1
|
Gutiérrez-Sandoval R, Gutiérrez-Castro F, Muñoz-Godoy N, Rivadeneira I, Sobarzo A, Alarcón L, Dorado W, Lagos A, Montenegro D, Muñoz I, Aguilera R, Iturra J, Krakowiak F, Peña-Vargas C, Toledo A. The Design of a Multistage Monitoring Protocol for Dendritic Cell-Derived Exosome (DEX) Immunotherapy: A Conceptual Framework for Molecular Quality Control and Immune Profiling. Int J Mol Sci 2025; 26:5444. [PMID: 40564910 DOI: 10.3390/ijms26125444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 06/01/2025] [Accepted: 06/03/2025] [Indexed: 06/28/2025] Open
Abstract
The increasing complexity of dendritic cell (DC)-derived exosome (DEX) immunotherapy demands structured monitoring protocols capable of translating molecular activity into actionable clinical outputs. This study proposes a standardized, multistage immunomonitoring framework designed to evaluate immune activation, cytokine polarization, and product integrity in DEX-based therapies. The protocol integrates open access methodologies-flow cytometry, cytometric bead array (CBA), and Western blotting-to assess CD69/CD25 activation, Th1/Th2/Th17 cytokine profiles, and vesicle identity across distinct checkpoints. These outputs are consolidated within the Structured Immunophenotypic Traceability Platform (STIP), which applies logic-based classifications (Type I-III) to support reproducible stratification of immune responses. Functional validation was performed through ex vivo co-culture models, enabling real-time interpretation of immune polarization, cytotoxic potential, and batch consistency. These outputs are supported by previous experimental validations published in Cancers and Biomedicines (2025), where PLPC and DC-derived vesicles demonstrated immunological consistency and a phenotypic stratification capacity. This approach provides a scalable monitoring structure that can support personalized treatment decisions, quality assurance workflows, and integration into regulatory documentation (e.g., CTD Module 5.3) for early-phase, non-pharmacodynamic immunotherapies. This conceptual protocol does not aim to demonstrate therapeutic efficacy but to provide a reproducible documentation framework for real-world immune monitoring and regulatory alignment in vesicle-based immunotherapy.
Collapse
Affiliation(s)
| | | | - Natalia Muñoz-Godoy
- Department of Cancer Research, Flowinmunocell-Bioexocell Group, 08028 Barcelona, Spain
| | - Ider Rivadeneira
- Department of Outreach and Engagement Programs for OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Adolay Sobarzo
- Departamento de Ciencias Biológicas y Químicas, Facultad de Ciencias, Universidad San Sebastián, Lientur 1457, Concepción 4080871, Chile
| | - Luis Alarcón
- Department of Outreach and Engagement Programs for OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Wilson Dorado
- Department of Outreach and Engagement Programs for OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Andy Lagos
- Department of Outreach and Engagement Programs for OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Diego Montenegro
- Department of Outreach and Engagement Programs for OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Ignacio Muñoz
- Department of Outreach and Engagement Programs for OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Rodrigo Aguilera
- Department of Outreach and Engagement Programs for OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | - Jordan Iturra
- Department of Outreach and Engagement Programs for OGRD Consortium, Charlestown KN0802, Saint Kitts and Nevis
| | | | | | - Andrés Toledo
- Department of Oncopathology, OGRD Alliance, Lewes, DE 19958, USA
| |
Collapse
|
2
|
Basyoni AE, Atta A, Salem MM, Mohamed TM. Harnessing exosomes for targeted drug delivery systems to combat brain cancer. Cancer Cell Int 2025; 25:150. [PMID: 40234973 PMCID: PMC12001718 DOI: 10.1186/s12935-025-03731-z] [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: 07/19/2024] [Accepted: 03/06/2025] [Indexed: 04/17/2025] Open
Abstract
Brain cancer remains a significant challenge in the field of oncology, primarily because of its aggressive nature and the limited treatment options available. Conventional therapies often fall short in effectively targeting tumor cells, while sparing healthy brain tissue from collateral damage. However, exosomes are now recognized as promising nanocarriers for targeted drug delivery. These naturally occurring extracellular vesicles can cross the blood-brain barrier and selectively interact with cancer cells. Utilizing exosomes as drug delivery vehicles offers a novel approach with significant potential for targeted therapy. By encapsulating therapeutic agents within exosomes, drugs can be specifically targeted to tumor cells, maximizing their impact whilst minimizing damage to healthy brain tissue. Furthermore, exosomes can be modified to display molecules that specifically recognize and bind to cancer cells, further enhancing their precision and efficacy. While exosome-based therapies show potential, scalability, purification, and clinical application challenges remain. The scalability of exosome production, purification, and modification techniques remains a hurdle that must be overcome for clinical translation. Additionally, the intricate interactions between the tumor microenvironment and exosomes necessitate further research to optimize therapeutic outcomes. The review explores applications and future perspectives of exosome-based therapies in advancing targeted brain cancer treatment.
