1
|
Shah RM, Jadhav SR, Bryant G, Kaur IP, Harding IH. On the formation and stability mechanisms of diverse lipid-based nanostructures for drug delivery. Adv Colloid Interface Sci 2025; 338:103402. [PMID: 39879887 DOI: 10.1016/j.cis.2025.103402] [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: 08/02/2024] [Revised: 12/02/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025]
Abstract
In the evolving landscape of nanotechnology and pharmaceuticals, lipid nanostructures have emerged as pivotal areas of research due to their unique ability to mimic biological membranes and encapsulate active molecules. These nanostructures offer promising avenues for drug delivery, vaccine development, and diagnostic applications. This comprehensive review explores the complex mechanisms underlying the formation and stability of various lipid nanostructures, including lipid liquid crystalline nanoparticles and solid lipid nanoparticles. Drawing upon a wide array of studies, we integrate current knowledge on the physicochemical properties of lipids that contribute to nanostructure formation, such as lipid composition, charge, and the role of environmental factors such as pH and ionic strength. We further discuss the stabilisation mechanisms that preserve the integrity and functionality of these nanostructures in biological systems, highlighting the influence of surface modification, PEGylation, and the incorporation of stabilising agents. Through a methodical examination of both classical theories and cutting-edge research, our review highlights the critical factors that dictate the self-assembly of lipids into nanostructures, the dynamics of their formation, and the interplay between different stabilising forces. The implications of these insights for the design of lipid-based delivery systems are vast, offering the potential to enhance the bioavailability of therapeutics, target specific tissues or cells, and minimise adverse effects. The integration of lipid nanostructures in pharmaceutical nanotechnology not only stands to revolutionise the delivery of therapeutic agents but also paves the way for innovative applications in targeted therapy, personalised medicine, and vaccine adjuvant development. By bridging the gap between fundamental biophysical studies and applied research, this review contributes to the ongoing discourse on lipid nanostructures, advocating for a multidisciplinary approach to harness their full potential.
Collapse
Affiliation(s)
- Rohan M Shah
- Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Australia; Iverson Health Innovation Research Institute, Swinburne University of Technology, Melbourne, Australia.
| | - Snehal R Jadhav
- Consumer Analytical Safety Sensory (CASS) Food Research Centre, School of Exercise and Nutrition Sciences, Deakin University, Melbourne, Australia
| | - Gary Bryant
- School of Science, STEM College, RMIT University, Melbourne, Australia
| | - Indu Pal Kaur
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
| | - Ian H Harding
- Department of Chemistry and Biotechnology, School of Science, Computing and Engineering Technologies, Swinburne University of Technology, Melbourne, Australia
| |
Collapse
|
2
|
Ding S, Alexander E, Liang H, Kulchar RJ, Singh R, Herzog RW, Daniell H, Leong KW. Synthetic and Biogenic Materials for Oral Delivery of Biologics: From Bench to Bedside. Chem Rev 2025. [PMID: 40168474 DOI: 10.1021/acs.chemrev.4c00482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
The development of nucleic acid and protein drugs for oral delivery has lagged behind their production for conventional nonoral routes. Over the past decade, the evolution of DNA- and RNA-based technologies combined with the innovation of state-of-the-art delivery vehicles for nucleic acids has brought rapid advancements to the biopharmaceutical field. Nucleic acid therapies have the potential to achieve long-lasting effects, or even cures, by inhibiting or editing genes, which is not possible with conventional small-molecule drugs. However, challenges and limitations must be addressed before these therapies can provide cures for chronic conditions and rare diseases, rather than only offering temporary relief. Nucleic acids and proteins face premature degradation in the acidic, enzyme-rich stomach environment and are rapidly cleared by the liver. To overcome these challenges, various delivery vehicles have been developed to transport therapeutic compounds to the intestines, where the active compounds are released and gut microbiota and mucosal immune system also play an important role. This review provides a comprehensive overview of the promises and pitfalls associated with the oral route of administration of biologics, current delivery systems, applications of orally delivered therapeutics, and the challenges and considerations for translation of nucleic acid and protein therapeutics into clinical practice.
Collapse
Affiliation(s)
- Suwan Ding
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Elena Alexander
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Huiyi Liang
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| | - Rachel J Kulchar
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Rahul Singh
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Roland W Herzog
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Henry Daniell
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 South 40th Street, Philadelphia, Pennsylvania 19104, United States
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, 500 West 120th Street, New York, New York 10027, United States
| |
Collapse
|
3
|
Lam JH, Sinsinbar G, Loo SY, Chia TW, Lee YJ, Fong JY, Chia YE, Penna RR, Liu S, Pascolo S, Schultheis K, Nallani M. Development of Thermostable and Immunogenic Block Copolymer Nanoparticles (BNPs) for mRNA Delivery. Biomacromolecules 2025. [PMID: 40163903 DOI: 10.1021/acs.biomac.4c01820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Combining an amphiphilic block copolymer polybutadiene-b-poly(ethylene glycol) (PBD-b-PEO), an ionizable lipid, a helper lipid, and cholesterol produces thermostable BNPs. Luciferase mRNA-BNPs can be stored for over 1 year at 4 °C with no evidence of degradation to the mRNA or nanocarrier. In vivo, mRNA-BNPs exhibit a greater affinity for secondary lymphoid organs than mRNA-lipid nanoparticles (LNPs) and are efficiently taken up by macrophages and dendritic cells. Freshly fabricated ovalbumin (OVA) mRNA-BNPs elicit robust OVA-specific IgG and functional memory CD8+ T cells that persist for at least 5 months. Immunogenicity remains intact after 24 weeks of storage at 4 °C. Anti-PEG antibodies are not boosted by the repeated administration of mRNA-BNPs, unlike mRNA-LNPs. Syrian hamsters vaccinated with SARS-CoV-2 spike mRNA-BNPs are protected against weight loss associated with infection and potently suppress pulmonary viral loads. Protective efficacy is comparable to that conferred by a Comirnaty biosimilar. Cumulatively, mRNA-BNPs are thermostable, immunogenic and possess the potential for clinical application.
Collapse
Affiliation(s)
| | | | - Ser Yue Loo
- ACM Biolabs Pte Ltd, Singapore 638075, Singapore
| | | | - Yan Jun Lee
- ACM Biolabs Pte Ltd, Singapore 638075, Singapore
| | - Jing Yi Fong
- ACM Biolabs Pte Ltd, Singapore 638075, Singapore
| | | | - Rocco Roberto Penna
- Department of Dermatology, University Hospital Zurich (USZ), University of Zurich (UZH), 8091 Zurich, Switzerland
- Faculty of Science, University of Zurich, 8006 Zurich, Switzerland
| | | | - Steve Pascolo
- Department of Dermatology, University Hospital Zurich (USZ), University of Zurich (UZH), 8091 Zurich, Switzerland
- Faculty of Science, University of Zurich, 8006 Zurich, Switzerland
| | | | - Madhavan Nallani
- ACM Biolabs Pte Ltd, Singapore 638075, Singapore
- ACM Biosciences AG, 4051 Basel, Switzerland
| |
Collapse
|
4
|
Razzaq S, Fatima I, Moafian Z, Rahdar A, Fathi-Karkan S, Kharaba Z, Shirzad M, Khan A, Pandey S. Nanomedicine innovations in colon and rectal cancer: advances in targeted drug and gene delivery systems. Med Oncol 2025; 42:113. [PMID: 40097759 DOI: 10.1007/s12032-025-02670-z] [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: 01/01/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Nanotechnology has revolutionized cancer diagnostics and therapy, offering unprecedented possibilities to overcome the constraints of conventional treatments. This study provides a detailed overview of the current progress and difficulties in the creation of nanostructured materials, with a specific emphasis on their use in drug and gene delivery systems. The study examines tactics that attempt to improve the effectiveness and safety of chemotherapeutic drugs such as doxorubicin (Dox) by focusing on the potential of antibody-drug conjugates and functionalized nanoparticles. Moreover, it clarifies the challenges encountered in administering nanoparticles orally for gastrointestinal treatments, emphasizing the crucial physicochemical properties that affect their behavior in the gastrointestinal system. This study highlights the transformational potential of nanostructured materials in precision oncology by examining advanced breakthroughs such cell membrane-camouflaged nanoparticles and inorganic nanoparticles designed for gastrointestinal disorders. The text investigates the processes involved in the absorption of nanoparticles and their destruction in lysosomes, revealing the many methods in which enterocytes take up these particles. This study strongly supports the use of advanced nanoparticle-based methods to reduce the harmful effects on the whole body and improve the effectiveness of therapy, based on a thorough examination of current experiments on animals and humans. The main objective of this paper is to provide a fundamental comprehension that will stimulate more investigation and practical use in the field of cancer nanomedicine, advancing its boundaries.
Collapse
Affiliation(s)
- Sobia Razzaq
- School of Pharmacy, University of Management and Technology, Lahore, Punjab, Pakistan
| | - Iqra Fatima
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Zeinab Moafian
- Department of Chemistry and Biochemistry, University of Delaware, Newark, USA
| | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol, 538-98615, Iran.
| | - Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran.
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 9414974877, Iran.
| | - Zelal Kharaba
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, UAE
| | - Maryam Shirzad
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ahmad Khan
- Department of Pharmacy, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| | - Sadanand Pandey
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, 173229, Himachal Pradesh, India.
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk, 38541, Republic of Korea.
| |
Collapse
|
5
|
Saha S, Tandon R, Sanku J, Kumari A, Shukla R, Srivastava N. siRNA-based Therapeutics in Hormone-driven Cancers: Advancements and benefits over conventional treatments. Int J Pharm 2025; 674:125463. [PMID: 40081431 DOI: 10.1016/j.ijpharm.2025.125463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/10/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025]
Abstract
Hormone-related cancers, also known as hormone-sensitive or hormone-dependent cancers, rely on hormones such as estrogen, testosterone, and progesterone for growth. These malignancies, including breast, pituitary, thyroid, ovarian, uterine, cervical, and prostate cancers, often exhibit accelerated progression in response to hormonal signaling. Small interfering RNA (siRNA) has emerged as a groundbreaking gene suppression therapy since the FDA approval of its first product in 2018. With over 200 ongoing clinical trials, siRNA is being actively explored as a targeted treatment for hormone-related cancers. Its ability to silence specific oncogenes offers significant advantages over conventional therapies, which are often associated with toxicity, resistance, and non-specific targeting. However, challenges in siRNA delivery remain a major barrier to its clinical translation, limiting its ability to reach target cells effectively. This review evaluates the potential of siRNA in hormone-related cancers, addressing the shortcomings of traditional treatments while examining novel strategies to enhance siRNA delivery and overcome tumor microenvironment obstacles. Notably, no existing literature comprehensively consolidates siRNA-based therapies for these cancers, emphasizing the importance of this manuscript in bridging current knowledge gaps and advancing the translational application of siRNA therapeutics.
Collapse
Affiliation(s)
- Sayani Saha
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Reetika Tandon
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Jhansi Sanku
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Anchala Kumari
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, India.
| |
Collapse
|
6
|
Wu J, Zuo J, Dou W, Wang K, Long J, Yu C, Miao Y, Liao Y, Li Y, Cao Y, Lu L, Jin Y, Zhang B, Yang J. Rapidly separable bubble microneedle-patch system present superior transdermal mRNA delivery efficiency. Int J Pharm 2025; 674:125427. [PMID: 40074159 DOI: 10.1016/j.ijpharm.2025.125427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
Traditional mRNA vaccine formulation loaded by lipid nanoparticle (mRNA-LNP) has several shortcomings in clinical application, including the need for cryopreservation, discomfort associated with intramuscular injections, and the risk of liver aggregation. Dissolvable microneedles (DMNs), as a novel transdermal drug delivery platform, can overcome the skin barrier to deliver drugs directly into the skin in a minimally invasive manner. However, mRNA-LNP is unstable and easily degraded during the solidification of DMN. In this study, we proposed to establish a rapidly dissolvable bubble microneedle patch (bMNP) system for the transdermal delivery of mRNA-LNP. We explored to use polyvinyl alcohol (PVA) and trehalose for the first time as matrix material for preparing microneedles. Our results demonstrate that the stability of the mRNA-LNP was obviously improved. The mRNA in this bMNP system can be stored at room temperature for at least one month. Furthermore, the existence of air bubbles between the needle tip and the dorsal scale of bMNP can achieve dorsal scale separation by applying shear force after inserting into subcutaneous tissue, and effectively target lymph nodes in vivo after releasing mRNA-LNP. Using mRNA that encodes the spike protein from SARS-CoV-2 as a test case, the rapidly separable bMNP system induced the production of significant levels of spike-specific IgG antibodies, neutralizing antibodies, and a Th1-polarized T cell response, providing an alternative route for mRNA delivery. Our research is expected to provide a promising transdermal drug delivery strategy that can improve mRNA vaccine accessibility.
