1
|
Jangavali SS, Hangargekar PB, Gangthade BU, Jadhav SA, Havelikar UA, Joshi AA. From lab to lifesaver: the rise of CAR T-cell therapy in oncology. J Egypt Natl Canc Inst 2025; 37:37. [PMID: 40377854 DOI: 10.1186/s43046-025-00262-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 02/03/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Recently, cancer treatment paradigms have shifted dramatically with the advent of immunotherapies, particularly chimeric antigen receptor (CAR) T-cell therapy. Despite it is revolutionary positive outcomes in treating hematologic malignancies, challenges such as severe toxicities, high treatment costs, and limited efficacy in solid tumors persist. This review highlights these limitations and the ongoing need for innovation in CAR T-cell therapy. MAIN BODY This manuscript provides a comprehensive review of most current advancements in CAR T-cell therapy, with a focus on targeting its immunotherapeutic principles, modification of T cells for Targeted cancer therapy using T cells, and clinical applications. It explores the key elements of CAR T-cell therapy, containing antigen recognition domain and intracellular signaling domains, which enable T cells to interact with cancer cells and exert cytotoxic effects. The review examines approved therapies, and ongoing clinical trials, Along with obstacles like cytokine release syndrome (CRS), neurotoxicity, along antigen escape mechanisms. Furthermore, innovations in cutting-edge CAR T-cell therapies and personalized treatment approaches are discussed, together with an emphasis on improving safety and efficacy. CONCLUSION The manuscript outlines the future outlook on integrating CAR T-cell therapy integrated with other treatments and exploring patient-specific approaches to revolutionize cancer care. This review aims to bridge the existing gaps in research, offering valuable insights for students and researchers in biomedical sciences and oncology.
Collapse
Affiliation(s)
- Shriyash S Jangavali
- Department of Pharmacy, ASPM's K. T. Patil College of Pharmacy, Dharashiv, 413501, Maharashtra, India
| | - Pallavi B Hangargekar
- Department of Quality Assurance, ASPM's K. T. Patil College of Pharmacy, Dharashiv, 413501, Maharashtra, India.
| | - Balasaheb U Gangthade
- Department of Pharmacy, ASPM's K. T. Patil College of Pharmacy, Dharashiv, 413501, Maharashtra, India
| | - Shreya A Jadhav
- Department of Pharmacy, ASPM's K. T. Patil College of Pharmacy, Dharashiv, 413501, Maharashtra, India
| | - Ujwal A Havelikar
- Department of Pharmacy, NIMS Institute of Pharmacy, Jaipur, Rajasthan, India
| | - Amol A Joshi
- Department of Pharmacognosy, ASPM's K. T. Patil College of Pharmacy, Dharashiv, 413501, Maharashtra, India
| |
Collapse
|
2
|
Li YR, Zhu Y, Halladay T, Yang L. In vivo CAR engineering for immunotherapy. Nat Rev Immunol 2025:10.1038/s41577-025-01174-1. [PMID: 40379910 DOI: 10.1038/s41577-025-01174-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2025] [Indexed: 05/19/2025]
Abstract
Chimeric antigen receptor (CAR)-engineered immune cell therapy represents an important advance in cancer treatments. However, the complex ex vivo cell manufacturing process and stringent patient selection criteria curtail its widespread use. In vivo CAR engineering is emerging as a promising off-the-shelf therapy, providing advantages such as streamlined production, elimination of patient-specific manufacturing, reduced costs and simplified logistics. A large set of preclinical findings has inspired further investigation into treatments for hard-to-treat diseases such as solid tumours and has facilitated the development of advanced products to enhance in vivo CAR engineering efficacy, the persistence of the cellular therapeutic and safety. In this Review, we summarize current in vivo CAR engineering strategies, including nanoparticle-based and viral delivery systems as well as bioinstructive implantable scaffolds, and discuss their advantages and disadvantages. Additionally, we provide a systematic comparison between in vivo and conventional ex vivo CAR engineering methods and address the challenges and future prospects of in vivo CAR engineering.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tyler Halladay
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Parveen S, Konde DV, Paikray SK, Tripathy NS, Sahoo L, Samal HB, Dilnawaz F. Nanoimmunotherapy: the smart trooper for cancer therapy. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002308. [PMID: 40230883 PMCID: PMC11996242 DOI: 10.37349/etat.2025.1002308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
Immunotherapy has gathered significant attention and is now a widely used cancer treatment that uses the body's immune system to fight cancer. Despite initial successes, its broader clinical application is hindered by limitations such as heterogeneity in patient response and challenges associated with the tumor immune microenvironment. Recent advancements in nanotechnology have offered innovative solutions to these barriers, providing significant enhancements to cancer immunotherapy. Nanotechnology-based approaches exhibit multifaceted mechanisms, including effective anti-tumor immune responses during tumorigenesis and overcoming immune suppression mechanisms to improve immune defense capacity. Nanomedicines, including nanoparticle-based vaccines, liposomes, immune modulators, and gene delivery systems, have demonstrated the ability to activate immune responses, modulate tumor microenvironments, and target specific immune cells. Success metrics in preclinical and early clinical studies, such as improved survival rates, enhanced tumor regression, and elevated immune activation indices, highlight the promise of these technologies. Despite these achievements, several challenges remain, including scaling up manufacturing, addressing off-target effects, and navigating regulatory complexities. The review emphasizes the need for interdisciplinary approaches to address these barriers, ensuring broader clinical adoption. It also provides insights into interdisciplinary approaches, advancements, and the transformative potential of nano-immunotherapy and promising results in checkpoint inhibitor delivery, nanoparticle-mediated photothermal therapy, immunomodulation as well as inhibition by nanoparticles and cancer vaccines.
