1
|
Renzi S, Digiacomo L, Pozzi D, Quagliarini E, Vulpis E, Giuli MV, Mancusi A, Natiello B, Pignataro MG, Canettieri G, Di Magno L, Pesce L, De Lorenzi V, Ghignoli S, Loconte L, Montone CM, Laura Capriotti A, Laganà A, Nicoletti C, Amenitsch H, Rossi M, Mura F, Parisi G, Cardarelli F, Zingoni A, Checquolo S, Caracciolo G. Structuring lipid nanoparticles, DNA, and protein corona into stealth bionanoarchitectures for in vivo gene delivery. Nat Commun 2024; 15:9119. [PMID: 39438484 PMCID: PMC11496629 DOI: 10.1038/s41467-024-53569-8] [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: 12/27/2023] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
Lipid nanoparticles (LNPs) play a crucial role in addressing genetic disorders, and cancer, and combating pandemics such as COVID-19 and its variants. Yet, the ability of LNPs to effectively encapsulate large-size DNA molecules remains elusive. This is a significant limitation, as the successful delivery of large-size DNA holds immense potential for gene therapy. To address this gap, the present study focuses on the design of PEGylated LNPs, incorporating large-sized DNA, departing from traditional RNA and ionizable lipids. The resultant LNPs demonstrate a unique particle morphology. These particles were further engineered with a DNA coating and plasma proteins. This multicomponent bionanoconstruct exhibits enhanced transfection efficiency and safety in controlled laboratory settings and improved immune system evasion in in vivo tests. These findings provide valuable insights for the design and development of bionanoarchitectures for large-size DNA delivery, opening new avenues for transformative gene therapies.
Collapse
Affiliation(s)
- Serena Renzi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Luca Digiacomo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniela Pozzi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Erica Quagliarini
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Elisabetta Vulpis
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Valeria Giuli
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Latina, Italy
| | - Angelica Mancusi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Bianca Natiello
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Gemma Pignataro
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Rome, Italy
| | | | - Laura Di Magno
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Luca Pesce
- NEST, Scuola Normale Superiore, Pisa, Italy
| | | | | | - Luisa Loconte
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | | | - Aldo Laganà
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | - Carmine Nicoletti
- Unit of Histology and Medical Embryology, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Rome, Italy
| | - Heinz Amenitsch
- Institute of Inorganic Chemistry, Graz University of Technology, Graz, Austria
| | - Marco Rossi
- Department of Basic and Applied Sciences for Engineering and Center for Nanotechnology Applied to Engineering (CNIS), Sapienza University of Rome, Rome, Italy
| | - Francesco Mura
- Department of Basic and Applied Sciences for Engineering and Center for Nanotechnology Applied to Engineering (CNIS), Sapienza University of Rome, Rome, Italy
| | - Giacomo Parisi
- Department of Basic and Applied Sciences for Engineering and Center for Nanotechnology Applied to Engineering (CNIS), Sapienza University of Rome, Rome, Italy
| | | | - Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| | - Saula Checquolo
- Department of Medico-Surgical Sciences and Biotechnology, Sapienza University of Rome, Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Latina, Italy.
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
2
|
McMillan C, Druschitz A, Rumbelow S, Borah A, Binici B, Rattray Z, Perrie Y. Tailoring lipid nanoparticle dimensions through manufacturing processes. RSC PHARMACEUTICS 2024; 1:841-853. [PMID: 39323767 PMCID: PMC11417672 DOI: 10.1039/d4pm00128a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 08/29/2024] [Indexed: 09/27/2024]
Abstract
Lipid nanoparticles (LNPs), most commonly recognised for their role in COVID-19 mRNA vaccines, are important delivery vehicles for nucleic acid (mRNA, siRNA) therapies. The physicochemical attributes, such as size, nucleic acid encapsulation and electric charge, may have a significant impact on the efficacy of these medicines. In this study, adjustments to aqueous to lipid phase ratios were assessed for their impact on LNP size and other critical quality attributes (CQAs). It was observed that minor adjustments of aqueous-to-organic lipid phase ratios can be used to precisely control the size of ALC-0315-formulated LNPs. This was then used to evaluate the impact of phase ratio and corresponding size ranges on the in vitro and in vivo expression of these LNPs. In HEK293 cells, larger LNPs led to higher expression of the mRNA cargo within the LNPs, with a linear correlation between size and expression. In THP-1 cells this preference for larger LNPs was observed up to 120 d.nm after which there was a fall in expression. In BALB/c mice, however, LNPs at the lowest phase ratio tested, >120 d.nm, showed reduced expression compared to those of range 60-120 d.nm, within which there was no significant difference between sizes. These results suggest a robustness of LNP expression up to 120 d.nm, larger than those <100 d.nm conventionally used in medicine.
