1
|
van Kammen C, van Hove H, Kapsokalyvas D, Greupink R, Schiffelers R, Lely T, Terstappen F. Targeted lipid nanoparticles to prevent trans-placental passage in the ex vivo human placental cotyledon perfusion model. Drug Deliv Transl Res 2025; 15:1985-1993. [PMID: 39402393 PMCID: PMC12037421 DOI: 10.1007/s13346-024-01715-6] [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] [Accepted: 09/19/2024] [Indexed: 04/29/2025]
Abstract
Medication use during pregnancy poses risks to both the mother and the fetus. These risks include an elevated potential for fetotoxicity due to placental drug transport. Nanomedicines offer a promising solution by potentially preventing trans-placental passage. Targeted nanomedicines could enhance safety and efficacy in treating maternal or placental pathophysiology. Our study investigates placental transfer kinetics of targeted lipid nanoparticles (LNPs) in an ex vivo human placenta cotyledon perfusion model. We collected human placentas for dual-side ex vivo placental perfusions. Targeted LNPs with a fluorescence tag were introduced into the maternal circuit of each placenta. To establish if there was trans-placental passage of LNPs to the fetal circuit, we collected samples from maternal and fetal circuits throughout the six hours of the perfusion. We determined the fluorescence signal using a multi-mode microplate reader and Multiphoton microscopy to localize the LNPs in the placenta tissue. Data from perfused placenta tissue showed no significant transfer of the fluorescently labeled LNPs across the placental barrier to the fetal circuit. Localization of targeted LNPs in tissue samples is mainly observed in the maternal blood space of the placenta. Our results suggest that targeted LNPs present a promising strategic approach to prevent trans-placental passage to the fetus. Our future perspectives involve investigating the efficacy of targeted LNPs as well as loading targeted LNPs with nucleic acid-based therapeutics to investigate their therapeutic potential.
Collapse
Affiliation(s)
- Caren van Kammen
- Department of Nanomedicine, LAB CDL Research, UMC Utrecht, Utrecht, The Netherlands.
| | - Hedwig van Hove
- Department of Pharmacy, Division of Pharmacology and Toxicology, Radboud UMC, Nijmegen, The Netherlands
| | - Dimitrios Kapsokalyvas
- Department of Genetics and Cell Biology, Maastricht University, Maastricht, The Netherlands
- Interdisciplinary Centre for Clinical Research IZKF, University Hospital RWTH Aachen, Aachen, Germany
| | - Rick Greupink
- Department of Pharmacy, Division of Pharmacology and Toxicology, Radboud UMC, Nijmegen, The Netherlands
| | - Raymond Schiffelers
- Department of Nanomedicine, LAB CDL Research, UMC Utrecht, Utrecht, The Netherlands
| | - Titia Lely
- Department of Obstetrics, Wilhemina Children's Hospital, UMC Utrecht, Utrecht, The Netherlands
| | - Fieke Terstappen
- Department of Obstetrics, Wilhemina Children's Hospital, UMC Utrecht, Utrecht, The Netherlands
- Department of Neonatology, Wilhemina Children's Hospital, UMC Utrecht, Utrecht, The Netherlands
| |
Collapse
|
2
|
Bae Y, Lee H, Lee JH, Yeo S, Mok H. Histidine Decapeptide-Incorporating Lipid Nanoparticles with Low Ionizable Lipids Proportion for Efficient Small Interfering RNA Delivery. Macromol Biosci 2025:e70005. [PMID: 40366254 DOI: 10.1002/mabi.202500165] [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: 03/08/2025] [Revised: 04/22/2025] [Indexed: 05/15/2025]
Abstract
The diversification of lipid compositions in lipid nanoparticles (LNPs) is crucial for expanding their clinical applications and overcoming current limitations. In this study, LNPs with varying lipid compositions are fabricated using three different mixing processes (pipette, vortex, and microfluidic mixing) for small interfering RNA (siRNA) delivery. While both siRNA and hydrophobic fluorescent dye are successfully incorporated within LNPs using pipette- and vortex-mixing, hydrophilic peptides cannot be encapsulated. Following optimization of ionizable lipid proportion via cost-efficient vortex-mixing method, LNPs with a lower ionizable lipid proportion (27.72%), termed LNP5, are selected and fabricated with histidine decapeptide (His10) during formulation via microfluidic mixing method to supplement the function of approximately half of the ionizable lipids by simple addition of His10. His10- incorporated LNP5 (LNP5H) exhibited a 1.6-fold increase in gene silencing efficiency, compared to conventional LNPs (cLNPs; ionizable lipid proportion of 47.95%). Furthermore, LNP5H maintained siRNA potency for 4 weeks when stored in a 1% sucrose solution at -70 °C. Taken together, it fabricates potent LNP5H with low proportion of ionizable lipids via fast and easy processes, which can be applied to a variety of siRNA therapeutics for their efficient intracellular delivery.
Collapse
Affiliation(s)
- Yeonho Bae
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Hyeondo Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Jun Hyuk Lee
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Sangho Yeo
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Hyejung Mok
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, 143-701, Republic of Korea
| |
Collapse
|
3
|
Lambart I, Zaryouh H, Audenaerde JV, Liu D, Quatannens D, Lion E, Schiller S, Geissler S, Smits E, Mäder K. Apolipoprotein E4 facilitates transfection of human monocyte-derived dendritic cells by lipid nanoparticles. Int J Pharm 2025:125720. [PMID: 40360096 DOI: 10.1016/j.ijpharm.2025.125720] [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: 03/06/2025] [Revised: 04/21/2025] [Accepted: 05/10/2025] [Indexed: 05/15/2025]
Abstract
The use of mRNA as a therapeutic drug class is a safe and fast alternative to viral vector or plasmid DNA therapies. Nevertheless, free mRNA will be rapidly degraded after administration to the body and only reach the cytosol of desired cells with difficulty. Lipid nanoparticles (LNP) safely deliver mRNA to cells of interest and can be used in the treatment of different diseases. Dendritic cells are the primary antigen-presenting cells and important for mRNA vaccine delivery. Efforts to increase LNP transfection of these cells are necessary and can be achieved by different approaches. Here, we present apolipoprotein E4 addition to LNP administration as one mean of increasing LNP-mediated eGFP mRNA delivery to dendritic cells. We also show some steps in the preparation method for LNP optimization using MS2 RNA as a novel model nucleic acid.
Collapse
Affiliation(s)
- Izabella Lambart
- Merck Healthcare KGaA, Global Drug Product Development, Orals Development, Darmstadt, Germany; Martin Luther University Halle-Wittenberg, Institute of Pharmacy, Faculty I of Natural Sciences, Halle/Saale, Germany.
| | - Hannah Zaryouh
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.
| | - Jonas Van Audenaerde
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.
| | - Dana Liu
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.
| | - Delphine Quatannens
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.
| | - Eva Lion
- Laboratory of Experimental Hematology (LEH), Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, Edegem, Belgium
| | - Stefan Schiller
- Merck Healthcare KGaA, Global Drug Product Development, Orals Development, Darmstadt, Germany.
| | - Simon Geissler
- Merck Healthcare KGaA, Global Drug Product Development, Orals Development, Darmstadt, Germany.
| | - Evelien Smits
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), Faculty of Medicine and Health Sciences, University of Antwerp, Wilrijk, Belgium.
| | - Karsten Mäder
- Martin Luther University Halle-Wittenberg, Institute of Pharmacy, Faculty I of Natural Sciences, Halle/Saale, Germany.