Collapse
Affiliation(s)
- Suphiya Parveen
- Department of Biotechnology and Genetics, School of Sciences, Jain (Deemed-to-be-University), Bengaluru 560027, Karnataka, India
| | - Dhanshree Vikrant Konde
- Department of Biotechnology and Genetics, School of Sciences, Jain (Deemed-to-be-University), Bengaluru 560027, Karnataka, India
| | - Safal Kumar Paikray
- School of Biotechnology, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| | - Nigam Sekhar Tripathy
- School of Biotechnology, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| | - Liza Sahoo
- School of Biotechnology, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| | - Himansu Bhusan Samal
- School of Pharmacy and Life Sciences, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| | - Fahima Dilnawaz
- School of Biotechnology, Centurion University of Technology and Management, Jatni 752050, Odisha, India
| |
Collapse
|
4
|
Seong J, Kim M, Yoo J, Mack DL, Lee JH, Joo J. Sustained Release of HIF-2α Inhibitors Using Biodegradable Porous Silicon Carriers for Enhanced Immunogenic Cell Death of Malignant Merkel Cell Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7449-7461. [PMID: 39834008 DOI: 10.1021/acsami.4c19961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Merkel cell carcinoma (MCC) is a rare but aggressive neuroendocrine skin cancer with limited treatment options, often associated with Merkel cell polyomavirus (MCPyV) and marked by hypoxic tumor microenvironments that promote resistance to therapies. Belzutifan, an FDA-approved hypoxia-inducible factor-2α (HIF-2α) inhibitor, has shown promise in inhibiting tumor growth; however, its clinical efficacy is hindered by its low solubility, rapid clearance, and limited bioavailability. In this study, we present a strategy using porous silicon (pSi) microparticles and nanoparticles as carriers for the sustained delivery of benzoate to MCC cells. The pSi carriers were engineered to securely encapsulate and gradually release belzutifan, overcoming the limitations of free drug administration. Microparticles provided sustained extracellular release, while nanoparticles enabled efficient intracellular delivery, enhancing HIF-2α inhibition. Moreover, the use of biodegradable silicon particles enables long-term consistent release of belzutifan over 10 days in vitro with a single dose administration in the tumor microenvironment, while free belzutifan is rapidly deactivated within 1 day postadministration. In vitro studies demonstrated significant immunogenic cell death (ICD) in MCC cells, marked by the cytosolic localization of HMGB1 and elevated expression of pro-inflammatory cytokines as well as strong upregulation of TLR9. Particularly, the increased TLR9 expression in both MCC cell lines with pSi carrier treatment reinforces immune activation through toll-like receptor signaling, enhancing both innate and adaptive immune responses within the tumor microenvironment. These findings indicate that pSi carriers not only enhance belzutifan's stability and release profile but also amplify antitumor immune responses within the tumor microenvironment. Our results suggest that belzutifan-loaded pSi carriers offer a potent and targeted therapeutic strategy for MCC, potentially addressing key challenges in cancer immunotherapy by combining HIF-2α inhibition with robust immune activation. This platform highlights the universal utility of pSi-based delivery systems to advance MCC treatment with implications for broader cancer therapies.
Collapse
Affiliation(s)
- Juyoung Seong
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Institute for Stem Cell and Regenerative Medicine (ISCRM), University of Washington, Seattle, Washington 98109, United States
| | - Minju Kim
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Jounghyun Yoo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - David L Mack
- Institute for Stem Cell and Regenerative Medicine (ISCRM), University of Washington, Seattle, Washington 98109, United States
- Department of Bioengineering, University of Washington, Seattle, Washington 98195, United States
- Department of Rehabilitation Medicine, University of Washington, Seattle, Washington 98195, United States
| | - Jung Hyun Lee
- Institute for Stem Cell and Regenerative Medicine (ISCRM), University of Washington, Seattle, Washington 98109, United States
- Department of Dermatology, School of Medicine, University of Washington, Seattle, Washington 98109, United States
| | - Jinmyoung Joo
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Graduate School of Health Science and Technology, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Center for Genomic Integrity, Institute for Basic Science, Ulsan 44919, Republic of Korea
| |
Collapse
|
5
|
Kushwaha N, Panjwani D, Patel S, Ahlawat P, Yadav MR, Patel AS. Emerging advances in nano-biomaterial assisted amyloid beta chimeric antigen receptor macrophages (CAR-M) therapy: reducing plaque burden in Alzheimer's disease. J Drug Target 2025; 33:185-205. [PMID: 39403775 DOI: 10.1080/1061186x.2024.2417012] [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: 08/04/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
Alzheimer's disease is the most common form, accounting for 60-70% of 55 million dementia cases. Even though the precise pathophysiology of AD is not completely understood, clinical trials focused on antibodies targeting aggregated forms of β amyloid (Aβ) have demonstrated that reducing amyloid plaques can arrest cognitive decline in patients in the early stages of AD. In this study, we provide an overview of current research and innovations for controlled release from nano-biomaterial-assisted chimeric antigen receptor macrophage (CAR-M) therapeutic strategies targeted at AD. Nano-bio materials, such as iron-oxide nanoparticles (IONPs), can be made selectively (Hp-Hb/mannose) to bind and take up Aβ plaques like CAR-M cells. By using nano-bio materials, both the delivery and stability of CAR-M cells in brain tissue can be improved to overcome the barriers of the BBB and enhance therapeutic effects. By enhancing the targeting capabilities and stability of CAR-M cells, mRNA-loaded nano-biomaterials can significantly improve the efficacy of immunotherapy for plaque reduction in AD. This novel strategy holds promise for translating preclinical successes into clinical applications, potentially revolutionising the management of AD.