Collapse
Affiliation(s)
- Abdullah E Basyoni
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Amira Atta
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Maha M Salem
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Tarek M Mohamed
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
3
|
Ljungström M, Oltra E. Methods for Extracellular Vesicle Isolation: Relevance for Encapsulated miRNAs in Disease Diagnosis and Treatment. Genes (Basel) 2025; 16:330. [PMID: 40149481 PMCID: PMC11942051 DOI: 10.3390/genes16030330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Extracellular vesicles (EVs) are nanovesicles that facilitate intercellular communication by carrying essential biomolecules under physiological and pathological conditions including microRNAs (miRNAs). They are found in various body fluids, such as blood, urine, and saliva, and their levels fluctuate with disease progression, making them valuable diagnostic tools. However, isolating EVs is challenging due to their small size and biological complexity. Here, we summarize the principles behind the most common EV isolation methods including ultracentrifugation, precipitation, immunoaffinity, sorting, ultrafiltration, size exclusion chromatography, and microfluidics while highlighting protocol strengths and weaknesses. We also review the main strategies to identify and quantify circulating miRNAs with a particular focus on EV-encapsulated miRNAs. Since these miRNAs hold special clinical interest derived from their superior stability and therapeutic potential, the information provided here should provide valuable guidance for future research initiatives in the promising field of disease diagnostic and treatment based on EV-encapsulated miRNAs.
Collapse
Affiliation(s)
- Maria Ljungström
- Escuela de Doctorado, School of Health Sciences, Catholic University of Valencia, 46001 Valencia, Spain;
| | - Elisa Oltra
- Department of Pathology, School of Health Sciences, Catholic University of Valencia, 46001 Valencia, Spain
| |
Collapse
|
4
|
Yang Y, Deng C, Aldali F, Huang Y, Luo H, Liu Y, Huang D, Cao X, Zhou Q, Xu J, Li Y, Chen H. Therapeutic Approaches and Potential Mechanisms of Small Extracellular Vesicles in Treating Vascular Dementia. Cells 2025; 14:409. [PMID: 40136659 PMCID: PMC11941715 DOI: 10.3390/cells14060409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Small extracellular vesicles (sEVs), including exosomes as a subtype, with a diameter typically less than 200 nm and originating from the endosomal system, are capable of transporting a diverse array of bioactive molecules, including proteins, nucleic acids, and lipids, thereby facilitating intercellular communication and modulating cellular functions. Vascular dementia (VaD) represents a form of cognitive impairment attributed to cerebrovascular disease, characterized by a complex and multifaceted pathophysiological mechanism. Currently, the therapeutic approach to VaD predominantly emphasizes symptom management, as no specific pharmacological treatment exists to cure the condition. Recent investigations have illuminated the significant role of sEVs in the pathogenesis of vascular dementia. This review seeks to provide a comprehensive analysis of the characteristics and functions of sEVs, with a particular focus on their involvement in vascular dementia and its underlying mechanisms. The objective is to advance the understanding of the interplays between sEVs and vascular dementia, thereby offering novel insights for future research and therapeutic strategies.
Collapse
Affiliation(s)
- Yujie Yang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Chunchu Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Fatima Aldali
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Yunjie Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Hongmei Luo
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Yizhou Liu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Danxia Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Xiaojian Cao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Qiuzhi Zhou
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Jia Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yajie Li
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
5
|
Lu M, Liu Y, Zhu J, Shang J, Bai L, Jin Z, Li W, Hu Y, Zheng X, Qian J. Mapping the intellectual structure and emerging trends on nanomaterials in colorectal cancer: a bibliometric analysis from 2003 to 2024. Front Oncol 2025; 14:1514581. [PMID: 39845318 PMCID: PMC11750690 DOI: 10.3389/fonc.2024.1514581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Background Colorectal cancer (CRC) is one of thes most prevalent malignant tumors worldwide. Current therapeutic strategies for CRC have limitations, while nanomaterials show significant potential for diagnosing and treating CRC. This study utilizes bibliometric analysis to evaluate the current status and trends in this field. Methods Research on nanomaterials in CRC from 2003 to 2024 was retrieved from the Web of Science Core Collection (WOSCC). Tools such as CiteSpace, VOSviewer, RStudio, GraphPad Prism, and Excel were used to analyze trends and hotspots, covering publication trends, countries, institutions, authors, journals, co-citation analysis, and keywords. Visual maps were created to forecast future developments. Results The analysis includes 3,683 publications by 17,261 authors from 3,721 institutions across 100 countries/regions, published in 840 journals. Global publications have steadily increased, particularly since 2018. China leads in publication volume and citations, with six of the top ten research institutions and seven of the ten most cited authors, while the United States excels in citation impact and academic centrality. Both countries currently dominate the field, underscoring the urgent need for enhanced international collaboration. Ramezani M and Abnous K lead in publication volume and H-index, while Siegel RL is highly cited. The International Journal of Nanomedicine has the highest publication volume, while the Journal of Controlled Release is the most cited. In addition to "colorectal cancer" and "nanoparticles," the most common keyword is "drug delivery." Emerging research areas such as "metal-organic frameworks (MOFs)" and "green synthesis" are gaining attention as leading hotspots. Conclusion This study offers an in-depth analysis of the application of nanomaterials in CRC, promoting interdisciplinary collaboration and advancing scientific progress in this field.
