1
|
Bhatt DK, Boerma A, Bustos SO, Otake AH, Murillo Carrasco AG, Reis PP, Chammas R, Daemen T, Andrade LNDS. Oncolytic alphavirus-induced extracellular vesicles counteract the immunosuppressive effect of melanoma-derived extracellular vesicles. Sci Rep 2025; 15:803. [PMID: 39755711 DOI: 10.1038/s41598-024-82331-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 12/04/2024] [Indexed: 01/06/2025] Open
Abstract
Extracellular vesicles (EVs)-mediated communication by cancer cells contributes towards the pro-tumoral reprogramming of the tumor microenvironment. Viral infection has been observed to alter the biogenesis and cargo of EVs secreted from host cells in the context of infectious biology. However, the impact of oncolytic viruses on the cargo and function of EVs released by cancer cells remains unknown. Here we show that upon oncolytic virotherapy with Semliki Forest virus-based replicon particles (rSFV), metastatic melanoma cells release EVs with a distinct biochemical profile and do not lead to suppression of immune cells. Specifically, we demonstrate that viral infection causes a differential loading of regulatory microRNAs (miRNAs) in EVs in addition to changes in their physical features. EVs derived from cancer cells potentially suppress splenocyte proliferation and induce regulatory macrophages. In contrast, EVs obtained from rSFV-infected cells did not exhibit such effects. Our results thus show that rSFV infection induces changes in the immunomodulatory properties of melanoma EVs, which may contribute to enhancing the therapeutic efficacy of virotherapy. Finally, our results show that the use of an oncolytic virus capable of a single-round of infection allows the analysis of EVs secreted from infected cells while preventing interference from extracellular virus particles.
Collapse
Affiliation(s)
- Darshak K Bhatt
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, 9713 AV, The Netherlands
- Center for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, CEP 01246-000, Brazil
| | - Annemarie Boerma
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, 9713 AV, The Netherlands
| | - Silvina Odete Bustos
- Center for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, CEP 01246-000, Brazil
- Comprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, Brazil
| | - Andréia Hanada Otake
- Center for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, CEP 01246-000, Brazil
- Comprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, Brazil
| | - Alexis Germán Murillo Carrasco
- Center for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, CEP 01246-000, Brazil
- Comprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, Brazil
| | - Patrícia Pintor Reis
- Department of Surgery and Orthopedics and Experimental Research Unity (UNIPEX), Faculdade de Medicina, Universidade Estadual Paulista (UNESP), Botucatu, 18618-687, Brazil
| | - Roger Chammas
- Center for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, CEP 01246-000, Brazil.
- Comprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, Brazil.
| | - Toos Daemen
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, 9713 AV, The Netherlands
| | - Luciana Nogueira de Sousa Andrade
- Center for Translational Research in Oncology (LIM/24), Instituto do Cancer do Estado de Sao Paulo, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, São Paulo, CEP 01246-000, Brazil.
- Comprehensive Center for Precision Oncology (C2PO), Universidade de Sao Paulo, São Paulo, Brazil.
| |
Collapse
|
2
|
Rixe O, Villano JL, Wesolowski R, Noonan AM, Puduvalli VK, Wise-Draper TM, Curry R, Yilmaz E, Cruze C, Ogretmen B, Tapolsky G, Takigiku R. A First-in-Human Phase I Study of BXQ-350, a First-in-Class Sphingolipid Metabolism Regulator, in Patients with Advanced/Recurrent Solid Tumors or High-Grade Gliomas. Clin Cancer Res 2024; 30:5053-5060. [PMID: 39264252 DOI: 10.1158/1078-0432.ccr-24-1721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/25/2024] [Accepted: 09/09/2024] [Indexed: 09/13/2024]
Abstract
PURPOSE BXQ-350, a nanovesicle formulation of saposin C, is an allosteric sphingolipid metabolism regulator that increases proapoptotic ceramide and decreases oncogenic sphingosine-1-phosphate levels. We conducted a first-in-human phase I study of BXQ-350. PATIENTS AND METHODS Adults (≥18 years old) with advanced/recurrent, treatment-refractory solid tumors or high-grade gliomas received BXQ-350 intravenously in five dose cohorts (0.7-2.4 mg/kg) in a 3+3 dose escalation and expansion design. The primary endpoints during dose escalation were dose-limiting toxicities and maximum tolerated dose; the primary objective in expansion parts was assessment of antitumor activity (RECIST v1.1/Response Assessment in Neuro-Oncology criteria). RESULTS Eighty-six patients were enrolled. Dose-limiting toxicities were not observed during dose escalation (n = 18), and a maximum tolerated dose was not identified. An additional 68 patients received the 2.4 mg/kg dose. Nine patients (10%) discontinued due to adverse events. The most common treatment-related adverse events were nausea (24%) and fatigue (23%). Eight patients had a progression-free survival of ≥6 months. Two of these achieved a partial response, and six had stable disease, among whom three had a reduction in ≥1 target lesion. Of those with progression-free survival of ≥6 months, seven remained on study for >12 months, five for >24 months, and after 7 years, two remained on study without disease progression. CONCLUSIONS BXQ-350 was well-tolerated as monotherapy at doses up to 2.4 mg/kg. It provided some lasting clinical benefit in patients with recurrent solid malignancies across several tumor types, consistent with a decreased systemic sphingosine-1-phosphate/ceramide metabolic rheostat. BXQ-350 warrants further clinical investigation alone and combined with standard of care for advanced solid tumors.
Collapse
Affiliation(s)
- Olivier Rixe
- University of New Mexico, Albuquerque, New Mexico
| | | | - Robert Wesolowski
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Anne M Noonan
- The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | | | | | | | | | | | - Besim Ogretmen
- Medical University of South Carolina, Hollings Cancer Center, Charleston, South Carolina
| | | | | |
Collapse
|
3
|
Chatterjee M, Gupta S, Nag S, Rehman I, Parashar D, Maitra A, Das K. Circulating Extracellular Vesicles: An Effective Biomarker for Cancer Progression. FRONT BIOSCI-LANDMRK 2024; 29:375. [PMID: 39614441 DOI: 10.31083/j.fbl2911375] [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: 08/01/2024] [Revised: 08/28/2024] [Accepted: 09/10/2024] [Indexed: 12/01/2024]
Abstract
Extracellular vesicles (EVs), the ubiquitous part of human biology, represent a small heterogenous, membrane-enclosed body that contains a diverse payload including genetic materials in the form of DNA, RNAs, small non-coding RNAs, etc. mostly mirroring their source of origin. Since, a vast majority of research has been conducted on how nucleic acids, proteins, lipids, and metabolites, associated with EVs can be effectively utilized to identify disease progression and therapeutic responses in cancer patients, EVs are increasingly being touted as valuable and reliable identifiers of cancer biomarkers in liquid biopsies. However, the lack of comprehensive clinical validation and effective standardization protocols severely limits its applications beyond the laboratories. The present review focuses on understanding the role of circulating EVs in different cancers and how they could potentially be treated as cancer biomarkers, typically due to the presence of bioactive molecules such as small non-coding RNAs, RNAs, DNA, proteins, etc., and their utilization for fine-tuning therapies. Here, we provide a brief general biology of EVs including their classification and subsequently discuss the source of circulatory EVs, the role of their associated payload as biomarkers, and how different cancers affect the level of circulatory EVs population.