Collapse
Affiliation(s)
- Jiayu Wu
- Department of Pharmacy & State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, People's Republic of China; Bioinformatics Center of AMMS, Beijing, People's Republic of China
| | - Jun Zuo
- Bioinformatics Center of AMMS, Beijing, People's Republic of China
| | - Wei Dou
- Department of Pharmacy & State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, People's Republic of China; Bioinformatics Center of AMMS, Beijing, People's Republic of China
| | - Ke Wang
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Jinrong Long
- Bioinformatics Center of AMMS, Beijing, People's Republic of China
| | - Changxiao Yu
- Bioinformatics Center of AMMS, Beijing, People's Republic of China
| | - Yiqi Miao
- Bioinformatics Center of AMMS, Beijing, People's Republic of China
| | - Yuqin Liao
- Bioinformatics Center of AMMS, Beijing, People's Republic of China
| | - Yanyan Li
- Bioinformatics Center of AMMS, Beijing, People's Republic of China
| | - Yiming Cao
- Bioinformatics Center of AMMS, Beijing, People's Republic of China
| | - Lu Lu
- Department of Pharmacy & State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, People's Republic of China
| | - Yiguang Jin
- Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.
| | - Bo Zhang
- Department of Pharmacy & State Key Laboratory of Complex Severe and Rare Disease, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, People's Republic of China.
| | - Jing Yang
- Bioinformatics Center of AMMS, Beijing, People's Republic of China.
| |
Collapse
|
7
|
Prajapati SK, Jain A, Bajpai M. Lipid-based nanoformulations in onychomycosis therapy: addressing challenges of current therapies and advancing treatment. RSC Adv 2025; 15:7799-7825. [PMID: 40070389 PMCID: PMC11895809 DOI: 10.1039/d5ra00387c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Onychomycosis significantly impacts approximately 20% of the global population. The physical barriers of the nail structure make fungal infections a persistent therapeutic challenge. Traditional approaches, including topical and oral antifungal agents, have limitations such as toxicities, low nail permeability, adverse effects, and high recurrence rates. Consequently, emerging lipid-based delivery systems have gained interest because of their potential to address these drawbacks. Nanostructured lipid carriers (NLCs), solid lipid nanoparticles (SLNs), liposomes, and transferosomes are innovative formulations that offer enhanced drug solubility, sustained release, and targeted delivery to the nail matrix. These lipid-mediated approaches have shown promise in overcoming the hurdles associated with conventional therapies, thereby improving treatment outcomes, patient compliance, and the overall quality of life. A comprehensive review focusing on the potential of lipid-based drug delivery systems in treating onychomycosis is lacking in the existing literature. This review explores various aspects of the clinical presentation of onychomycosis, available treatments, challenges associated with treatment, formulation science related to lipid-based vehicles and their applications, highlighted by the promising aspects of these novel formulations, and provides insights into clinical developments. In addition, the regulatory perspective is critical to such development, and approval is discussed, particularly in managing regulatory compliance complexities to ensure successful implementation. The holistic approach provides a comprehensive basis for determining lipid-based drug delivery systems' state-of-the-art and future scope.
Collapse
Affiliation(s)
| | - Ankit Jain
- Department of Pharmacy, Birla Institute of Technology and Science-Pilani Pilani Campus Pilani India
| | - Meenakshi Bajpai
- Institute of Pharmaceutical Research, GLA University Mathura India
| |
Collapse
|
8
|
Yoo S, Faisal M, Bae SH, Youn K, Park HJ, Kwon SP, Hwang IK, Lee J, Kim HJ, Nam JH, Keum G, Bang EK. Novel Less Toxic, Lymphoid Tissue-Targeted Lipid Nanoparticles Containing a Vitamin B5-Derived Ionizable Lipid for mRNA Vaccine Delivery. Adv Healthc Mater 2025; 14:e2403366. [PMID: 39502027 PMCID: PMC11912100 DOI: 10.1002/adhm.202403366] [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: 09/05/2024] [Revised: 10/23/2024] [Indexed: 03/18/2025]
Abstract
Following their approval by the Food and Drug Administration, lipid nanoparticles (LNPs) have emerged as promising tools for delivering mRNA vaccines and therapeutics. Ionizable lipids are among the essential components of LNPs, as they play crucial roles in encapsulating mRNA and facilitating its release into the cytosol. In this study, 17 innovative ionizable lipids using vitamin B5 are designed as the core structure, aiming to reduce toxicity, to maintain vaccine efficiency, and to ensure synthetic feasibility. The top-performing LNP in terms of mRNA vaccine delivery in the mouse model is LNP 5097, which is generated by incorporating ionizable lipid I97. mRNA⊂LNP 5097 demonstrates favorable structural and physicochemical properties, high mRNA transfection efficiency, and long-term stability. Moreover, mRNA⊂LNP 5097 specifically delivers the mRNA to the spleen and lymph nodes in model mice, induces balanced Th1/Th2 immune responses, and elicits the production of high levels of neutralizing antibodies with low toxicity. The findings here suggest the high utility of LNP 5097, which includes novel vitamin B5-derived ionizable lipids with reduced toxicity, in mRNA vaccine research for both infectious diseases and cancer.
Collapse
Affiliation(s)
- Soyeon Yoo
- Brain Technology Research Center, Brain Science Research Division, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Muhammad Faisal
- Brain Technology Research Center, Brain Science Research Division, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Seo-Hyeon Bae
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43 Jibong-ro, Bucheon, Gyeonggi-do, 14662, Republic of Korea
- BK21 four Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon, Gyeonggi-do, 14662, South Korea
| | - Kounghwa Youn
- Brain Technology Research Center, Brain Science Research Division, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, 7-13 Kyungheedae-ro 6-gil, Dongdaemun-gu, Seoul, 02447, Republic of Korea
| | - Hyo-Jung Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43 Jibong-ro, Bucheon, Gyeonggi-do, 14662, Republic of Korea
- BK21 four Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon, Gyeonggi-do, 14662, South Korea
| | - Sung Pil Kwon
- Brain Technology Research Center, Brain Science Research Division, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Il Kwon Hwang
- Brain Technology Research Center, Brain Science Research Division, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Department of Chemical and Biological Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Jisun Lee
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43 Jibong-ro, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Hyeong Jun Kim
- Medicinal Materials Research Center, Biomedical Research Division, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Jae-Hwan Nam
- Department of Medical and Biological Sciences, The Catholic University of Korea, 43 Jibong-ro, Bucheon, Gyeonggi-do, 14662, Republic of Korea
- BK21 four Department of Biotechnology, The Catholic University of Korea, 43 Jibong-ro, Bucheon, Gyeonggi-do, 14662, South Korea
- SML Biopharm, Gwangmyeong Station M Cluster, 17 Deokan-ro 104beon-gil, Gwangmyeong-si, Gyeonggi-do, 14353, Republic of Korea
| | - Gyochang Keum
- Brain Technology Research Center, Brain Science Research Division, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Eun-Kyoung Bang
- Brain Technology Research Center, Brain Science Research Division, Korea Institute of Science and Technology, 5 Hwarang-ro 14-gil, Seongbuk-gu, Seoul, 02792, Republic of Korea
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, 7-13 Kyungheedae-ro 6-gil, Dongdaemun-gu, Seoul, 02447, Republic of Korea
- Department of Chemical and Biological Engineering, Korea University, 145 Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| |
Collapse
|
9
|
Torisu T, Maeda A, Ito S, Uchiyama S. Protein aggregation in the frozen state induced by dropping stress. Eur J Pharm Sci 2025; 205:106996. [PMID: 39722424 DOI: 10.1016/j.ejps.2024.106996] [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: 09/26/2024] [Revised: 12/12/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
The formation of protein aggregates, which can be immunogenic and lower the efficacy and safety of protein drugs, has been an issue in biopharmaceutical development for more than a decade. Although protein drugs are often shipped as frozen material, the effect of the accidental dropping of frozen proteins, which can occur during shipping and handling, on the physical stability has not been studied. Here, a frozen Fc fusion protein was subjected to dropping stress and the increase in the aggregate concentration was evaluated. Significant increases in micron-sized aggregates were observed at -30 °C (p ≤ 0.01), but not at -60 °C. Proteins adsorbed on the vial surfaces were not remarkably detached by the action of dropping and were not the primary cause of the increase in micron-sized aggregates. When the vials were dropped, local heat generation occurred and this led to local freeze-thaw stress that induced protein aggregation. Poloxamer-188, which is known to mitigate aggregation caused by freeze-thaw stress, effectively prevented the aggregation caused by the dropping stress in the frozen state at -30 °C. In addition, rapid freezing could suppress the aggregation caused by the dropping stress. The results demonstrated that dropping stress reduced the stability of proteins even in the frozen state, and they provide new insights into the formulation and freezing processes to prevent protein aggregation caused by dropping stress in the frozen state.
Collapse
Affiliation(s)
- Tetsuo Torisu
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan.
| | - Ayuko Maeda
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shuhei Ito
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Susumu Uchiyama
- Department of Biotechnology, Graduate School of Engineering, Osaka University, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Exploratory Research Center on Life and Living Systems, National Institutes of Natural Sciences, 5-1 Higashiyama, Myodaiji, Okazaki, Aichi 444-8787, Japan.
| |
Collapse
|
10
|
Talbot A, de Koning-Ward TF, Layton D. Left out in the cold - inequity in infectious disease control due to cold chain disparity. Vaccine 2025; 45:126648. [PMID: 39708516 DOI: 10.1016/j.vaccine.2024.126648] [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: 04/29/2024] [Revised: 11/28/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
Vaccines and diagnostic tools stand out as among the most influential advancements in public health, credited with averting an estimated 6 million deaths annually and substantially alleviating the burden of infectious disease. Despite this progress, the global imperative to prevent, detect, and combat infectious disease persists. Regrettably, hundreds of thousands of lives are still lost due to inadequate access to vaccines and diagnostics. A critical obstacle in accessibility lies in the requirement of reliable cold chain for their transportation and storage, a resource that remains inadequate in many regions, particularly in the developing world. Various factors, including socio-economic disparities, biological complexities, and manufacturing processes, exert significant influence on the availability and integrity of vaccines and diagnostic materials. This review aims to explore the multifaceted issue of inequality in access to disease control tools, examining the vulnerabilities of vaccines and diagnostics while also investigating recent advancements that offer promising solutions to improve thermal stability.
Collapse
Affiliation(s)
- Aimee Talbot
- CSIRO, Health and Biosecurity, Australian Centre for Disease Preparedness, Geelong, Victoria, Australia; School of Medicine, Deakin University, Geelong, Victoria, Australia; Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Victoria, Australia
| | - Tania F de Koning-Ward
- School of Medicine, Deakin University, Geelong, Victoria, Australia; Institute for Mental and Physical Health and Clinical Translation, Deakin University, Geelong, Victoria, Australia
| | - Daniel Layton
- CSIRO, Health and Biosecurity, Australian Centre for Disease Preparedness, Geelong, Victoria, Australia.
| |
Collapse
|
11
|
Dattani S, Li X, Lampa C, Barriscale A, Damadzadeh B, Lechuga-Ballesteros D, Jasti BR. Development of Spray-Dried Micelles, Liposomes, and Solid Lipid Nanoparticles for Enhanced Stability. Pharmaceutics 2025; 17:122. [PMID: 39861769 PMCID: PMC11768165 DOI: 10.3390/pharmaceutics17010122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Objectives: Micelles, liposomes, and solid lipid nanoparticles (SLNs) are promising drug delivery vehicles; however, poor aqueous stability requires post-processing drying methods for maintaining long-term stability. The objective of this study was to compare the potential of lipid-based micelles, liposomes, and SLNs for producing stable re-dispersible spray-dried powders with trehalose or a combination of trehalose and L-leucine. This study provides novel insights into the implementation of spray drying as a technique to enhance long-term stability for these lipid-based nanocarriers. Methods: Aqueous dispersions of LDV-targeted micelles, liposomes, and SLNs loaded with paclitaxel (PTX) were converted into re-dispersible powders using spray drying. The physicochemical properties of the nanocarriers were determined via scanning electron microscopy (SEM), Karl Fischer titration, differential scanning calorimetry (DSC), and dynamic light scattering (DLS). Short-term stability of all nanocarrier formulations was compared by measuring particle size, polydispersity index (PDI), and paclitaxel retention over 7 days at room temperature and at 4 °C. Results: Paclitaxel-loaded micelles, liposomes, and SLN formulations were successfully converted into well-dispersed spray-dried powders with acceptable yields (71.5 to 83.5%), low moisture content (<2%), and high transition temperatures (95.1 to 100.8 °C). SEM images revealed differences in morphology, where nanocarriers spray-dried with trehalose or a combination of trehalose and L-leucine produced smooth or corrugated particle surfaces, respectively. Reconstituted spray-dried nanocarriers maintained their nanosize and paclitaxel content over 7 days at 4 °C. Conclusions: The results of this study demonstrate the potential for the development of spray-dried lipid-based nanocarriers for long-term stability.
Collapse
Affiliation(s)
- Shradha Dattani
- Department of Pharmaceutics and Medicinal Chemistry, University of the Pacific, Stockton, CA 95211, USA
| | - Xiaoling Li
- Department of Pharmaceutics and Medicinal Chemistry, University of the Pacific, Stockton, CA 95211, USA
| | - Charina Lampa
- Inhalation Product Development, PT&D AstraZeneca, LLC, South San Francisco, CA 94080, USA
| | - Amanda Barriscale
- Inhalation Product Development, PT&D AstraZeneca, LLC, South San Francisco, CA 94080, USA
| | - Behzad Damadzadeh
- Inhalation Product Development, PT&D AstraZeneca, LLC, South San Francisco, CA 94080, USA
| | | | - Bhaskara R Jasti
- Department of Pharmaceutics and Medicinal Chemistry, University of the Pacific, Stockton, CA 95211, USA
| |
Collapse
|
12
|
Bustos Araya ME, Ricart AN, Calpena Campmany AC, Prohens R, Miñarro Carmona M. Development and Characterization of Lyophilized Chondroitin Sulfate-Loaded Solid Lipid Nanoparticles: Encapsulation Efficiency and Stability. Pharmaceutics 2025; 17:86. [PMID: 39861734 PMCID: PMC11768393 DOI: 10.3390/pharmaceutics17010086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
This study explores the development and characterization of lyophilized chondroitin sulfate (CHON)-loaded solid lipid nanoparticles (SLN) as an innovative platform for advanced drug delivery. Background/Objectives: Solid lipid nanoparticles are increasingly recognized for their biocompatibility, their ability to encapsulate diverse compounds, their capacity to enhance drug stability, their bioavailability, and their therapeutic efficacy. Methods: CHON, a naturally occurring glycosaminoglycan with anti-inflammatory and regenerative properties, was integrated into SLN formulations using the hot microemulsion technique. Two formulations (SLN-1 and SLN-2) were produced and optimized by evaluating critical physicochemical properties such as particle size, zeta potential, encapsulation efficiency (EE%), and stability. The lyophilization process, with the addition of various cryoprotectants, revealed trehalose to be the most effective agent in maintaining nanoparticle integrity and functional properties. Results: Morphological analyses using transmission electron microscopy (TEM) and atomic force microscopy (AFM) confirmed the dimensions of the nanoscales and their structural uniformity. Differential scanning calorimetry (DSC) and X-ray diffraction (XRD) revealed minimal excipient interaction with CHON, ensuring formulation stability. Stability studies under different environmental conditions highlighted that SLN-2 is the most stable formulation, maintaining superior encapsulation efficiency (≥88%) and particle size consistency over time. Conclusions: These findings underscore the potential of CHON-loaded SLNs as promising candidates for targeted, sustained-release therapies in the treatment of inflammatory and degenerative diseases.
