1
|
Shi Y, Mao J, Wang S, Ma S, Luo L, You J. Pharmaceutical strategies for optimized mRNA expression. Biomaterials 2025; 314:122853. [PMID: 39342919 DOI: 10.1016/j.biomaterials.2024.122853] [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: 06/13/2024] [Revised: 09/19/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
Messenger RNA (mRNA)-based immunotherapies and protein in situ production therapies hold great promise for addressing theoretically all the diseases characterized by aberrant protein levels. The safe, stable, and precise delivery of mRNA to target cells via appropriate pharmaceutical strategies is a prerequisite for its optimal efficacy. In this review, we summarize the structural characteristics, mode of action, development prospects, and limitations of existing mRNA delivery systems from a pharmaceutical perspective, with an emphasis on the impacts from formulation adjustments and preparation techniques of non-viral vectors on mRNA stability, target site accumulation and transfection efficiency. In addition, we introduce strategies for synergistical combination of mRNA and small molecules to augment the potency or mitigate the adverse effects of mRNA therapeutics. Lastly, we delve into the challenges impeding the development of mRNA drugs while exploring promising avenues for future advancements.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Jiapeng Mao
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Sijie Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China
| | - Siyao Ma
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, 166 Qiutaobei Road, Hangzhou, Zhejiang, 310017, PR China
| | - Lihua Luo
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China.
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, Zhejiang, 310058, PR China; State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, 79 Qingchun Road, Shangcheng District, Hangzhou, Zhejiang, 310006, PR China; The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 QingChun Road, Hangzhou, Zhejiang, 310000, PR China; Jinhua Institute of Zhejiang University, 498 Yiwu Street, Jinhua, Zhejiang, 321299, PR China.
| |
Collapse
|
2
|
Vaidya K, Regan MS, Lin J, Houle J, Gupta A, Stopka SA, Agar NYR, Hammond PT, Boehnke N. Pooled Nanoparticle Screening Using a Chemical Barcoding Approach. Angew Chem Int Ed Engl 2025; 64:e202420052. [PMID: 39714325 PMCID: PMC11773315 DOI: 10.1002/anie.202420052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
We report the development of a small molecule-based barcoding platform for pooled screening of nanoparticle delivery. Using aryl halide-based tags (halocodes), we achieve high-sensitivity detection via gas chromatography coupled with mass spectrometry or electron capture. This enables barcoding and tracking of nanoparticles with minimal halocode concentrations and without altering their physicochemical properties. To demonstrate the utility of our platform for pooled screening, we synthesized a halocoded library of polylactide-co-glycolide (PLGA) nanoparticles and quantified uptake in ovarian cancer cells in a pooled manner. Our findings correlate with conventional fluorescence-based assays. Additionally, we demonstrate the potential of halocodes for spatial mapping of nanoparticles using mass spectrometry imaging (MSI). Halocoding presents an accessible and modular nanoparticle screening platform capable of quantifying delivery of pooled nanocarrier libraries in a range of biological settings.
Collapse
Affiliation(s)
- Katherine Vaidya
- Department of Chemical Engineering and Materials ScienceUniversity of Minnesota Twin CitiesMinneapolis, MNUSA
| | - Michael S. Regan
- Department of Neurosurgery Brigham and Women's HospitalHarvard Medical SchoolBoston, MAUSA
| | - James Lin
- Department of Chemical Engineering and Materials ScienceUniversity of Minnesota Twin CitiesMinneapolis, MNUSA
| | - Jenna Houle
- Department of BioengineeringMassachusetts Institute of TechnologyCambridge, MAUSA
| | - Aanchal Gupta
- Department of Chemical Engineering and Materials ScienceUniversity of Minnesota Twin CitiesMinneapolis, MNUSA
| | - Sylwia A. Stopka
- Department of Neurosurgery Brigham and Women's HospitalHarvard Medical SchoolBoston, MAUSA
| | - Nathalie Y. R. Agar
- Department of Neurosurgery Brigham and Women's HospitalHarvard Medical SchoolBoston, MAUSA
- Department of Radiology Brigham and Women's HospitalHarvard Medical SchoolBoston, MAUSA
- Department of Cancer Biology Dana-Farber Cancer InstituteHarvard Medical SchoolBoston, MAUSA
| | - Paula T. Hammond
- Department of Chemical EngineeringMassachusetts Institute of TechnologyCambridge, MAUSA
- Koch Institute for Integrative Cancer Research at MITMassachusetts Institute of TechnologyCambridge, MAUSA
| | - Natalie Boehnke
- Department of Chemical Engineering and Materials ScienceUniversity of Minnesota Twin CitiesMinneapolis, MNUSA
| |
Collapse
|
3
|
Chettri D, Satapathy BP, Yadav R, Uttam V, Jain A, Prakash H. CAR-macrophages: tailoring cancer immunotherapy. Front Immunol 2025; 15:1532833. [PMID: 39877364 PMCID: PMC11772431 DOI: 10.3389/fimmu.2024.1532833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Affiliation(s)
- Dewan Chettri
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, India
| | - Bibhu Prasad Satapathy
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, India
| | - Rohit Yadav
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, India
| | - Vivek Uttam
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, India
| | - Aklank Jain
- Non-Coding RNA and Cancer Biology Laboratory, Department of Zoology, Central University of Punjab, Bathinda, India
| | - Hridayesh Prakash