| |
Collapse
|
4
|
Misra B, Hughes KA, Pentz WH, Surface M, Geldenhuys WJ, Bobbala S. TLR7-Adjuvanted Ionizable Lipid Nanoparticles for mRNA Vaccine Delivery. AAPS J 2025; 27:80. [PMID: 40281311 DOI: 10.1208/s12248-025-01073-2] [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: 01/03/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
Ionizable lipid nanoparticles (LNPs) are clinically relevant non-viral vectors that allow intracellular delivery of mRNA vaccines to immune cells. To fight against notorious pathogens and cancer, mRNA vaccines necessitate the addition of an adjuvant to induce strong and durable cell-mediated immune responses. Adjuvants that stimulate Toll-like receptor 7 (TLR7) induce the secretion of type I interferons and proinflammatory cytokines, vital for generating strong immune responses. However, the intracellular delivery of TLR7 adjuvants to precisely stimulate the endosomal TLR7 receptor remains a huge challenge. This issue can be addressed by exploiting ionizable LNP platforms, which can encapsulate and carry mRNA vaccines and small molecule hydrophobic adjuvants to immune cells. CL347 is a potent lipid-based adjuvant that selectively stimulates the TLR7 receptor. In this study, we developed ionizable LNPs incorporating SM102 and CL347 adjuvant as the ionizable lipid and TLR7 adjuvant, respectively. CL347-SM102 LNPs exhibited particle sizes of less than 150 nm with spherical morphology and mRNA encapsulation efficiency of greater than 95%. In vivo studies showed a two-fold increase in IFN-γ producing CD4 and CD8 T cells in the lymphoid organs of the mice immunized with adjuvanted LNPs compared to the non-adjuvanted LNPs. Human PBMCs treated with adjuvanted LNPs exhibited significantly higher CD40 expression and pro-inflammatory cytokine (IL-6 and IFN-γ) secretion than non-adjuvanted LNPs. Together, these results suggest the potential of ionizable LNPs as a platform for concurrent delivery of mRNA and adjuvants for prophylactic and therapeutic vaccine applications.
Collapse
Affiliation(s)
- Bishal Misra
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - Krystal A Hughes
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - William H Pentz
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, 26506, USA
- School of Medicine, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - Morgan Surface
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - Werner J Geldenhuys
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, 26506, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, West Virginia, 26506, USA.
| |
Collapse
|
5
|
Kim H, Simpson GG, Fei F, Garris C, Weissleder R. Fluorinated Ribonucleocarbohydrate Nanoparticles Allow Ultraefficient mRNA Delivery and Protein Expression in Tumor-Associated Myeloid Cells. J Am Chem Soc 2025; 147:11766-11776. [PMID: 40135499 PMCID: PMC11987029 DOI: 10.1021/jacs.4c14474] [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: 10/16/2024] [Revised: 02/02/2025] [Accepted: 02/05/2025] [Indexed: 03/27/2025]
Abstract
Ribonucleic acids (RNA) are commonly formulated into lipid nanoparticles (LNP) for in vivo use, but challenges exist with systemic delivery and low in vivo expression efficiency. Inspired by ribonucleoprotein complexes in cells, we created synthetic ribonucleocarbohydrate (RNC) complexes based on cyclodextrin nanoparticles with ferrocenyl fluorocarbons capable of carrying mRNA and additional small-molecule drug payloads, facilitating lysosomal escape and immune stimulation all in the same nanoparticle. We show that this strategy results in highly efficient myeloid cell targeting (dendritic cells and MHC expressing macrophages) and protein expression following systemic administration. The RNC platform should have broad applications in vaccine development, immunosuppression, and immunostimulation for various diseases.
Collapse
Affiliation(s)
- Hyung
Shik Kim
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN, 5206, Boston, Massachusetts 02114, United States
| | - Grant Gerald Simpson
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN, 5206, Boston, Massachusetts 02114, United States
| | - Fan Fei
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN, 5206, Boston, Massachusetts 02114, United States
| | - Christopher Garris
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN, 5206, Boston, Massachusetts 02114, United States
- Department
of Pathology, Massachusetts General Hospital, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Ralph Weissleder
- Center
for Systems Biology, Massachusetts General
Hospital, 185 Cambridge Street, CPZN, 5206, Boston, Massachusetts 02114, United States
- Department
of Systems Biology, Harvard Medical School, 200 Longwood Avenue, Boston, Massachusetts 02115, United States
| |
Collapse
|
6
|
Szebeni J. Expanded Spectrum and Increased Incidence of Adverse Events Linked to COVID-19 Genetic Vaccines: New Concepts on Prophylactic Immuno-Gene Therapy, Iatrogenic Orphan Disease, and Platform-Inherent Challenges. Pharmaceutics 2025; 17:450. [PMID: 40284445 PMCID: PMC12029998 DOI: 10.3390/pharmaceutics17040450] [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: 02/12/2025] [Revised: 03/17/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
The mRNA- and DNA-based "genetic" COVID-19 vaccines can induce a broad range of adverse events (AEs), with statistics showing significant variation depending on the timing and data analysis methods used. Focusing only on lipid nanoparticle-enclosed mRNA (mRNA-LNP) vaccines, this review traces the evolution of statistical conclusions on the prevalence of AEs and incidents associated with these vaccines, from initial underestimation of atypical, severe toxicities to recent claims suggesting the possible contribution of COVID-19 vaccinations to the excess deaths observed in many countries over the past few years. Among hundreds of different AEs listed in Pfizer's pharmacovigilance survey, the present analysis categorizes the main symptoms according to organ systems, with nearly all of them being affected. Using data from the US Vaccine Adverse Event Reporting System and a global vaccination dataset, a comparison of the prevalence and incidence rates of AEs induced by genetic versus flu vaccines revealed an average 26-fold increase in AEs with the use of genetic vaccines. The difference is especially pronounced in the case of severe 'Brighton-listed' AEs, which are also observed in COVID-19 and post-COVID conditions. Among these, the increases in incidence rates relative to flu vaccines, given as x-fold rises, were 1152x, 455x, 226x, 218x, 162x, 152x, and 131x for myocarditis, thrombosis, death, myocardial infarction, tachycardia, dyspnea, and hypertension, respectively. The review delineates the concept that genetic vaccines can be regarded as prophylactic immuno-gene therapies and that the observed chronic disabling AEs might be categorized as iatrogenic orphan diseases. It also examines the unique vaccine characteristics that could be causally related to abnormal immune responses which potentially lead to adverse events and complications. These new insights may contribute to improving the safety of this platform technology and assessing the risk/benefit balance of various products.
Collapse
Affiliation(s)
- Janos Szebeni
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, 1089 Budapest, Hungary;
- SeroScience LCC, 1125 Budapest, Hungary
- Translational Nanobioscience Research Center, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
7
|
Lindsay S, Hussain M, Binici B, Perrie Y. Exploring the Challenges of Lipid Nanoparticle Development: The In Vitro-In Vivo Correlation Gap. Vaccines (Basel) 2025; 13:339. [PMID: 40333261 PMCID: PMC12031360 DOI: 10.3390/vaccines13040339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND/OBJECTIVES The development of lipid nanoparticles (LNPs) as delivery platforms for nucleic acids has revolutionised possibilities for both therapeutic and vaccine applications. However, emerging studies highlight challenges in achieving reliable in vitro-in vivo correlation (IVIVC), which delays the translation of experimental findings into clinical applications. This study investigates these potential discrepancies by evaluating the physicochemical properties, in vitro efficacy (across three commonly used cell lines), and in vivo performance (mRNA expression and vaccine efficacy) of four LNP formulations. METHODS LNPs composed of DSPC, cholesterol, a PEGylated lipid, and one of four ionizable lipids (SM-102, ALC-0315, MC3, or C12-200) were manufactured using microfluidics. RESULTS All formulations exhibited comparable physicochemical properties, as expected (size 70-100 nm, low PDI, near-neutral zeta potential, and high mRNA encapsulation). In vitro studies demonstrated variable LNP-mediated mRNA expression in both immortalised and immune cells, with SM-102 inducing significantly higher protein expression (p < 0.05) than the other formulations in immortalised and immune cells. However, in vivo results revealed that ALC-0315 and SM-102-based LNPs achieved significantly (p < 0.05) higher protein expression without a significant difference between them, while MC3- and C12-200-based LNPs exhibited lower expression levels. As vaccine formulations, all LNPs elicited strong immune responses with no significant differences among them. CONCLUSIONS These findings highlight the complexities of correlating in vitro and in vivo outcomes in LNP development and demonstrate the importance of holistic evaluation strategies to optimise their clinical translation.