Collapse
Affiliation(s)
- Madhura Chatterjee
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, 741251 Kalyani, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, 281406 Mathura, India
| | - Sayoni Nag
- Department of Biotechnology, Brainware University, 700125 Barasat, India
| | - Ishita Rehman
- Department of Biotechnology, The Neotia University, 743368 Parganas, India
| | - Deepak Parashar
- Department of Medicine, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Arindam Maitra
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, 741251 Kalyani, India
| | - Kaushik Das
- Biotechnology Research and Innovation Council-National Institute of Biomedical Genomics, 741251 Kalyani, India
| |
Collapse
|
4
|
Xia Y, Zhang J, Liu G, Wolfram J. Immunogenicity of Extracellular Vesicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403199. [PMID: 38932653 DOI: 10.1002/adma.202403199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/30/2024] [Indexed: 06/28/2024]
Abstract
Extracellular vesicles (EVs) are promising next-generation therapeutics and drug delivery systems due to demonstrated safety and efficacy in preclinical models and early-stage clinical trials. There is an urgent need to address the immunogenicity of EVs (beyond the apparent lack of immunotoxicity) to advance clinical development. To date, few studies have assessed unintended immunological recognition of EVs. An in-depth understanding of EV-induced immunogenicity and clearance is necessary to develop effective therapeutic strategies, including approaches to mitigate immunological recognition when undesired. This article summarizes various factors involved in the potential immunogenicity of EVs and strategies to reduce immunological recognition for improved therapeutic benefit.
Collapse
Affiliation(s)
- Yutian Xia
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Jianzhong Zhang
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Gang Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Joy Wolfram
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD, 4072, Australia
| |
Collapse
|
5
|
Xiao L, Zhang L, Guo C, Xin Q, Gu X, Jiang C, Wu J. "Find Me" and "Eat Me" signals: tools to drive phagocytic processes for modulating antitumor immunity. Cancer Commun (Lond) 2024; 44:791-832. [PMID: 38923737 PMCID: PMC11260773 DOI: 10.1002/cac2.12579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Phagocytosis, a vital defense mechanism, involves the recognition and elimination of foreign substances by cells. Phagocytes, such as neutrophils and macrophages, rapidly respond to invaders; macrophages are especially important in later stages of the immune response. They detect "find me" signals to locate apoptotic cells and migrate toward them. Apoptotic cells then send "eat me" signals that are recognized by phagocytes via specific receptors. "Find me" and "eat me" signals can be strategically harnessed to modulate antitumor immunity in support of cancer therapy. These signals, such as calreticulin and phosphatidylserine, mediate potent pro-phagocytic effects, thereby promoting the engulfment of dying cells or their remnants by macrophages, neutrophils, and dendritic cells and inducing tumor cell death. This review summarizes the phagocytic "find me" and "eat me" signals, including their concepts, signaling mechanisms, involved ligands, and functions. Furthermore, we delineate the relationships between "find me" and "eat me" signaling molecules and tumors, especially the roles of these molecules in tumor initiation, progression, diagnosis, and patient prognosis. The interplay of these signals with tumor biology is elucidated, and specific approaches to modulate "find me" and "eat me" signals and enhance antitumor immunity are explored. Additionally, novel therapeutic strategies that combine "find me" and "eat me" signals to better bridge innate and adaptive immunity in the treatment of cancer patients are discussed.
Collapse
Affiliation(s)
- Lingjun Xiao
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
| | - Louqian Zhang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
| | - Ciliang Guo
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
| | - Qilei Xin
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| | - Xiaosong Gu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| | - Chunping Jiang
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| | - Junhua Wu
- State Key Laboratory of Pharmaceutical BiotechnologyNational Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing UniversityNanjingJiangsuP. R. China
- Jinan Microecological Biomedicine Shandong LaboratoryJinanShandongP. R. China
| |
Collapse
|
6
|
Mittal S, Kumar S, Gupta P, Singh M, Chaluvally-Raghavan P, Pradeep S. Protocol for the isolation of tumor cell-derived extracellular vesicles followed by in vivo metastasis assessment in a murine ovarian cancer model. STAR Protoc 2024; 5:102943. [PMID: 38470912 PMCID: PMC10945248 DOI: 10.1016/j.xpro.2024.102943] [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: 11/01/2023] [Revised: 01/10/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Extracellular vesicles (EVs) play a crucial role in facilitating communication between cancer cells and their immediate or remote microenvironments, thereby promoting the extensive spread of cancer throughout the body. In this context, we present a protocol for the isolation of tumor cell-derived EVs followed by in vivo metastasis assessment in a murine ovarian cancer model. We describe steps for the isolation and characterization of EVs from ID8 cells, development of a metastatic mouse model, and sample preparation for flow cytometry. For complete details on the use and execution of this protocol, please refer to Gupta et al.1.
Collapse
Affiliation(s)
- Sonam Mittal
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sudhir Kumar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Prachi Gupta
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Mona Singh
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Pradeep Chaluvally-Raghavan
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sunila Pradeep
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
7
|
Rai A, Claridge B, Lozano J, Greening DW. The Discovery of Extracellular Vesicles and Their Emergence as a Next-Generation Therapy. Circ Res 2024; 135:198-221. [PMID: 38900854 DOI: 10.1161/circresaha.123.323054] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
From their humble discovery as cellular debris to cementing their natural capacity to transfer functional molecules between cells, the long-winded journey of extracellular vesicles (EVs) now stands at the precipice as a next-generation cell-free therapeutic tool to revolutionize modern-day medicine. This perspective provides a snapshot of the discovery of EVs to their emergence as a vibrant field of biology and the renaissance they usher in the field of biomedical sciences as therapeutic agents for cardiovascular pathologies. Rapid development of bioengineered EVs is providing innovative opportunities to overcome biological challenges of natural EVs such as potency, cargo loading and enhanced secretion, targeting and circulation half-life, localized and sustained delivery strategies, approaches to enhance systemic circulation, uptake and lysosomal escape, and logistical hurdles encompassing scalability, cost, and time. A multidisciplinary collaboration beyond the field of biology now extends to chemistry, physics, biomaterials, and nanotechnology, allowing rapid development of designer therapeutic EVs that are now entering late-stage human clinical trials.
Collapse
Affiliation(s)
- Alin Rai
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.R., B.C., J.L., D.W.G.)
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia (A.R., J.L., D.W.G.)
- Baker Department of Cardiometabolic Health, University of Melbourne, Victoria, Australia (A.R., D.W.G.)
- Central Clinical School, Monash University, Melbourne, Victoria, Australia (A.R., D.W.G.)
| | - Bethany Claridge
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.R., B.C., J.L., D.W.G.)
| | - Jonathan Lozano
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.R., B.C., J.L., D.W.G.)
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia (A.R., J.L., D.W.G.)
| | - David W Greening
- Molecular Proteomics, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia (A.R., B.C., J.L., D.W.G.)
- Baker Department of Cardiovascular Research Translation and Implementation, La Trobe University, Melbourne, Victoria, Australia (A.R., J.L., D.W.G.)
- Baker Department of Cardiometabolic Health, University of Melbourne, Victoria, Australia (A.R., D.W.G.)
- Central Clinical School, Monash University, Melbourne, Victoria, Australia (A.R., D.W.G.)
| |
Collapse
|
8
|
Geethadevi A, Ku Z, Tsaih SW, Parashar D, Kadamberi IP, Xiong W, Deng H, George J, Kumar S, Mittal S, Zhang N, Pradeep S, An Z, Chaluvally-Raghavan P. Blocking Oncostatin M receptor abrogates STAT3 mediated integrin signaling and overcomes chemoresistance in ovarian cancer. NPJ Precis Oncol 2024; 8:127. [PMID: 38839865 PMCID: PMC11153533 DOI: 10.1038/s41698-024-00593-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 04/30/2024] [Indexed: 06/07/2024] Open
Abstract
Chemotherapy such as cisplatin is widely used to treat ovarian cancer either before or after surgical debulking. However, cancer relapse due to chemotherapy resistance is a major challenge in the treatment of ovarian cancer. The underlying mechanisms related to chemotherapy resistance remain largely unclear. Therefore, identification of effective therapeutic strategies is urgently needed to overcome therapy resistance. Transcriptome-based analysis, in vitro studies and functional assays identified that cisplatin-resistant ovarian cancer cells express high levels of OSMR compared to cisplatin sensitive cells. Furthermore, OSMR expression associated with a module of integrin family genes and predominantly linked with integrin αV (ITGAV) and integrin β3 (ITGB3) for cisplatin resistance. Using ectopic expression and knockdown approaches, we proved that OSMR directly regulates ITGAV and ITGB3 gene expression through STAT3 activation. Notably, targeting OSMR using anti-OSMR human antibody inhibited the growth and metastasis of ovarian cancer cells and sensitized cisplatin treatment. Taken together, our results underscore the pivotal role of OSMR as a requirement for cisplatin resistance in ovarian cancer. Notably, OSMR fostered the expression of a distinct set of integrin genes, which in turn resulted into a crosstalk between OSMR and integrins for signaling activation that is critical for cisplatin resistance. Therefore, targeting OSMR emerges as a promising and viable strategy to reverse cisplatin-resistance in ovarian cancer.