Collapse
Affiliation(s)
- Marta E. Bustos Araya
- Instituto de Investigaciones Farmacéuticas, Facultad de Farmacia, Universidad de Costa Rica, San José 11501, Costa Rica;
| | - Anna Nardi Ricart
- Pharmacy, Pharmaceutical Technology and Physico-Chemical Department, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.C.C.C.); (M.M.C.)
| | - Ana C. Calpena Campmany
- Pharmacy, Pharmaceutical Technology and Physico-Chemical Department, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.C.C.C.); (M.M.C.)
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, 08028 Barcelona, Spain
| | - Rafel Prohens
- Laboratory of Organic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain;
| | - Montserrat Miñarro Carmona
- Pharmacy, Pharmaceutical Technology and Physico-Chemical Department, University of Barcelona, Av. Joan XXIII, 27-31, 08028 Barcelona, Spain; (A.C.C.C.); (M.M.C.)
- IDIBELL-UB Research Group, Pharmacotherapy, Pharmacogenomics and Pharmaceutical Technology, Avinguda Granvia, 199-203, L’Hospitalet de Llobregat, 08908 Barcelona, Spain
| |
Collapse
|
13
|
Gulati GK, Simpson AC, MacMillen Z, Krieger K, Sharma S, Erasmus JH, Reed SG, Davie JW, Avril M, Khandhar AP. Preclinical development of lyophilized self-replicating RNA vaccines for COVID-19 and malaria with improved long-term thermostability. J Control Release 2025; 377:81-92. [PMID: 39547422 DOI: 10.1016/j.jconrel.2024.11.023] [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: 06/18/2024] [Revised: 10/02/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Messenger RNA (mRNA) vaccines against COVID-19 have demonstrated high efficacy and rapid deployment capability to target emerging infectious diseases. However, the need for ultra-low temperature storage made the distribution of LNP/mRNA vaccines to regions with limited resources impractical. This study explores the use of lyophilization to enhance the stability of self-replicating mRNA (repRNA) vaccines, allowing for their storage at non-freezing temperatures such as 2-8 °C or room temperature (25 °C). We lyophilized repRNA molecules complexed to a novel cationic emulsion delivery system, LION™, with different sugar-based lyoprotectants to identify candidates that provided the best vaccine integrity and effectiveness after being thermally stressed. For screening, we used repRNA encoding the reporter protein secreted embryonic alkaline phosphatase (SEAP) and for proof-of-concept, we used repRNA vaccines encoding SARS-CoV-2 full-length spike (WA-1 isolate) or full-length surface protein circumsporozoite (CS) of Plasmodium yoelii (Py). We found that lyophilization of LION/repRNA with sucrose provided the best colloidal stability, preserved in vitro expression, and induced equivalent antigen-specific antibody responses in mice compared to freshly prepared liquid LION/repRNA. Furthermore, lyophilized vaccines were stable for at least one week at 25 °C and at least one year at 2-8 °C. The cumulative analysis of stability-determining physicochemical data, in vitro potency, and in vivo immunogenicity in mice enabled the selection of a lead lyophilized composition containing 10 % w/v sucrose as the lyoprotectant. The data presented here provide a foundation for the clinical evaluation of next-generation thermostable repRNA vaccines that will enable more equitable vaccine access globally.
Collapse
Affiliation(s)
- Gaurav K Gulati
- HDT Bio, 1150 Eastlake Ave E Suite 200A, Seattle, WA 98109, USA.
| | | | - Zachary MacMillen
- MalarVx, Inc, 1551 Eastlake Ave E, Suite 100, Seattle, WA 98102, USA
| | - Kyle Krieger
- HDT Bio, 1150 Eastlake Ave E Suite 200A, Seattle, WA 98109, USA
| | - Shibbu Sharma
- HDT Bio, 1150 Eastlake Ave E Suite 200A, Seattle, WA 98109, USA
| | - Jesse H Erasmus
- HDT Bio, 1150 Eastlake Ave E Suite 200A, Seattle, WA 98109, USA
| | - Steven G Reed
- HDT Bio, 1150 Eastlake Ave E Suite 200A, Seattle, WA 98109, USA
| | - James W Davie
- MalarVx, Inc, 1551 Eastlake Ave E, Suite 100, Seattle, WA 98102, USA
| | - Marion Avril
- MalarVx, Inc, 1551 Eastlake Ave E, Suite 100, Seattle, WA 98102, USA
| | - Amit P Khandhar
- HDT Bio, 1150 Eastlake Ave E Suite 200A, Seattle, WA 98109, USA.
| |
Collapse
|
14
|
Ruchi R, Raman GM, Kumar V, Bahal R. Evolution of antisense oligonucleotides: navigating nucleic acid chemistry and delivery challenges. Expert Opin Drug Discov 2025; 20:63-80. [PMID: 39653607 PMCID: PMC11823135 DOI: 10.1080/17460441.2024.2440095] [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: 09/13/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024]
Abstract
INTRODUCTION Antisense oligonucleotide (ASO) was established as a viable therapeutic option for genetic disorders. ASOs can target RNAs implicated in various diseases, including upregulated mRNA and pre-mRNA undergoing abnormal alternative splicing events. Therapeutic applications of ASOs have been proven with the Food and Drug Administration approval of several drugs in recent years. Earlier enzymatic stability and delivery remains a big challenge for ASOs. Introducing new chemical modifications and new formulations resolving the issues related to the nuclease stability and delivery of the ASOs. Excitingly, ASOs-based bioconjugates that target the hepatocyte have gained much attraction. Efforts are ongoing to increase the therapeutic application of the ASOs to the extrahepatic tissue as well. AREA COVERED We have briefly discussed the mechanism of ASOs, the development of new chemistries, and delivery strategies for ASO-based drug discovery and development. The discussion focuses more on the already approved ASOs and those in the clinical development stage. EXPERT OPINION To expand the clinical application of ASOs, continuous effort is required to develop precise delivery strategies for targeting extrahepatic tissue to minimize the off-target effects.
Collapse
Affiliation(s)
- Ruchi Ruchi
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Govind Mukesh Raman
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
- Farmington High School, Farmington, CT, USA
| | - Vikas Kumar
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Raman Bahal
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| |
Collapse
|
15
|
Kafle U, Truong HQ, Nguyen CTG, Meng F. Development of Thermally Stable mRNA-LNP Delivery Systems: Current Progress and Future Prospects. Mol Pharm 2024; 21:5944-5959. [PMID: 39529245 DOI: 10.1021/acs.molpharmaceut.4c00826] [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] [Indexed: 11/16/2024]
Abstract
The success of mRNA-LNP-based COVID-19 vaccines opens a new era for mRNA-LNP-based therapy. This breakthrough is expected to catalyze the development of more mRNA-LNP-based medicines, not only for preventive vaccines but also for therapeutic purposes. Despite the promising outlook, there are fundamental challenges impeding the progress and widespread application of mRNA-LNP formulations. One of the significant challenges is their thermal instability, requiring these products to be stored at ultralow temperatures for long-term stability. The specific requirements present significant challenges for the storage, transportation, and distribution of mRNA-LNP formulations. To effectively prepare for future infectious disease outbreaks and broaden the application of mRNA-LNP-based therapies for other illnesses, improving the thermostability of mRNA-LNP formulations is critical. In this review, we discuss the potential factors contributing to the thermal instability of mRNA-LNP formulations and examine the roles of key components such as ionizable lipids, cholesterol, pH, buffers, and stabilizing agents like sugars in maintaining their thermal stability, with the goal of providing insights that can guide the future development of thermally stable mRNA-LNP formulations.
Collapse
Affiliation(s)
- Urmila Kafle
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, Massachusetts 01854, United States
| | - Hoang Quan Truong
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, Massachusetts 01854, United States
| | - Cao Thuy Giang Nguyen
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, Massachusetts 01854, United States
| | - Fanfei Meng
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, 3 Solomont Way, Lowell, Massachusetts 01854, United States
| |
Collapse
|
16
|
Haque MA, Shrestha A, Mikelis CM, Mattheolabakis G. Comprehensive analysis of lipid nanoparticle formulation and preparation for RNA delivery. Int J Pharm X 2024; 8:100283. [PMID: 39309631 PMCID: PMC11415597 DOI: 10.1016/j.ijpx.2024.100283] [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: 05/02/2024] [Revised: 08/21/2024] [Accepted: 09/07/2024] [Indexed: 09/25/2024] Open
Abstract
Nucleic acid-based therapeutics are a common approach that is increasingly popular for a wide spectrum of diseases. Lipid nanoparticles (LNPs) are promising delivery carriers that provide RNA stability, with strong transfection efficiency, favorable and tailorable pharmacokinetics, limited toxicity, and established translatability. In this review article, we describe the lipid-based delivery systems, focusing on lipid nanoparticles, the need of their use, provide a comprehensive analysis of each component, and highlight the advantages and disadvantages of the existing manufacturing processes. We further summarize the ongoing and completed clinical trials utilizing LNPs, indicating important aspects/questions worth of investigation, and analyze the future perspectives of this significant and promising therapeutic approach.
Collapse
Affiliation(s)
- Md. Anamul Haque
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Archana Shrestha
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| | - Constantinos M. Mikelis
- Laboratory of Molecular Pharmacology, Department of Pharmacy, University of Patras, Patras 26504, Greece
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - George Mattheolabakis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71201, USA
| |
Collapse
|
17
|
Athaydes Seabra Ferreira H, Ricardo Aluotto Scalzo Júnior S, Kelton Santos de Faria K, Henrique Costa Silva G, Túllio Rodrigues Alves M, Oliveira Lobo A, Pires Goulart Guimarães P. Cryoprotectant optimization for enhanced stability and transfection efficiency of pDNA-loaded ionizable lipid nanoparticles. Int J Pharm 2024; 665:124696. [PMID: 39265853 DOI: 10.1016/j.ijpharm.2024.124696] [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: 05/10/2024] [Revised: 08/01/2024] [Accepted: 09/09/2024] [Indexed: 09/14/2024]
Abstract
Advances in gene therapy, exemplified by mRNA vaccines against COVID-19, highlight the importance of lipid nanoparticles (LNPs) for nucleic acid delivery despite challenging storage conditions. Substituting mRNA with pDNA in LNPs may enhance stability and efficacy, yet maintaining LNP stability poses challenges, particularly during freeze-drying. Cryoprotectants offer potential to mitigate destabilization, improving LNP properties and in vivo performance. Here, we evaluated the effects of different concentrations of various cryoprotectants on the freeze-drying process of pDNA-loaded LNPs, assessing their physicochemical characteristics and transfection efficiency. Stability was examined under various storage conditions, confirming biological efficacy post-storage. Our results highlight the role of cryoprotectants in optimizing freeze-drying for the extended shelf life of nucleic acid-loaded LNPs. Trehalose emerged as an efficient cryoprotectant, maintaining LNP stability after the freeze-drying process for up to 2 years, with diameters and transfection efficiency comparable to fresh formulations. These findings demonstrate the optimized concentration of cryoprotectants to sustain LNP stability despite freeze-drying and prolonged storage, providing valuable insights for nucleic acid-based therapies.
Collapse
Affiliation(s)
- Heloísa Athaydes Seabra Ferreira
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Sérgio Ricardo Aluotto Scalzo Júnior
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Kevin Kelton Santos de Faria
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Gabriel Henrique Costa Silva
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Marco Túllio Rodrigues Alves
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil
| | - Anderson Oliveira Lobo
- Department of Materials Engineering, Federal University of Piauí, Teresina, 64049-550, Piauí, Brazil
| | - Pedro Pires Goulart Guimarães
- Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, 31270-901 Minas Gerais, Brazil.
| |
Collapse
|
18
|
Gordon A, Li B, Witten J, Nguyen H, Anderson DG. Inhalable Dry Powders for Lung mRNA Delivery. Adv Healthc Mater 2024; 13:e2400509. [PMID: 39352052 PMCID: PMC11582499 DOI: 10.1002/adhm.202400509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/05/2024] [Indexed: 10/03/2024]
Abstract
Despite great promise, application of mRNA therapeutics in the lung has proven challenging. Many groups have reported success instilling liquid mRNA formulations in animal models, but direct intratracheal administration of large liquid quantities to the human lung presents significant safety and distribution concerns. To accomplish safe and effective mRNA delivery to the lung, formulations must be prepared for dosing via inhalation. An inhaled mRNA delivery system for the lung must be both robust enough to survive inhalation conditions and potent enough to deliver mRNA upon reaching the lung. In this work dry powder lipid nanoparticle formulations are developed, using spray-freeze-drying, to produce stable, biologically active, inhalable dry powders for mRNA delivery. The final powders have suitable aerosolization properties, with mean mass aerodynamic diameter (MMAD) of 3-4 microns, and fine particle fraction (FPF) ≈40%, allowing for efficient mRNA delivery to the deep lung following inhalation. Importantly, the formulations developed here are suitable for use with different ionizable lipids. Four different ionizable lipid-based formulations are evaluated as powders, and all exhibit in vivo pulmonary mRNA delivery equal to that of instilled liquid formulations. These results lay promising groundwork for the eventual development of an inhalable mRNA dry powder therapeutic.