- Amity Centre for Translational Research, Amity University, Noida, India
| |
Collapse
|
4
|
Chen Z, Shu J, Hu Y, Mei H. Synergistic integration of mRNA-LNP with CAR-engineered immune cells: Pioneering progress in immunotherapy. Mol Ther 2024; 32:3772-3792. [PMID: 39295145 PMCID: PMC11573621 DOI: 10.1016/j.ymthe.2024.09.019] [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: 04/26/2024] [Revised: 08/18/2024] [Accepted: 09/13/2024] [Indexed: 09/21/2024] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy has emerged as a revolutionary approach in the treatment of malignancies. Despite its remarkable successes, this field continues to grapple with challenges such as scalability, safety concerns, limited therapeutic effect, in vivo persistence, and the need for precise control over CAR expression. In the post-pandemic era of COVID-19 vaccine immunization, the application of messenger RNA (mRNA) encapsulated within lipid nanoparticles (LNPs) has recently garnered significant attention as a potential solution to address these challenges. This review delves into the dynamic landscape of mRNA-LNP technology and its potential implications for CAR-engineered immune cell-based immunotherapy.
Collapse
Affiliation(s)
- Zhaozhao Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Jinhui Shu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China.
| |
Collapse
|
5
|
Carvalho HMB, Fidalgo TAS, Acúrcio RC, Matos AI, Satchi-Fainaro R, Florindo HF. Better, Faster, Stronger: Accelerating mRNA-Based Immunotherapies With Nanocarriers. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2017. [PMID: 39537215 DOI: 10.1002/wnan.2017] [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: 06/15/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Messenger ribonucleic acid (mRNA) therapeutics are attracting attention as promising tools in cancer immunotherapy due to their ability to leverage the in vivo expression of all known protein sequences. Even small amounts of mRNA can have a powerful effect on cancer vaccines by promoting the synthesis of tumor-specific antigens (TSA) or tumor-associated antigens (TAA) by antigen-presenting cells (APC). These antigens are then presented to T cells, eliciting strong antitumor immune stimulation. The potential of mRNA can be further enhanced by expressing immunomodulatory agents, such as cytokines, antibodies, and chimeric antigen receptors (CAR), enhancing tumor immunity. Recent research also explores mRNA-encoded tumor death inducers or tumor microenvironment (TME) modulators. Despite its promise, the clinical translation of mRNA-based anticancer strategies faces challenges, including inefficient targeted delivery in vivo, failure of endosomal escape, and inadequate intracellular mRNA release, resulting in poor transfection efficiencies. Inspired by the approval of lipid nanoparticle-loaded mRNA vaccines against coronavirus disease 2019 (COVID-19) and the encouraging outcomes of mRNA-based cancer therapies in trials, innovative nonviral nanotechnology delivery systems have been engineered. These aim to advance mRNA-based cancer immunotherapies from research to clinical application. This review summarizes recent preclinical and clinical progress in lipid and polymeric nanomedicines for delivering mRNA-encoded antitumor therapeutics, including cytokines and antibody-based immunotherapies, cancer vaccines, and CAR therapies. It also addresses advanced delivery systems for direct oncolysis or TME reprogramming and highlights key challenges in translating these therapies to clinical use, exploring future perspectives, including the role of artificial intelligence and machine learning in their development.
Collapse
Affiliation(s)
- Henrique M B Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Tiago A S Fidalgo
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Rita C Acúrcio
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Ana I Matos
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
6
|
Wei PS, Thota N, John G, Chang E, Lee S, Wang Y, Ma Z, Tsai YH, Mei KC. Enhancing RNA-lipid nanoparticle delivery: Organ- and cell-specificity and barcoding strategies. J Control Release 2024; 375:366-388. [PMID: 39179112 PMCID: PMC11972657 DOI: 10.1016/j.jconrel.2024.08.030] [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: 04/24/2024] [Revised: 07/25/2024] [Accepted: 08/19/2024] [Indexed: 08/26/2024]
Abstract
Recent advancements in RNA therapeutics highlight the critical need for precision gene delivery systems that target specific organs and cells. Lipid nanoparticles (LNPs) have emerged as key vectors in delivering mRNA and siRNA, offering protection against enzymatic degradation, enabling targeted delivery and cellular uptake, and facilitating RNA cargo release into the cytosol. This review discusses the development and optimization of organ- and cell-specific LNPs, focusing on their design, mechanisms of action, and therapeutic applications. We explore innovations such as DNA/RNA barcoding, which facilitates high-throughput screening and precise adjustments in formulations. We address major challenges, including improving endosomal escape, minimizing off-target effects, and enhancing delivery efficiencies. Notable clinical trials and recent FDA approvals illustrate the practical applications and future potential of LNP-based RNA therapies. Our findings suggest that while considerable progress has been made, continued research is essential to resolve existing limitations and bridge the gap between preclinical and clinical evaluation of the safety and efficacy of RNA therapeutics. This review highlights the dynamic progress in LNP research. It outlines a roadmap for future advancements in RNA-based precision medicine.