Collapse
Affiliation(s)
- Anjali Geethadevi
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zhiqiang Ku
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Shirng-Wern Tsaih
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Medicine, Division of Hematology & Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ishaque P Kadamberi
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Wei Xiong
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Hui Deng
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Jasmine George
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sudhir Kumar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Sonam Mittal
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Sunila Pradeep
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA.
| | - Pradeep Chaluvally-Raghavan
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, WI, USA.
- Medical College of Wisconsin Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA.
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
9
|
Rahmat JN, Liu J, Chen T, Li Z, Zhang Y. Engineered biological nanoparticles as nanotherapeutics for tumor immunomodulation. Chem Soc Rev 2024; 53:5862-5903. [PMID: 38716589 DOI: 10.1039/d3cs00602f] [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: 06/05/2024]
Abstract
Biological nanoparticles, or bionanoparticles, are small molecules manufactured in living systems with complex production and assembly machinery. The products of the assembly systems can be further engineered to generate functionalities for specific purposes. These bionanoparticles have demonstrated advantages such as immune system evasion, minimal toxicity, biocompatibility, and biological clearance. Hence, bionanoparticles are considered the new paradigm in nanoscience research for fabricating safe and effective nanoformulations for therapeutic purposes. Harnessing the power of the immune system to recognize and eradicate malignancies is a viable strategy to achieve better therapeutic outcomes with long-term protection from disease recurrence. However, cancerous tissues have evolved to become invisible to immune recognition and to transform the tumor microenvironment into an immunosuppressive dwelling, thwarting the immune defense systems and creating a hospitable atmosphere for cancer growth and progression. Thus, it is pertinent that efforts in fabricating nanoformulations for immunomodulation are mindful of the tumor-induced immune aberrations that could render cancer nanotherapy inoperable. This review systematically categorizes the immunosuppression mechanisms, the regulatory immunosuppressive cellular players, and critical suppressive molecules currently targeted as breakthrough therapies in the clinic. Finally, this review will summarize the engineering strategies for affording immune moderating functions to bionanoparticles that tip the tumor microenvironment (TME) balance toward cancer elimination, a field still in the nascent stage.
Collapse
Affiliation(s)
- Juwita N Rahmat
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117585, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119074, Singapore
| | - Jiayi Liu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Taili Chen
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - ZhiHong Li
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yong Zhang
- Department of Biomedical Engineering, College of Engineering, The City University of Hong Kong, Hong Kong SAR.
| |
Collapse
|
10
|
Ye Z, Chen W, Li G, Huang J, Lei J. Tissue-derived extracellular vesicles in cancer progression: mechanisms, roles, and potential applications. Cancer Metastasis Rev 2024; 43:575-595. [PMID: 37851319 DOI: 10.1007/s10555-023-10147-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 10/03/2023] [Indexed: 10/19/2023]
Abstract
Extracellular vesicles (EVs) are small lipid bilayer-enclosed vesicles that mediate vital cellular communication by transferring cargo between cells. Among these, tissue-derived extracellular vesicles (Ti-EVs) stand out due to their origin from the tissue microenvironment, providing a more accurate reflection of changes in this setting. This unique advantage makes Ti-EVs valuable in investigating the intricate relationship between extracellular vesicles and cancer progression. Despite considerable research efforts exploring the association between Ti-EVs and cancers, a comprehensive clustering or grouping of these studies remains lacking. In this review, we aim to fill this gap by presenting a comprehensive synthesis of the mechanisms underlying Ti-EV generation, release, and transport within cancer tissues. Moreover, we delve into the pivotal roles that Ti-EVs play in cancer progression, shedding light on their potential as diagnostic and therapeutic tools. The review culminates in the construction of a comprehensive functional spectrum of Ti-EVs, providing a valuable reference for future research endeavors. By summarizing the current state of knowledge on Ti-EVs and their significance in tumor biology, this work contributes to a deeper understanding of cancer microenvironment dynamics and opens up avenues for harnessing Ti-EVs in diagnostic and therapeutic applications.
Collapse
Affiliation(s)
- Ziyang Ye
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenjie Chen
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Genpeng Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jing Huang
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianyong Lei
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
11
|
Abyadeh M, Mirshahvaladi S, Kashani SA, Paulo JA, Amirkhani A, Mehryab F, Seydi H, Moradpour N, Jodeiryjabarzade S, Mirzaei M, Gupta V, Shekari F, Salekdeh GH. Proteomic profiling of mesenchymal stem cell-derived extracellular vesicles: Impact of isolation methods on protein cargo. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e159. [PMID: 38947171 PMCID: PMC11212298 DOI: 10.1002/jex2.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/01/2024] [Accepted: 05/15/2024] [Indexed: 07/02/2024]
Abstract
Extracellular vesicles (EVs) are nanosized vesicles with a lipid bilayer that are secreted by cells and play a critical role in cell-to-cell communication. Despite the promising reports regarding their diagnostic and therapeutic potential, the utilization of EVs in the clinical setting is limited due to insufficient information about their cargo and a lack of standardization in isolation and analysis methods. Considering protein cargos in EVs as key contributors to their therapeutic potency, we conducted a tandem mass tag (TMT) quantitative proteomics analysis of three subpopulations of mesenchymal stem cell (MSC)-derived EVs obtained through three different isolation techniques: ultracentrifugation (UC), high-speed centrifugation (HS), and ultracentrifugation on sucrose cushion (SU). Subsequently, we checked EV marker expression, size distribution, and morphological characterization, followed by bioinformatic analysis. The bioinformatic analysis of the proteome results revealed that these subpopulations exhibit distinct molecular and functional characteristics. The choice of isolation method impacts the proteome of isolated EVs by isolating different subpopulations of EVs. Specifically, EVs isolated through the high-speed centrifugation (HS) method exhibited a higher abundance of ribosomal and mitochondrial proteins. Functional apoptosis assays comparing isolated mitochondria with EVs isolated through different methods revealed that HS-EVs, but not other EVs, induced early apoptosis in cancer cells. On the other hand, EVs isolated using the sucrose cushion (SU) and ultracentrifugation (UC) methods demonstrated a higher abundance of proteins primarily involved in the immune response, cell-cell interactions and extracellular matrix interactions. Our analyses unveil notable disparities in proteins and associated biological functions among EV subpopulations, underscoring the importance of meticulously selecting isolation methods and resultant EV subpopulations based on the intended application.