Collapse
Affiliation(s)
- Man Lu
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yi Liu
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jin Zhu
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jiarong Shang
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Lu Bai
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhichao Jin
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenting Li
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yue Hu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xia Zheng
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jun Qian
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
6
|
Sergazy S, Seydahmetova R, Gulyayev A, Shulgau Z, Aljofan M. The Role of Exosomes in Cancer Progression and Therapy. BIOLOGY 2025; 14:27. [PMID: 39857258 PMCID: PMC11763171 DOI: 10.3390/biology14010027] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/03/2024] [Accepted: 12/16/2024] [Indexed: 01/27/2025]
Abstract
Exosomes are small extracellular vesicles and are crucial in intercellular communication. Interestingly, tumor-derived exosomes carry oncogenic molecules, such as proteins and microRNAs, which can reprogram recipient cells, promote angiogenesis, and stimulate cancer pre-metastatic niche, supporting cancer growth and metastasis. On the other hand, their biocompatibility, stability, and ability to cross biological barriers make them attractive candidates for drug delivery. Recent advances have shown the potential for exosomes to be used in early disease detection and in targeted drug therapy by delivering therapeutic agents specifically to tumor sites. Despite the promising applications, a number of challenges remain, including exosome isolation and characterization, as well as their inherent heterogeneity. Thus, the current review aims to describe the roles of exosomes in health and disease, and discuss the challenges that hinder their development into becoming useful medical tools.
Collapse
Affiliation(s)
- Shynggys Sergazy
- LLP VICTUS PHARM, Astana 010000, Kazakhstan; (S.S.); (R.S.); (A.G.); (Z.S.)
- National Laboratory Astana, Center for Life Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Roza Seydahmetova
- LLP VICTUS PHARM, Astana 010000, Kazakhstan; (S.S.); (R.S.); (A.G.); (Z.S.)
| | - Alexandr Gulyayev
- LLP VICTUS PHARM, Astana 010000, Kazakhstan; (S.S.); (R.S.); (A.G.); (Z.S.)
- National Laboratory Astana, Center for Life Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Zarina Shulgau
- LLP VICTUS PHARM, Astana 010000, Kazakhstan; (S.S.); (R.S.); (A.G.); (Z.S.)
- National Laboratory Astana, Center for Life Sciences, Nazarbayev University, Astana 010000, Kazakhstan
| | - Mohamad Aljofan
- National Laboratory Astana, Center for Life Sciences, Nazarbayev University, Astana 010000, Kazakhstan
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana 010000, Kazakhstan
| |
Collapse
|
7
|
Leung LL, Qu X, Chen B, Chan JYK. Extracellular vesicles in liquid biopsies: there is hope for oral squamous cell carcinoma. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2024; 5:639-659. [PMID: 39811735 PMCID: PMC11725428 DOI: 10.20517/evcna.2024.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/29/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025]
Abstract
Current approaches to oral cancer diagnosis primarily involve physical examination, tissue biopsy, and advanced computer-aided imaging techniques. However, despite these advances, patient survival rates have not significantly improved. Hence, there is a critical need to develop minimally invasive tools with high sensitivity and specificity to improve patient survival and quality of life. Liquid biopsy is a non-invasive, real-time method for predicting cancer status and potentially serves as a biomarker source for treatment response. Liquid biopsy includes rich biologically relevant components, such as circulating tumor cells, circulating tumor DNA, and extracellular vesicles (EVs). EVs are particularly intriguing due to their relatively high abundance in most biofluids, with the potential to identify specific cargo derived from circulating tumor EVs. Moreover, normal cells in lymph nodes can uptake EVs, fostering a pre-metastatic microenvironment that facilitates lymph node metastases - a common occurrence in oral cancers. This review encompasses English language publications over the last twenty years, focusing on methods for isolating EVs from saliva, blood, and lymphatic fluids, as well as the collection methods employed. Seventeen cases met the inclusion criteria according to ISEV guidelines, including 10 saliva cases, 6 blood cases, and 1 lymphatic fluid case. This review also highlighted research gaps in oral squamous cell carcinoma (OSCC) EVs, including a lack of multi-omics studies and the exploration of potential EV markers for drug resistance, as well as a notable underutilization of microfluidic technologies to translate liquid biopsy EV findings into clinical applications.
