutic Potential. Int J Mol Sci 2021; 22:ijms222312916. [PMID: 34884721 PMCID: PMC8657458 DOI: 10.3390/ijms222312916] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/24/2021] [Accepted: 11/24/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy is fast rising as a prominent new pillar of cancer treatment, harnessing the immune system to fight against numerous types of cancer. Rho-kinase (ROCK) pathway is involved in diverse cellular activities, and is therefore the target of interest in various diseases at the cellular level including cancer. Indeed, ROCK is well-known for its involvement in the tumor cell and tumor microenvironment, especially in its ability to enhance tumor cell progression, migration, metastasis, and extracellular matrix remodeling. Importantly, ROCK is also considered to be a novel and effective modulator of immune cells, although further studies are needed. In this review article, we describe the various activities of ROCK and its potential to be utilized in cancer treatment, particularly in cancer immunotherapy, by shining a light on its activities in the immune system.
Collapse
Affiliation(s)
- Seohyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea; (S.K.); (S.A.K.); (J.H.)
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Seong A. Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea; (S.K.); (S.A.K.); (J.H.)
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Jihoon Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea; (S.K.); (S.A.K.); (J.H.)
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea; (S.K.); (S.A.K.); (J.H.)
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
- Correspondence:
| |
Collapse
|
41
|
Targeting small GTPases and their downstream pathways with intracellular macromolecule binders to define alternative therapeutic strategies in cancer. Biochem Soc Trans 2021; 49:2021-2035. [PMID: 34623375 DOI: 10.1042/bst20201059] [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: 07/21/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 11/17/2022]
Abstract
The RAS superfamily of small GTPases regulates major physiological cellular processes. Mutation or deregulation of these small GTPases, their regulators and/or their effectors are associated with many diseases including cancer. Hence, targeting these classes of proteins is an important therapeutic strategy in cancer. This has been recently achieved with the approval of the first KRASG12C covalent inhibitors for the clinic. However, many other mutants and small GTPases are still considered as 'undruggable' with small molecule inhibitors because of a lack of well-defined pocket(s) at their surface. Therefore, alternative therapeutic strategies have been developed to target these proteins. In this review, we discuss the use of intracellular antibodies and derivatives - reagents that bind their antigen inside the cells - for the discovery of novel inhibitory mechanisms, targetable features and therapeutic strategies to inhibit small GTPases and their downstream pathways. These reagents are also versatile tools used to better understand the biological mechanisms regulated by small GTPases and to accelerate the drug discovery process.
Collapse
|
42
|
Crosas-Molist E, Samain R, Kohlhammer L, Orgaz J, George S, Maiques O, Barcelo J, Sanz-Moreno V. RhoGTPase Signalling in Cancer Progression and Dissemination. Physiol Rev 2021; 102:455-510. [PMID: 34541899 DOI: 10.1152/physrev.00045.2020] [Citation(s) in RCA: 140] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rho GTPases are a family of small G proteins that regulate a wide array of cellular processes related to their key roles controlling the cytoskeleton. On the other hand, cancer is a multi-step disease caused by the accumulation of genetic mutations and epigenetic alterations, from the initial stages of cancer development when cells in normal tissues undergo transformation, to the acquisition of invasive and metastatic traits, responsible for a large number of cancer related deaths. In this review, we discuss the role of Rho GTPase signalling in cancer in every step of disease progression. Rho GTPases contribute to tumour initiation and progression, by regulating proliferation and apoptosis, but also metabolism, senescence and cell stemness. Rho GTPases play a major role in cell migration, and in the metastatic process. They are also involved in interactions with the tumour microenvironment and regulate inflammation, contributing to cancer progression. After years of intensive research, we highlight the importance of relevant models in the Rho GTPase field, and we reflect on the therapeutic opportunities arising for cancer patients.