Collapse
Affiliation(s)
| | | | | | - Yvonne Perrie
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0RE, UK; (S.L.); (M.H.); (B.B.)
| |
Collapse
|
8
|
DeFranciscis V, Amabile G, Kortylewski M. Clinical applications of oligonucleotides for cancer therapy. Mol Ther 2025:S1525-0016(25)00172-8. [PMID: 40045578 DOI: 10.1016/j.ymthe.2025.02.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/17/2025] Open
Abstract
Oligonucleotide therapeutics (ONTs) represent a rapidly evolving modality for cancer treatment, capitalizing on their ability to modulate gene expression with high specificity. With more than 20 nucleic acid-based therapies that gained regulatory approval, advances in chemical modifications, sequence optimization, and novel delivery systems have propelled ONTs from research tools to clinical realities. ONTs, including siRNAs, antisense oligonucleotides, saRNA, miRNA, aptamers, and decoys, offer promising solutions for targeting previously "undruggable" molecules, such as transcription factors, and enhancing cancer immunotherapy by overcoming tumor immune evasion. The promise of ONT application in cancer treatment is exemplified by the recent FDA approval of the first oligonucleotide-based treatment to myeloproliferative disease. At the same time, there are challenges in delivering ONTs to specific tissues, mitigating off-target effects, and improving cellular uptake and endosomal release. This review provides a comprehensive overview of ONTs in clinical trials, emerging delivery strategies, and innovative therapeutic approaches, emphasizing the role of ONTs in immunotherapy and addressing hurdles that hinder their clinical translation. By examining advances and remaining challenges, we highlight opportunities for ONTs to revolutionize oncology and enhance patient outcomes.
Collapse
Affiliation(s)
- Vittorio DeFranciscis
- National Research Council, Institute of Genetic and Biomedical Research, Milan, Italy
| | | | - Marcin Kortylewski
- Department of Immuno-Oncology, Beckman Research Institute at City of Hope National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
9
|
Somu Naidu G, Rampado R, Sharma P, Ezra A, Kundoor GR, Breier D, Peer D. Ionizable Lipids with Optimized Linkers Enable Lung-Specific, Lipid Nanoparticle-Mediated mRNA Delivery for Treatment of Metastatic Lung Tumors. ACS NANO 2025; 19:6571-6587. [PMID: 39912611 PMCID: PMC11841047 DOI: 10.1021/acsnano.4c18636] [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: 12/23/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
Lipid nanoparticles (LNPs) have emerged as a groundbreaking delivery system for vaccines and therapeutic mRNAs. Ionizable lipids are the most pivotal component of LNPs due to their ability to electrostatically interact with mRNA, allowing its encapsulation while concurrently enabling its endosomal escape following cellular internalization. Thus, extensive research has been performed to optimize the ionizable lipid structure and to develop formulations that are well tolerated and allow efficient targeting of different organs that result in a high and sustained mRNA expression. However, one facet of the ionizable lipids' structure has been mostly overlooked: the linker segment between the ionizable headgroup and their tails. Here, we screened a rationally designed library of ionizable lipids with different biodegradable linkers. We extensively characterized LNPs formulated using these ionizable lipids and elucidated how these minor structural changes in the ionizable lipids structure radically influenced the LNPs' biodistribution in vivo. We showed how the use of amide and urea linkers can modulate the LNPs' pKa, resulting in an improved specificity for lung transfection. Finally, we demonstrated how one of these lipids (lipid 35) that form LNPs entrapping a bacterial toxin [pseudomonas exotoxin A (mmPE)] in the form of an mRNA reduced tumor burden and significantly increased the survival of mice with lung metastasis.
Collapse
Affiliation(s)
- Gonna Somu Naidu
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Riccardo Rampado
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Pharmaceutical Sciences, University of
Padova, Padova 35131, Italy
| | - Preeti Sharma
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Assaf Ezra
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Govinda Reddy Kundoor
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Dor Breier
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Dan Peer
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| |
Collapse
|
10
|
Koren L, Koren A, Likić R, Katanec T. Revolutionizing Dentistry: Preclinical Insights and Future Applications of mRNA Vaccines in Dentistry-A Narrative Review. Dent J (Basel) 2025; 13:79. [PMID: 39996953 PMCID: PMC11854559 DOI: 10.3390/dj13020079] [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: 11/27/2024] [Revised: 01/16/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Recent advances in mRNA vaccine technology, accelerated by the global COVID-19 pandemic, have generated significant interest in their applications beyond infectious diseases. Dentistry has emerged as a promising field for exploring the potential of mRNA-based therapies in preventing and treating oral diseases. Objectives: This narrative review aims to evaluate the current status of mRNA vaccine development and its preclinical applications in oral health, focusing on periodontal disease, dental caries, regenerative medicine, implantology, and oral cancer. Methods: The review synthesizes findings from preclinical studies, including research conducted in animal models and in vitro, to assess the potential of mRNA-based therapies to modulate immune responses and promote tissue regeneration in the oral cavity. Clinical trials were only mentioned in the context of broader areas of mRNA vaccine implementation such as oncology and immunotherapy. Results: The preclinical studies highlight the capacity of mRNA vaccines to enhance the body's immune response and facilitate tissue repair processes. Despite these promising results, challenges persist in delivering mRNA vaccines effectively within the complex oral environment. These challenges include vaccine stability, delivery mechanisms, and the modulation of immune responses. Conclusions: While mRNA vaccines offer significant promise for revolutionizing oral health care, they face notable limitations concerning safety, efficacy, and clinical feasibility. Overcoming these obstacles through further research is essential to unlock their full translational potential and ensure their safe and effective integration into dental practice.
Collapse
Affiliation(s)
- Luciana Koren
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.K.); (A.K.)
| | - Andro Koren
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia; (L.K.); (A.K.)
| | - Robert Likić
- Unit for Clinical Pharmacology, Department of Internal Medicine, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia;
| | - Tomislav Katanec
- Department of Oral Surgery, School of Dental Medicine Zagreb, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia
| |
Collapse
|
11
|
Hofbauer SI, Fink LA, Young RE, Vijayakumar T, Nelson KM, Bellopede N, Alameh MG, Weissman D, Gleghorn JP, Riley RS. Cytokine mRNA Delivery and Local Immunomodulation in the Placenta using Lipid Nanoparticles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.637086. [PMID: 39974923 PMCID: PMC11839073 DOI: 10.1101/2025.02.07.637086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
During pregnancy, the maternal immune system adapts to balance tolerance of the semi-allogenic fetus while protecting the fetus from pathogens. Dysregulated immune activity at the maternal-fetal interface contributes to pregnancy complications, such as recurrent pregnancy loss and preeclampsia. Compared to healthy placentas, preeclamptic placentas exhibit increased pro-inflammatory signaling, including a predominance of inflammatory macrophages, leading to impaired tissue remodeling and restricted blood flow. However, the precise mechanisms driving this immune imbalance remain poorly understood, in part due to the lack of tools to probe individual pathways. Here, we use lipid nanoparticles (LNPs) to deliver cytokine-encoded mRNA to placental cells, called trophoblasts, enabling local immunomodulation. LNP-mediated delivery of IL-4 and IL-13 mRNA induced cytokine secretion by trophoblasts, leading to polarization of primary human monocytes toward anti-inflammatory phenotypes. Notably, lowering the mRNA dose increased expression of alternatively-activated macrophage markers, revealing an inverse relationship between dose and polarization status. Intravenous injection of LNPs in pregnant mice achieved placental secretion of IL-4 and IL-13 with minimal changes to pro-inflammatory cytokines in the serum. These findings establish LNPs as a tool for local immunomodulation in the placenta, offering a strategy to study and treat immune dysfunction in pregnancy and in other inflammatory conditions.