Collapse
Affiliation(s)
- Nishabh Kushwaha
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Drishti Panjwani
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Shruti Patel
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Priyanka Ahlawat
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Mange Ram Yadav
- Research and Development Cell, Parul University, Vadodara, India
| | - Asha S Patel
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| |
Collapse
|
6
|
Yun Y, Kim S, Lee SN, Cho HY, Choi JW. Nanomaterial-based detection of circulating tumor cells and circulating cancer stem cells for cancer immunotherapy. NANO CONVERGENCE 2024; 11:56. [PMID: 39671082 PMCID: PMC11645384 DOI: 10.1186/s40580-024-00466-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024]
Abstract
Nanomaterials have emerged as transformative tools for detecting circulating tumor cells (CTCs) and circulating cancer stem cells (CCSCs), significantly enhancing cancer diagnostics and immunotherapy. Nanomaterials, including those composed of gold, magnetic materials, and silica, have enhanced the sensitivity, specificity, and efficiency of isolating these rare cells from blood. These developments are of paramount importance for the early detection of cancer and for providing real-time insights into metastasis and treatment resistance, which are essential for the development of personalized immunotherapies. The combination of nanomaterial-based platforms with phenotyping techniques, such as Raman spectroscopy and microfluidics, enables researchers to enhance immunotherapy protocols targeting specific CTC and CCSC markers. Nanomaterials also facilitate the targeted delivery of immunotherapeutic agents, including immune checkpoint inhibitors and therapeutic antibodies, directly to tumor cells. This synergistic approach has the potential to enhance therapeutic efficacy and mitigate the risk of metastasis and relapse. In conclusion, this review critically examines the use of nanomaterial-driven detection systems for detecting CTCs and CCSCs, their application in immunotherapy, and suggests future directions, highlighting their potential to transform the integration of diagnostics and treatment, thereby paving the way for more precise and personalized cancer therapies.
Collapse
Affiliation(s)
- Yeochan Yun
- Department of Bio and Fermentation Convergence Technology, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul, 02707, Republic of Korea
| | - Seewoo Kim
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea
| | - Sang-Nam Lee
- Uniance Gene Inc., 273, Digital-ro, Guro-gu, Seoul, 08381, Republic of Korea.
| | - Hyeon-Yeol Cho
- Department of Bio and Fermentation Convergence Technology, Kookmin University, 77 Jeongneung-ro, Seongbuk-gu, Seoul, 02707, Republic of Korea.
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, 35 Baekbeom-ro, Mapo-gu, Seoul, 04107, Republic of Korea.
| |
Collapse
|
7
|
Tiwari A, Tiwari V, Sharma A, Marrisetti AL, Kumar M, Rochani A, Kaushik D, Mittal V, Jyothi S R, Ali H, Hussain MS, Gupta G. Unlocking the potential: integrating phytoconstituents and nanotechnology in skin cancer therapy - A comprehensive review. JOURNAL OF COMPLEMENTARY & INTEGRATIVE MEDICINE 2024:jcim-2024-0338. [PMID: 39668578 DOI: 10.1515/jcim-2024-0338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/14/2024] [Indexed: 12/14/2024]
Abstract
Skin carcinoma, which includes basal cell carcinoma (BCC), squamous cell carcinoma (SCC), and melanoma, is influenced by various factors such as genetic predisposition, chemical exposures, immune system imbalances, and ultraviolet (UV) radiation. This review delves into the mechanisms behind the development of these cancers, exploring the therapeutic potential of microbial, plant derived compounds and nanoparticles in advancing skin cancer treatments. Special attention is given to the cytotoxic effects of anti-neoplastic agents from microbial sources on different cancer cell lines, particularly melanoma. Additionally, the review highlights the role of phytochemicals - such as quercetin, resveratrol, and curcumin alongside vitamins, terpenoids, and sulforaphane, in management of skin cancers through mechanisms like apoptosis induction and cell cycle regulation. Recent advancements in nanotechnology-based drug delivery systems, including NP and microemulsion formulations, are also discussed for their enhanced ability to specifically target cancer cells. The diverse roles of NPs in skin cancer therapy, especially in terms of targeted drug delivery and immune modulation, are reviewed. These innovative NPs formulations have showed improved skin penetration and tumor-specific delivery, reduced systemic toxicity and enhanced therapeutic effectiveness.
Collapse
Affiliation(s)
- Abhishek Tiwari
- Department of Pharmaceutical Chemistry, Amity Institute of Pharmacy, Amity University Uttar Pradesh, Lucknow Campus, Noida, Uttar Pradesh, India
- Amity University Uttar Pradesh, Sector 125, Noida 201313, Uttar Pradesh, India
| | - Varsha Tiwari
- Department of Pharmacognosy Chemistry, Amity Institute of Pharmacy, Lucknow Campus, Lucknow, India
- Amity University Uttar Pradesh, Sector 125, Noida 201313, Uttar Pradesh, India
| | - Ajay Sharma
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, India
| | - Arya Lakshmi Marrisetti
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Pushp Vihar, New Delhi, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Ankit Rochani
- Wegmans School of Pharmacy, St John Fisher University, Rochester, NY, USA
| | - Deepak Kaushik
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Vineet Mittal
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Renuka Jyothi S
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India
- Department of Pharmacology, Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| | - Md Sadique Hussain
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, UAE
| |
Collapse
|
8
|
Joyce P, Allen CJ, Alonso MJ, Ashford M, Bradbury MS, Germain M, Kavallaris M, Langer R, Lammers T, Peracchia MT, Popat A, Prestidge CA, Rijcken CJF, Sarmento B, Schmid RB, Schroeder A, Subramaniam S, Thorn CR, Whitehead KA, Zhao CX, Santos HA. A translational framework to DELIVER nanomedicines to the clinic. NATURE NANOTECHNOLOGY 2024; 19:1597-1611. [PMID: 39242807 DOI: 10.1038/s41565-024-01754-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 07/09/2024] [Indexed: 09/09/2024]
Abstract
Nanomedicines have created a paradigm shift in healthcare. Yet fundamental barriers still exist that prevent or delay the clinical translation of nanomedicines. Critical hurdles inhibiting clinical success include poor understanding of nanomedicines' physicochemical properties, limited exposure in the cell or tissue of interest, poor reproducibility of preclinical outcomes in clinical trials, and biocompatibility concerns. Barriers that delay translation include industrial scale-up or scale-down and good manufacturing practices, funding and navigating the regulatory environment. Here we propose the DELIVER framework comprising the core principles to be realized during preclinical development to promote clinical investigation of nanomedicines. The proposed framework comes with design, experimental, manufacturing, preclinical, clinical, regulatory and business considerations, which we recommend investigators to carefully review during early-stage nanomedicine design and development to mitigate risk and enable timely clinical success. By reducing development time and clinical trial failure, it is envisaged that this framework will help accelerate the clinical translation and maximize the impact of nanomedicines.