Collapse
Affiliation(s)
- Pu-Sheng Wei
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Nagasri Thota
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Greshma John
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Evelyn Chang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Sunjae Lee
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Yuanjun Wang
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Zitao Ma
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Yu-Hsuan Tsai
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA
| | - Kuo-Ching Mei
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Binghamton, Johnson City, New York, NY 13790, USA.
| |
Collapse
|
7
|
Vaidya K, Regan MS, Lin J, Houle J, Stopka SA, Agar NYR, Hammond PT, Boehnke N. Pooled nanoparticle screening using a chemical barcoding approach. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.24.614746. [PMID: 39386478 PMCID: PMC11463557 DOI: 10.1101/2024.09.24.614746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
We report the development of a small molecule-based barcoding platform for pooled screening of nanoparticle delivery. Using aryl halide-based tags (halocodes), we achieve high-sensitivity detection via gas chromatography coupled with mass spectrometry or electron capture. This enables barcoding and tracking of nanoparticles with minimal halocode concentrations and without altering their physicochemical properties. To demonstrate the utility of our platform for pooled screening, we synthesized a halocoded library of polylactide-co-glycolide (PLGA) nanoparticles and quantified uptake in ovarian cancer cells in a pooled manner. Our findings correlate with conventional fluorescence-based assays. Additionally, we demonstrate the potential of halocodes for spatial mapping of nanoparticles using mass spectrometry imaging (MSI). Halocoding presents an accessible and modular nanoparticle screening platform capable of quantifying delivery of pooled nanocarrier libraries in a range of biological settings.
Collapse
|
8
|
Jain M, Yu X, Schneck JP, Green JJ. Nanoparticle Targeting Strategies for Lipid and Polymer-Based Gene Delivery to Immune Cells In Vivo. SMALL SCIENCE 2024; 4:2400248. [PMID: 40212067 PMCID: PMC11935263 DOI: 10.1002/smsc.202400248] [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: 05/20/2024] [Revised: 06/15/2024] [Indexed: 04/13/2025] Open
Abstract
Lipid nanoparticles and polymeric nanoparticles are promising biomaterial platforms for robust intracellular DNA and mRNA delivery, highlighted by the widespread use of nanoparticle- (NP) based mRNA vaccines to help end the COVID-19 pandemic. Recent research has sought to adapt this nanotechnology to transfect and engineer immune cells in vivo. The immune system is an especially appealing target due to its involvement in many different diseases, and ex vivo-engineered immune cell therapies like chimeric antigen receptor (CAR) T therapy have already demonstrated remarkable clinical success in certain blood cancers. Although gene delivery can potentially address some of the cost and manufacturing concerns associated with current autologous immune cell therapies, transfecting immune cells in vivo is challenging. Not only is extrahepatic NP delivery to lymphoid organs difficult, but immune cells like T cells have demonstrated particular resistance to transfection. Despite these challenges, the modular nature of NPs allows researchers to examine critical structure-function relationships between a particle's properties and its ability to specifically engineer immune cells in vivo. Herein, several nanomaterial components are outlined, including targeting ligands, nucleic acid cargo, chemical properties, physical properties, and the route of administration to specifically target NPs to immune cells for optimal in vivo transfection.
Collapse
Affiliation(s)
- Manav Jain
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
| | - Xinjie Yu
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Department of Chemical & Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| | - Jonathan P. Schneck
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for Cell EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Departments of Pathology and MedicineJohns Hopkins University School of MedicineBaltimoreMD21231USA
| | - Jordan J. Green
- Department of Biomedical EngineeringJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Institute for NanoBioTechnology, and Translational Tissue Engineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Johns Hopkins Translational ImmunoEngineering CenterJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Department of Chemical & Biomolecular EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
- Department of OncologyThe Sidney Kimmel Comprehensive Cancer CenterThe Bloomberg∼Kimmel Institute for Cancer ImmunotherapyJohns Hopkins University School of MedicineBaltimoreMD21231USA
- Departments of Ophthalmology, Neurosurgery, and Materials Science & EngineeringJohns Hopkins UniversityBaltimoreMD21218USA
| |
Collapse
|