Collapse
Affiliation(s)
- Akiva Gordon
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Bowen Li
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Jacob Witten
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Hong Nguyen
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Daniel G Anderson
- David H Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Statistics, University of Michigan, Ann Arbor, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| |
Collapse
|
19
|
Isaac I, Shaikh A, Bhatia M, Liu Q, Park S, Bhattacharya C. Tetrahydropyrimidine Ionizable Lipids for Efficient mRNA Delivery. ACS NANO 2024; 18:29045-29058. [PMID: 39393001 PMCID: PMC11781979 DOI: 10.1021/acsnano.4c10154] [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] [Indexed: 10/13/2024]
Abstract
Lipid nanoparticles (LNPs) have emerged as an effective and promising technology for messenger RNA (mRNA) delivery, offering a potential solution to physiological barriers and providing an alternative approach to gene therapy without the drawbacks associated with viral delivery. However, efficiently delivering mRNA remains a significant challenge in nucleic acid-based therapies due to the limitations of current LNP platforms in achieving optimal endosomal escape and mRNA release, which largely relies on finding a suitable ionizable lipid. Additionally, the synthesis of these ionizable lipids involves multiple chemical reactions, often making the process time-consuming and difficult to translate. In this study, we employed a facile, catalyst-free, and versatile one-pot multicomponent reaction (MCR) to develop a library of ionizable lipids featuring a pharmacologically significant tetrahydropyrimidine (THP) backbone, tailored for enhanced mRNA delivery. A library of 26 THP ionizable lipids was systematically synthesized in just 3 h and formulated with luciferase mRNA for initial in vitro screening. The THP LNPs exhibited tunable particle sizes, favorable ζ-potentials, and high encapsulation efficiencies. Among them, THP1 demonstrated the highest transfection efficiency both in vitro and in vivo after intramuscular administration, comparable to DLin-MC3-DMA (MC3), a conventional benchmark. Further optimization of THP1 with phospholipids significantly enhanced intramuscular mRNA delivery and showed sustained protein expression in vivo for up to 5 days. More importantly, it demonstrated successful intravenous delivery in a dose-dependent manner with minimal toxicity, as indicated by hematological, histopathological, and proinflammatory cytokine assessments. Furthermore, THP1 LNPs also demonstrated the ability to edit genes in specific liver tissues in a tdTomato transgenic mouse model, highlighting their precision and utility in targeted therapeutic applications. These findings position THP1 LNPs as promising candidates for advancing mRNA-based therapies, with significant implications for clinical translation in vaccine delivery and CRISPR/Cas9-mediated gene editing in the liver.
Collapse
Affiliation(s)
- Ivan Isaac
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, Nevada 89154, United States
| | - Altab Shaikh
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, Nevada 89154, United States
| | - Mayurakkhi Bhatia
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, Nevada 89154, United States
| | - Qian Liu
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, Las Vegas, Nevada 89154, United States
- School of Life Sciences, College of Sciences, University of Nevada, Las Vegas, Las Vegas, Nevada 89154, United States
| | - Seungman Park
- Department of Mechanical Engineering, University of Nevada, Las Vegas, Las Vegas, Nevada 89154, United States
- Interdisciplinary Biomedical Engineering Program, University of Nevada, Las Vegas, Las Vegas, Nevada 89154, United States
| | - Chandrabali Bhattacharya
- Department of Chemistry and Biochemistry, University of Nevada, Las Vegas, Las Vegas, Nevada 89154, United States
- Interdisciplinary Biomedical Engineering Program, University of Nevada, Las Vegas, Las Vegas, Nevada 89154, United States
| |
Collapse
|
20
|
Hara S, Arase S, Sano S, Suzuki T, Mizogaki I, Sato S, Ukai K. Anion exchange-HPLC method for evaluating the encapsulation efficiency of mRNA-loaded lipid nanoparticles using analytical quality by design. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1247:124317. [PMID: 39303519 DOI: 10.1016/j.jchromb.2024.124317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/28/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
Lipid nanoparticles (LNPs) are emerging nucleic acid delivery systems in the development of mRNA therapeutics such as the severe acute respiratory syndrome coronavirus 2 vaccines. However, a suitable analytical method for evaluating the encapsulation efficiency (EE) of the LNPs is required to ensure drug efficacy, as current analytical methods exhibit throughput issues and require long analysis times. Hence, we developed and validated an anion-exchange HPLC method using Analytical Quality by Design. Three critical method parameters (CMPs) were identified using risk assessment and Design of Experiments: column temperature, flow rate, and sodium perchlorate concentration. The CMPs were optimized using Face-Centered Central Composite Design. The discriminating power of the optimized HPLC method and RiboGreen assay was comparable. The main advantage of this method is that LNPs can be directly injected into the HPLC system without bursting the LNPs loaded with encapsulated poly(A). The optimized HPLC method was validated as robust, high-throughput, and sufficiently sensitive according to the ICH Q2 guidelines. We believe our findings could promote efficient LNPs-based drug development.
Collapse
Affiliation(s)
- Shoki Hara
- Formulation Research, Pharmaceutical Science and Technologies Unit, Pharmaceutical Profiling & Development Function, Eisai Co., Ltd., 1 Kawashimatakehaya-machi, Kakamigahara, Gifu 501-6195, Japan.
| | - Shuntaro Arase
- Formulation Research, Pharmaceutical Science and Technologies Unit, Pharmaceutical Profiling & Development Function, Eisai Co., Ltd., 1 Kawashimatakehaya-machi, Kakamigahara, Gifu 501-6195, Japan
| | - Syusuke Sano
- Formulation Research, Pharmaceutical Science and Technologies Unit, Pharmaceutical Profiling & Development Function, Eisai Co., Ltd., 1 Kawashimatakehaya-machi, Kakamigahara, Gifu 501-6195, Japan
| | - Takuya Suzuki
- Formulation Research, Pharmaceutical Science and Technologies Unit, Pharmaceutical Profiling & Development Function, Eisai Co., Ltd., 1 Kawashimatakehaya-machi, Kakamigahara, Gifu 501-6195, Japan
| | - Iori Mizogaki
- Formulation Research, Pharmaceutical Science and Technologies Unit, Pharmaceutical Profiling & Development Function, Eisai Co., Ltd., 1 Kawashimatakehaya-machi, Kakamigahara, Gifu 501-6195, Japan
| | - Shinya Sato
- Formulation Research, Pharmaceutical Science and Technologies Unit, Pharmaceutical Profiling & Development Function, Eisai Co., Ltd., 1 Kawashimatakehaya-machi, Kakamigahara, Gifu 501-6195, Japan
| | - Koji Ukai
- Formulation Research, Pharmaceutical Science and Technologies Unit, Pharmaceutical Profiling & Development Function, Eisai Co., Ltd., 1 Kawashimatakehaya-machi, Kakamigahara, Gifu 501-6195, Japan
| |
Collapse
|
21
|
Kiaie SH, Zangi AR, Sheibani M, Hemmati S, Baradaran B, Valizadeh H. Novel synthesized ionizable lipid for LNP-mediated P2X7siRNA to inhibit migration and induce apoptosis of breast cancer cells. Purinergic Signal 2024; 20:533-546. [PMID: 38436880 PMCID: PMC11377399 DOI: 10.1007/s11302-024-09989-8] [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: 07/10/2023] [Accepted: 01/23/2024] [Indexed: 03/05/2024] Open
Abstract
The development of ionizable lipid (IL) was necessary to enable the effective formulation of small interfering RNA (siRNA) to inhibit P2X7 receptors (P2X7R), a key player in tumor proliferation, apoptosis, and metastasis. In this way, the synthesis and utility of IL for enhancing cellular uptake of lipid nanoparticles (LNP) improve the proper delivery of siRNA-LNPs for knockdown overexpression of P2X7R. Therefore, to evaluate the impact of P2X7 knockdown on breast cancer (BC) migration and apoptosis, a branched and synthesized ionizable lipid (SIL) was performed for efficient transfection of LNP with siRNA for targeting P2X7 receptors (siP2X7) in mouse 4T-1 cells. Following synthesis and structural analysis of SIL, excellent characterization of the LNP was achieved (Z-average 126.8 nm, zeta-potential - 12.33, PDI 0.16, and encapsulation efficiency 85.35%). Afterward, the stability of the LNP was evaluated through an analysis of the leftover composition, and toxic concentration values for SIL and siP2X7 were determined. Furthermore, siP2X7-LNP cellular uptake in the formulation was assessed via confocal microscopy. Following determining the optimal dose (45 pmol), wound healing analysis was assessed using scratch assay microscopy, and apoptosis was evaluated using flow cytometry. The use of the innovative branched SIL in the formulation of siP2X7-LNP resulted in significant inhibition of migration and induction of apoptosis in 4T-1 cells due to improved cellular uptake. Subsequently, the innovative SIL represents a critical role in efficiently delivering siRNA against murine triple-negative breast cancer cells (TNBC) using LNP formulation, resulting in significant efficacy.
Collapse
Affiliation(s)
- Seyed Hossein Kiaie
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Rajabi Zangi
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Razi Drug Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Salar Hemmati
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center and School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hadi Valizadeh
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
22
|
Chaudhary AA, Fareed M, Khan SUD, Alneghery LM, Aslam M, Alex A, Rizwanullah M. Exploring the therapeutic potential of lipid-based nanoparticles in the management of oral squamous cell carcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:1223-1246. [PMID: 39465011 PMCID: PMC11502080 DOI: 10.37349/etat.2024.00272] [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: 06/08/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a highly malignant and invasive tumor with significant mortality and morbidity. Current treatment modalities such as surgery, radiotherapy, and chemotherapy encounter significant limitations, such as poor targeting, systemic toxicity, and drug resistance. There is an urgent need for novel therapeutic strategies that offer targeted delivery, enhanced efficacy, and reduced side effects. The advent of lipid-based nanoparticles (LNPs) offers a promising tool for OSCC therapy, potentially overcoming the limitations of current therapeutic approaches. LNPs are composed of biodegradable and biocompatible lipids, which minimize the risk of toxicity and adverse effects. LNPs can encapsulate hydrophobic drugs, improving their solubility and stability in the biological environment, thereby enhancing their bioavailability. LNPs demonstrate significantly higher ability to encapsulate lipophilic drugs than other nanoparticle types. LNPs offer excellent storage stability, minimal drug leakage, and controlled drug release, making them highly effective nanoplatforms for the delivery of chemotherapeutic agents. Additionally, LNPs can be modified by complexing them with specific target ligands on their surface. This surface modification allows the active targeting of LNPs to the tumors in addition to the passive targeting mechanism. Furthermore, the PEGylation of LNPs improves their hydrophilicity and enhances their biological half-life by reducing clearance by the reticuloendothelial system. This review aims to discuss current treatment approaches and their limitations, as well as recent advancements in LNPs for better management of OSCC.
Collapse
Affiliation(s)
- Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammad Fareed
- College of Medicine, AlMaarefa University, Diriyah, Riyadh 11597, Saudi Arabia
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Lina M Alneghery
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mohammed Aslam
- Pharmacy Department, Tishk International University, Erbil 44001, Kurdistan Region, Iraq
| | - Arockia Alex
- Molecular and Nanobiotechnology Laboratory (MNBL), Department of Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| | - Md Rizwanullah
- Drug Delivery and Nanomedicine Unit, Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Chennai 602105, Tamil Nadu, India
| |
Collapse
|
23
|
Zhang Z, Cheng D, Luo W, Hu D, Yang T, Hu K, Liang L, Liu W, Hu J. Molecular Dynamics Simulation of Lipid Nanoparticles Encapsulating mRNA. Molecules 2024; 29:4409. [PMID: 39339404 PMCID: PMC11433737 DOI: 10.3390/molecules29184409] [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: 08/25/2024] [Revised: 09/14/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
mRNA vaccines have shown great potential in responding to emerging infectious diseases, with their efficacy and stability largely dependent on the delivery vehicles-lipid nanoparticles (LNPs). This study aims to explore the mechanisms by which LNPs encapsulate mRNA, as well as the effects of different N/P ratios and acid types in nucleic acid solutions on the structure and properties of LNPs, using the ethanol solvent injection method as the encapsulation technique. Six systems were designed, based on the composition and proportions of the existing mRNA vaccine mRNA-1273, and molecular dynamics (MD) simulations were employed to investigate the self-assembly process of LNPs. Ethanol was used as a solvent instead of pure water to better mimic experimental conditions. The results indicate that lipid components self-assemble into nanoparticles under neutral conditions, with the ionizable lipid SM-102 predominantly concentrating in the core of the particles. Upon mixing with nucleic acids in acidic conditions, LNPs undergo disassembly, during which protonated SM-102 encapsulates mRNA through electrostatic interactions, forming stable hydrogen bonds. Cluster structure analysis revealed that the four lipid components of LNPs are distributed sequentially from the outside inwards as DMG-PEG 2000, DSPC, cholesterol, and protonated SM-102. Moreover, LNPs constructed under low pH or low N/P ratios using citric acid exhibited larger volumes and more uniform distribution. These findings provide a scientific basis for further designing and optimizing LNP components to enhance the efficacy of mRNA vaccine encapsulation.