Collapse
Affiliation(s)
- Morteza Abyadeh
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Shahab Mirshahvaladi
- Macquarie Medical School, School of MedicineHealth and Human Sciences, Macquarie UniversitySydneyNew South WalesAustralia
| | - Sara Assar Kashani
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Motor Neuron Disease Research Centre, Faculty of Medicine, Health and Human SciencesMacquarie UniversitySydneyNew South WalesAustralia
| | - Joao A. Paulo
- Department of Cell BiologyHarvard Medical SchoolBostonMassachusettsUSA
| | - Ardeshir Amirkhani
- Australian Proteome Analysis FacilityMacquarie UniversitySydneyNew South WalesAustralia
| | - Fatemeh Mehryab
- Advanced Therapy Medicinal Product Technology Development Center, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | - Homeyra Seydi
- Advanced Therapy Medicinal Product Technology Development Center, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Department of BiologyUniversity of Science and CultureTehranIran
| | | | | | - Mehdi Mirzaei
- Macquarie Medical School, School of MedicineHealth and Human Sciences, Macquarie UniversitySydneyNew South WalesAustralia
| | - Vivek Gupta
- Macquarie Medical School, School of MedicineHealth and Human Sciences, Macquarie UniversitySydneyNew South WalesAustralia
| | - Faezeh Shekari
- Department of Stem Cells and Developmental Biology, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
- Advanced Therapy Medicinal Product Technology Development Center, Cell Science Research CenterRoyan Institute for Stem Cell Biology and Technology, ACECRTehranIran
| | | |
Collapse
|
12
|
Di Niro L, Linders AC, Glynn T, Pegtel DM, Siderius M, Crudden C, Smit MJ. G protein-coupled receptors: a gateway to targeting oncogenic EVs? EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:233-248. [PMID: 39698539 PMCID: PMC11648488 DOI: 10.20517/evcna.2024.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/29/2024] [Accepted: 05/14/2024] [Indexed: 12/20/2024]
Abstract
Dysregulated intercellular communication is a key feature driving cancer progression. Recently, extracellular vesicles (EVs) have added a new channel to this dense communication network. Despite solid evidence that EVs are central mediators of dysregulated signaling in onco-pathological settings, this has yet to be translated into clinically actionable strategies. The heterogeneity of EV cargo molecules, plasticity of biogenesis routes, and large overlap with their role in physiological communication, complicate a potential targeting strategy. However, recent work has linked EV biology to perhaps the "most druggable" proteins - G protein-coupled receptors (GPCRs). GPCR targeting accounts for ~60% of drugs in development and more than a third of all currently approved drugs, spanning almost all areas of medicine. Although several GPCRs have been linked to cancer initiation and progression, relatively few agents have made it into oncological regimes, suggesting that their potential is underexploited. Herein, we examine the molecular mechanisms linking GPCRs to EV communication in cancer settings. We propose that GPCRs hold potential in the search for EV-targeting in oncology.
Collapse
Affiliation(s)
- Lotte Di Niro
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Amber C. Linders
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Thomas Glynn
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - D. Michiel Pegtel
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Marco Siderius
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Caitrin Crudden
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| | - Martine J. Smit
- Department of Chemistry and Pharmaceutical Sciences, Division of Medicinal Chemistry, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081 HV, The Netherlands
| |
Collapse
|
13
|
Sun H, Hu B, Wu C, Jiang T. Targeting the SPHK1/S1P/S1PR2 axis ameliorates GH-secreted pituitary adenoma progression. Eur J Clin Invest 2024; 54:e14117. [PMID: 37888843 DOI: 10.1111/eci.14117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 09/30/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND Growth hormone-secreted pituitary adenoma (GHPA) is a prominent subtype of pituitary adenoma (PA) associated with progressive somatic disfigurement, various complications, and elevated mortality rates. Existing treatment options have limited efficacy, highlighting the urgent need for novel pharmacological interventions. Previous studies have revealed that sphingosine kinase 1 (SphK1)/sphingosine-1-phosphate (S1P)/S1P receptors (S1PRs) signalling have critical roles in the tumour microenvironment, but their role in GHPA remains unclear. METHODS We performed integrative analyses including bioinformatics analyses, functional studies, and clinical validation to investigate the pathological roles of SPHK1/S1P and evaluated the effectiveness of the S1P receptor 2 (S1PR2) inhibitor JTE-013 in GHPA treatment. RESULTS SPHK1/S1P signalling is abnormally expressed in patients with GHPA. Knockdown of SPHK1 suppresses S1P-mediated cell proliferation in GH3 Cells. Mechanistically, S1P inhibits apoptosis and autophagy while promoting the secretion of Growth Hormone (GH) by binding to the S1P receptor subtype 2 (S1PR2) in GH3 cells. Moreover, the function of S1PR2 in GH3 cells is mediated by the downstream Akt-Creb pathway. We then identify the S1PR2 as a novel target for therapeutic intervention in GHPA. Systemic administration of the potent and selective S1PR2 antagonist, JTE-013, significantly reduces both tumour size and GH secretion. Importantly, we identify preoperative serum S1P levels as a biomarker predicting poor prognosis in GHPA patients at follow-up. CONCLUSION Our study shows that blocking SPHK1/S1P/S1PR2 axis can ameliorate the progression of GHPA, providing evidence of a promising therapeutic target for GHPA.
Collapse
Affiliation(s)
- Heng Sun
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Hunan, Changsha, China
| | - Biao Hu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Hunan, Changsha, China
| | - Chunli Wu
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Hunan, Changsha, China
| | - Tiejian Jiang
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Hunan, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Hunan, Changsha, China
| |
Collapse
|
14
|
Lyu C, Sun H, Sun Z, Liu Y, Wang Q. Roles of exosomes in immunotherapy for solid cancers. Cell Death Dis 2024; 15:106. [PMID: 38302430 PMCID: PMC10834551 DOI: 10.1038/s41419-024-06494-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 02/03/2024]
Abstract
Although immunotherapy has made breakthrough progress, its efficacy in solid tumours remains unsatisfactory. Exosomes are the main type of extracellular vesicles that can deliver various intracellular molecules to adjacent or distant cells and organs, mediating various biological functions. Studies have found that exosomes can both activate the immune system and inhibit the immune system. The antigen and major histocompatibility complex (MHC) carried in exosomes make it possible to develop them as anticancer vaccines. Exosomes derived from blood, urine, saliva and cerebrospinal fluid can be used as ideal biomarkers in cancer diagnosis and prognosis. In recent years, exosome-based therapy has made great progress in the fields of drug transportation and immunotherapy. Here, we review the composition and sources of exosomes in the solid cancer immune microenvironment and further elaborate on the potential mechanisms and pathways by which exosomes influence immunotherapy for solid cancers. Moreover, we summarize the potential clinical application prospects of engineered exosomes and exosome vaccines in immunotherapy for solid cancers. Eventually, these findings may open up avenues for determining the potential of exosomes for diagnosis, treatment, and prognosis in solid cancer immunotherapy.
Collapse
Affiliation(s)
- Cong Lyu
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Haifeng Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Yang Liu
- Department of Radiotherapy, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China.
| |
Collapse
|
15
|
Rome S, Tacconi S. High-fat diets: You are what you eat….your extracellular vesicles too! J Extracell Vesicles 2024; 13:e12382. [PMID: 38151475 PMCID: PMC10752826 DOI: 10.1002/jev2.12382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/13/2023] [Accepted: 11/10/2023] [Indexed: 12/29/2023] Open
Abstract
Recent works indicate that the lipid composition of extracellular vesicles (EVs) can modify their biological functions and their incorporation into recipient cells. In particular high-fat diets affect EV biogenesis, EV lipid composition, EV targeting and consequently the cross-talk between tissues. This review connects different research topics to show that a vicious circle is established during the development of high-fat diet-induced obesity, connecting the alteration of lipid metabolism, the composition of extracellular vesicles and the spread of deleterious lipids between tissues, which participates in NAFLD/NASH and diabetes development. According to the studies described in this review, it is urgent to take an interest in this question as the modulation of EV lipid composition could be an important factor to take into account during the therapeutic management of patients suffering from metabolic syndrome and related pathologies such as obesity and diabetes. Furthermore, as lipid modification of EVs is a strategy currently being tested to enable better integration into their target tissue or cell, it is important to consider the impact of these lipid modifications on the homeostasis of these targets.
Collapse
Affiliation(s)
- Sophie Rome
- CarMeN Laboratory, INSERM 1060‐INRAE 1397, Department of Human Nutrition, Lyon Sud HospitalUniversity of LyonLyonFrance
| | - Stefano Tacconi
- CarMeN Laboratory, INSERM 1060‐INRAE 1397, Department of Human Nutrition, Lyon Sud HospitalUniversity of LyonLyonFrance
| |
Collapse
|
16
|
Xiao Y, Wu M, Xue C, Wang Y. Recent Advances in the Development of Membrane-derived Vesicles for Cancer Immunotherapy. Curr Drug Deliv 2024; 21:403-420. [PMID: 37143265 DOI: 10.2174/1567201820666230504120841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 03/01/2023] [Accepted: 03/13/2023] [Indexed: 05/06/2023]
Abstract
The surface proteins on cell membranes enable the cells to have different properties, such as high biocompatibility, surface modifiability, and homologous targeting ability. Cell-membrane-derived vesicles have features identical to those of their parental cells, which makes them one of the most promising materials for drug delivery. Recently, as a result of the impressive effects of immunotherapy in cancer treatment, an increasing number of researchers have used cell-membrane-derived vesicles to enhance immune responses. To be more specific, the membrane vesicles derived from immune cells, tumor cells, bacteria, or engineered cells have the antigen presentation capacity and can trigger strong anti-tumor effects of the immune system. In this review, we first indicated a brief description of the vesicles and then introduced the detection technology and drug-loading methods for them. Secondly, we concluded the characteristics and applications of vesicles derived from different sources in cancer immunotherapy.