Collapse
Affiliation(s)
| | - Amy Druschitz
- Croda International Plc and Avanti Polar Lipids Alabaster AL USA
| | - Stephen Rumbelow
- Croda International Plc and Avanti Polar Lipids Alabaster AL USA
| | | | | | | | | |
Collapse
|
3
|
Vinales I, Silva-Espinoza JC, Medina BA, Urbay JEM, Beltran MA, Salinas DE, Ramirez-Ramos MA, Maldonado RA, Poon W, Penichet ML, Almeida IC, Michael K. Selective Transfection of a Transferrin Receptor-Expressing Cell Line with DNA-Lipid Nanoparticles. ACS OMEGA 2024; 9:39533-39545. [PMID: 39346819 PMCID: PMC11425831 DOI: 10.1021/acsomega.4c03541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/26/2024] [Accepted: 08/07/2024] [Indexed: 10/01/2024]
Abstract
Despite considerable progress in using lipid nanoparticle (LNP) vehicles for gene delivery, achieving selective transfection of specific cell types remains a significant challenge, hindering the advancement of new gene or gene-editing therapies. Although LNPs have been equipped with ligands aimed at targeting specific cellular receptors, achieving complete selectivity continues to be elusive. The exact reasons for this limited selectivity are not fully understood, as cell targeting involves a complex interplay of various cellular factors. Assessing how much ligand/receptor binding contributes to selectivity is challenging due to these additional influencing factors. Nonetheless, such data are important for developing new nanocarriers and setting realistic expectations for selectivity. Here, we have quantified the selective, targeted transfection using two uniquely engineered cell lines that eliminate unpredictable and interfering cellular influences. We have compared the targeted transfection of Chinese ovary hamster (CHO) cells engineered to express the human transferrin receptor 1 (hTfR1), CHO-TRVb-hTfR1, with CHO cells that completely lack any transferrin receptor, CHO-TRVb-neo cells (negative control). Thus, the two cell lines differ only in the presence/absence of hTfR1. The transfection was performed with pDNA-encapsulating LNPs equipped with the DT7 peptide ligand that specifically binds to hTfR1 and enables targeted transfection. The LNP's pDNA encoded for the monomeric GreenLantern (mGL) reporter protein, whose fluorescence was used to quantify transfection. We report a novel LNP composition designed to achieve an optimal particle size and ζ-potential, efficient pDNA encapsulation, hTfR1-targeting capability, and sufficient polyethylene glycol sheltering to minimize random cell targeting. The transfection efficiency was quantified in both cell lines separately through flow cytometry based on the expression of the fluorescent gene product. Our results demonstrated an LNP dose-dependent mGL expression, with a 5-fold preference for the CHO-TRVb-hTfR1 when compared to CHO-TRVb-neo. In another experiment, when both cell lines were mixed at a 1:1 ratio, the DT7-decorated LNP achieved a 3-fold higher transfection of the CHO-TRVb-hTfR1 over the CHO-TRVb-neo cells. Based on the low-level transfection of the CHO-TRVb-neo cells in both experiments, our results suggest that 17-25% of the transfection occurred in a nonspecific manner. The observed transfection selectivity for the CHO-TRVb-hTfR1 cells was based entirely on the hTfR1/DT7 interaction. This work showed that the platform of two engineered cell lines which differ only in the hTfR1 can greatly facilitate the development of LNPs with hTfR1-targeting ligands.