Collapse
Affiliation(s)
- Paul Joyce
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia.
| | - Christine J Allen
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), IDIS Research Institute, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Marianne Ashford
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | - Michelle S Bradbury
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | | | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Research Centre, School of Clinical Medicine, Faculty of Medicine and Health UNSW, Sydney, New South Wales, Australia
- UNSW Australian Centre for Nanomedicine, Faculty of Engineering, University of New South Wales (UNSW), Sydney, New South Wales, Australia
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany
- Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIOABCD), RWTH Aachen University Hospital, Aachen, Germany
| | | | - Amirali Popat
- School of Pharmacy, The University of Queensland, Woolloongabba, Queensland, Australia
| | - Clive A Prestidge
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | | | - Bruno Sarmento
- IiS - Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
- INEB - Institute for Biomedical Engineering, University of Porto, Porto, Portugal
| | - Ruth B Schmid
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Avi Schroeder
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion - Israel Institute of Technology, Haifa, Israel
| | - Santhni Subramaniam
- Centre for Pharmaceutical Innovation, UniSA Clinical and Health Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Chelsea R Thorn
- BioTherapeutics Pharmaceutical Sciences, Pfizer, Andover, MA, USA
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Chun-Xia Zhao
- School of Chemical Engineering, Faculty of Sciences, Engineering and Technology, University of Adelaide, Adelaide, South Australia, Australia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Queensland, Australia
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
- The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen, Groningen, The Netherlands.
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
9
|
Feng F, Shen J, Qi Q, Zhang Y, Ni S. Empowering brain tumor management: chimeric antigen receptor macrophage therapy. Theranostics 2024; 14:5725-5742. [PMID: 39310093 PMCID: PMC11413779 DOI: 10.7150/thno.98290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/17/2024] [Indexed: 09/25/2024] Open
Abstract
Brain tumors pose formidable challenges in oncology due to the intricate biology and the scarcity of effective treatment modalities. The emergence of immunotherapy has opened new avenues for innovative therapeutic strategies. Chimeric antigen receptor, originally investigated in T cell-based therapy, has now expanded to encompass macrophages, presenting a compelling avenue for augmenting anti-tumor immune surveillance. This emerging frontier holds promise for advancing the repertoire of therapeutic options against brain tumors, offering potential breakthroughs in combating the formidable malignancies of the central nervous system. Tumor-associated macrophages constitute a substantial portion, ranging from 30% to 50%, of the tumor tissue and exhibit tumor-promoting phenotypes within the immune-compromised microenvironment. Constructing CAR-macrophages can effectively repolarize M2-type macrophages towards an M1-type phenotype, thereby eliciting potent anti-tumor effects. CAR-macrophages can recruit T cells to the brain tumor site, thereby orchestrating a remodeling of the immune niche to effectively inhibit tumor growth. In this review, we explore the potential limitations as well as strategies for optimizing CAR-M therapy, offering insights into the future direction of this innovative therapeutic approach.
Collapse
Affiliation(s)
| | | | - Qichao Qi
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Yulin Zhang
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| | - Shilei Ni
- Department of Neurosurgery, Qilu Hospital and Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, 107 Wenhua Xi Road, Jinan, 250012, Shandong, China
| |
Collapse
|
10
|
Li J, Chen P, Ma W. The next frontier in immunotherapy: potential and challenges of CAR-macrophages. Exp Hematol Oncol 2024; 13:76. [PMID: 39103972 PMCID: PMC11302330 DOI: 10.1186/s40164-024-00549-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024] Open
Abstract
Chimeric antigen receptor macrophage (CAR-MΦ) represents a significant advancement in immunotherapy, especially for treating solid tumors where traditional CAR-T therapies face limitations. CAR-MΦ offers a promising approach to target and eradicate tumor cells by utilizing macrophages' phagocytic and antigen-presenting abilities. However, challenges such as the complex tumor microenvironment (TME), variability in antigen expression, and immune suppression limit their efficacy. This review addresses these issues, exploring mechanisms of CAR-MΦ action, optimal construct designs, and interactions within the TME. It also delves into the ex vivo manufacturing challenges of CAR-MΦ, discussing autologous and allogeneic sources and the importance of stringent quality control. The potential synergies of integrating CAR-MΦ with existing cancer therapies like checkpoint inhibitors and conventional chemotherapeutics are examined to highlight possible enhanced treatment outcomes. Furthermore, regulatory pathways for CAR-MΦ therapies are scrutinized alongside established protocols for CAR-T cells, identifying unique considerations essential for clinical trials and market approval. Proposed safety monitoring frameworks aim to manage potential adverse events, such as cytokine release syndrome, crucial for patient safety. Consolidating current research and clinical insights, this review seeks to refine CAR-MΦ therapeutic applications, overcome barriers, and suggest future research directions to transition CAR-MΦ therapies from experimental platforms to standard cancer care options.