Collapse
Affiliation(s)
- Eva Crosas-Molist
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Remi Samain
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Leonie Kohlhammer
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jose Orgaz
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom.,Instituto de Investigaciones Biomédicas 'Alberto Sols', CSIC-UAM, 28029, Madrid, Spain
| | - Samantha George
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Oscar Maiques
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Jaume Barcelo
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
43
|
Martin TD, Patel RS, Cook DR, Choi MY, Patil A, Liang AC, Li MZ, Haigis KM, Elledge SJ. The adaptive immune system is a major driver of selection for tumor suppressor gene inactivation. Science 2021; 373:1327-1335. [PMID: 34529489 DOI: 10.1126/science.abg5784] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
During tumorigenesis, tumors must evolve to evade the immune system and do so by disrupting the genes involved in antigen processing and presentation or up-regulating inhibitory immune checkpoint genes. We performed in vivo CRISPR screens in syngeneic mouse tumor models to examine requirements for tumorigenesis both with and without adaptive immune selective pressure. In each tumor type tested, we found a marked enrichment for the loss of tumor suppressor genes (TSGs) in the presence of an adaptive immune system relative to immunocompromised mice. Nearly one-third of TSGs showed preferential enrichment, often in a cancer- and tissue-specific manner. These results suggest that clonal selection of recurrent mutations found in cancer is driven largely by the tumor’s requirement to avoid the adaptive immune system.
Collapse
MESH Headings
- Animals
- CRISPR-Cas Systems
- Carcinogenesis
- Cell Line, Tumor
- Chemokine CCL2/metabolism
- Female
- GTP-Binding Protein alpha Subunits, G12-G13/genetics
- GTP-Binding Protein alpha Subunits, G12-G13/metabolism
- Gene Silencing
- Genes, Tumor Suppressor
- Humans
- Immune Evasion/genetics
- Mammary Neoplasms, Experimental/genetics
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, SCID
- Neoplasm Transplantation
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Selection, Genetic
- Tumor Microenvironment
Collapse
Affiliation(s)
- Timothy D Martin
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Rupesh S Patel
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Danielle R Cook
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Mei Yuk Choi
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Ajinkya Patil
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Anthony C Liang
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Mamie Z Li
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Kevin M Haigis
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Stephen J Elledge
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
44
|
New insights into exosome mediated tumor-immune escape: Clinical perspectives and therapeutic strategies. Biochim Biophys Acta Rev Cancer 2021; 1876:188624. [PMID: 34487817 DOI: 10.1016/j.bbcan.2021.188624] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 12/13/2022]
Abstract
Recent advances in extracellular vesicle biology have uncovered a substantial role in maintaining cell homeostasis in health and disease conditions by mediating intercellular communication, thus catching the scientific community's attention worldwide. Extracellular microvesicles, some called exosomes, functionally transfer biomolecules such as proteins and non-coding RNAs from one cell to another, influencing the local environment's biology. Although numerous advancements have been made in treating cancer patients with immune therapy, controlling the disease remains a challenge in the clinic due to tumor-driven interference with the immune response and inability of immune cells to clear cancer cells from the body. The present review article discusses the recent findings and knowledge gaps related to the role of exosomes derived from tumors and the tumor microenvironment cells in tumor escape from immunosurveillance. Further, we highlight examples where exosomal non-coding RNAs influence immune cells' response within the tumor microenvironment and favor tumor growth and progression. Therefore, exosomes can be used as a therapeutic target for the treatment of human cancers.
Collapse
|
45
|
Choi Y, Nam GH, Kim GB, Kim S, Kim YK, Kim SA, Kim HJ, Lee EJ, Kim IS. Nanocages displaying SIRP gamma clusters combined with prophagocytic stimulus of phagocytes potentiate anti-tumor immunity. Cancer Gene Ther 2021; 28:960-970. [PMID: 34349240 DOI: 10.1038/s41417-021-00372-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/10/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
Antigen-presenting cells (APCs), including macrophages and dendritic cells (DCs), play a crucial role in bridging innate and adaptive immunity; thereby, innate immune checkpoint blockade-based therapy is an attractive approach for the induction of sustainable tumor-specific immunity. The interaction between the cluster of differentiation 47 (CD47) on tumor and signal-regulatory protein alpha (SIRPα) on phagocytic cells inhibits the phagocytic function of APCs, acting as a "don't eat me" signal. Accordingly, CD47 blockade is known to increase tumor cell phagocytosis, eliciting tumor-specific CD8+ T-cell immunity. Here, we introduced a nature-derived nanocage to deliver SIRPγ for blocking of antiphagocytic signaling through binding to CD47 and combined it with prophagocytic stimuli using a metabolic reprogramming reagent for APCs (CpG-oligodeoxynucleotides). Upon delivering the clustered SIRPγ variant, the nanocage showed enhanced CD47 binding profiles on tumor cells, thereby promoting active engulfment by phagocytes. Moreover, combination with CpG potentiated the prophagocytic ability, leading to the establishment of antitumorigenic surroundings. This combination treatment could competently inhibit tumor growth by invigorating APCs and CD8+ T-cells in TMEs in B16F10 orthotopic tumor models, known to be resistant to CD47-targeting therapeutics. Collectively, enhanced delivery of an innate immune checkpoint antagonist with metabolic modulation stimuli of immune cells could be a promising strategy for arousing immune responses against cancer.