Collapse
|
12
|
Mok CKP, Tang YS, Tan CW, Chong KC, Chen C, Sun Y, Yiu K, Ling KC, Chan KK, Hui DS. Comparison of safety and immunogenicity in the elderly after receiving either Comirnaty or Spikevax monovalent XBB1.5 COVID-19 vaccine. J Infect 2025; 90:106374. [PMID: 39657850 DOI: 10.1016/j.jinf.2024.106374] [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: 07/17/2024] [Revised: 10/13/2024] [Accepted: 12/04/2024] [Indexed: 12/12/2024]
Abstract
BACKGROUND The emergence of SARS-CoV-2 variants necessitates ongoing evaluation of vaccine performance. This study evaluates and compares the safety and immunogenicity of the Comirnaty and Spikevax monovalent XBB.1.5 COVID-19 vaccines in an elderly population. METHODS Altogether, 129 elderly individuals were recruited between 2 January and 3 February 2024, and received a booster dose of either Comirnaty (n=59) or Spikevax (n=70) monovalent XBB.1.5 COVID-19 vaccine. Blood samples were collected at before and one month after vaccination. Immunogenicity was assessed by measuring the percentage of IFNγ+CD4+ and IFNγ+CD8+ T cells, and neutralizing antibody titers (NT50) using a surrogate virus neutralization test (sVNT). Adverse reactions were recorded and analyzed. FINDINGS Both vaccines significantly increased the percentage of IFNγ+CD8+ T cells against XBB.1.5 and wild-type (WT) SARS-CoV-2 at one-month post-vaccination. Spikevax induced a significantly higher percentages of IFNγ+CD8+ and CD4+ T cells against XBB.1.5 than Comirnaty (p<0.001). The proportion of participants showing a positive T cell response to XBB1.5 after vaccination was higher in the Spikevax group (64.3% CD8, 71.4% CD4) than in the Comirnaty group (42.4% CD8, 57.6% CD4). Spikevax also elicited higher NT50 levels against XBB1.5, JN.1 and the latest variant KP.2 than Comirnaty (XBB1.5: p<0.01; KP.2: p<0.05). Fever was more common in the Spikevax group (fever: p=0.006). However, all side effects were short-term and resolved on their own. INTERPRETATION Both vaccines induce neutralizing antibody to XBB1.5, JN.1 and KP.2. Specifically, Spikevax induces higher cellular and humoral immune responses than Comirnaty in the elderly, but it is also associated with a higher incidence of fever. These findings can guide public health strategies for vaccinating the elderly population.
Collapse
Affiliation(s)
- Chris Ka Pun Mok
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong; SH Ho Research Centre for Emerging Infectious Diseases, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Yun Sang Tang
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Chee Wah Tan
- Program in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore; Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ka Chun Chong
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong; Centre for Health Systems and Policy Research, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Chunke Chen
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Yuanxin Sun
- The Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Karen Yiu
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - Kwun Cheung Ling
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - Ken Kp Chan
- Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong
| | - David S Hui
- SH Ho Research Centre for Emerging Infectious Diseases, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong; Department of Medicine & Therapeutics, The Chinese University of Hong Kong, Hong Kong.
| |
Collapse
|
13
|
Wang X, Fan R, Mu M, Zhou L, Zou B, Tong A, Guo G. Harnessing nanoengineered CAR-T cell strategies to advance solid tumor immunotherapy. Trends Cell Biol 2024:S0962-8924(24)00252-6. [PMID: 39721923 DOI: 10.1016/j.tcb.2024.11.010] [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: 08/21/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024]
Abstract
The efficacy and safety of chimeric antigen receptor (CAR) T cell therapy is still inconclusive in solid tumor treatment. Recently, nanotechnology has emerged as a potent strategy to reshape CAR-T cell therapy with promising outcomes. This review aims to discuss the significant potential of nano-engineered CAR-T cell therapy in addressing existing challenges, including CAR-T cell engineering evolution, tumor microenvironment (TME) modulation, and precise CAR-T cell therapy (precise targeting, monitoring, and activation), under the main consideration of clinical translation. It also focuses on the growing trend of technological convergence within this domain, such as mRNA therapeutics, organoids, neoantigen, and artificial intelligence. Moreover, safety management of nanomedicine is seriously emphasized to facilitate clinical translation.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China; West China School of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Rangrang Fan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Min Mu
- Department of Radiation Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liangxue Zhou
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bingwen Zou
- Department of Radiation Oncology, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Aiping Tong
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Gang Guo
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
14
|
Catenacci L, Rossi R, Sechi F, Buonocore D, Sorrenti M, Perteghella S, Peviani M, Bonferoni MC. Effect of Lipid Nanoparticle Physico-Chemical Properties and Composition on Their Interaction with the Immune System. Pharmaceutics 2024; 16:1521. [PMID: 39771501 PMCID: PMC11728546 DOI: 10.3390/pharmaceutics16121521] [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: 10/17/2024] [Revised: 11/19/2024] [Accepted: 11/24/2024] [Indexed: 01/16/2025] Open
Abstract
Lipid nanoparticles (LNPs) have shown promise as a delivery system for nucleic acid-based therapeutics, including DNA, siRNA, and mRNA vaccines. The immune system plays a critical role in the response to these nanocarriers, with innate immune cells initiating an early response and adaptive immune cells mediating a more specific reaction, sometimes leading to potential adverse effects. Recent studies have shown that the innate immune response to LNPs is mediated by Toll-like receptors (TLRs) and other pattern recognition receptors (PRRs), which recognize the lipid components of the nanoparticles. This recognition can trigger the activation of inflammatory pathways and the production of cytokines and chemokines, leading to potential adverse effects such as fever, inflammation, and pain at the injection site. On the other hand, the adaptive immune response to LNPs appears to be primarily directed against the protein encoded by the mRNA cargo, with little evidence of an ongoing adaptive immune response to the components of the LNP itself. Understanding the relationship between LNPs and the immune system is critical for the development of safe and effective nucleic acid-based delivery systems. In fact, targeting the immune system is essential to develop effective vaccines, as well as therapies against cancer or infections. There is a lack of research in the literature that has systematically studied the factors that influence the interaction between LNPs and the immune system and further research is needed to better elucidate the mechanisms underlying the immune response to LNPs. In this review, we discuss LNPs' composition, physico-chemical properties, such as size, shape, and surface charge, and the protein corona formation which can affect the reactivity of the immune system, thus providing a guide for the research on new formulations that could gain a favorable efficacy/safety profile.
Collapse
Affiliation(s)
- Laura Catenacci
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Rachele Rossi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Francesca Sechi
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Daniela Buonocore
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Milena Sorrenti
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Sara Perteghella
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| | - Marco Peviani
- Department of Biology and Biotechnology “L. Spallanzani”, University of Pavia, 27100 Pavia, Italy;
| | - Maria Cristina Bonferoni
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy; (L.C.); (R.R.); (F.S.); (M.S.); (M.C.B.)
| |
Collapse
|
15
|
Berger S, Zeyn Y, Wagner E, Bros M. New insights for the development of efficient DNA vaccines. Microb Biotechnol 2024; 17:e70053. [PMID: 39545748 PMCID: PMC11565620 DOI: 10.1111/1751-7915.70053] [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: 04/25/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Despite the great potential of DNA vaccines for a broad range of applications, ranging from prevention of infections, over treatment of autoimmune and allergic diseases to cancer immunotherapies, the implementation of such therapies for clinical treatment is far behind the expectations up to now. The main reason is the poor immunogenicity of DNA vaccines in humans. Consequently, the improvement of the performance of DNA vaccines in vivo is required. This mini-review provides an overview of the current state of DNA vaccines and the various strategies to enhance the immunogenic potential of DNA vaccines, including (i) the optimization of the DNA construct itself regarding size, nuclear transfer and transcriptional regulation; (ii) the use of appropriate adjuvants; and (iii) improved delivery, for example, by careful choice of the administration route, physical methods such as electroporation and nanomaterials that may allow cell type-specific targeting. Moreover, combining nanoformulated DNA vaccines with other immunotherapies and prime-boost strategies may help to enhance success of treatment.