Collapse
Affiliation(s)
- Jing Li
- The Affiliated Hospital of Qingdao University, Qingdao, 266003, Shandong, China
| | - Ping Chen
- Fujian Institute of Hematology, Fujian Provincial Key Laboratory of Hematology, Union Hospital, Fujian Medical University Fuzhou, Fujian, 350001, China
| | - Wenxue Ma
- Sanford Stem Cell Institute, Moores Cancer Center, University of California San Diego, CA, 92093, La Jolla, USA.
| |
Collapse
|
11
|
Bui TA, Mei H, Sang R, Ortega DG, Deng W. Advancements and challenges in developing in vivo CAR T cell therapies for cancer treatment. EBioMedicine 2024; 106:105266. [PMID: 39094262 PMCID: PMC11345408 DOI: 10.1016/j.ebiom.2024.105266] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 07/08/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
The Chimeric Antigen Receptor (CAR) T cell therapy has emerged as a ground-breaking immunotherapeutic approach in cancer treatment. To overcome the complexity and high manufacturing cost associated with current ex vivo CAR T cell therapy products, alternative strategies to produce CAR T cells directly in the body have been developed in recent years. These strategies involve the direct infusion of CAR genes via engineered nanocarriers or viral vectors to generate CAR T cells in situ. This review offers a comprehensive overview of recent advancements in the development of T cell-targeted CAR generation in situ. Additionally, it identifies the challenges associated with in vivo CAR T method and potential strategies to overcome these issues.
Collapse
Affiliation(s)
- Thuy Anh Bui
- School of Biomedical Engineering, University of Technology Sydney, Ultimo, NSW 2007, Australia; Whitlam Orthopaedic Research Centre, Ingham Institute for Applied Medical Research, Liverpool, NSW 2170, Australia; School of Clinical Medicine, Faculty of Medicine, University of New South Wales Sydney, Kensington, NSW 2052, Australia
| | - Haoqi Mei
- School of Biomedical Engineering, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Rui Sang
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, UNSW Sydney, NSW 2052, Australia
| | - David Gallego Ortega
- School of Biomedical Engineering, University of Technology Sydney, Ultimo, NSW 2007, Australia; Garvan Institute of Medical Research, The Kinghorn Cancer Centre, Darlinghurst, NSW 2010, Australia; School of Clinical Medicine, Faculty of Medicine, University of New South Wales Sydney, Kensington, NSW 2052, Australia
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Ultimo, NSW 2007, Australia; Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics, Faculty of Engineering, UNSW Sydney, NSW 2052, Australia.
| |
Collapse
|
12
|
Metanat Y, Viktor P, Amajd A, Kaur I, Hamed AM, Abed Al-Abadi NK, Alwan NH, Chaitanya MVNL, Lakshmaiya N, Ghildiyal P, Khalaf OM, Ciongradi CI, Sârbu I. The paths toward non-viral CAR-T cell manufacturing: A comprehensive review of state-of-the-art methods. Life Sci 2024; 348:122683. [PMID: 38702027 DOI: 10.1016/j.lfs.2024.122683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/11/2024] [Accepted: 04/28/2024] [Indexed: 05/06/2024]
Abstract
Although CAR-T cell therapy has emerged as a game-changer in cancer immunotherapy several bottlenecks limit its widespread use as a front-line therapy. Current protocols for the production of CAR-T cells rely mainly on the use of lentiviral/retroviral vectors. Nevertheless, according to the safety concerns around the use of viral vectors, there are several regulatory hurdles to their clinical use. Large-scale production of viral vectors under "Current Good Manufacturing Practice" (cGMP) involves rigorous quality control assessments and regulatory requirements that impose exorbitant costs on suppliers and as a result, lead to a significant increase in the cost of treatment. Pursuing an efficient non-viral method for genetic modification of immune cells is a hot topic in cell-based gene therapy. This study aims to investigate the current state-of-the-art in non-viral methods of CAR-T cell manufacturing. In the first part of this study, after reviewing the advantages and disadvantages of the clinical use of viral vectors, different non-viral vectors and the path of their clinical translation are discussed. These vectors include transposons (sleeping beauty, piggyBac, Tol2, and Tc Buster), programmable nucleases (ZFNs, TALENs, and CRISPR/Cas9), mRNA, plasmids, minicircles, and nanoplasmids. Afterward, various methods for efficient delivery of non-viral vectors into the cells are reviewed.
Collapse
Affiliation(s)
- Yekta Metanat
- Faculty of Medicine, Zahedan University of Medical Sciences, Sistan and Baluchestan Province, Iran
| | - Patrik Viktor
- Óbuda University, Karoly Keleti faculty, Tavaszmező u. 15-17, H-1084 Budapest, Hungary
| | - Ayesha Amajd
- Faculty of Transport and Aviation Engineering, Silesian University of Technology, Krasińskiego 8 Street, 40-019 Katowice, Poland
| | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bangalore, Karnataka, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan-303012, India
| | | | | | | | - M V N L Chaitanya
- School of pharmaceutical sciences, Lovely Professional University, Jalandhar-Delhi G.T. Road, Phagwara, Punjab - 144411, India
| | | | - Pallavi Ghildiyal
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | | | - Carmen Iulia Ciongradi
- 2nd Department of Surgery-Pediatric Surgery and Orthopedics, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania.