Collapse
Affiliation(s)
- Yoonjeong Choi
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Gi-Hoon Nam
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gi Beom Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Seohyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Yoon Kyoung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Seong A Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ha-Jeong Kim
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Jung Lee
- Department of Chemical Engineering, School of Applied Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea. .,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea.
| |
Collapse
|
46
|
Zhou J, Yang Q, Lu L, Tuo Z, Shou Z, Cheng J. PLK1 Inhibition Induces Immunogenic Cell Death and Enhances Immunity against NSCLC. Int J Med Sci 2021; 18:3516-3525. [PMID: 34522178 PMCID: PMC8436107 DOI: 10.7150/ijms.60135] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/02/2021] [Indexed: 01/22/2023] Open
Abstract
PLK1 inhibitors were shown, in vitro and in vivo, to possess inhibitory activities against non-small cell lung cancer (NSCLC), and such inhibition has been proven by clinical trials. However, it remains unclear whether and how the immune microenvironment is associated with the action. In this study, we found that inhibiting PLK1 could alter the tumor immune microenvironment by increasing DC maturation, and enriching T cells infiltration. PLK1 inhibitors, serving as immunogenic cell death (ICD) inducers, indirectly activated DCs, instead of directly acting on DC cells, through the surface expression of costimulatory molecules on and enhanced phagocytosis by DCs. Furthermore, upon targeting PLK1, tumor cells that had undergone ICD were converted into an endogenous vaccine, which triggered the immune memory responses and protected the mice from tumor challenge. Collectively, these results suggested that the PLK1 inhibitor might function as an immune modulator in antitumor treatment.
Collapse
Affiliation(s)
- Jie Zhou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P. R. China
| | - Qifan Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P. R. China
| | - Lisen Lu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P. R. China
| | - Zhan Tuo
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P. R. China
| | - Zhexing Shou
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P. R. China
| | - Jing Cheng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P. R. China
| |
Collapse
|
47
|
Nam GH, Kwon M, Jung H, Ko E, Kim SA, Choi Y, Song SJ, Kim S, Lee Y, Kim GB, Han J, Woo J, Cho Y, Jeong C, Park SY, Roberts TM, Cho YB, Kim IS. Statin-mediated inhibition of RAS prenylation activates ER stress to enhance the immunogenicity of KRAS mutant cancer. J Immunother Cancer 2021; 9:jitc-2021-002474. [PMID: 34330763 PMCID: PMC8327837 DOI: 10.1136/jitc-2021-002474] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2021] [Indexed: 12/16/2022] Open
Abstract
Background Statins preferentially promote tumor-specific apoptosis by depleting isoprenoid such as farnesyl pyrophosphate and geranylgeranyl pyrophosphate. However, statins have not yet been approved for clinical cancer treatment due, in part, to poor understanding of molecular determinants on statin sensitivity. Here, we investigated the potential of statins to elicit enhanced immunogenicity of KRAS-mutant (KRASmut) tumors. Methods The immunogenicity of treated cancer cells was determined by western blot, flow cytometry and confocal microscopy. The immunotherapeutic efficacy of mono or combination therapy using statin was assessed in KRASmut tumor models, including syngeneic colorectal cancer and genetically engineered lung and pancreatic tumors. Using NanoString analysis, we analyzed how statin influenced the gene signatures associated with the antigen presentation of dendritic cells in vivo and evaluated whether statin could induce CD8+ T-cell immunity. Multiplex immunohistochemistry was performed to better understand the complicated tumor-immune microenvironment. Results Statin-mediated inhibition of KRAS prenylation provoked severe endoplasmic reticulum (ER) stress by attenuating the anti-ER stress effect of KRAS mutation, thereby resulting in the immunogenic cell death (ICD) of KRASmut cancer cells. Moreover, statin-mediated ICD enhanced the cross-priming ability of dendritic cells, thereby provoking CD8+ T-cell immune responses against KRASmut tumors. Combination therapy using statin and oxaliplatin, an ICD inducer, significantly enhanced the immunogenicity of KRASmut tumors and promoted tumor-specific immunity in syngeneic and genetically engineered KRASmut tumor models. Along with immune-checkpoint inhibitors, the abovementioned combination therapy overcame resistance to PD-1 blockade therapies, improving the survival rate of KRASmut tumor models. Conclusions Our findings suggest that KRAS mutation could be a molecular target for statins to elicit potent tumor-specific immunity.