Collapse
Affiliation(s)
- Simone Berger
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Yanira Zeyn
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Center for NanoScienceLudwig‐Maximilians‐Universität (LMU) MunichMunichGermany
| | - Matthias Bros
- Department of DermatologyUniversity Medical Center of the Johannes Gutenberg University (JGU) MainzMainzGermany
| |
Collapse
|
16
|
Pardi N, Krammer F. mRNA vaccines for infectious diseases - advances, challenges and opportunities. Nat Rev Drug Discov 2024; 23:838-861. [PMID: 39367276 DOI: 10.1038/s41573-024-01042-y] [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] [Accepted: 08/27/2024] [Indexed: 10/06/2024]
Abstract
The concept of mRNA-based vaccines emerged more than three decades ago. Groundbreaking discoveries and technological advancements over the past 20 years have resolved the major roadblocks that initially delayed application of this new vaccine modality. The rapid development of nucleoside-modified COVID-19 mRNA vaccines demonstrated that this immunization platform is easy to develop, has an acceptable safety profile and can be produced at a large scale. The flexibility and ease of antigen design have enabled mRNA vaccines to enter development for a wide range of viruses as well as for various bacteria and parasites. However, gaps in our knowledge limit the development of next-generation mRNA vaccines with increased potency and safety. A deeper understanding of the mechanisms of action of mRNA vaccines, application of novel technologies enabling rational antigen design, and innovative vaccine delivery strategies and vaccination regimens will likely yield potent novel vaccines against a wide range of pathogens.
Collapse
Affiliation(s)
- Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
17
|
Sharma P, Breier D, Peer D. Immunogenic amines on lipid nanoparticles. Nat Biomed Eng 2024; 8:1332-1333. [PMID: 39443742 DOI: 10.1038/s41551-024-01265-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Affiliation(s)
- Preeti Sharma
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel, Tel Aviv, Israel
| | - Dor Breier
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel, Tel Aviv, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, Shmunis School for Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel.
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel, Tel Aviv, Israel.
| |
Collapse
|
18
|
Parvin N, Mandal TK, Joo SW. The Impact of COVID-19 on RNA Therapeutics: A Surge in Lipid Nanoparticles and Alternative Delivery Systems. Pharmaceutics 2024; 16:1366. [PMID: 39598489 PMCID: PMC11597542 DOI: 10.3390/pharmaceutics16111366] [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: 10/02/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/29/2024] Open
Abstract
The COVID-19 pandemic has significantly accelerated progress in RNA-based therapeutics, particularly through the successful development and global rollout of mRNA vaccines. This review delves into the transformative impact of the pandemic on RNA therapeutics, with a strong focus on lipid nanoparticles (LNPs) as a pivotal delivery platform. LNPs have proven to be critical in enhancing the stability, bioavailability, and targeted delivery of mRNA, facilitating the unprecedented success of vaccines like those developed by Pfizer-BioNTech and Moderna. Beyond vaccines, LNP technology is being explored for broader therapeutic applications, including treatments for cancer, rare genetic disorders, and infectious diseases. This review also discusses emerging RNA delivery systems, such as polymeric nanoparticles and viral vectors, which offer alternative strategies to overcome existing challenges related to stability, immune responses, and tissue-specific targeting. Additionally, we examine the pandemic's influence on regulatory processes, including the fast-tracked approvals for RNA therapies, and the surge in research funding that has spurred further innovation in the field. Public acceptance of RNA-based treatments has also grown, laying the groundwork for future developments in personalized medicine. By providing an in-depth analysis of these advancements, this review highlights the long-term impact of COVID-19 on the evolution of RNA therapeutics and the future of precision drug delivery technologies.
Collapse
Affiliation(s)
| | - Tapas K. Mandal
- School of Mechanical Engineering, School of Basic Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Sang-Woo Joo
- School of Mechanical Engineering, School of Basic Science, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
19
|
Tang X, Ding S, Yang S, Cheng Y, Liu H, Chen K, Han X. Polysorbate 80-containing ionizable lipid nanoparticles for mRNA delivery. Biomater Sci 2024; 12:5573-5581. [PMID: 39297400 DOI: 10.1039/d4bm00523f] [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: 10/23/2024]
Abstract
Ionizable lipid nanoparticles have demonstrated remarkable success as mRNA vaccine carriers and represent one of the most promising gene drug delivery vehicles. However, polyethylene glycol (PEG), one of the major components, can cause immunogenic reactions, anaphylaxis and increased blood clearance, leading to toxic side effects and reduced efficacy. In this study, we utilize polysorbate 80 (PS80) as a PEG alternative in formulating eGFP mRNA-loaded ionizable lipid nanoparticles (PS80-iLNPs), aiming to enhance stealth properties, uptake efficiency, and biosafety. Our findings revealed that PS80-iLNPs enhanced the stealthiness and resistance to serum interference. Compared to PEG-containing ionizable lipid nanoparticles (PEG-iLNPs), PS80-iLNPs showed a 1.14-fold increase in stealthiness. Moreover, at a total lipid concentration of 50 μg mL-1, PS80-iLNPs exhibited 1.12 times higher cell viability compared to PEG-iLNPs. Notably, under serum interference, PEG-iLNPs showed a 44.97% uptake reduction, whereas PS80-iLNPs exhibited a modest 30.55% decrease, underscoring its superior serum resistance. This work demonstrated that PS80 could serve as a suitable substitute for PEG, thus signifying an excellent basis for the development of PEG-free ionizable lipid nanoparticles.
Collapse
Affiliation(s)
- Xuefeng Tang
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| | - Shixiao Ding
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| | - Shilin Yang
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| | - Yuqiao Cheng
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| | - Hanyu Liu
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| | - Kexin Chen
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| | - Xiaojun Han
- State Key Laboratory of Urban Water Resource and Environment, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150001, China.
| |
Collapse
|
20
|
Tarab-Ravski D, Stotsky-Oterin L, Elisha A, Kundoor GR, Ramishetti S, Hazan-Halevy I, Haas H, Peer D. The future of genetic medicines delivered via targeted lipid nanoparticles to leukocytes. J Control Release 2024; 376:286-302. [PMID: 39401676 DOI: 10.1016/j.jconrel.2024.10.014] [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: 07/05/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024]
Abstract
Genetic medicines hold vast therapeutic potential, offering the ability to silence or induce gene expression, knock out genes, and even edit DNA fragments. Applying these therapeutic modalities to leukocytes offers a promising path for treating various conditions yet overcoming the obstacles of specific and efficient delivery to leukocytes remains a major bottleneck in their clinical translation. Lipid nanoparticles (LNPs) have emerged as the leading delivery system for nucleic acids due to their remarkable versatility and ability to improve their in vivo stability, pharmacokinetics, and therapeutic benefits. Equipping LNPs with targeting moieties can promote their specific cellular uptake and internalization to leukocytes, making targeted LNPs (tLNPs) an inseparable part of developing leukocyte-targeted gene therapy. However, despite the significant advancements in research, genetic medicines for leukocytes using targeted delivery approaches have not been translated into the clinic yet. Herein, we discuss the important aspects of designing tLNPs and highlight the considerations for choosing an appropriate bioconjugation strategy and targeting moiety. Furthermore, we provide our insights on limiting challenges and identify key areas for further research to advance these exciting therapies for patient care.
Collapse
Affiliation(s)
- Dana Tarab-Ravski
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Lior Stotsky-Oterin
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Aviad Elisha
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Govinda Reddy Kundoor
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | | | - Inbal Hazan-Halevy
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Heinrich Haas
- NeoVac Ltd. 127 Olympic Ave., OX14 4SA, Milton Park, Oxfordshire, UK; Department of Biopharmaceutics and Pharmaceutical Technology, Johannes Gutenberg-University, Mainz, Germany
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise, Faculty of Life Science, Tel Aviv University, Tel Aviv, Israel; Department of Materials Science and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
21
|
Whitehead AJ, Woodring T, Klein BS. Immunity to fungi and vaccine considerations. Cell Host Microbe 2024; 32:1681-1690. [PMID: 39389032 PMCID: PMC11980782 DOI: 10.1016/j.chom.2024.09.011] [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: 08/23/2024] [Accepted: 09/16/2024] [Indexed: 10/12/2024]
Abstract
Fungal disease poses a growing threat to public health that our current antifungal therapies are not well equipped to meet. As the population of immunocompromised hosts expands, and ecological changes favor the emergence of fungal pathogens, the development of new antifungal agents, including vaccines, becomes a global priority. Here, we summarize recent advancements in the understanding of fungal pathogenesis, key features of the host antifungal immune response, and how these findings could be leveraged to design novel approaches to deadly fungal disease.