| | - Ioan Sârbu
- 2nd Department of Surgery-Pediatric Surgery and Orthopedics, "Grigore T. Popa" University of Medicine and Pharmacy, 700115 Iași, Romania.
| |
Collapse
|
13
|
Shin HE, Han JH, Shin S, Bae GH, Son B, Kim TH, Park HH, Park CG, Park W. M1-polarized macrophage-derived cellular nanovesicle-coated lipid nanoparticles for enhanced cancer treatment through hybridization of gene therapy and cancer immunotherapy. Acta Pharm Sin B 2024; 14:3169-3183. [PMID: 39027257 PMCID: PMC11252390 DOI: 10.1016/j.apsb.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 01/25/2024] [Accepted: 02/07/2024] [Indexed: 07/20/2024] Open
Abstract
Optimum genetic delivery for modulating target genes to diseased tissue is a major obstacle for profitable gene therapy. Lipid nanoparticles (LNPs), considered a prospective vehicle for nucleic acid delivery, have demonstrated efficacy in human use during the COVID-19 pandemic. This study introduces a novel biomaterial-based platform, M1-polarized macrophage-derived cellular nanovesicle-coated LNPs (M1-C-LNPs), specifically engineered for a combined gene-immunotherapy approach against solid tumor. The dual-function system of M1-C-LNPs encapsulates Bcl2-targeting siRNA within LNPs and immune-modulating cytokines within M1 macrophage-derived cellular nanovesicles (M1-NVs), effectively facilitating apoptosis in cancer cells without impacting T and NK cells, which activate the intratumoral immune response to promote granule-mediating killing for solid tumor eradication. Enhanced retention within tumor was observed upon intratumoral administration of M1-C-LNPs, owing to the presence of adhesion molecules on M1-NVs, thereby contributing to superior tumor growth inhibition. These findings represent a promising strategy for the development of targeted and effective nanoparticle-based cancer genetic-immunotherapy, with significant implications for advancing biomaterial use in cancer therapeutics.
Collapse
Affiliation(s)
- Ha Eun Shin
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
| | - Jun-Hyeok Han
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
- Deparment of Inteligent Precision Healthcare Convergence, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
| | - Seungyong Shin
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
| | - Ga-Hyun Bae
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
- Department of MetaBioHealth, SKKU Institute for Convergence, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
| | - Boram Son
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Tae-Hyung Kim
- Department of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hee Ho Park
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Chun Gwon Park
- Deparment of Inteligent Precision Healthcare Convergence, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
- Department of Biomedical Engineering, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
- Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Wooram Park
- Department of Integrative Biotechnology, Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
- Department of MetaBioHealth, SKKU Institute for Convergence, SKKU, Suwon, Gyeonggi 16419, Republic of Korea
- Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| |
Collapse
|
14
|
Khawar MB, Afzal A, Si Y, Sun H. Steering the course of CAR T cell therapy with lipid nanoparticles. J Nanobiotechnology 2024; 22:380. [PMID: 38943167 PMCID: PMC11212433 DOI: 10.1186/s12951-024-02630-1] [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: 03/11/2024] [Accepted: 06/09/2024] [Indexed: 07/01/2024] Open
Abstract
Lipid nanoparticles (LNPs) have proven themselves as transformative actors in chimeric antigen receptor (CAR) T cell therapy, surpassing traditional methods and addressing challenges like immunogenicity, reduced toxicity, and improved safety. Promising preclinical results signal a shift toward safer and more effective CAR T cell treatments. Ongoing research aims to validate these findings in clinical trials, marking a new era guided by LNPs utility in CAR therapy. Herein, we explore the preference for LNPs over traditional methods, highlighting the versatility of LNPs and their effective delivery of nucleic acids. Additionally, we address key challenges in clinical considerations, heralding a new era in CAR T cell therapy.
Collapse
Affiliation(s)
- Muhammad Babar Khawar
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research Yangzhou, Yangzhou, China
- Applied Molecular Biology and Biomedicine Lab, Department of Zoology, University of Narowal, Narowal, Pakistan
| | - Ali Afzal
- Shenzhen Institute of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
- Molecular Medicine and Cancer Therapeutics Lab, Department of Zoology, Faculty of Sciences and Technology, University of Central Punjab, Lahore, Pakistan
| | - Yue Si
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research Yangzhou, Yangzhou, China
| | - Haibo Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, China.
- Jiangsu Key Laboratory of Experimental & Translational Non-Coding RNA Research Yangzhou, Yangzhou, China.
| |
Collapse
|
15
|
Nie S, Qin Y, Ou L, Chen X, Li L. In Situ Reprogramming of Immune Cells Using Synthetic Nanomaterials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310168. [PMID: 38229527 DOI: 10.1002/adma.202310168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/12/2024] [Indexed: 01/18/2024]
Abstract
In the past decade, adoptive cell therapy with chimeric antigen receptor-T (CAR-T) cells has revolutionized cancer treatment. However, the complexity and high costs involved in manufacturing current adoptive cell therapy greatly inhibit its widespread availability and access. To address this, in situ cell therapy, which directly reprograms immune cells inside the body, has recently been developed as a promising alternative. Here, an overview of the recent progress in the development of synthetic nanomaterials is provided to deliver plasmid DNA or mRNA for in situ reprogramming of T cells and macrophages, focusing especially on in situ CAR therapies. Also, the main challenges for in situ immune cell reprogramming are discussed and some approaches to overcome these barriers to fulfill the clinical applications are proposed.