Collapse
Affiliation(s)
- Gi-Hoon Nam
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Minsu Kwon
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Hanul Jung
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Eunbyeol Ko
- Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Seong A Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Yoonjeong Choi
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Su Jeong Song
- Institute for Future Medicine, Samsung Medical Center, Seoul, Republic of Korea
| | - Seohyun Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Yeji Lee
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Gi Beom Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Jihoon Han
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Jiwan Woo
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Yakdol Cho
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Cherlhyun Jeong
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.,KHU-KIST Department of Converging Science and Technology, Kyunghee University, Seoul 02447, Republic of Korea
| | - Seung-Yoon Park
- Department of Biochemistry, School of Medicine, Dongguk University, Gyeongju 38066, Republic of Korea
| | - Thomas M Roberts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Yong Beom Cho
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea .,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - In-San Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea .,KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| |
Collapse
|
48
|
Li C, Zhang Y, Yan S, Zhang G, Wei W, Qi Z, Li B. Alternol triggers immunogenic cell death via reactive oxygen species generation. Oncoimmunology 2021; 10:1952539. [PMID: 34350063 PMCID: PMC8296969 DOI: 10.1080/2162402x.2021.1952539] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Alternol is a naturally occurring compound that exerts antitumor activity in several cancers. However, whether Alternol induces antitumor immune response remains unknown. In this study, we investigated whether Alternol induced immunogenic cell death (ICD) in prostate cancer cells. Alternol triggered ICD in prostate cancer cells, as evidenced by the release of damage-associated molecular patterns (DAMPs) (i.e., calreticulin, CALR; high mobility group protein B1, HMGB1; and adenosine triphosphate, ATP) and pro-inflammatory cytokine (i.e., interleukin [IL]-1α, IL-1β, IL-6, and IL-8) expression. Alternol facilitated tumor-associated antigen uptake and cross-presentation, CD8 + T-cell priming, and T-cell infiltration in tumor-draining lymph nodes (LNs) and tumors. The presence of Alternol fostered antitumor immune response in vivo, resulting in delayed tumor growth and prolonged survival. Moreover, inhibition of reactive oxygen species (ROS) generation blocked Alternol-induced upregulation of pre-inflammation cytokines, endoplasmic reticulum (ER) stress, and consequent antitumor immune response. Overall, our data indicate that Alternol triggers ICD in prostate cancer cells, which is mediated by ROS generation.