Collapse
Affiliation(s)
- Zhigang Zhang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Dazhi Cheng
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Wenqin Luo
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Donling Hu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Tiantian Yang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Kaixuan Hu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Li Liang
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Wei Liu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Jianping Hu
- Key Laboratory of Medicinal and Edible Plants Resources Development of Sichuan Education Department, School of Pharmacy, Chengdu University, Chengdu 610106, China
- School of Life Science, Leshan Normal University, Leshan 614004, China
| |
Collapse
|
24
|
Beckert N, Dietrich A, Hubbuch J. RP-CAD for Lipid Quantification: Systematic Method Development and Intensified LNP Process Characterization. Pharmaceuticals (Basel) 2024; 17:1217. [PMID: 39338379 PMCID: PMC11435201 DOI: 10.3390/ph17091217] [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: 07/08/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Lipid nanoparticles (LNPs) and their versatile nucleic acid payloads bear great potential as delivery systems. Despite their complex lipid composition, their quality is primarily judged by particle characteristics and nucleic acid encapsulation. In this study, we present a holistic reversed-phase (RP)-charged aerosol detection (CAD)-based method developed for commonly used LNP formulations, allowing for intensified LNP and process characterization. We used an experimental approach for power function value (PFV) optimization termed exploratory calibration, providing a single PFV (1.3) in an appropriate linearity range for all six lipids. Followed by the procedure of method calibration and validation, linearity (10-400 ng, R2 > 0.996), precision, accuracy, and robustness were effectively proven. To complement the commonly determined LNP attributes and to evaluate the process performance across LNP processing, the developed RP-CAD method was applied in a process parameter study varying the total flow rate (TFR) during microfluidic mixing. The RP-CAD method revealed a constant lipid molar ratio across processing but identified deviations in the theoretical lipid content and general lipid loss, which were both, however, entirely TFR-independent. The deviations in lipid content could be successfully traced back to the lipid stock solution preparation. In contrast, the observed lipid loss was attributable to the small-scale dialysis following microfluidic mixing. Overall, this study establishes a foundation for employing RP-CAD for lipid quantification throughout LNP processing, and it highlights the potential to extend its applicability to other LNPs, process parameter studies, or processes such as cross-flow filtration.
Collapse
Affiliation(s)
| | | | - Jürgen Hubbuch
- Institute of Process Engineering in Life Sciences—Section IV: Biomolecular Separation Engineering, Karlsruhe Institute of Technology (KIT), 76131 Karlsruhe, Germany; (N.B.); (A.D.)
| |
Collapse
|
25
|
Feng S, Chen T, Zhang Y, Lu C. mRNA Fragmentation Pattern Detected by SHAPE. Curr Issues Mol Biol 2024; 46:10249-10258. [PMID: 39329962 PMCID: PMC11431040 DOI: 10.3390/cimb46090610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/12/2024] [Accepted: 09/14/2024] [Indexed: 09/28/2024] Open
Abstract
The success of messenger RNA (mRNA) vaccines in controlling COVID-19 has warranted further developments in new technology. Currently, their quality control process largely relies on low-resolution electrophoresis for detecting chain breaks. Here, we present an approach using multi-primer reverse transcription sequencing (MPRT-seq) to identify degradation fragments in mRNA products. Using this in-house-made mRNA containing two antigens and untranslated regions (UTRs), we analyzed the mRNA completeness and degradation pattern at a nucleotide resolution. We then analyzed the sensitive base sequence and its correlation with the secondary structure. Our MPRT-seq mapping shows that certain sequences on the 5' of bulge-stem-loop structures can result in preferential chain breaks. Our results agree with commonly used capillary electrophoresis (CE) integrity analysis but at a much higher resolution, and can improve mRNA stability by providing information to remove sensitive structures or sequences in the mRNA sequence design.
Collapse
Affiliation(s)
| | | | | | - Changrui Lu
- College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, China; (S.F.); (T.C.); (Y.Z.)
| |
Collapse
|
26
|
Wang T, Yu T, Li W, Liu Q, Sung TC, Higuchi A. Design and lyophilization of mRNA-encapsulating lipid nanoparticles. Int J Pharm 2024; 662:124514. [PMID: 39067550 DOI: 10.1016/j.ijpharm.2024.124514] [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: 04/27/2024] [Revised: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
The remarkable success of two FDA-approved mRNA-encapsulating vaccines (Comirnaty® and Spikevax®) indicated the importance of lipid nanoparticles (LNPs) delivery systems in clinical use. Currently, mRNA-encapsulating LNPs (mRNA-LNPs) vaccines are stored as frozen liquid at low or ultralow temperatures. We designed lyophilized LNPs utilizing FDA-approved lipids to expedite the clinical application of our developed lyophilized mRNA-LNPs in the future. The key parameters of sucrose concentration and the selection and molar ratio of the four lipids in these vaccines were optimized for long-term stability with high transfection efficiency after lyophilization. We demonstrated that 8.7% sucrose is the optimal cryoprotectant concentration to maintain the transfection efficiency of lyophilized mRNA-LNPs. Optimal lipid formulations with high transfection efficiency both before and after lyophilization were screened using an orthogonal experimental design. The ratios of distearoylphosphatidylcholine (DSPC)/cholesterol and the selection of the ionizable and PEGylated lipids are the main factors influencing the long-term stability of mRNA-LNPs. Comparative mouse transfection experiments showed that the optimal lyophilized mRNA-LNPs maintained high mRNA expression after lyophilization, predominantly in the spleen or liver, with no expression in the kidneys or eyes. Our studies demonstrated the importance of the sucrose concentration and of the selection and molar ratio of the four lipids composing LNPs for maintaining mRNA-LNP stability under lyophilization and for long-term storage under mild conditions.
Collapse
Affiliation(s)
- Ting Wang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang 325027, China
| | - Tao Yu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang 325027, China
| | - Wanqi Li
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang 325027, China
| | - Qian Liu
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang 325027, China
| | - Tzu-Cheng Sung
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang 325027, China
| | - Akon Higuchi
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, No. 270, Xueyuan Road, Wenzhou, Zhejiang 325027, China; Department of Chemical and Materials Engineering, National Central University, No. 300, Jhongda RD., Jhongli, Taoyuan 32001, Taiwan; R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taoyuan 320, Taiwan.
| |
Collapse
|
27
|
Chen S, Deng Z, Ji D. Advances in the development of lipid nanoparticles for ophthalmic therapeutics. Biomed Pharmacother 2024; 178:117108. [PMID: 39067162 DOI: 10.1016/j.biopha.2024.117108] [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: 05/22/2024] [Revised: 06/28/2024] [Accepted: 07/07/2024] [Indexed: 07/30/2024] Open
Abstract
Previously, researchers have employed Lipid nanoparticles (LNPs) to directly encapsulate medicines. In the realm of gene therapy, researchers have begun to employ lipid nanoparticles to encapsulate nucleic acids such as messenger RNA, small interfering RNA, and plasmid DNA, which are known as nucleic acid lipid nanoparticles. Recent breakthroughs in LNP-based medicine have provided significant prospects for the treatment of ocular disorders, such as corneal, choroidal, and retinal diseases. The use of LNP as a delivery mechanism for medicines and therapeutic genes can increase their effectiveness while avoiding undesired immune reactions. However, LNP-based medicines may pose ocular concerns. In this review, we discuss the general framework of LNP. Additionally, we review adjustable approaches and evaluate their possible risks. In addition, we examine newly described ocular illnesses in which LNP was utilized as a delivery mechanism. Finally, we provide perspectives for solving these potential issues.
Collapse
Affiliation(s)
- Shen Chen
- The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhihong Deng
- Department of Ophthalmology, the Third Xiangya Hospital, Central South University, Changsha, China.
| | - Dan Ji
- Department of Ophthalmology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China; Department of Ophthalmology, Xiangya Hospital, Central South University, Hunan Key Laboratory of Ophthalmology, Changsha, China.
| |
Collapse
|
28
|
Eygeris Y, Henderson MI, Curtis AG, Jozic A, Stoddard J, Reynaga R, Chirco KR, Su GLN, Neuringer M, Lauer AK, Ryals RC, Sahay G. Preformed Vesicle Approach to LNP Manufacturing Enhances Retinal mRNA Delivery. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400815. [PMID: 38738752 PMCID: PMC11661498 DOI: 10.1002/smll.202400815] [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: 02/01/2024] [Revised: 04/20/2024] [Indexed: 05/14/2024]
Abstract
Complete encapsulation of nucleic acids by lipid-based nanoparticles (LNPs) is often thought to be one of the main prerequisites for successful nucleic acid delivery, as the lipid environment protects mRNA from degradation by external nucleases and assists in initiating delivery processes. However, delivery of mRNA via a preformed vesicle approach (PFV-LNPs) defies this precondition. Unlike traditional LNPs, PFV-LNPs are formed via a solvent-free mixing process, leading to a superficial mRNA localization. While demonstrating low encapsulation efficiency in the RiboGreen assay, PFV-LNPs improved delivery of mRNA to the retina by up to 50% compared to the LNP analogs across several benchmark formulations, suggesting the utility of this approach regardless of the lipid composition. Successful mRNA and gene editors' delivery is observed in the retinal pigment epithelium and photoreceptors and validated in mice, non-human primates, and human retinal organoids. Deploying PFV-LNPs in gene editing experiments result in a similar extent of gene editing compared to analogous LNP (up to 3% on genomic level) in the Ai9 reporter mouse model; but, remarkably, retinal tolerability is significantly improved for PFV-LNP treatment. The study findings indicate that the LNP formulation process can greatly influence mRNA transfection and gene editing outcomes, improving LNP treatment safety without sacrificing efficacy.
Collapse
Affiliation(s)
- Yulia Eygeris
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201 USA
| | - Michael I. Henderson
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201 USA
| | - Allison G. Curtis
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
| | - Antony Jozic
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201 USA
| | - Jonathan Stoddard
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Rene Reynaga
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Kathleen R. Chirco
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
| | - Grace Li-Na Su
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
| | - Martha Neuringer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Andreas K. Lauer
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Renee C. Ryals
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Portland, Oregon 97201 USA
- Department of Ophthalmology, Casey Eye Institute, Oregon Health & Science University, Portland, Oregon 97201 USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon 97201
| |
Collapse
|
29
|
Gao Y, Yang L, Li Z, Peng X, Li H. mRNA vaccines in tumor targeted therapy: mechanism, clinical application, and development trends. Biomark Res 2024; 12:93. [PMID: 39217377 PMCID: PMC11366172 DOI: 10.1186/s40364-024-00644-3] [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: 06/04/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024] Open
Abstract
Malignant tumors remain a primary cause of human mortality. Among the various treatment modalities for neoplasms, tumor vaccines have consistently shown efficacy and promising potential. These vaccines offer advantages such as specificity, safety, and tolerability, with mRNA vaccines representing promising platforms. By introducing exogenous mRNAs encoding antigens into somatic cells and subsequently synthesizing antigens through gene expression systems, mRNA vaccines can effectively induce immune responses. Katalin Karikó and Drew Weissman were awarded the 2023 Nobel Prize in Physiology or Medicine for their great contributions to mRNA vaccine research. Compared with traditional tumor vaccines, mRNA vaccines have several advantages, including rapid preparation, reduced contamination, nonintegrability, and high biodegradability. Tumor-targeted therapy is an innovative treatment modality that enables precise targeting of tumor cells, minimizes damage to normal tissues, is safe at high doses, and demonstrates great efficacy. Currently, targeted therapy has become an important treatment option for malignant tumors. The application of mRNA vaccines in tumor-targeted therapy is expanding, with numerous clinical trials underway. We systematically outline the targeted delivery mechanism of mRNA vaccines and the mechanism by which mRNA vaccines induce anti-tumor immune responses, describe the current research and clinical applications of mRNA vaccines in tumor-targeted therapy, and forecast the future development trends of mRNA vaccine application in tumor-targeted therapy.
Collapse
Affiliation(s)
- Yu Gao
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China
| | - Zhenning Li
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Liaoning Province Key Laboratory of Oral Disease, Shenyang, 110001, China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032, China.
| |
Collapse
|
30
|
Choi W, Kohane DS. Hybrid Nanoparticle-Hydrogel Systems for Drug Delivery Depots and Other Biomedical Applications. ACS NANO 2024; 18:22780-22792. [PMID: 39140388 PMCID: PMC11926886 DOI: 10.1021/acsnano.4c06888] [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] [Indexed: 08/15/2024]
Abstract
Hydrogel-based depots typically tend to remain where injected and have excellent biocompatibility but are relatively poor at controlling drug release. Nanoparticles (NPs) typically have the opposite properties. The smaller the NPs are, the more likely they are to leave the site of injection. Their biocompatibility is variable depending on the material but can be poor. However, NPs can be good at controlling drug release. In these and other properties, combining NPs and hydrogels can leverage their advantages and negate their disadvantages. This review highlights the rationale for hybrid NP-hydrogel systems in drug delivery, the basic methods of producing them, and examples where combining the two systems addressed specific problems.
Collapse
Affiliation(s)
- Wonmin Choi
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
31
|
Khan MS, Fatima M, Wahab S, Khalid M, Kesharwani P. Gallic acid loaded self-nano emulsifying hydrogel-based drug delivery system against onychomycosis. Nanomedicine (Lond) 2024; 19:2065-2083. [PMID: 39143900 PMCID: PMC11485813 DOI: 10.1080/17435889.2024.2386923] [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: 05/09/2024] [Accepted: 07/29/2024] [Indexed: 08/16/2024] Open
Abstract
Aim: To developed and investigate gallic acid (GA) loaded self-nanoemulsifying drug delivery systems (SNEDDS) for treating onychomycosis via transungual route.Materials & methods: The SNEDDS were prepared by direct dispersion technique and were evaluated for characteristics parameters using Fourier transform infrared, differential scanning calorimetry, confocal microscopy, transmission electron microscopy and zeta sizer. Furthermore, the safety of prepared formulation was evaluated via Hen's egg test-chorioallantoic membrane study and stability was confirmed using different parameters. Also, its effectiveness was evaluated against fungal strain Trichophyton mentagrophytes.Results: The SNEDDS displayed a particle size of 199.8 ± 4.21 nm and a zeta potential; of -22.75 ± 2.09 mV. Drug release study illustrated a sustained release pattern with a release of 70.34 ± 0.20% over a period of 24 h. The penetration across the nail plate was found to be 1.59 ± 0.002 µg/mg and 0.97 ± 0.001 µg/mg for GA loaded SNEDDS and GA solution respectively. An irritation score of 0.52 ± 0.005 and 3.84 ± 0.001 was reported for GA loaded SNEDDS hydrogel and GA solution, indicating a decrease in the drug's irritation potential from slightly irritating to non irritating due to its entrapment within the SNEDDS.Conclusion: GA loaded SNEDDS has potential to address limitations of conventional treatments, enhancing the drug's efficacy and reducing the likelihood of resistance in the treatment of Onychomycosis.