Collapse
Affiliation(s)
- Yuai Xiao
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Minliang Wu
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chunyu Xue
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yuchong Wang
- Department of Plastic Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| |
Collapse
|
17
|
Zhang C, Qin M. Extracellular vesicles targeting tumor microenvironment in ovarian cancer. Int J Biol Macromol 2023; 252:126300. [PMID: 37573911 DOI: 10.1016/j.ijbiomac.2023.126300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/17/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
Ovarian cancer (OC) is a prevalent neoplastic condition affecting women. Extracellular vesicles (EVs), nano-sized membrane vesicles, are secreted by various cells in both physiological and pathological states. The profound interplay between EVs and the tumor microenvironment (TME) in ovarian cancer is crucial. In this review, we explores the pivotal role of EVs in facilitating intercellular communication between cancer cells and the TME, emphasizing the potential of EVs as promising diagnostic markers and innovative therapeutic targets for ovarian cancer. The comprehensive analysis outlines the specific mechanisms by which EVs engage in communication with the constituents of the TME, including the modulation of tumor growth through EVs carrying matrix metalloproteinases (MMPs) and EV-mediated inhibition of angiogenesis, among other factors. Additionally, the we discuss the potential clinical applications of EVs that target the TME in ovarian cancer, encompassing the establishment of novel treatment strategies and the identification of novel biomarkers for early detection and prognosis. Finally, this review identifies novel strategies for therapeutic interventions, such as utilizing EVs as carriers for drug delivery and targeting specific EV-mediated signaling pathways. In summary, this manuscript offers valuable insights into the role of EVs in ovarian cancer and highlights the significance of comprehending intercellular communication in the realm of cancer biology.
Collapse
Affiliation(s)
- Chunmei Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, China
| | - Meiying Qin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
18
|
Chang YC, Liu HP, Chuang HL, Liao JW, Kao PL, Chan HL, Chen TH, Wang YC. Feline mammary carcinoma-derived extracellular vesicle promotes liver metastasis via sphingosine kinase-1-mediated premetastatic niche formation. Lab Anim Res 2023; 39:27. [PMID: 37941082 PMCID: PMC10634095 DOI: 10.1186/s42826-023-00180-5] [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: 08/08/2023] [Revised: 10/26/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Feline mammary carcinoma (FMC) is one of the most prevalent malignancies of female cats. FMC is highly metastatic and thus leads to poor disease outcomes. Among all metastases, liver metastasis occurs in about 25% of FMC patients. However, the mechanism underlying hepatic metastasis of FMC remains largely uncharacterized. RESULTS Herein, we demonstrate that FMC-derived extracellular vesicles (FMC-EVs) promotes the liver metastasis of FMC by activating hepatic stellate cells (HSCs) to prime a hepatic premetastatic niche (PMN). Moreover, we provide evidence that sphingosine kinase 1 (SK1) delivered by FMC-EV was pivotal for the activation of HSC and the formation of hepatic PMN. Depletion of SK1 impaired cargo sorting in FMC-EV and the EV-potentiated HSC activation, and abolished hepatic colonization of FMC cells. CONCLUSIONS Taken together, our findings uncover a previously uncharacterized mechanism underlying liver-metastasis of FMC and provide new insights into prognosis and treatment of this feline malignancy.
Collapse
Affiliation(s)
- Yi-Chih Chang
- Department of Medical Laboratory Science & Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Hao-Ping Liu
- Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, 145 Xingda Rd., South Dist., Taichung, 40227, Taiwan
| | - Pei-Ling Kao
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, 145 Xingda Rd., South Dist., Taichung, 40227, Taiwan
| | - Hsun-Lung Chan
- Veterinary Research Institute, Ministry of Agriculture, Zhunan, Taiwan
| | - Ter-Hsin Chen
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, 145 Xingda Rd., South Dist., Taichung, 40227, Taiwan
| | - Yu-Chih Wang
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, 145 Xingda Rd., South Dist., Taichung, 40227, Taiwan.
| |
Collapse
|
19
|
Das K, Paul S, Ghosh A, Gupta S, Mukherjee T, Shankar P, Sharma A, Keshava S, Chauhan SC, Kashyap VK, Parashar D. Extracellular Vesicles in Triple-Negative Breast Cancer: Immune Regulation, Biomarkers, and Immunotherapeutic Potential. Cancers (Basel) 2023; 15:4879. [PMID: 37835573 PMCID: PMC10571545 DOI: 10.3390/cancers15194879] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype accounting for ~10-20% of all human BC and is characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) amplification. Owing to its unique molecular profile and limited targeted therapies, TNBC treatment poses significant challenges. Unlike other BC subtypes, TNBC lacks specific molecular targets, rendering endocrine therapies and HER2-targeted treatments ineffective. The chemotherapeutic regimen is the predominant systemic treatment modality for TNBC in current clinical practice. However, the efficacy of chemotherapy in TNBC is variable, with response rates varying between a wide range of patients, and the emerging resistance further adds to the difficulties. Furthermore, TNBC exhibits a higher mutational burden and is acknowledged as the most immunogenic of all BC subtypes. Consequently, the application of immune checkpoint inhibition has been investigated in TNBC, yielding promising outcomes. Recent evidence identified extracellular vesicles (EVs) as an important contributor in the context of TNBC immunotherapy. In view of the extraordinary ability of EVs to transfer bioactive molecules, such as proteins, lipids, DNA, mRNAs, and small miRNAs, between the cells, EVs are considered a promising diagnostic biomarker and novel drug delivery system among the prospects for immunotherapy. The present review provides an in-depth understanding of how EVs influence TNBC progression, its immune regulation, and their contribution as a predictive biomarker for TNBC. The final part of the review focuses on the recent key advances in immunotherapeutic strategies for better understanding the complex interplay between EVs and the immune system in TNBC and further developing EV-based targeted immunotherapies.
Collapse
Affiliation(s)
- Kaushik Das
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Subhojit Paul
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700012, India; (S.P.); (A.G.)
| | - Arnab Ghosh
- School of Biological Sciences, Indian Association for the Cultivation of Science, Jadavpur, Kolkata 700012, India; (S.P.); (A.G.)
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura 281406, India;
| | - Tanmoy Mukherjee
- School of Medicine, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Prem Shankar
- Department of Neurobiology, The University of Texas Medical Branch, Galveston, TX 77555, USA or
| | - Anshul Sharma
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, The University of Texas at Tyler Health Science Center, Tyler, TX 75708, USA;
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (S.C.C.); (V.K.K.)
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Vivek Kumar Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; (S.C.C.); (V.K.K.)