Collapse
Affiliation(s)
| | | | | | - Jason YK. Chan
- Department of Otorhinolaryngology, Head and Neck Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong 00000, China
| |
Collapse
|
8
|
Li X, Lai Y, Wan G, Zou J, He W, Yang P. Approved natural products-derived nanomedicines for disease treatment. Chin J Nat Med 2024; 22:1100-1116. [PMID: 39725511 DOI: 10.1016/s1875-5364(24)60726-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Indexed: 12/28/2024]
Abstract
In recent years, there has been an increasing emphasis on exploring innovative drug delivery approaches due to the limitations of conventional therapeutic strategies, such as inadequate drug targeting, insufficient therapeutic efficacy, and significant adverse effects. Nanomedicines have emerged as a promising solution with notable advantages, including extended drug circulation, targeted delivery, and improved bioavailability, potentially enhancing the clinical treatment of various diseases. Natural products/materials-derived nanomedicines, characterized by their natural therapeutic efficacy, superior biocompatibility, and safety profile, play a crucial role in nanomedicine-based treatments. This review provides a comprehensive overview of currently approved natural products-derived nanomedicines, emphasizing the essential properties of natural products-derived drug carriers, their applications in clinical diagnosis and treatment, and the current therapeutic potential and challenges. The aim is to offer guidance for the application and further development of these innovative therapeutic approaches.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Yaoyao Lai
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Guanghan Wan
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China.
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 2111198, China.
| |
Collapse
|
9
|
Bahreyni A, Mohamud Y, Ashraf Nouhegar S, Zhang J, Luo H. Synergistic Viro-chemoimmunotherapy in Breast Cancer Enabled by Bioengineered Immunostimulatory Exosomes and Dual-Targeted Coxsackievirus B3. ACS NANO 2024; 18:4241-4255. [PMID: 38278522 PMCID: PMC10851665 DOI: 10.1021/acsnano.3c09491] [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: 09/29/2023] [Revised: 12/22/2023] [Accepted: 12/27/2023] [Indexed: 01/28/2024]
Abstract
Breast cancer's immunosuppressive environment hinders effective immunotherapy, but oncolytic viruses hold promise for addressing this challenge by targeting tumor cells and altering the microenvironment. Yet, neutralizing antibodies and immune clearance impede their clinical utility. This study explored microRNA-modified coxsackievirus B3 (miR-CVB3), an innovative oncolytic virus, and its potential in breast cancer treatment. It investigated miR-CVB3's impact on immune-related proteins and utilized exosomes as both protective shields and delivery carriers. Results demonstrated miR-CVB3's capacity to reshape immune-related protein profiles toward a more immunostimulatory state and enhance exosome-mediated immune cell activation. Notably, cancer cell-released exosomes encapsulating miR-CVB3 (ExomiR-CVB3) maintained its antitumor cytotoxicity and bolstered its immunostimulatory effects. Moreover, ExomiR-CVB3 shielded miR-CVB3 from neutralizing antibodies and rapid immune clearance when it was systemically administered. Building on these findings, ExomiR-CVB3 was engineered with the AS1411 aptamer and doxorubicin (ExomiR-CVB3/DoxApt), enhancing therapeutic efficacy. This notable approach, combining genomic modification, aptamer surface decoration, and doxorubicin addition, demonstrated safe delivery of CVB3 to cancer cells. Comprehensive in vitro and in vivo analyses revealed selective breast cancer cell targeting, cell death induction, and significant immune cell infiltration within the tumor microenvironment while sparing healthy organs. In summary, this study highlights ExomiR-CVB3/DoxApt as a pioneering breast cancer treatment strategy adaptable for diverse cancer types, offering a potent and versatile approach to reshaping cancer immunotherapy.
Collapse
Affiliation(s)
- Amirhossein Bahreyni
- Centre
for Heart Lung Innovation, St Paul’s
Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department
of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Yasir Mohamud
- Centre
for Heart Lung Innovation, St Paul’s
Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department
of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Sanaz Ashraf Nouhegar
- Centre
for Heart Lung Innovation, St Paul’s
Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department
of Chemistry, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Jingchun Zhang
- Centre
for Heart Lung Innovation, St Paul’s
Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department
of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Honglin Luo
- Centre
for Heart Lung Innovation, St Paul’s
Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department
of Pathology and Laboratory of Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|