Collapse
Affiliation(s)
- Mohammad Sameer Khan
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Mahak Fatima
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha62529, Saudi Arabia
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, 110062, India
| |
Collapse
|
32
|
Ekmekcioglu A, Gok O, Oz-Arslan D, Erdal MS, Yagan Uzuner Y, Muftuoglu M. Mitochondria-Targeted Liposomes for Drug Delivery to Tumor Mitochondria. Pharmaceutics 2024; 16:950. [PMID: 39065647 PMCID: PMC11280384 DOI: 10.3390/pharmaceutics16070950] [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: 06/05/2024] [Revised: 07/01/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
The special bilayer structure of mitochondrion is a promising therapeutic target in the diagnosis and treatment of diseases such as cancer and metabolic diseases. Nanocarriers such as liposomes modified with mitochondriotropic moieties can be developed to send therapeutic molecules to mitochondria. In this study, DSPE-PEG-TPP polymer conjugate was synthesized and used to prepare mitochondria-targeted liposomes (TPPLs) to improve the therapeutic index of chemotherapeutic agents functioning in mitochondria and reduce their side effects. Doxorubicin (Dox) loaded-TPPL and non-targeted PEGylated liposomes (PPLs) were prepared and compared based on physicochemical properties, morphology, release profile, cellular uptake, mitochondrial localization, and anticancer effects. All formulations were spherically shaped with appropriate size, dispersity, and zeta potential. The stability of the liposomes was favorable for two months at 4 °C. TPPLs localize to mitochondria, whereas PPLs do not. The empty TPPLs and PPLs were not cytotoxic to HCT116 cells. The release kinetics of Dox-loaded liposomes showed that Dox released from TPPLs was higher at pH 5.6 than at pH 7.4, which indicates a higher accumulation of the released drug in the tumor environment. The half-maximal inhibitory concentration of Dox-loaded TPPLs and PPLs was 1.62-fold and 1.17-fold lower than that of free Dox due to sustained drug release, respectively. The reactive oxygen species level was significantly increased when HCT116 cells were treated with Dox-loaded TPPLs. In conclusion, TPPLs may be promising carriers for targeted drug delivery to tumor mitochondria.
Collapse
Affiliation(s)
- Aysegul Ekmekcioglu
- Institute of Health Sciences, Department of Medical Biotechnology, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | - Ozgul Gok
- Faculty of Engineering and Natural Sciences, Department of Biomedical Engineering, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | - Devrim Oz-Arslan
- School of Medicine, Department of Biophysics, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | - Meryem Sedef Erdal
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Istanbul University, 34116 Istanbul, Turkey;
| | - Yasemin Yagan Uzuner
- Faculty of Pharmacy, Department of Pharmaceutical Technology, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | - Meltem Muftuoglu
- Institute of Health Sciences, Department of Medical Biotechnology, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
- Faculty of Engineering and Natural Sciences, Department of Molecular Biology and Genetics, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey
| |
Collapse
|
33
|
He X, Payne TJ, Takanashi A, Fang Y, Kerai SD, Morrow JP, Al-Wassiti H, Pouton CW, Kempe K. Tailored Monoacyl Poly(2-oxazoline)- and Poly(2-oxazine)-Lipids as PEG-Lipid Alternatives for Stabilization and Delivery of mRNA-Lipid Nanoparticles. Biomacromolecules 2024; 25:4591-4603. [PMID: 38918933 DOI: 10.1021/acs.biomac.4c00651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
The successful use of lipid nanoparticles (LNPs) for clinical development of the COVID-19 mRNA vaccines marked a breakthrough in mRNA-LNP therapeutics. As one of the vital components of LNPs, poly(ethylene glycol)-lipid conjugates (PEG-lipids) influence particle biophysical properties and stability, as well as interactions within biological environments. Reports suggesting that anti-PEG antibodies can be detected quite commonly within the human population raise concerns that PEG content in commercial LNP products could further stimulate immune responses to PEG. The presence of anti-PEG antibodies has been linked to accelerated clearance of LNPs, potentially a source of variability in the biological response to mRNA-LNP products. This motivated us to explore potential PEG alternatives. Herein, we report physicochemical and biological properties of mRNA-LNPs assembled using poly(2-oxazoline) (POx)- and poly(2-oxazine) (POz)-based polymer-lipid conjugates. Notably, we investigated monoacyl lipids as alternatives to diacyl lipids. mRNA-LNPs produced using monoacyl POx/POz-lipids displayed comparable biophysical characteristics and cytocompatibility. Delivery of reporter mRNA resulted in similar transfection efficiencies, in both adherent and suspension cells, and in mice, compared to PEG-lipid equivalents. Our results suggest that monoacyl POx/POz-lipid-containing LNPs are promising candidates for the development of PEG-free LNP-based therapeutic products.
Collapse
Affiliation(s)
- Xie He
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Thomas J Payne
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Asuka Takanashi
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Yunji Fang
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Simran D Kerai
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Joshua P Morrow
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Hareth Al-Wassiti
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Colin W Pouton
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Kristian Kempe
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
- Materials Science and Engineering, Monash University, Clayton, VIC 3800, Australia
| |
Collapse
|
34
|
Okezue MA, Uche C, Adebola A, Byrn SR. A Quality by Design Approach for Optimizing Solid Lipid Nanoparticles of Bedaquiline for Improved Product Performance. AAPS PharmSciTech 2024; 25:152. [PMID: 38954218 DOI: 10.1208/s12249-024-02873-z] [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: 04/23/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024] Open
Abstract
Bedaquiline (BQ) solid lipid nanoparticles (SLNs), which have previously been formulated for parenteral administration, have a risk of patient non-compliance in treating tuberculosis. This research presents a strategy to develop BQ SLNs for oral delivery to improve patient adherence, The upper and lower levels for the formulation excipients were generated from screening experiments. Using 4 input factors (BQ, lecithin, Tween 80, and PEG), a full factorial design from 3 × 2x2 × 2 experiments was randomly arranged to investigate 3 response variables: Particle size distribution (PSD), polydispersity index (PdI), and zeta potential (ZP). High shear homogenization was used to mix the solvent and aqueous phases, with 15% sucrose as a cryoprotectant. The response variables were assessed using a zeta sizer while TEM micrographs confirmed the PSD data. Solid-state assessments were conducted using powdered X-ray diffraction and scanning electron microscopy (SEM) imaging. A comparative invitro assessment was used to determine drug release from an equivalent dose of BQ free base powder and BQ-SLN, both packed in hard gelatin capsules. The sonicated formulations obtained significant effects for PSD, PdI, and ZP. The p-values (0.0001 for PdI, 0.0091 for PSD) for BQ as an independent variable in the sonicated formulation were notably higher than those in the unsonicated formulation (0.1336 for PdI, 0.0117 for PSD). The SEM images were between 100 - 400 nm and delineated nanocrystals of BQ embedded in the lipid matrix. The SLN formulation provides higher drug levels over the drug's free base; a similarity factor (f2 = 18.3) was estimated from the dissolution profiles.
Collapse
Affiliation(s)
- Mercy A Okezue
- Industrial and Molecular Pharmaceutics Department, Purdue University, West Lafayette, Indiana, 47907, USA.
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, Michigan, USA.
| | - Chidi Uche
- School of Agricultural & Biological Engineering, Biotechnology Innovation & Regulatory Science, Purdue University, West Lafayette, Indiana, 47907, USA
| | - Adekoya Adebola
- Department of Public Health, Texila American University, Georgetown, Guyana
| | - Stephen R Byrn
- Industrial and Molecular Pharmaceutics Department, Purdue University, West Lafayette, Indiana, 47907, USA.
| |
Collapse
|
35
|
Pula W, Ganugula R, Esposito E, Ravi Kumar MNV, Arora M. Engineered urolithin A-laden functional polymer-lipid hybrid nanoparticles prevent cisplatin-induced proximal tubular injury in vitro. Eur J Pharm Biopharm 2024; 200:114334. [PMID: 38768764 PMCID: PMC11262884 DOI: 10.1016/j.ejpb.2024.114334] [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: 03/29/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 05/22/2024]
Abstract
Functional polymer-lipid hybrid nanoparticles (H-NPs) are a promising class of nanocarriers that combine the benefits of polymer and lipid nanoparticles, offering biocompatibility, structural stability, high loading capacity, and, most importantly, superior surface functionalization. Here, we report the synthesis and design of highly functional H-NPs with specificity toward the transferrin receptor (TfR), using a small molecule ligand, gambogic acid (GA). A fluorescence study revealed the molecular orientation of H-NPs, where the lipid-dense core is surrounded by a polymer exterior, functionalized with GA. Urolithin A, an immunomodulator and anti-inflammatory agent, served as a model drug-like compound to prepare H-NPs via traditional emulsion-based techniques, where H-NPs led to smaller particles (132 nm) and superior entrapment efficiencies (70 % at 10 % drug loading) compared to GA-conjugated polymeric nanoparticles (P-NPs) (157 nm and 52 % entrapment efficiency) and solid lipid nanoparticles (L-NPs) (186 nm and 29 % entrapment efficiency). H-NPs showed superior intracellular accumulation compared to individual NPs using human small intestinal epithelial (FHs 74) cells. The in vitro efficacy was demonstrated by flow cytometry analysis, in which UA-laden H-NPs showed excellent anti-inflammatory properties in cisplatin-induced injury in healthy human proximal tubular cell (HK2) model by decreasing the TLR4, NF-κβ, and IL-β expression. This preliminary work highlights the potential of H-NPs as a novel functional polymer-lipid drug delivery system, establishing the foundation for future research on its therapeutic potential in addressing chemotherapy-induced acute kidney injury in cancer patients.
Collapse
Affiliation(s)
- W Pula
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Fossato di Mortara, 19-44121 Ferrara, Italy; The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States
| | - R Ganugula
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States; Division of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL 35401, United States; Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35401, United States; Department of Biological Sciences, The University of Alabama, SEC 1325, Tuscaloosa, AL 35487, United States
| | - E Esposito
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Via Fossato di Mortara, 19-44121 Ferrara, Italy
| | - M N V Ravi Kumar
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States; Division of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL 35401, United States; Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35401, United States; Department of Biological Sciences, The University of Alabama, SEC 1325, Tuscaloosa, AL 35487, United States; Chemical and Biological Engineering, University of Alabama, SEC 3448, Tuscaloosa, AL 35487, United States; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States; Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - M Arora
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States; Division of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL 35401, United States; Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35401, United States; Department of Biological Sciences, The University of Alabama, SEC 1325, Tuscaloosa, AL 35487, United States.
| |
Collapse
|
36
|
Alwani S, Wasan EK, Badea I. Solid Lipid Nanoparticles for Pulmonary Delivery of Biopharmaceuticals: A Review of Opportunities, Challenges, and Delivery Applications. Mol Pharm 2024; 21:3084-3102. [PMID: 38828798 DOI: 10.1021/acs.molpharmaceut.4c00128] [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] [Indexed: 06/05/2024]
Abstract
Biopharmaceuticals such as nucleic acids, proteins, and peptides constitute a new array of treatment modalities for chronic ailments. Invasive routes remain the mainstay of administering biopharmaceuticals due to their labile nature in the biological environment. However, it is not preferred for long-term therapy due to the lack of patient adherence and clinical suitability. Therefore, alternative routes of administration are sought to utilize novel biopharmaceutical therapies to their utmost potential. Nanoparticle-mediated pulmonary delivery of biologics can facilitate both local and systemic disorders. Solid lipid nanoparticles (SLNs) afford many opportunities as pulmonary carriers due to their physicochemical stability and ability to incorporate both hydrophilic and hydrophobic moieties, thus allowing novel combinatorial drug/gene therapies. These applications include pulmonary infections, lung cancer, and cystic fibrosis, while systemic delivery of biomolecules, like insulin, is also attractive for the treatment of chronic ailments. This Review explores physiological and particle-associated factors affecting pulmonary delivery of biopharmaceuticals. It compares the advantages and limitations of SLNs as pulmonary nanocarriers along with design improvements underway to overcome these limitations. Current research illustrating various SLN designs to deliver proteins, peptides, plasmids, oligonucleotides, siRNA, and mRNA is also summarized.
Collapse
Affiliation(s)
- Saniya Alwani
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Saskatoon, S7N 5E5 Saskatchewan, Canada
| | - Ellen K Wasan
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Saskatoon, S7N 5E5 Saskatchewan, Canada
| | - Ildiko Badea
- College of Pharmacy and Nutrition, University of Saskatchewan, 107 Wiggins Road, Health Sciences Building, Saskatoon, S7N 5E5 Saskatchewan, Canada
| |
Collapse
|
37
|
Karekar N, Reid Cahn A, Morla-Folch J, Saffon A, Ward RW, Ananthanarayanan A, Teunissen AJP, Bhardwaj N, Vabret N. Protocol for the development of mRNA lipid nanoparticle vaccines and analysis of immunization efficiency in mice. STAR Protoc 2024; 5:103087. [PMID: 38795353 PMCID: PMC11144802 DOI: 10.1016/j.xpro.2024.103087] [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: 02/05/2024] [Revised: 04/07/2024] [Accepted: 05/02/2024] [Indexed: 05/27/2024] Open
Abstract
Here, we present a protocol for the development of mRNA-loaded lipid nanoparticle (LNP) vaccines for target antigen sequences of interest. We describe key steps required to design and synthesize mRNA constructs, their LNP encapsulation, and mouse immunization. We then detail quality control assays to determine RNA purity, guidelines to measure RNA immunogenicity using in vitro reporter systems, and a technique to evaluate antigen-specific T cell responses following immunization.