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Deepak Parashar
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
20
|
Beyoğlu D, Schwalm S, Semmo N, Huwiler A, Idle JR. Hepatitis C Virus Infection Upregulates Plasma Phosphosphingolipids and Endocannabinoids and Downregulates Lysophosphoinositols. Int J Mol Sci 2023; 24:ijms24021407. [PMID: 36674922 PMCID: PMC9864155 DOI: 10.3390/ijms24021407] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/12/2023] Open
Abstract
A mass spectrometry-based lipidomic investigation of 30 patients with chronic hepatitis C virus (HCV) infection and 30 age- and sex-matched healthy blood donor controls was undertaken. The clustering and complete separation of these two groups was found by both unsupervised and supervised multivariate data analyses. Three patients who had spontaneously cleared the virus and three who were successfully treated with direct-acting antiviral drugs remained within the HCV-positive metabotype, suggesting that the metabolic effects of HCV may be longer-lived. We identified 21 metabolites that were upregulated in plasma and 34 that were downregulated (p < 1 × 10-16 to 0.0002). Eleven members of the endocannabinoidome were elevated, including anandamide and eight fatty acid amides (FAAs). These likely activated the cannabinoid receptor GPR55, which is a pivotal host factor for HCV replication. FAAH1, which catabolizes FAAs, reduced mRNA expression. Four phosphosphingolipids, d16:1, d18:1, d19:1 sphingosine 1-phosphate, and d18:0 sphinganine 1-phosphate, were increased, together with the mRNA expression for their synthetic enzyme SPHK1. Among the most profoundly downregulated plasma lipids were several lysophosphatidylinositols (LPIs) from 3- to 3000-fold. LPIs are required for the synthesis of phosphatidylinositol 4-phosphate (PI4P) pools that are required for HCV replication, and LPIs can also activate the GPR55 receptor. Our plasma lipidomic findings shed new light on the pathobiology of HCV infection and show that a subset of bioactive lipids that may contribute to liver pathology is altered by HCV infection.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
- Hepatology Research Group, Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Stephanie Schwalm
- Pharmazentrum Frankfurt/ZAFES, Institute of General Pharmacology and Toxicology, University Hospital, Goethe University Frankfurt am Main, D-60590 Frankfurt am Main, Germany
- Institute of Pharmacology, Inselspital, INO-F, University of Bern, CH-3010 Bern, Switzerland
| | - Nasser Semmo
- Hepatology Research Group, Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Andrea Huwiler
- Institute of Pharmacology, Inselspital, INO-F, University of Bern, CH-3010 Bern, Switzerland
- Correspondence: (A.H.); (J.R.I.)
| | - Jeffrey R. Idle
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA
- Hepatology Research Group, Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
- Correspondence: (A.H.); (J.R.I.)
| |
Collapse
|
21
|
Guo L, Tang X, Wong SW, Guo A, Lin Y, Kwok HF. Regulation of IFN-γ-mediated PD-L1 expression by MYC in colorectal cancer with wild-type KRAS and TP53 and its clinical implications. Front Pharmacol 2022; 13:1022129. [PMID: 36582540 PMCID: PMC9792609 DOI: 10.3389/fphar.2022.1022129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction: In the tumor microenvironment, interferon gamma (IFN-γ) secreted by tumor infiltrating lymphocytes can upregulate programmed cell death 1 ligand 1 (PD-L1) expression in many cancers. The present study evaluated the expression of PD-L1 in selected colorectal cancer cell lines with IFN-γ treatment and explored the correlation between programmed cell death 1 ligand 1 expression and KRAS/TP53 mutation status. Methods: The selected colorectal cancer cell lines had known KRAS mutations or TP53 mutations. TCGA data analysis were used to investigate the correlation between overall survival of patient with anti-PD-1/PD-L1 immunotherapy and KRAS/TP53 mutation status. Besides, the correlation between PD-L1 expression and KRAS/TP53 mutation status were also investigated by using TCGA data analysis. In vitro experiments were used to explore the mechanism underlying KRAS- and TP53-related PD-L1 expression. Results: Firstly, TCGA data analysis for gene expression and overall survival and an in vitro study revealed that the wild-type KRAS/TP53 cell lines exhibited hyperresponsiveness to interferon gamma exposure and correlated with better survival in patients receiving anti-PD-1/PD-L1 treatment. Secondly, experimental data revealed that interferon gamma induced the upregulation of programmed cell death 1 ligand 1 mainly through regulating MYC in wild-type KRAS and TP53 colorectal cancers. Discussion: Our findings revealed that the response to anti-PD-1/PD-L1 cancer immunotherapy frequently happened in wild-type KRAS and TP53 colorectal cancers, which were also found to show higher programmed cell death 1 ligand 1 expression. Our results indicate that the wild-type KRAS/TP53 colorectal cancer cell lines may respond better to interferon gamma treatment, which causes increased programmed cell death 1 ligand 1 expression and may be a mechanism underlying the better responses to anti-PD-1/PD-L1 therapies in wild-type KRAS and wild-type TP53 colorectal cancer. Furthermore, the experimental results suggest that interferon gamma regulated programmed cell death 1 ligand 1 expression through the regulation of MYC, which may further affect the response to PD-1/PD-L1 cancer immunotherapy. These results suggest a novel potential treatment strategy for enhancing the efficacy of PD-1/PD-L1 blockade immunotherapy in most colorectal cancer patients.
Collapse
Affiliation(s)
- Libin Guo
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China,MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
| | - Xiaoqiong Tang
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China,Central Laboratory at the Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Sin Wa Wong
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China
| | - Anyuan Guo
- Center for Artificial Intelligence Biology, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Lin
- Key Laboratory of Optoelectronic Science and Technology for Medicine of Ministry of Education, College of Life Sciences, Fujian Normal University, Fuzhou, China,Central Laboratory at the Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, China,*Correspondence: Yao Lin, ; Hang Fai Kwok,
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China,MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR, China,*Correspondence: Yao Lin, ; Hang Fai Kwok,
| |
Collapse
|
22
|
Reale A, Khong T, Spencer A. Extracellular Vesicles and Their Roles in the Tumor Immune Microenvironment. J Clin Med 2022; 11:jcm11236892. [PMID: 36498469 PMCID: PMC9737553 DOI: 10.3390/jcm11236892] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/15/2022] [Accepted: 11/15/2022] [Indexed: 11/24/2022] Open
Abstract
Tumor cells actively incorporate molecules (e.g., proteins, lipids, RNA) into particles named extracellular vesicles (EVs). Several groups have demonstrated that EVs can be transferred to target (recipient) cells, making EVs an important means of intercellular communication. Indeed, EVs are able to modulate the functions of target cells by reprogramming signaling pathways. In a cancer context, EVs promote the formation of a supportive tumor microenvironment (TME) and (pre)metastatic niches. Recent studies have revealed that immune cells, tumor cells and their secretome, including EVs, promote changes in the TME and immunosuppressive functions of immune cells (e.g., natural killer, dendritic cells, T and B cells, monocytes, macrophages) that allow tumor cells to establish and propagate. Despite the growing knowledge on EVs and on their roles in cancer and as modulators of the immune response/escape, the translation into clinical practice remains in its early stages, hence requiring improved translational research in the EVs field. Here, we comprehensively review the current knowledge and most recent research on the roles of EVs in tumor immune evasion and immunosuppression in both solid tumors and hematological malignancies. We also highlight the clinical utility of EV-mediated immunosuppression targeting and EV-engineering. Importantly, we discuss the controversial role of EVs in cancer biology, current limitations and future perspectives to further the EV knowledge into clinical practice.
Collapse
Affiliation(s)
- Antonia Reale
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University—Alfred Health, Melbourne, VIC 3004, Australia
- Correspondence: (A.R.); (A.S.)
| | - Tiffany Khong
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University—Alfred Health, Melbourne, VIC 3004, Australia
| | - Andrew Spencer
- Myeloma Research Group, Australian Centre for Blood Diseases, Central Clinical School, Monash University—Alfred Health, Melbourne, VIC 3004, Australia
- Malignant Haematology and Stem Cell Transplantation, Department of Haematology, Alfred Hospital, Melbourne, VIC 3004, Australia
- Department of Clinical Hematology, Monash University, Melbourne, VIC 3004, Australia
- Correspondence: (A.R.); (A.S.)
| |
Collapse
|
23
|
Onuma T, Asare-Werehene M, Yoshida Y, Tsang BK. Exosomal Plasma Gelsolin Is an Immunosuppressive Mediator in the Ovarian Tumor Microenvironment and a Determinant of Chemoresistance. Cells 2022; 11:cells11203305. [PMID: 36291171 PMCID: PMC9600545 DOI: 10.3390/cells11203305] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/15/2022] [Accepted: 10/18/2022] [Indexed: 12/03/2022] Open
Abstract
Ovarian Cancer (OVCA) is the most fatal gynecologic cancer and has a 5-year survival rate less than 45%. This is mainly due to late diagnosis and drug resistance. Overexpression of plasma gelsolin (pGSN) is key contributing factor to OVCA chemoresistance and immunosuppression. Gelsolin (GSN) is a multifunctional protein that regulates the activity of actin filaments by cleavage, capping, and nucleation. Generally, it plays an important role in cytoskeletal remodeling. GSN has three isoforms: cytosolic GSN, plasma GSN (pGSN), and gelsolin-3. Exosomes containing pGSN are released and contribute to the progression of OVCA. This review describes how pGSN overexpression inhibits chemotherapy-induced apoptosis and triggers positive feedback loops of pGSN expression. It also describes the mechanisms by which exosomal pGSN promotes apoptosis and dysfunction in tumor-killing immune cells. A discussion on the potential of pGSN as a prognostic, diagnostic, and therapeutic marker is also presented herein.