Collapse
Affiliation(s)
- Alexander J Whitehead
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Therese Woodring
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Bruce S Klein
- Department of Pediatrics, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Medicine, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
22
|
Li J, Foged C. Evaluating the breadth of nucleic acid-based payloads delivered in lipid nanoparticles to establish fundamental differences in development. Expert Opin Drug Deliv 2024; 21:1441-1461. [PMID: 39387233 DOI: 10.1080/17425247.2024.2409142] [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: 03/19/2024] [Revised: 08/29/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
INTRODUCTION Nucleic acid (NA)-based therapeutics have shown great potential for downregulating or augmenting gene expression, and for promising applications, e.g., protein-replacement therapy and vaccination, a comprehensive understanding of the requirements for their targeted delivery to specific tissues or cells is needed. AREAS COVERED In this review, we discuss clinical applications of four representative types of NA-based therapeutics, i.e. antisense oligonucleotides, small interfering RNA, messenger RNA, and circular RNA, with a focus on the lipid nanoparticle (LNP) technology used for intracellular delivery. The in vivo fate of LNPs is discussed to improve the understanding of trafficking of nanomedicines at the systemic and cellular levels. In addition, NA-based vaccines are discussed, focusing on targeting antigen-presenting cells and immune activation. EXPERT OPINION Optimization of delivery systems for NA-based therapeutics is mainly focused on the standard requirements of prolonged systemic circulation and enhancing endosomal escape. Depending on the final destination in specific target tissues or cells, strategies should be adjusted to achieve the desired biodistribution of NA-based payloads. More studies relating to the pharmacokinetics of both cargo and carrier are encouraged, because their in vivo fates may differ, considering the possibility of premature cargo release before reaching the target.
Collapse
Affiliation(s)
- Jinjin Li
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Ø, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen Ø, Denmark
| |
Collapse
|
23
|
Tapescu I, Madsen PJ, Lowenstein PR, Castro MG, Bagley SJ, Fan Y, Brem S. The transformative potential of mRNA vaccines for glioblastoma and human cancer: technological advances and translation to clinical trials. Front Oncol 2024; 14:1454370. [PMID: 39399167 PMCID: PMC11466887 DOI: 10.3389/fonc.2024.1454370] [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: 06/25/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Originally devised for cancer control, mRNA vaccines have risen to the forefront of medicine as effective instruments for control of infectious disease, notably their pivotal role in combating the COVID-19 pandemic. This review focuses on fundamental aspects of the development of mRNA vaccines, e.g., tumor antigens, vector design, and precise delivery methodologies, - highlighting key technological advances. The recent, promising success of personalized mRNA vaccines against pancreatic cancer and melanoma illustrates the potential value for other intractable, immunologically resistant, solid tumors, such as glioblastoma, as well as the potential for synergies with a combinatorial, immunotherapeutic approach. The impact and progress in human cancer, including pancreatic cancer, head and neck cancer, bladder cancer are reviewed, as are lessons learned from first-in-human CAR-T cell, DNA and dendritic cell vaccines targeting glioblastoma. Going forward, a roadmap is provided for the transformative potential of mRNA vaccines to advance cancer immunotherapy, with a particular focus on the opportunities and challenges of glioblastoma. The current landscape of glioblastoma immunotherapy and gene therapy is reviewed with an eye to combinatorial approaches harnessing RNA science. Preliminary preclinical and clinical data supports the concept that mRNA vaccines could be a viable, novel approach to prolong survival in patients with glioblastoma.
Collapse
Affiliation(s)
- Iulia Tapescu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Peter J. Madsen
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Neurosurgery, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, United States
| | - Pedro R. Lowenstein
- Department of Neurosurgery, The University of Michigan, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, United States
- Department of Biomedical Engineering, The University of Michigan, Ann Arbor, MI, United States
| | - Maria G. Castro
- Department of Neurosurgery, The University of Michigan, Ann Arbor, MI, United States
- Department of Cell and Developmental Biology, The University of Michigan, Ann Arbor, MI, United States
| | - Stephen J. Bagley
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| | - Yi Fan
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, United States
| | - Steven Brem
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Neurosurgery, University of Pennsylvania, Philadelphia, PA, United States
- Glioblastoma Translational Center of Excellence, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
24
|
Lu RM, Hsu HE, Perez SJLP, Kumari M, Chen GH, Hong MH, Lin YS, Liu CH, Ko SH, Concio CAP, Su YJ, Chang YH, Li WS, Wu HC. Current landscape of mRNA technologies and delivery systems for new modality therapeutics. J Biomed Sci 2024; 31:89. [PMID: 39256822 PMCID: PMC11389359 DOI: 10.1186/s12929-024-01080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 08/20/2024] [Indexed: 09/12/2024] Open
Abstract
Realizing the immense clinical potential of mRNA-based drugs will require continued development of methods to safely deliver the bioactive agents with high efficiency and without triggering side effects. In this regard, lipid nanoparticles have been successfully utilized to improve mRNA delivery and protect the cargo from extracellular degradation. Encapsulation in lipid nanoparticles was an essential factor in the successful clinical application of mRNA vaccines, which conclusively demonstrated the technology's potential to yield approved medicines. In this review, we begin by describing current advances in mRNA modifications, design of novel lipids and development of lipid nanoparticle components for mRNA-based drugs. Then, we summarize key points pertaining to preclinical and clinical development of mRNA therapeutics. Finally, we cover topics related to targeted delivery systems, including endosomal escape and targeting of immune cells, tumors and organs for use with mRNA vaccines and new treatment modalities for human diseases.
Collapse
Affiliation(s)
- Ruei-Min Lu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Hsiang-En Hsu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | | | - Monika Kumari
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan
| | - Guan-Hong Chen
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Ming-Hsiang Hong
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Yin-Shiou Lin
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Ching-Hang Liu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Shih-Han Ko
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | | | - Yi-Jen Su
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan
| | - Yi-Han Chang
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Wen-Shan Li
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan.
- Institute of Chemistry, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan.
| | - Han-Chung Wu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan.
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan.
| |
Collapse
|
25
|
Fan Y, Rigas D, Kim LJ, Chang FP, Zang N, McKee K, Kemball CC, Yu Z, Winkler P, Su WC, Jessen P, Hura GL, Chen T, Koenig SG, Nagapudi K, Leung D, Yen CW. Physicochemical and structural insights into lyophilized mRNA-LNP from lyoprotectant and buffer screenings. J Control Release 2024; 373:727-737. [PMID: 39059500 DOI: 10.1016/j.jconrel.2024.07.052] [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: 06/06/2024] [Revised: 07/21/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
The surge in RNA therapeutics has revolutionized treatments for infectious diseases like COVID-19 and shows the potential to expand into other therapeutic areas. However, the typical requirement for ultra-cold storage of mRNA-LNP formulations poses significant logistical challenges for global distribution. Lyophilization serves as a potential strategy to extend mRNA-LNP stability while eliminating the need for ultra-cold supply chain logistics. Although recent advancements have demonstrated the promise of lyophilization, the choice of lyoprotectant is predominately focused on sucrose, and there remains a gap in comprehensive evaluation and comparison of lyoprotectants and buffers. Here, we aim to systematically investigate the impact of a diverse range of excipients including oligosaccharides, polymers, amino acids, and various buffers, on the quality and performance of lyophilized mRNA-LNPs. From the screening of 45 mRNA-LNP formulations under various lyoprotectant and buffer conditions for lyophilization, we identified previously unexplored formulation compositions, e.g., polyvinylpyrrolidone (PVP) in Tris or acetate buffers, as promising alternatives to the commonly used oligosaccharides to maintain the physicochemical stability of lyophilized mRNA-LNPs. Further, we delved into how physicochemical and structural properties influence the functionality of lyophilized mRNA-LNPs. Leveraging high-throughput small-angle X-ray scattering (SAXS) and cryogenic transmission electron microscopy (cryo-TEM), we showed that there is complex interplay between mRNA-LNP structural features and cellular translation efficacy. We also assessed innate immune responses of the screened mRNA-LNPs in human peripheral blood mononuclear cells (PBMCs), and showed minimal alterations of cytokine secretion profiles induced by lyophilized formulations. Our results provide valuable insights into the structure-activity relationship of lyophilized formulations of mRNA-LNP therapeutics, paving the way for rational design of these formulations. This work creates a foundation for a comprehensive understanding of mRNA-LNP properties and in vitro performance change resulting from lyophilization.