Collapse
Affiliation(s)
- Neha Karekar
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ashley Reid Cahn
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Judit Morla-Folch
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Alexis Saffon
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ross W Ward
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Aparna Ananthanarayanan
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Abraham J P Teunissen
- Biomedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nina Bhardwaj
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Hematology/Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Nicolas Vabret
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
38
|
Chu R, Wang Y, Kong J, Pan T, Yang Y, He J. Lipid nanoparticles as the drug carrier for targeted therapy of hepatic disorders. J Mater Chem B 2024; 12:4759-4784. [PMID: 38682294 DOI: 10.1039/d3tb02766j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
The liver, a complex and vital organ in the human body, is susceptible to various diseases, including metabolic disorders, acute hepatitis, cirrhosis, and hepatocellular carcinoma. In recent decades, these diseases have significantly contributed to global morbidity and mortality. Currently, liver transplantation remains the most effective treatment for hepatic disorders. Nucleic acid therapeutics offer a selective approach to disease treatment through diverse mechanisms, enabling the regulation of relevant genes and providing a novel therapeutic avenue for hepatic disorders. It is expected that nucleic acid drugs will emerge as the third generation of pharmaceuticals, succeeding small molecule drugs and antibody drugs. Lipid nanoparticles (LNPs) represent a crucial technology in the field of drug delivery and constitute a significant advancement in gene therapies. Nucleic acids encapsulated in LNPs are shielded from the degradation of enzymes and effectively delivered to cells, where they are released and regulate specific genes. This paper provides a comprehensive review of the structure, composition, and applications of LNPs in the treatment of hepatic disorders and offers insights into prospects and challenges in the future development of LNPs.
Collapse
Affiliation(s)
- Runxuan Chu
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| | - Yi Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tung, Hong Kong SAR, P. R. China.
| | - Jianglong Kong
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tung, Hong Kong SAR, P. R. China.
| | - Ting Pan
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
- Department of Pharmaceutics School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Yani Yang
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| | - Jun He
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| |
Collapse
|
39
|
Gagliardi A, Chiarella E, Voci S, Ambrosio N, Celano M, Cristina Salvatici M, Cosco D. DIFUCOSIN: DIclofenac sodium salt loaded FUCOidan-SericIN nanoparticles for the management of chronic inflammatory diseases. Int J Pharm 2024; 655:124034. [PMID: 38531433 DOI: 10.1016/j.ijpharm.2024.124034] [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: 03/07/2024] [Accepted: 03/20/2024] [Indexed: 03/28/2024]
Abstract
The current investigation emphasizes the use of fucoidan and sericin as dual-role biomaterials for obtaining novel nanohybrid systems for the delivery of diclofenac sodium (DS) and the potential treatment of chronic inflammatory diseases. The innovative formulations containing 4 mg/ml of fucoidan and 3 mg/ml of sericin showed an average diameter of about 200 nm, a low polydispersity index (0.17) and a negative surface charge. The hybrid nanosystems demonstrated high stability at various pHs and temperatures, as well as in both saline and glucose solutions. The Rose Bengal assay evidenced that fucoidan is the primary modulator of relative surface hydrophobicity with a two-fold increase of this parameter when compared to sericin nanoparticles. The interaction between the drug and the nanohybrids was confirmed through FT-IR analysis. Moreover, the release profile of DS from the colloidal systems showed a prolonged and constant drug leakage over time both at pH 5 and 7. The DS-loaded nanohybrids (DIFUCOSIN) induced a significant decrease of IL-6 and IL-1β with respect to the active compound in human chondrocytes evidencing a synergistic action of the individual components of nanosystems and the drug and demonstrating the potential application of the proposed nanomedicine for the treatment of inflammation.
Collapse
Affiliation(s)
- Agnese Gagliardi
- Department of Health Sciences, University "Magna Græcia", 88100 Catanzaro, Italy
| | - Emanuela Chiarella
- Department of Experimental and Clinical Medicine, University "Magna Græcia", 88100 Catanzaro, Italy
| | - Silvia Voci
- Department of Health Sciences, University "Magna Græcia", 88100 Catanzaro, Italy
| | - Nicola Ambrosio
- Department of Health Sciences, University "Magna Græcia", 88100 Catanzaro, Italy
| | - Marilena Celano
- Department of Health Sciences, University "Magna Græcia", 88100 Catanzaro, Italy
| | - Maria Cristina Salvatici
- Institute of Chemistry of Organometallic Compounds (ICCOM)-Electron Microscopy Centre (Ce.M.E.), National Research Council (CNR), 50019, Sesto Fiorentino, Firenze, Italy
| | - Donato Cosco
- Department of Health Sciences, University "Magna Græcia", 88100 Catanzaro, Italy.
| |
Collapse
|
40
|
Opsomer L, Jana S, Mertens I, Cui X, Hoogenboom R, Sanders NN. Efficient in vitro and in vivo transfection of self-amplifying mRNA with linear poly(propylenimine) and poly(ethylenimine-propylenimine) random copolymers as non-viral carriers. J Mater Chem B 2024; 12:3927-3946. [PMID: 38563779 DOI: 10.1039/d3tb03003b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Messenger RNA (mRNA) based vaccines have been introduced worldwide to combat the Covid-19 pandemic. These vaccines consist of non-amplifying mRNA formulated in lipid nanoparticles (LNPs). Consequently, LNPs are considered benchmark non-viral carriers for nucleic acid delivery. However, the formulation and manufacturing of these mRNA-LNP nanoparticles are expensive and time-consuming. Therefore, we used self-amplifying mRNA (saRNA) and synthesized novel polymers as alternative non-viral carrier platform to LNPs, which enable a simple, rapid, one-pot formulation of saRNA-polyplexes. Our novel polymer-based carrier platform consists of randomly concatenated ethylenimine and propylenimine comonomers, resulting in linear, poly(ethylenimine-ran-propylenimine) (L-PEIx-ran-PPIy) copolymers with controllable degrees of polymerization. Here we demonstrate in multiple cell lines, that our saRNA-polyplexes show comparable to higher in vitro saRNA transfection efficiencies and higher cell viabilities compared to formulations with Lipofectamine MessengerMAX™ (LFMM), a commercial, lipid-based carrier considered to be the in vitro gold standard carrier. This is especially true for our in vitro best performing saRNA-polyplexes with N/P 5, which are characterised with a size below 100 nm, a positive zeta potential, a near 100% encapsulation efficiency, a high retention capacity and the ability to protect the saRNA from degradation mediated by RNase A. Furthermore, an ex vivo hemolysis assay with pig red blood cells demonstrated that the saRNA-polyplexes exhibit negligible hemolytic activity. Finally, a bioluminescence-based in vivo study was performed over a 35-day period, and showed that the polymers result in a higher and prolonged bioluminescent signal compared to naked saRNA and L-PEI based polyplexes. Moreover, the polymers show different expression profiles compared to those of LNPs, with one of our new polymers (L-PPI250) demonstrating a higher sustained expression for at least 35 days after injection.
Collapse
Affiliation(s)
- Lisa Opsomer
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium.
| | - Somdeb Jana
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, 9000 Ghent, Belgium.
| | - Ine Mertens
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, 9000 Ghent, Belgium.
| | - Xiaole Cui
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium.
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Ghent University, 9000 Ghent, Belgium.
| | - Niek N Sanders
- Laboratory of Gene Therapy, Department of Veterinary and Biosciences, Faculty of Veterinary Medicine, Ghent University, B-9820 Merelbeke, Belgium.
- Cancer Research Institute (CRIG), Ghent University, B-9000 Ghent, Belgium
| |
Collapse
|
41
|
Zhou L, Godse S, Sinha N, Kodidela S, Singh U, Kumar S. Darunavir Nanoformulation Suppresses HIV Pathogenesis in Macrophages and Improves Drug Delivery to the Brain in Mice. Pharmaceutics 2024; 16:555. [PMID: 38675216 PMCID: PMC11054602 DOI: 10.3390/pharmaceutics16040555] [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: 03/04/2024] [Revised: 04/01/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Although antiretroviral therapy (ART) can suppress peripheral HIV, patients still suffer from neuroHIV due to insufficient levels of ART drugs in the brain. Hence, this study focuses on developing a poly lactic-co-glycolic acid (PLGA) nanoparticle-based ART drug delivery system for darunavir (DRV) using an intranasal route that can overcome the limitation of drug metabolic stability and blood-brain barrier (BBB) permeability. The physicochemical properties of PLGA-DRV were characterized. The results indicated that PLGA-DRV formulation inhibits HIV replication in U1 macrophages directly and in the presence of the BBB without inducing cytotoxicity. However, the PLGA-DRV did not inhibit HIV replication more than DRV alone. Notably, the total antioxidant capacity remained unchanged upon treatment with both DRV or PLGA-DRV in U1 cells. Compared to DRV alone, PLGA-DRV further decreased reactive oxygen species, suggesting a decrease in oxidative stress by the formulation. Oxidative stress is generally increased by HIV infection, leading to increased inflammation. Although the PLGA-DRV formulation did not further reduce the inflammatory response, the formulation did not provoke an inflammatory response in HIV-infected U1 macrophages. As expected, in vitro experiments showed higher DRV permeability by PLGA-DRV than DRV alone to U1 macrophages. Importantly, in vivo experiments, especially using intranasal administration of PLGA-DRV in wild-type mice, demonstrated a significant increase in the brain-to-plasma ratio of DRV compared to the free DRV. Overall, findings from this study attest to the potential of the PLGA-DRV nanoformulation in reducing HIV pathogenesis in macrophages and enhancing drug delivery to the brain, offering a promising avenue for treating HIV-related neurological disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Santosh Kumar
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, 881 Madison Ave., Memphis, TN 38163, USA (S.G.); (U.S.)
| |
Collapse
|
42
|
De A, Kang JH, Sauraj, Lee OH, Ko YT. Optimizing long-term stability of siRNA using thermoassemble ionizable reverse pluronic-Bcl2 micelleplexes. Int J Biol Macromol 2024; 264:130783. [PMID: 38471603 DOI: 10.1016/j.ijbiomac.2024.130783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/26/2024] [Accepted: 03/08/2024] [Indexed: 03/14/2024]
Abstract
Thermosassemble Ionizable Reverse Pluronic (TIRP) platform stands out for its distinctive combination of thermoassemble and ionizable features, effectively overcoming challenges in previous siRNA delivery systems. This study opens up a formation for long-term stabilization, and high loading of siRNA, specifically crafted for targeting oncogenic pathways. TIRP-Bcl2 self-assembles into a unique micelle structure with a nanodiameter of 75.8 ± 5.7 nm, efficiently encapsulating Bcl2 siRNA while maintaining exceptional colloidal stability at 4 °C for 8 months, along with controlled release profiles lasting 180 h. The dual ionizable headgroup enhance the siRNA loading and the revers pluronic unique structural orientation enhance the stability of the siRNA. The thermoassemble of TIRP-Bcl2 facilitates flexi-rigid response to mild hyperthermia, enhancing deep tissue penetration and siRNA release in the tumor microenvironment. This responsive behavior improves intracellular uptake and gene silencing efficacy in cancer cells. TIRP, with its smaller particle size and reverse pluronic nature, efficiently transports siRNA across the blood-brain barrier, holding promise for revolutionizing glioblastoma (GBM) treatment. TIRP-Bcl2 shows significant potential for precise, personalized therapies, promising prolonged siRNA delivery and in vitro/in vivo stability. This research opens avenues for further exploration and clinical translation of this innovative nanocarrier system across different cancers.
Collapse
Affiliation(s)
- Anindita De
- College of Pharmacy, Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, South Korea..
| | - Ji Hee Kang
- College of Pharmacy, Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, South Korea
| | - Sauraj
- College of Pharmacy, Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, South Korea
| | - O Hyun Lee
- College of Pharmacy, Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, South Korea
| | - Young Tag Ko
- College of Pharmacy, Gachon Institute of Pharmaceutical Science, Gachon University, Incheon 21936, South Korea..
| |
Collapse
|
43
|
Hsia T, Chen Y. RNA-encapsulating lipid nanoparticles in cancer immunotherapy: From pre-clinical studies to clinical trials. Eur J Pharm Biopharm 2024; 197:114234. [PMID: 38401743 DOI: 10.1016/j.ejpb.2024.114234] [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: 11/19/2023] [Revised: 01/29/2024] [Accepted: 02/14/2024] [Indexed: 02/26/2024]
Abstract
Nanoparticle-based delivery systems such as RNA-encapsulating lipid nanoparticles (RNA LNPs) have dramatically advanced in function and capacity over the last few decades. RNA LNPs boast of a diverse array of external and core configurations that enhance targeted delivery and prolong circulatory retention, advancing therapeutic outcomes. Particularly within the realm of cancer immunotherapies, RNA LNPs are increasingly gaining prominence. Pre-clinical in vitro and in vivo studies have laid a robust foundation for new and ongoing clinical trials that are actively enrolling patients for RNA LNP cancer immunotherapy. This review explores RNA LNPs, starting from their core composition to their external membrane formulation, set against a backdrop of recent clinical breakthroughs. We further elucidate the LNP delivery avenues, broach the prevailing challenges, and contemplate the future perspectives of RNA LNP-mediated immunotherapy.