Collapse
Affiliation(s)
- Toshimichi Onuma
- Department of Obstetrics & Gynecology, Faculty of Medicine & Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine & the Centre for Infection, Immunity and Inflammation (CI3), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Department of Obstetrics and Gynecology, University of Fukui, Fukui 910-8507, Japan
| | - Meshach Asare-Werehene
- Department of Obstetrics & Gynecology, Faculty of Medicine & Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine & the Centre for Infection, Immunity and Inflammation (CI3), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
| | - Yoshio Yoshida
- Department of Obstetrics and Gynecology, University of Fukui, Fukui 910-8507, Japan
- Correspondence: (Y.Y.); (B.K.T.)
| | - Benjamin K. Tsang
- Department of Obstetrics & Gynecology, Faculty of Medicine & Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON K1H 8L1, Canada
- Department of Cellular and Molecular Medicine & the Centre for Infection, Immunity and Inflammation (CI3), Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
- Chronic Disease Program, Ottawa Hospital Research Institute, Ottawa, ON K1H 8L6, Canada
- Correspondence: (Y.Y.); (B.K.T.)
| |
Collapse
|
24
|
McAlarnen LA, Gupta P, Singh R, Pradeep S, Chaluvally-Raghavan P. Extracellular vesicle contents as non-invasive biomarkers in ovarian malignancies. Mol Ther Oncolytics 2022; 26:347-359. [PMID: 36090475 PMCID: PMC9420349 DOI: 10.1016/j.omto.2022.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Ovarian cancer most commonly presents at an advanced stage where survival is approximately 30% compared with >80% if diagnosed and treated before disease spreads. Diagnostic capabilities have progressed from surgical staging via laparotomy to image-guided biopsies and immunohistochemistry staining, along with advances in technology and medicine. Despite improvements in diagnostic capabilities, population-level screening for ovarian cancer is not recommended. Extracellular vesicles (EVs) are 40–150 nm structures formed when the cellular lipid bilayer invaginates. These structures function in cell signaling, immune responses, cancer progression, and establishing the tumor microenvironment. EVs are found in nearly every bodily fluid, including serum, plasma, ascites, urine, and effusion fluid, and contain molecular cargo from their cell of origin. This cargo can be analyzed to yield information about a possible malignancy. In this review we describe how the cargo of EVs has been studied as biomarkers in ovarian cancer. We bring together studies analyzing evidence for various cargos as ovarian cancer biomarkers. Then, we describe the role of EVs in modulation of the tumor microenvironment. This review also summarizes the therapeutic and translational potential of EVs for their optimal utilization as non-invasive biomarkers for novel treatments against cancer.
Collapse
|
25
|
Parashar D, Singh A, Gupta S, Sharma A, Sharma MK, Roy KK, Chauhan SC, Kashyap VK. Emerging Roles and Potential Applications of Non-Coding RNAs in Cervical Cancer. Genes (Basel) 2022; 13:genes13071254. [PMID: 35886037 PMCID: PMC9317009 DOI: 10.3390/genes13071254] [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: 05/31/2022] [Revised: 07/05/2022] [Accepted: 07/06/2022] [Indexed: 12/06/2022] Open
Abstract
Cervical cancer (CC) is a preventable disease using proven interventions, specifically prophylactic vaccination, pervasive disease screening, and treatment, but it is still the most frequently diagnosed cancer in women worldwide. Patients with advanced or metastatic CC have a very dismal prognosis and current therapeutic options are very limited. Therefore, understanding the mechanism of metastasis and discovering new therapeutic targets are crucial. New sequencing tools have given a full visualization of the human transcriptome's composition. Non-coding RNAs (NcRNAs) perform various functions in transcriptional, translational, and post-translational processes through their interactions with proteins, RNA, and even DNA. It has been suggested that ncRNAs act as key regulators of a variety of biological processes, with their expression being tightly controlled under physiological settings. In recent years, and notably in the past decade, significant effort has been made to examine the role of ncRNAs in a variety of human diseases, including cancer. Therefore, shedding light on the functions of ncRNA will aid in our better understanding of CC. In this review, we summarize the emerging roles of ncRNAs in progression, metastasis, therapeutics, chemo-resistance, human papillomavirus (HPV) regulation, metabolic reprogramming, diagnosis, and as a prognostic biomarker of CC. We also discussed the role of ncRNA in the tumor microenvironment and tumor immunology, including cancer stem cells (CSCs) in CC. We also address contemporary technologies such as antisense oligonucleotides, CRISPR-Cas9, and exosomes, as well as their potential applications in targeting ncRNAs to manage CC.
Collapse
Affiliation(s)
- Deepak Parashar
- Department of Obstetrics and Gynecology, Medical College of Wisconsin, Milwaukee, MI 53226, USA
- Correspondence: (D.P.); (V.K.K.); Tel.: +1-414-439-8089 (D.P.); +1-956-296-1738 (V.K.K.)
| | - Anupam Singh
- Department of Biotechnology, GLA University, Mathura 281406, Uttar Pradesh, India; (A.S.); (S.G.)
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura 281406, Uttar Pradesh, India; (A.S.); (S.G.)
| | - Aishwarya Sharma
- Sri Siddhartha Medical College and Research Center, Tumkur 572107, Karnataka, India;
| | - Manish K. Sharma
- Department of Biotechnology, IP College, Bulandshahr 203001, Uttar Pradesh, India;
| | - Kuldeep K. Roy
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun 248007, Uttarakhand, India;
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA;
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Vivek K. Kashyap
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA;
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
- Correspondence: (D.P.); (V.K.K.); Tel.: +1-414-439-8089 (D.P.); +1-956-296-1738 (V.K.K.)
| |
Collapse
|
26
|
Zhang H, Cai YH, Ding Y, Zhang G, Liu Y, Sun J, Yang Y, Zhan Z, Iliuk A, Gu Z, Gu Y, Tao WA. Proteomics, Phosphoproteomics and Mirna Analysis of Circulating Extracellular Vesicles through Automated and High-Throughput Isolation. Cells 2022; 11:2070. [PMID: 35805153 PMCID: PMC9265938 DOI: 10.3390/cells11132070] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/22/2022] [Accepted: 06/24/2022] [Indexed: 12/28/2022] Open
Abstract
Extracellular vesicles (EVs) play an important role in the diagnosis and treatment of diseases because of their rich molecular contents involved in intercellular communication, regulation, and other functions. With increasing efforts to move the field of EVs to clinical applications, the lack of a practical EV isolation method from circulating biofluids with high throughput and good reproducibility has become one of the biggest barriers. Here, we introduce a magnetic bead-based EV enrichment approach (EVrich) for automated and high-throughput processing of urine samples. Parallel enrichments can be performed in 96-well plates for downstream cargo analysis, including EV characterization, miRNA, proteomics, and phosphoproteomics analysis. We applied the instrument to a cohort of clinical urine samples to achieve reproducible identification of an average of 17,000 unique EV peptides and an average of 2800 EV proteins in each 1 mL urine sample. Quantitative phosphoproteomics revealed 186 unique phosphopeptides corresponding to 48 proteins that were significantly elevated in prostate cancer patients. Among them, multiple phosphoproteins were previously reported to associate with prostate cancer. Together, EVrich represents a universal, scalable, and simple platform for EV isolation, enabling downstream EV cargo analyses for a broad range of research and clinical applications.