Collapse
Affiliation(s)
- Yuchen Fan
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| | - Diamanda Rigas
- Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Lee Joon Kim
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Lab, Berkeley, CA 94020, USA
| | - Feng-Peng Chang
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Nanzhi Zang
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Kristina McKee
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Christopher C Kemball
- Biochemical and Cellular Pharmacology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Zhixin Yu
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Pascal Winkler
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Wan-Chih Su
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Pierce Jessen
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Greg L Hura
- Molecular Biophysics and Integrated Bioimaging Division, Lawrence Berkeley National Lab, Berkeley, CA 94020, USA; Chemistry and Biochemistry Department, University of California Santa Cruz, Santa Cruz, CA 95064, USA
| | - Tao Chen
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Stefan G Koenig
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Karthik Nagapudi
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Dennis Leung
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Chun-Wan Yen
- Synthetic Molecule Pharmaceutical Sciences, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
26
|
Chen D, Gu X, Nurzat Y, Xu L, Li X, Wu L, Jiao H, Gao P, Zhu X, Yan D, Li S, Xue C. Writers, readers, and erasers RNA modifications and drug resistance in cancer. Mol Cancer 2024; 23:178. [PMID: 39215288 PMCID: PMC11363509 DOI: 10.1186/s12943-024-02089-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
Drug resistance in cancer cells significantly diminishes treatment efficacy, leading to recurrence and metastasis. A critical factor contributing to this resistance is the epigenetic alteration of gene expression via RNA modifications, such as N6-methyladenosine (m6A), N1-methyladenosine (m1A), 5-methylcytosine (m5C), 7-methylguanosine (m7G), pseudouridine (Ψ), and adenosine-to-inosine (A-to-I) editing. These modifications are pivotal in regulating RNA splicing, translation, transport, degradation, and stability. Governed by "writers," "readers," and "erasers," RNA modifications impact numerous biological processes and cancer progression, including cell proliferation, stemness, autophagy, invasion, and apoptosis. Aberrant RNA modifications can lead to drug resistance and adverse outcomes in various cancers. Thus, targeting RNA modification regulators offers a promising strategy for overcoming drug resistance and enhancing treatment efficacy. This review consolidates recent research on the role of prevalent RNA modifications in cancer drug resistance, with a focus on m6A, m1A, m5C, m7G, Ψ, and A-to-I editing. Additionally, it examines the regulatory mechanisms of RNA modifications linked to drug resistance in cancer and underscores the existing limitations in this field.
Collapse
Affiliation(s)
- Di Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, 471000, Henan, China
| | - Yeltai Nurzat
- State Key Laboratory of Respiratory Disease, Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lixia Xu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xueyuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Lixin Wu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Henan Jiao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Peng Gao
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xuqiang Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Shaohua Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| | - Chen Xue
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
27
|
Masarwy R, Stotsky-Oterin L, Elisha A, Hazan-Halevy I, Peer D. Delivery of nucleic acid based genome editing platforms via lipid nanoparticles: Clinical applications. Adv Drug Deliv Rev 2024; 211:115359. [PMID: 38857763 DOI: 10.1016/j.addr.2024.115359] [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: 03/30/2024] [Revised: 05/17/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
CRISPR/Cas technology presents a promising approach for treating a wide range of diseases, including cancer and genetic disorders. Despite its potential, the translation of CRISPR/Cas into effective in-vivo gene therapy encounters challenges, primarily due to the need for safe and efficient delivery mechanisms. Lipid nanoparticles (LNPs), FDA-approved for RNA delivery, show potential for delivering also CRISPR/Cas, offering the capability to efficiently encapsulate large mRNA molecules with single guide RNAs. However, achieving precise targeting in-vivo remains a significant obstacle, necessitating further research into optimizing LNP formulations. Strategies to enhance specificity, such as modifying LNP structures and incorporating targeting ligands, are explored to improve organ and cell type targeting. Furthermore, the development of base and prime editing technology presents a potential breakthrough, offering precise modifications without generating double-strand breaks (DSBs). Prime editing, particularly when delivered via targeted LNPs, holds promise for treating diverse diseases safely and precisely. This review assesses both the progress made and the persistent challenges faced in using LNP-encapsulated CRISPR-based technologies for therapeutic purposes, with a particular focus on clinical translation.
Collapse
Affiliation(s)
- Razan Masarwy
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lior Stotsky-Oterin
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Aviad Elisha
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Inbal Hazan-Halevy
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.
| | - Dan Peer
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
28
|
Shah N, Soma SR, Quaye MB, Mahmoud D, Ahmed S, Malkoochi A, Obaid G. A Physiochemical, In Vitro, and In Vivo Comparative Analysis of Verteporfin-Lipid Conjugate Formulations: Solid Lipid Nanoparticles and Liposomes. ACS APPLIED BIO MATERIALS 2024; 7:4427-4441. [PMID: 38934648 PMCID: PMC11253097 DOI: 10.1021/acsabm.4c00316] [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: 03/05/2024] [Revised: 06/10/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
VisudyneⓇ, a liposomal formulation of verteporfin (benzoporphyrin derivative; BPD), is the only nanomedicine approved to date for photodynamic therapy (PDT). We have previously demonstrated that BPD conjugated to the lysophospholipid 1-arachidoyl-2-hydroxy-sn-glycero-3-phosphocholine (BPD-PC) exhibits the greatest physical stability in liposomes, while maintaining cancer cell phototoxicity, from a panel of BPD lipid conjugates evaluated. In this study, we prepared 1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC)-based solid lipid nanoparticles (LNPs) that stably entrap BPD-PC, which resemble the composition of the SpikevaxⓇ Moderna COVID-19 vaccine, and compared them to a DPPC based liposomal formulation (Lipo BPD-PC). We evaluated the photochemical, optical, and phototherapeutic properties of both formulations. We also investigated the in vivo distribution and tumor microdistribution of both formulations. Our results demonstrated that Lipo BPD-PC is able to generate 17% more singlet oxygen than LNP BPD-PC, while interestingly, LNP BPD-PC is able to produce 76% more hydroxyl radicals and/or peroxynitrite anion. Importantly, only 28% of BPD-PC leaches out of the LNP BPD-PC formulation during 7 days of incubation in serum at 37 °C, while 100% of BPD-PC leaches out of the Lipo BPD-PC formulation under the same conditions. Despite these differences, there was no significant difference in cellular uptake of BPD-PC or phototoxicity in CT1BA5 murine pancreatic cancer cells (derived from a genetically engineered mouse model). Interestingly, PDT using LNP BPD-PC was more efficient at inducing immunogenic cell death (calreticulin membrane translocation) than Lipo BPD-PC when using IC25 and IC50 PDT doses. In vivo studies revealed that CT1BA5 tumor fluorescence signals from BPD-PC were 2.41-fold higher with Lipo BPD-PC than with LNP BPD-PC; however, no significant difference was observed in tumor tissue selectivity or tumor penetration. As such, we present LNP BPD-PC as a unique and more stable nanoplatform to carry BPD lipid conjugates, such as BPD-PC, with a potential for future photodynamic immune priming studies and multiagent drug delivery.