Collapse
Affiliation(s)
- Changlin Li
- Institute of Precision Medicine, Jining Medical University, JiningChina
- Department of Urology, The University of Kansas Medical Center, Kansas City, KSUSA
| | - Ying Zhang
- Institute of Precision Medicine, Jining Medical University, JiningChina
| | - Siyuan Yan
- Institute of Precision Medicine, Jining Medical University, JiningChina
| | - Guoan Zhang
- Institute of Cancer Pathology Research, Jining Medical University, Jining, China
| | - Wei Wei
- Center for Experimental Medicine, School of Public Health, Jining Medical University, Jining, China
| | - Zhi Qi
- Department of Histology and Embryology, School of Medicine, Nankai University, Tianjin, China
| | - Benyi Li
- Department of Urology, The University of Kansas Medical Center, Kansas City, KSUSA
| |
Collapse
|
49
|
Engineered macrophages as near-infrared light activated drug vectors for chemo-photodynamic therapy of primary and bone metastatic breast cancer. Nat Commun 2021; 12:4310. [PMID: 34262026 PMCID: PMC8280231 DOI: 10.1038/s41467-021-24564-0] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
Patients with primary and bone metastatic breast cancer have significantly reduced survival and life quality. Due to the poor drug delivery efficiency of anti-metastasis therapy and the limited response rate of immunotherapy for breast cancer, effective treatment remains a formidable challenge. In this work, engineered macrophages (Oxa(IV)@ZnPc@M) carrying nanomedicine containing oxaliplatin prodrug and photosensitizer are designed as near-infrared (NIR) light-activated drug vectors, aiming to achieve enhanced chemo/photo/immunotherapy of primary and bone metastatic tumors. Oxa(IV)@ZnPc@M exhibits an anti-tumor M1 phenotype polarization and can efficiently home to primary and bone metastatic tumors. Additionally, therapeutics inside Oxa(IV)@ZnPc@M undergo NIR triggered release, which can kill primary tumors via combined chemo-photodynamic therapy and induce immunogenic cell death simultaneously. Oxa(IV)@ZnPc@M combined with anti-PD-L1 can eliminate primary and bone metastatic tumors, activate tumor-specific antitumor immune response, and improve overall survival with limited systemic toxicity. Therefore, this all-in-one macrophage provides a treatment platform for effective therapy of primary and bone metastatic tumors.
Collapse
|
50
|
Yang H, Han X, Hao Z. An Immune-Gene-Based Classifier Predicts Prognosis in Patients With Cervical Squamous Cell Carcinoma. Front Mol Biosci 2021; 8:679474. [PMID: 34291084 PMCID: PMC8289438 DOI: 10.3389/fmolb.2021.679474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/21/2021] [Indexed: 01/10/2023] Open
Abstract
Objective: Immunity plays a vital role in the human papilloma virus (HPV) persistent infection, and closely associates with occurrence and development of cervical squamous cell carcinoma (CSCC). Herein, we performed an integrated bioinformatics analysis to establish an immune-gene signature and immune-associated nomogram for predicting prognosis of CSCC patients. Methods: The list of immunity-associated genes was retrieved from ImmPort database. The gene and clinical information of CSCC patients were obtained from The Cancer Genome Atlas (TCGA) website. The immune gene signature for predicting overall survival (OS) of CSCC patients was constructed using the univariate Cox-regression analysis, random survival forests, and multivariate Cox-regression analysis. This signature was externally validated in GSE44001 cohort from Gene Expression Omnibus (GEO). Then, based on the established signature and the TCGA cohort with the corresponding clinical information, a nomogram was constructed and evaluated via Cox regression analysis, concordance index (C-index), receiver operating characteristic (ROC) curves, calibration plots and decision curve analyses (DCAs). Results: A 5-immune-gene prognostic signature for CSCC was established. Low expression of ICOS, ISG20 and high expression of ANGPTL4, SBDS, LTBR were risk factors for CSCC prognosis indicating poor OS. Based on this signature, the OS was significantly worse in high-risk group than in low-risk group (p-value < 0.001), the area under curves (AUCs) for 1-, 3-, 5-years OS were, respectively, 0.784, 0.727, and 0.715. A nomogram incorporating the risk score of signature and the clinical stage was constructed. The C-index of this nomogram was 0.76. AUC values were 0.811, 0.717, and 0.712 for 1-, 3-, 5-years OS. The nomogram showed good calibration and gained more net benefits than the 5-immune-gene signature and the clinical stage. Conclusion: The 5-immune-gene signature may serve as a novel, independent predictor for prognosis in patients with CSCC. The nomogram incorporating the signature risk score and clinical stage improved the predictive performance than the signature and clinical stage alone for predicting 1-year OS.
Collapse
Affiliation(s)
- Huixia Yang
- Department of Gynecology and Obstetrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaoyan Han
- Department of Gynecology and Obstetrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zengping Hao
- Department of Gynecology and Obstetrics, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|