Collapse
Affiliation(s)
- Hao Zhang
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
- EVLiXiR Biotech, Nanjing 210032, China
| | - Yu-Han Cai
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
| | - Yajie Ding
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
| | - Guiyuan Zhang
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
- Bell Mountain Molecular MedTech Institute, Nanjing 210032, China
| | - Yufeng Liu
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
- Bell Mountain Molecular MedTech Institute, Nanjing 210032, China
| | - Jie Sun
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
| | - Yuchen Yang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China; (Y.Y.); (Y.G.)
| | - Zhen Zhan
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
| | - Anton Iliuk
- Tymora Analytical Operations, West Lafayette, IN 47906, USA;
| | - Zhongze Gu
- State Key Laboratory of Bioelectronics, National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing 210096, China; (H.Z.); (Y.-H.C.); (Y.D.); (G.Z.); (Y.L.); (J.S.); (Z.Z.); (Z.G.)
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing 210029, China; (Y.Y.); (Y.G.)
| | - W. Andy Tao
- Tymora Analytical Operations, West Lafayette, IN 47906, USA;
- Department of Chemistry and Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
27
|
Luo D, Liao S, Liu Y, Lin Y, Li Y, Liao X. Holliday Cross-Recognition Protein HJURP: Association With the Tumor Microenvironment in Hepatocellular Carcinoma and With Patient Prognosis. Pathol Oncol Res 2022; 28:1610506. [PMID: 35783358 PMCID: PMC9248293 DOI: 10.3389/pore.2022.1610506] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 05/31/2022] [Indexed: 12/04/2022]
Abstract
Background: Hepatocellular carcinoma is the most common type of primary liver cancer, and it is associated with poor prognosis. It often fails to respond to immunotherapy, highlighting the need to identify genes that are associated with the tumor microenvironment and may be good therapeutic targets. We and others have shown that the Holliday cross-recognition protein HJURP can promote the proliferation, migration, and invasion by hepatocellular carcinoma cells, and that HJURP overexpression is associated with poor survival. Here we explored the potential relationship between HJURP and the tumor microenvironment in hepatocellular carcinoma. Methods: We used the Immuno-Oncology-Biological-Research (IOBR) software package to analyze the potential roles of HJURP in the tumor microenvironment. Using single-cell RNA sequencing data, we identified the cell clusters expressing abundant HJURP, then linked some of these clusters to certain bioprocesses using Gene Set Enrichment Analysis (GSEA). We validated the differential expression of HJURP in tumor-infiltrating CD8+ T cells, sorted by flow cytometry into populations based on the expression level of PD-1. We used weighted gene co-expression network analysis (WGCNA) to identify immunity-related genes whose expression strongly correlated with that of HJURP. The function of these genes was validated based on enrichment in Gene Ontology (GO) terms, and they were used to establish a prognosis prediction model. Results: IOBR analysis suggested that HJURP is significantly related to the immunosuppressive tumor microenvironment and was significantly related to T cells, dendritic cells, and B cells. Based on single-cell RNA sequencing, HJURP was strongly expressed in T cells, erythrocytes, and B cells from normal liver tissues, as well as in CD8+ T cells, dendritic cells, and one cluster of hepatocytes in hepatocellular carcinoma tissues. Malignant hepatocytes strongly expressing HJURP were associated with the downregulation of immune bioprocesses. HJURP expression was significantly higher in CD8+ T cells strongly expressing PD-1 than in those expressing no or intermediate levels of PD1. WGCNA identified two module eigengenes (comprising 397 and 84 genes) related to the tumor microenvironment. We identified 24 hub genes and confirmed that they were related to immune regulation. A prognostic risk score model based on expression of HJURP, PPT1, PML, and CLEC7A showed moderate ability to predict survival. Conclusion:HJURP is associated with tumor-infiltrating immune cells, immune checkpoints, and immune suppression in hepatocellular carcinoma. HJURP-related genes involved in immune responses may be useful for predicting patient prognosis.
Collapse
Affiliation(s)
- Dongcheng Luo
- Department of First Chemotherapy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Sina Liao
- Department of First Chemotherapy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yu Liu
- Department of First Chemotherapy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Youzhi Lin
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yongqiang Li
- Department of First Chemotherapy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - XiaoLi Liao
- Department of First Chemotherapy, Guangxi Medical University Cancer Hospital, Nanning, China
- *Correspondence: XiaoLi Liao,
| |
Collapse
|
28
|
Wang Y, Fan J, Chen T, Xu L, Liu P, Xiao L, Wu T, Zhou Q, Zheng Q, Liu C, Chan FL, Wu D. A novel ferroptosis-related gene prognostic index for prognosis and response to immunotherapy in patients with prostate cancer. Front Endocrinol (Lausanne) 2022; 13:975623. [PMID: 36034466 PMCID: PMC9399637 DOI: 10.3389/fendo.2022.975623] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/13/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Prostate cancer (PCa) is among the leading causes of cancer death worldwide. Ferroptosis refers to an iron-dependent form of regulated cell death and is involved in prostate tumorigenesis. A few ferroptosis-related gene signatures have been developed to predict the prognosis for PCa patients. However, previous signatures were typically established based on biochemical recurrence-free survival, which has proven not to be a good surrogate for overall survival (OS). This study aimed to construct a novel ferroptosis-related gene prognostic index (FRGPI) to predict disease-free survival (DFS) and response to immunotherapy for PCa patients after radical prostatectomy. METHODS Gene expression and clinicopathological data on PCa patients were obtained from the TCGA database. Ferroptosis-related hub genes associated with DFS of PCa patients were identified by an in-depth bioinformatics analysis using a novel and comprehensive algorithm based on functional enrichment, consensus clustering, weighted gene co-expression network analysis (WGCNA), and protein-protein interaction (PPI) network construction. The FRGPI was established on the basis of the genes selected using multivariate cox regression analysis and further validated in two additional PCa cohorts. Next, the clinicopathological, molecular, and immune profiles were characterized and compared between FRGPI-high and FRGPI-low subgroups. Finally, the predictive role of the FRGPI in response to immunotherapy was estimated using a metastatic urothelial cancer cohort treated with an anti-PD-L1 agent. RESULTS The FRGPI was constructed based on four genes (E2F1, CDC20, TYMS, and NUP85), and FRGPI-high patients had worse DFS than FRGPI-low patients. Multivariate cox regression analysis revealed that FRGPI could act as an independent prognostic factor for PCa patients after radical prostatectomy. A prognostic nomogram comprising the FRGPI and other clinicopathological parameters was established to predict the DFS for PCa patients quantitatively. In addition, comprehensive results demonstrated that high FRGPI scores showed a significantly positive correlation with worse clinicopathological features, higher mutation counts, increased frequency of copy number variations (CNVs), higher homologous recombination deficiency (HRD) and immune scores, higher mRNAsi, and more importantly, enhanced sensitivity to immunotherapy. CONCLUSIONS FRGPI is not only a promising and robust prognostic biomarker, but also a potential indicator of immunotherapeutic outcomes for PCa patients after radical prostatectomy.
Collapse
Affiliation(s)
- Yuliang Wang
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, China
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Jiaqi Fan
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, China
| | - Tao Chen
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Lele Xu
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Pengyu Liu
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Lijia Xiao
- Department of Clinical Laboratory Medicine Center, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Tao Wu
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Qingchun Zhou
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Qingyou Zheng
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Chunxiao Liu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Chunxiao Liu, ; Franky Leung Chan, ; Dinglan Wu,
| | - Franky Leung Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- *Correspondence: Chunxiao Liu, ; Franky Leung Chan, ; Dinglan Wu,
| | - Dinglan Wu
- Shenzhen Key Laboratory of Viral Oncology, The Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, China
- *Correspondence: Chunxiao Liu, ; Franky Leung Chan, ; Dinglan Wu,
| |
Collapse
|