Collapse
Affiliation(s)
- Shihong Nie
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuyang Qin
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Liyuan Ou
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Ling Li
- West China School of Public Health and West China Fourth Hospital, and State Key Laboratory of Biotherapy, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
16
|
Hu J, Ascierto P, Cesano A, Herrmann V, Marincola FM. Shifting the paradigm: engaging multicellular networks for cancer therapy. J Transl Med 2024; 22:270. [PMID: 38475820 PMCID: PMC10936124 DOI: 10.1186/s12967-024-05043-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 11/01/2023] [Indexed: 03/14/2024] Open
Abstract
Most anti-cancer modalities are designed to directly kill cancer cells deploying mechanisms of action (MOAs) centered on the presence of a precise target on cancer cells. The efficacy of these approaches is limited because the rapidly evolving genetics of neoplasia swiftly circumvents the MOA generating therapy-resistant cancer cell clones. Other modalities engage endogenous anti-cancer mechanisms by activating the multi-cellular network (MCN) surrounding neoplastic cells in the tumor microenvironment (TME). These modalities hold a better chance of success because they activate numerous types of immune effector cells that deploy distinct cytotoxic MOAs. This in turn decreases the chance of developing treatment-resistance. Engagement of the MCN can be attained through activation of immune effector cells that in turn kill cancer cells or when direct cancer killing is complemented by the production of proinflammatory factors that secondarily recruit and activate immune effector cells. For instance, adoptive cell therapy (ACT) supplements cancer cell killing with the release of homeostatic and pro-inflammatory cytokines by the immune cells and damage associated molecular patterns (DAMPs) by dying cancer cells. The latter phenomenon, referred to as immunogenic cell death (ICD), results in an exponential escalation of anti-cancer MOAs at the tumor site. Other approaches can also induce exponential cancer killing by engaging the MCN of the TME through the release of DAMPs and additional pro-inflammatory factors by dying cancer cells. In this commentary, we will review the basic principles that support emerging paradigms likely to significantly improve the efficacy of anti-cancer therapy.
Collapse
Affiliation(s)
- Joyce Hu
- Sonata Therapeutics, Watertown, MA, 02472, USA.
| | - Paolo Ascierto
- Cancer Immunotherapy and Innovative Therapy, National Tumor Institute, Fondazione G. Pascale, 80131, Naples, Italy
| | | | | | | |
Collapse
|
17
|
Pandey R, Chiu CC, Wang LF. Immunotherapy Study on Non-small-Cell Lung Cancer (NSCLC) Combined with Cytotoxic T Cells and miRNA34a. Mol Pharm 2024; 21:1364-1381. [PMID: 38291993 PMCID: PMC10915804 DOI: 10.1021/acs.molpharmaceut.3c01040] [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: 11/02/2023] [Revised: 01/18/2024] [Accepted: 01/18/2024] [Indexed: 02/01/2024]
Abstract
Immunotherapy has emerged as a promising approach for cancer treatment, and the use of microRNAs (miRNAs) as therapeutic agents has gained significant attention. In this study, we investigated the effectiveness of immunotherapy utilizing miRNA34a and Jurkat T cells in inducing cell death in non-small-cell lung cancer cells, specifically A549 cells. Moreover, we explored the impact of Jurkat T cell activation and miRNA34a delivery using iron oxide nanorods (IONRs) on the killing of cancer cells. A549 cells were cocultured with both activated and inactivated Jurkat T cells, both before and after the delivery of miRNA34a. Surprisingly, our results revealed that even inactive Jurkat T cells were capable of inducing cell death in cancer cells. This unexpected observation suggested the presence of alternative mechanisms by which Jurkat T cells can exert cytotoxic effects on cancer cells. We stimulated Jurkat T cells using anti-CD3/CD28 and analyzed their efficacy in killing A549 compared to that of the inactive Jurkat T cells in conjunction with miRNA34a. Our findings indicated that the activation of Jurkat T cells significantly enhanced their cytotoxic potential against cancer cells compared to their inactive counterparts. The combined treatment of A549 cells with activated Jurkat T cells and miRNA34a demonstrated the highest level of cancer cell death, suggesting a synergistic effect between Jurkat T cell activation and miRNA therapy. Besides the apoptosis mechanism for the Jurkat T cells' cytotoxic effects on A549 cells, we furthermore investigated the ferroptosis pathway, which was found to have an impact on the cancer cell killing due to the presence of miRNA34a and IONRs as the delivery agent inside the cancer cells.
Collapse
Affiliation(s)
- Richa Pandey
- Department
of Medicinal and Applied Chemistry, Kaohsiung
Medical University, No. 100 Shih-Chuan first Road, Kaohsiung 80708, Taiwan
| | - Chien-Chih Chiu
- Department
of Biotechnology, Kaohsiung Medical University, No. 100 Shih-Chuan first Road, Kaohsiung 80708, Taiwan
- Department
of Medical Research, Kaohsiung Medical University
Hospital, No.100 Tzyou
first Road, Kaohsiung 80708, Taiwan
| | - Li-Fang Wang
- Department
of Medicinal and Applied Chemistry, Kaohsiung
Medical University, No. 100 Shih-Chuan first Road, Kaohsiung 80708, Taiwan
- Department
of Medical Research, Kaohsiung Medical University
Hospital, No.100 Tzyou
first Road, Kaohsiung 80708, Taiwan
- Institute
of Medical Science and Technology, National
Sun Yat-Sen University, No.70 Lien-Hai Road, Kaohsiung 804201, Taiwan
| |
Collapse
|
18
|
Foley CR, Swan SL, Swartz MA. Engineering Challenges and Opportunities in Autologous Cellular Cancer Immunotherapy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:188-198. [PMID: 38166251 PMCID: PMC11155266 DOI: 10.4049/jimmunol.2300642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/18/2023] [Indexed: 01/04/2024]
Abstract
The use of a patient's own immune or tumor cells, manipulated ex vivo, enables Ag- or patient-specific immunotherapy. Despite some clinical successes, there remain significant barriers to efficacy, broad patient population applicability, and safety. Immunotherapies that target specific tumor Ags, such as chimeric Ag receptor T cells and some dendritic cell vaccines, can mount robust immune responses against immunodominant Ags, but evolving tumor heterogeneity and antigenic downregulation can drive resistance. In contrast, whole tumor cell vaccines and tumor lysate-loaded dendritic cell vaccines target the patient's unique tumor antigenic repertoire without prior neoantigen selection; however, efficacy can be weak when lower-affinity clones dominate the T cell pool. Chimeric Ag receptor T cell and tumor-infiltrating lymphocyte therapies additionally face challenges related to genetic modification, T cell exhaustion, and immunotoxicity. In this review, we highlight some engineering approaches and opportunities to these challenges among four classes of autologous cell therapies.