Collapse
Affiliation(s)
- Tiffaney Hsia
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
44
|
Hoover E, Ruggiero OM, Swingler RN, Day ES. FZD7-Targeted Nanoparticles to Enhance Doxorubicin Treatment of Triple-Negative Breast Cancer. ACS OMEGA 2024; 9:14323-14335. [PMID: 38559981 PMCID: PMC10976388 DOI: 10.1021/acsomega.3c10275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 04/04/2024]
Abstract
Doxorubicin (DOX) is a chemotherapy agent commonly used to treat triple-negative breast cancer (TNBC), but it has insufficient efficacy against the disease and considerable toxicity due to its off-target delivery. To improve the specificity of DOX for TNBC, we encapsulated it in poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) coated with antibodies against Frizzled7 (FZD7), a receptor that is overexpressed on TNBC cells and which is a key activator of the Wnt signaling pathway. In vitro studies show that DOX encapsulation does not hinder its ability to localize to the nucleus in human TNBC cell cultures and that DOX delivered via NPs induces apoptosis and DNA damage via H2A.X phosphorylation to the same degree as freely delivered DOX. FZD7-targeted NPs delivering DOX caused significantly greater inhibition of metabolic activity and led to a smaller cell population following treatment when compared to freely delivered DOX or DOX-loaded NPs coated only with poly(ethylene glycol) (PEG). The FZD7 antibodies additionally provided significant levels of Wnt pathway inhibition, as demonstrated by an increase in β-catenin phosphorylation, indicative of β-catenin destruction and downregulation. These results show that FZD7-targeted platforms have great promise for improving the therapeutic window of otherwise toxic chemotherapies like DOX in TNBC and other cancers that display the overexpression of FZD7 receptors.
Collapse
Affiliation(s)
- Elise
C. Hoover
- Department
of Biomedical Engineering, University of
Delaware, Newark, Delaware 19713, United States
| | - Olivia M. Ruggiero
- Department
of Biomedical Engineering, University of
Delaware, Newark, Delaware 19713, United States
| | - Rachel N. Swingler
- Department
of Biomedical Engineering, University of
Delaware, Newark, Delaware 19713, United States
| | - Emily S. Day
- Department
of Biomedical Engineering, University of
Delaware, Newark, Delaware 19713, United States
- Department
of Materials Science and Engineering, University
of Delaware, Newark, Delaware 19716, United States
- Helen
F. Graham Cancer Center and Research Institute, Newark, Delaware 19713, United States
| |
Collapse
|
45
|
Kim LJ, Shin D, Leite WC, O’Neill H, Ruebel O, Tritt A, Hura GL. Simple Scattering: Lipid nanoparticle structural data repository. Front Mol Biosci 2024; 11:1321364. [PMID: 38584701 PMCID: PMC10998447 DOI: 10.3389/fmolb.2024.1321364] [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: 10/13/2023] [Accepted: 02/19/2024] [Indexed: 04/09/2024] Open
Abstract
Lipid nanoparticles (LNPs) are being intensively researched and developed to leverage their ability to safely and effectively deliver therapeutics. To achieve optimal therapeutic delivery, a comprehensive understanding of the relationship between formulation, structure, and efficacy is critical. However, the vast chemical space involved in the production of LNPs and the resulting structural complexity make the structure to function relationship challenging to assess and predict. New components and formulation procedures, which provide new opportunities for the use of LNPs, would be best identified and optimized using high-throughput characterization methods. Recently, a high-throughput workflow, consisting of automated mixing, small-angle X-ray scattering (SAXS), and cellular assays, demonstrated a link between formulation, internal structure, and efficacy for a library of LNPs. As SAXS data can be rapidly collected, the stage is set for the collection of thousands of SAXS profiles from a myriad of LNP formulations. In addition, correlated LNP small-angle neutron scattering (SANS) datasets, where components are systematically deuterated for additional contrast inside, provide complementary structural information. The centralization of SAXS and SANS datasets from LNPs, with appropriate, standardized metadata describing formulation parameters, into a data repository will provide valuable guidance for the formulation of LNPs with desired properties. To this end, we introduce Simple Scattering, an easy-to-use, open data repository for storing and sharing groups of correlated scattering profiles obtained from LNP screening experiments. Here, we discuss the current state of the repository, including limitations and upcoming changes, and our vision towards future usage in developing our collective knowledge base of LNPs.
Collapse
Affiliation(s)
- Lee Joon Kim
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - David Shin
- David Shin Consulting, Berkeley, CA, United States
| | - Wellington C. Leite
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Hugh O’Neill
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, TN, United States
| | - Oliver Ruebel
- Scientific Data Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Andrew Tritt
- Applied Mathematics and Computational Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
| | - Greg L. Hura
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, United States
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, CA, United States
| |
Collapse
|
46
|
Bristow P, Schantz K, Moosbrugger Z, Martin K, Liebenberg H, Steimle S, Xiao Q, Percec V, Wilner SE. Aptamer-Targeted Dendrimersomes Assembled from Azido-Modified Janus Dendrimers "Clicked" to DNA. Biomacromolecules 2024; 25:1541-1549. [PMID: 38394608 PMCID: PMC10934268 DOI: 10.1021/acs.biomac.3c01108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024]
Abstract
Amphiphilic Janus dendrimers (JDs), synthetic alternatives to lipids, have the potential to expand the scope of nanocarrier delivery systems. JDs self-assemble into vesicles called dendrimersomes, encapsulate both hydrophobic cargo and nucleic acids, and demonstrate enhanced stability in comparison to lipid nanoparticles (LNPs). Here, we report the ability to enhance the cellular uptake of Janus dendrimersomes using DNA aptamers. Azido-modified JDs were synthesized and conjugated to alkyne-modified DNAs using copper-catalyzed azide alkyne cycloaddition. DNA-functionalized JDs form nanometer-sized dendrimersomes in aqueous solution via thin film hydration. These vesicles, now displaying short DNAs, are then hybridized to transferrin receptor binding DNA aptamers. Aptamer-targeted dendrimersomes show improved cellular uptake as compared to control vesicles via fluorescence microscopy and flow cytometry. This work demonstrates the versatility of using click chemistry to conjugate functionalized JDs with biologically relevant molecules and the feasibility of targeting DNA-modified dendrimersomes for drug delivery applications.
Collapse
Affiliation(s)
- Paige Bristow
- Department
of Chemistry, Ursinus College, Collegeville, Pennsylvania 19426, United States
| | - Kyle Schantz
- Department
of Chemistry, Ursinus College, Collegeville, Pennsylvania 19426, United States
| | - Zoe Moosbrugger
- Department
of Chemistry, Ursinus College, Collegeville, Pennsylvania 19426, United States
| | - Kailey Martin
- Department
of Chemistry, Ursinus College, Collegeville, Pennsylvania 19426, United States
| | - Haley Liebenberg
- Department
of Chemistry, Ursinus College, Collegeville, Pennsylvania 19426, United States
| | - Stefan Steimle
- Department
of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19014, United States
| | - Qi Xiao
- Roy
& Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19014, United States
| | - Virgil Percec
- Roy
& Diana Vagelos Laboratories, Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19014, United States
| | - Samantha E. Wilner
- Department
of Chemistry, Ursinus College, Collegeville, Pennsylvania 19426, United States
| |
Collapse
|
47
|
Williamson GL, Bachelder EM, Ainslie KM. Clinical and Preclinical Methods of Heat-Stabilization of Human Vaccines. Mol Pharm 2024; 21:1015-1026. [PMID: 38288698 PMCID: PMC11607726 DOI: 10.1021/acs.molpharmaceut.3c00844] [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] [Indexed: 03/05/2024]
Abstract
Vaccines have historically faced challenges regarding stability, especially in regions lacking a robust cold chain infrastructure. This review delves into established and emergent techniques to improve the thermostability of vaccines. We discuss the widely practiced lyophilization method, effectively transforming liquid vaccine formulations into a solid powdered state, enhancing storage and transportation ability. However, potential protein denaturation during lyophilization necessitates alternative stabilization methods. Cryoprotectants, namely, starch and sugar molecules, have shown promise in protecting vaccine antigens and adjuvants from denaturation and augmenting the stability of biologics during freeze-drying. Biomineralization, a less studied yet innovative approach, utilizes inorganic or organic-inorganic hybrids to encapsulate biological components of vaccines with a particular emphasis on metal-organic coordination polymers. Encapsulation in organic matrices to form particles or microneedles have also been studied in the context of vaccine thermostability, showing some ability to store outside the cold-chain. Unfortunately, few of these techniques have advanced to clinical trials that evaluate differences in storage conditions. Nonetheless, early trials suggest that alternative storage techniques are viable and emphasize the need for more comprehensive studies. This review underscores the pressing need for heat-stable vaccines, especially in light of the increasing global distribution challenges. Combining traditional methods with novel approaches holds promise for the future adaptability of vaccine distribution and use.
Collapse
Affiliation(s)
- Grace L. Williamson
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA
| | - Eric M. Bachelder
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA
| | - Kristy M. Ainslie
- Division of Pharmacoengineering & Molecular Pharmaceutics, Eshelman School of Pharmacy, UNC, Chapel Hill, NC, USA
- Department of Biomedical Engineering, NC State/UNC, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, School of Medicine, UNC, Chapel Hill, NC, USA
| |
Collapse
|
48
|
Vander Straeten A, Sarmadi M, Daristotle JL, Kanelli M, Tostanoski LH, Collins J, Pardeshi A, Han J, Varshney D, Eshaghi B, Garcia J, Forster TA, Li G, Menon N, Pyon SL, Zhang L, Jacob-Dolan C, Powers OC, Hall K, Alsaiari SK, Wolf M, Tibbitt MW, Farra R, Barouch DH, Langer R, Jaklenec A. A microneedle vaccine printer for thermostable COVID-19 mRNA vaccines. Nat Biotechnol 2024; 42:510-517. [PMID: 37095347 PMCID: PMC10593912 DOI: 10.1038/s41587-023-01774-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/30/2023] [Indexed: 04/26/2023]
Abstract
Decentralized manufacture of thermostable mRNA vaccines in a microneedle patch (MNP) format could enhance vaccine access in low-resource communities by eliminating the need for a cold chain and trained healthcare personnel. Here we describe an automated process for printing MNP Coronavirus Disease 2019 (COVID-19) mRNA vaccines in a standalone device. The vaccine ink is composed of lipid nanoparticles loaded with mRNA and a dissolvable polymer blend that was optimized for high bioactivity by screening formulations in vitro. We demonstrate that the resulting MNPs are shelf stable for at least 6 months at room temperature when assessed using a model mRNA construct. Vaccine loading efficiency and microneedle dissolution suggest that efficacious, microgram-scale doses of mRNA encapsulated in lipid nanoparticles could be delivered with a single patch. Immunizations in mice using manually produced MNPs with mRNA encoding severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein receptor-binding domain stimulate long-term immune responses similar to those of intramuscular administration.
Collapse
Affiliation(s)
- Aurélien Vander Straeten
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Morteza Sarmadi
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - John L Daristotle
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Maria Kanelli
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lisa H Tostanoski
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Joe Collins
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Apurva Pardeshi
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jooli Han
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dhruv Varshney
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Behnaz Eshaghi
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Johnny Garcia
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Timothy A Forster
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Gary Li
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nandita Menon
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sydney L Pyon
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Linzixuan Zhang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Catherine Jacob-Dolan
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Olivia C Powers
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kevin Hall
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Shahad K Alsaiari
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Morris Wolf
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | - Mark W Tibbitt
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, Zurich, Switzerland
| | | | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Robert Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Ana Jaklenec
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
49
|
Zhai J, Cote T, Chen Y. Challenges and advances of the stability of mRNA delivery therapeutics. NUCLEIC ACID INSIGHTS 2024; 1:101-113. [PMID: 38903876 PMCID: PMC11189690 DOI: 10.18609/nai.2024.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
mRNA therapeutics have garnered significant attention in the biomedical realm, showing immense potential across a spectrum of applications from COVID-19 to cancer treatments. Their ability to trigger precise protein expression, particularly in genome editing, is pivotal in minimizing off-target effects. At the core of mRNA therapy lies a dual-component system, comprising the mRNA itself and a delivery vehicle. The breakthrough success of novel COVID-19 vaccines has catapulted lipid nanoparticles to prominence as the preferred delivery vehicle. However, despite their US FDA approval and efficacy, lipid nanoparticles face a significant challenge: poor stability at room temperature, which limits their applications in various geographic regions with disparities in infrastructure and technology. This review aims to dissect the issue of stability inherent in lipid nanoparticles and other mRNA delivery platforms such as polymer-based materials and protein derivative materials. We herein endeavor to unravel the factors contributing to their instability and explore potential strategies to enhance their stability. By doing so, we provide a comprehensive analysis of the current landscape of mRNA delivery systems, highlighting both their successes and limitations, and paving the way for future advancements in this rapidly evolving field.
Collapse
Affiliation(s)
- Jin Zhai
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Trystin Cote
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Yupeng Chen
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
50
|
Jiang AY, Witten J, Raji IO, Eweje F, MacIsaac C, Meng S, Oladimeji FA, Hu Y, Manan RS, Langer R, Anderson DG. Combinatorial development of nebulized mRNA delivery formulations for the lungs. NATURE NANOTECHNOLOGY 2024; 19:364-375. [PMID: 37985700 PMCID: PMC10954414 DOI: 10.1038/s41565-023-01548-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 10/16/2023] [Indexed: 11/22/2023]
Abstract
Inhaled delivery of mRNA has the potential to treat a wide variety of diseases. However, nebulized mRNA lipid nanoparticles (LNPs) face several unique challenges including stability during nebulization and penetration through both cellular and extracellular barriers. Here we develop a combinatorial approach addressing these barriers. First, we observe that LNP formulations can be stabilized to resist nebulization-induced aggregation by altering the nebulization buffer to increase the LNP charge during nebulization, and by the addition of a branched polymeric excipient. Next, we synthesize a combinatorial library of ionizable, degradable lipids using reductive amination, and evaluate their delivery potential using fully differentiated air-liquid interface cultured primary lung epithelial cells. The final combination of ionizable lipid, charge-stabilized formulation and stability-enhancing excipient yields a significant improvement in lung mRNA delivery over current state-of-the-art LNPs and polymeric nanoparticles.
Collapse
Affiliation(s)
- Allen Y Jiang
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jacob Witten
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Idris O Raji
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA
| | - Feyisayo Eweje
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard/MIT MD-PhD Program, Boston, MA, USA
| | - Corina MacIsaac
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sabrina Meng
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Favour A Oladimeji
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yizong Hu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rajith S Manan
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Daniel G Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Anesthesiology, Boston Children's Hospital, Boston, MA, USA.
- Harvard and MIT Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|