Collapse
Affiliation(s)
- Nimit Shah
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| | - Siddharth Reddy Soma
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| | - Maxwell Bortei Quaye
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| | - Doha Mahmoud
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| | - Sarah Ahmed
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| | - Ashritha Malkoochi
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| | - Girgis Obaid
- Department
of Bioengineering, University of Texas at
Dallas, Richardson, Texas 75080, United States
| |
Collapse
|
29
|
Cao X, Su L, Chen H. A potent bioreducible ionizable lipid nanoparticle enables siRNA delivery for retinal neovascularization inhibition. Eur J Pharm Biopharm 2024; 199:114296. [PMID: 38636882 DOI: 10.1016/j.ejpb.2024.114296] [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: 02/09/2024] [Revised: 03/25/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Small interfering RNA (siRNA) is emerging as a promising treatment for retinal neovascularization due to its specific inhibition of the expression of target genes. However, the clinical translation of siRNA drugs is hindered by the efficiency and safety of delivery vectors. Here, we describe the properties of a new bioreducible ionizable lipid nanoparticle (LNP) 2N12H, which is based on a rationally designed novel ionizable lipid called 2N12B. 2N12H exhibited degradation in response to the mimic cytoplasmic glutathione condition and ionization with a pKa value of 6.5, which remaining neutral at pH 7.4. At a nitrogen to phosphorus ratio of 5, 2N12H efficiently encapsulated and protected siRNA from degradation. Compared to the commercial vehicle Lipofectamine 2000, 2N12H demonstrated similar silencing efficiency and improved safety in the in vitro cell experiments. 2N12H/siVEGFA reduced the expression of VEGFA in retinal pigment epithelium cells and mouse retina, consequently suppressing cell migration and retinal neovascularization. In the mouse model, the therapeutic effect of 2N12H/siVEGFA was comparable to that of the clinical drug ranibizumab. Together, these results suggest the potential of this novel ionizable LNP to facilitate the development of nonviral ocular gene delivery systems.
Collapse
Affiliation(s)
- Xiaowen Cao
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Lili Su
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Hao Chen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
30
|
Li Z, Amaya L, Ee A, Wang SK, Ranjan A, Waymouth RM, Chang HY, Wender PA. Organ- and Cell-Selective Delivery of mRNA In Vivo Using Guanidinylated Serinol Charge-Altering Releasable Transporters. J Am Chem Soc 2024; 146:14785-14798. [PMID: 38743019 DOI: 10.1021/jacs.4c02704] [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] [Indexed: 05/16/2024]
Abstract
Selective RNA delivery is required for the broad implementation of RNA clinical applications, including prophylactic and therapeutic vaccinations, immunotherapies for cancer, and genome editing. Current polyanion delivery relies heavily on cationic amines, while cationic guanidinium systems have received limited attention due in part to their strong polyanion association, which impedes intracellular polyanion release. Here, we disclose a general solution to this problem in which cationic guanidinium groups are used to form stable RNA complexes upon formulation but at physiological pH undergo a novel charge-neutralization process, resulting in RNA release. This new delivery system consists of guanidinylated serinol moieties incorporated into a charge-altering releasable transporter (GSer-CARTs). Significantly, systematic variations in structure and formulation resulted in GSer-CARTs that exhibit highly selective mRNA delivery to the lung (∼97%) and spleen (∼98%) without targeting ligands. Illustrative of their breadth and translational potential, GSer-CARTs deliver circRNA, providing the basis for a cancer vaccination strategy, which in a murine model resulted in antigen-specific immune responses and effective suppression of established tumors.
Collapse
Affiliation(s)
- Zhijian Li
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Laura Amaya
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, California 94305, United States
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Aloysius Ee
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Department of Materials Science & Engineering, Stanford University, Stanford, California 94305, United States
| | - Sean K Wang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, California 94305, United States
- Department of Ophthalmology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Alok Ranjan
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Robert M Waymouth
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, California 94305, United States
- Howard Hughes Medical Institute, Stanford University, Stanford, California 94305, United States
| | - Paul A Wender
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Department of Chemical and Systems Biology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
31
|
Omo-Lamai S, Wang Y, Patel MN, Essien EO, Shen M, Majumdar A, Espy C, Wu J, Channer B, Tobin M, Murali S, Papp TE, Maheshwari R, Wang L, Chase LS, Zamora ME, Arral ML, Marcos-Contreras OA, Myerson JW, Hunter CA, Tsourkas A, Muzykantov V, Brodsky I, Shin S, Whitehead KA, Gaskill P, Discher D, Parhiz H, Brenner JS. Lipid Nanoparticle-Associated Inflammation is Triggered by Sensing of Endosomal Damage: Engineering Endosomal Escape Without Side Effects. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589801. [PMID: 38659905 PMCID: PMC11042321 DOI: 10.1101/2024.04.16.589801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Lipid nanoparticles (LNPs) have emerged as the dominant platform for RNA delivery, based on their success in the COVID-19 vaccines and late-stage clinical studies in other indications. However, we and others have shown that LNPs induce severe inflammation, and massively aggravate pre-existing inflammation. Here, using structure-function screening of lipids and analyses of signaling pathways, we elucidate the mechanisms of LNP-associated inflammation and demonstrate solutions. We show that LNPs' hallmark feature, endosomal escape, which is necessary for RNA expression, also directly triggers inflammation by causing endosomal membrane damage. Large, irreparable, endosomal holes are recognized by cytosolic proteins called galectins, which bind to sugars on the inner endosomal membrane and then regulate downstream inflammation. We find that inhibition of galectins abrogates LNP-associated inflammation, both in vitro and in vivo . We show that rapidly biodegradable ionizable lipids can preferentially create endosomal holes that are smaller in size and reparable by the endosomal sorting complex required for transport (ESCRT) pathway. Ionizable lipids producing such ESCRT-recruiting endosomal holes can produce high expression from cargo mRNA with minimal inflammation. Finally, we show that both routes to non-inflammatory LNPs, either galectin inhibition or ESCRT-recruiting ionizable lipids, are compatible with therapeutic mRNAs that ameliorate inflammation in disease models. LNPs without galectin inhibition or biodegradable ionizable lipids lead to severe exacerbation of inflammation in these models. In summary, endosomal escape induces endosomal membrane damage that can lead to inflammation. However, the inflammation can be controlled by inhibiting galectins (large hole detectors) or by using biodegradable lipids, which create smaller holes that are reparable by the ESCRT pathway. These strategies should lead to generally safer LNPs that can be used to treat inflammatory diseases.
Collapse
|
32
|
Chiesa E, Caimi A, Bellotti M, Giglio A, Conti B, Dorati R, Auricchio F, Genta I. Effect of Micromixer Design on Lipid Nanocarriers Manufacturing for the Delivery of Proteins and Nucleic Acids. Pharmaceutics 2024; 16:507. [PMID: 38675169 PMCID: PMC11054535 DOI: 10.3390/pharmaceutics16040507] [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: 03/11/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Lipid-based nanocarriers have emerged as helpful tools to deliver sensible biomolecules such as proteins and oligonucleotides. To have a fast and robust microfluidic-based nanoparticle synthesis method, the setup of versatile equipment should allow for the rapid transfer to scale cost-effectively while ensuring tunable, precise and reproducible nanoparticle attributes. The present work aims to assess the effect of different micromixer geometries on the manufacturing of lipid nanocarriers taking into account the influence on the mixing efficiency by changing the fluid-fluid interface and indeed the mass transfer. Since the geometry of the adopted micromixer varies from those already published, a Design of Experiment (DoE) was necessary to identify the operating (total flow, flow rate ratio) and formulation (lipid concentration, lipid molar ratios) parameters affecting the nanocarrier quality. The suitable application of the platform was investigated by producing neutral, stealth and cationic liposomes, using DaunoXome®, Myocet®, Onivyde® and Onpattro® as the benchmark. The effect of condensing lipid (DOTAP, 3-10-20 mol%), coating lipids (DSPE-PEG550 and DSPE-PEG2000), as well as structural lipids (DSPC, eggPC) was pointed out. A very satisfactory encapsulation efficiency, always higher than 70%, was successfully obtained for model biomolecules (myoglobin, short and long nucleic acids).
Collapse
Affiliation(s)
- Enrica Chiesa
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100 Pavia, Italy; (E.C.); (A.G.); (B.C.); (R.D.)
| | - Alessandro Caimi
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy; (A.C.); (M.B.)
| | - Marco Bellotti
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy; (A.C.); (M.B.)
| | - Alessia Giglio
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100 Pavia, Italy; (E.C.); (A.G.); (B.C.); (R.D.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100 Pavia, Italy; (E.C.); (A.G.); (B.C.); (R.D.)
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100 Pavia, Italy; (E.C.); (A.G.); (B.C.); (R.D.)
| | - Ferdinando Auricchio
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy; (A.C.); (M.B.)
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, V.le Taramelli 12, 27100 Pavia, Italy; (E.C.); (A.G.); (B.C.); (R.D.)
| |
Collapse
|