Collapse
Affiliation(s)
- Colleen R. Foley
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Sheridan L. Swan
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
| | - Melody A. Swartz
- Pritzker School of Molecular Engineering, University of Chicago, Chicago, Illinois
- Committee on Immunology, University of Chicago, Chicago, Illinois
- Ben May Department of Cancer Research, University of Chicago, Chicago, Illinois
| |
Collapse
|
19
|
Mashima R, Takada S, Miyamoto Y. RNA-Based Therapeutic Technology. Int J Mol Sci 2023; 24:15230. [PMID: 37894911 PMCID: PMC10607345 DOI: 10.3390/ijms242015230] [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: 09/11/2023] [Revised: 10/09/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
RNA-based therapy has been an expanding area of clinical research since the COVID-19 outbreak. Often, its comparison has been made to DNA-based gene therapy, such as adeno-associated virus- and lentivirus-mediated therapy. These DNA-based therapies show persistent expression, with maximized therapeutic efficacy. However, accumulating data indicate that proper control of gene expression is occasionally required. For example, in cancer immunotherapy, cytokine response syndrome is detrimental for host animals, while excess activation of the immune system induces supraphysiological cytokines. RNA-based therapy seems to be a rather mild therapy, and it has room to fit unmet medical needs, whereas current DNA-based therapy has unclear issues. This review focused on RNA-based therapy for cancer immunotherapy, hematopoietic disorders, and inherited disorders, which have received attention for possible clinical applications.
Collapse
Affiliation(s)
- Ryuichi Mashima
- Department of Clinical Laboratory Medicine, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Shuji Takada
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Yoshitaka Miyamoto
- Department of Maternal-Fetal Biology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| |
Collapse
|
20
|
Han J, Lim J, Wang CPJ, Han JH, Shin HE, Kim SN, Jeong D, Lee SH, Chun BH, Park CG, Park W. Lipid nanoparticle-based mRNA delivery systems for cancer immunotherapy. NANO CONVERGENCE 2023; 10:36. [PMID: 37550567 PMCID: PMC10406775 DOI: 10.1186/s40580-023-00385-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 07/23/2023] [Indexed: 08/09/2023]
Abstract
Cancer immunotherapy, which harnesses the power of the immune system, has shown immense promise in the fight against malignancies. Messenger RNA (mRNA) stands as a versatile instrument in this context, with its capacity to encode tumor-associated antigens (TAAs), immune cell receptors, cytokines, and antibodies. Nevertheless, the inherent structural instability of mRNA requires the development of effective delivery systems. Lipid nanoparticles (LNPs) have emerged as significant candidates for mRNA delivery in cancer immunotherapy, providing both protection to the mRNA and enhanced intracellular delivery efficiency. In this review, we offer a comprehensive summary of the recent advancements in LNP-based mRNA delivery systems, with a focus on strategies for optimizing the design and delivery of mRNA-encoded therapeutics in cancer treatment. Furthermore, we delve into the challenges encountered in this field and contemplate future perspectives, aiming to improve the safety and efficacy of LNP-based mRNA cancer immunotherapies.
Collapse
Affiliation(s)
- Jieun Han
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea
- Institute of Biotechnology and Bioengineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Jaesung Lim
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University, Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Chi-Pin James Wang
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University, Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Jun-Hyeok Han
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University, Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Ha Eun Shin
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Se-Na Kim
- MediArk, Chungdae-ro 1, Seowon-gu, Cheongju, Chungcheongbuk, 28644, Republic of Korea
| | - Dooyong Jeong
- R&D center of HLB Pharmaceutical Co., Ltd., Hwaseong, Gyeonggi, 18469, Republic of Korea
| | - Sang Hwi Lee
- R&D center of HLB Pharmaceutical Co., Ltd., Hwaseong, Gyeonggi, 18469, Republic of Korea
| | - Bok-Hwan Chun
- R&D center of HLB Pharmaceutical Co., Ltd., Hwaseong, Gyeonggi, 18469, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea.
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University, Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea.
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea.
| | - Wooram Park
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea.
- Institute of Biotechnology and Bioengineering, College of Biotechnology and Bioengineering, Sungkyunkwan University, Seobu-ro 2066, Suwon, Gyeonggi, 16419, Republic of Korea.
| |
Collapse
|
21
|
Yun CH. Relationship between Tissue Regeneration and Immune Modulation. Tissue Eng Regen Med 2023; 20:325-327. [PMID: 37079197 PMCID: PMC10219898 DOI: 10.1007/s13770-023-00544-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023] Open
Affiliation(s)
- Cheol-Heui Yun
- Department of Agricultural Biotechnology, and Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
- Center for Food and Bioconvergence, and Interdisciplinary Programs in Agricultural Genomics, Seoul National University, Seoul, 08826, Republic of Korea.
- Institutes of Green Bio Science and Technology, Seoul National University, Pyeongchang, Gangwon-Do, 25354, Republic of Korea.
| |
Collapse
|