Collapse
Affiliation(s)
- Irodiel Vinales
- Department
of Chemistry and Biochemistry, University
of Texas at El Paso, El Paso, Texas 79968, United States
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79968, United States
| | - Juan Carlos Silva-Espinoza
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79968, United States
- Department
of Biological Sciences, University of Texas
at El Paso, El Paso, Texas 79968, United States
| | - Bryan A. Medina
- Department
of Chemistry and Biochemistry, University
of Texas at El Paso, El Paso, Texas 79968, United States
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79968, United States
| | - Juan E. M. Urbay
- Department
of Chemistry and Biochemistry, University
of Texas at El Paso, El Paso, Texas 79968, United States
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79968, United States
| | - Miguel A. Beltran
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79968, United States
- Department
of Biological Sciences, University of Texas
at El Paso, El Paso, Texas 79968, United States
| | - Dante E. Salinas
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79968, United States
- Department
of Biological Sciences, University of Texas
at El Paso, El Paso, Texas 79968, United States
| | - Marco A. Ramirez-Ramos
- Department
of Chemistry and Biochemistry, University
of Texas at El Paso, El Paso, Texas 79968, United States
| | - Rosa A. Maldonado
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79968, United States
- Department
of Biological Sciences, University of Texas
at El Paso, El Paso, Texas 79968, United States
| | - Wilson Poon
- Department
of Metallurgical, Materials and Biomedical Engineering, University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Manuel L. Penichet
- Division
of Surgical Oncology, Department of Surgery, David Geffen School of
Medicine, University of California, Los
Angeles (UCLA), Los Angeles, California 90095, United States
- Department
of Microbiology, Immunology and Molecular Genetics, David Geffen School
of Medicine, University of California, Los
Angeles (UCLA), Los Angeles, California 90095, United States
- California
Nanosystems Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- The Molecular
Biology Institute, University of California,
Los Angeles, Los Angeles, California 90095, United States
- Jonsson Comprehensive
Cancer Center, University of California,
Los Angeles, Los Angeles, California 90095, United States
| | - Igor C. Almeida
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79968, United States
- Department
of Biological Sciences, University of Texas
at El Paso, El Paso, Texas 79968, United States
| | - Katja Michael
- Department
of Chemistry and Biochemistry, University
of Texas at El Paso, El Paso, Texas 79968, United States
- Border
Biomedical Research Center, University of
Texas at El Paso, El Paso, Texas 79968, United States
| |
Collapse
|
4
|
Kachanov A, Kostyusheva A, Brezgin S, Karandashov I, Ponomareva N, Tikhonov A, Lukashev A, Pokrovsky V, Zamyatnin AA, Parodi A, Chulanov V, Kostyushev D. The menace of severe adverse events and deaths associated with viral gene therapy and its potential solution. Med Res Rev 2024; 44:2112-2193. [PMID: 38549260 DOI: 10.1002/med.22036] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 08/09/2024]
Abstract
Over the past decade, in vivo gene replacement therapy has significantly advanced, resulting in market approval of numerous therapeutics predominantly relying on adeno-associated viral vectors (AAV). While viral vectors have undeniably addressed several critical healthcare challenges, their clinical application has unveiled a range of limitations and safety concerns. This review highlights the emerging challenges in the field of gene therapy. At first, we discuss both the role of biological barriers in viral gene therapy with a focus on AAVs, and review current landscape of in vivo human gene therapy. We delineate advantages and disadvantages of AAVs as gene delivery vehicles, mostly from the safety perspective (hepatotoxicity, cardiotoxicity, neurotoxicity, inflammatory responses etc.), and outline the mechanisms of adverse events in response to AAV. Contribution of every aspect of AAV vectors (genomic structure, capsid proteins) and host responses to injected AAV is considered and substantiated by basic, translational and clinical studies. The updated evaluation of recent AAV clinical trials and current medical experience clearly shows the risks of AAVs that sometimes overshadow the hopes for curing a hereditary disease. At last, a set of established and new molecular and nanotechnology tools and approaches are provided as potential solutions for mitigating or eliminating side effects. The increasing number of severe adverse reactions and, sadly deaths, demands decisive actions to resolve the issue of immune responses and extremely high doses of viral vectors used for gene therapy. In response to these challenges, various strategies are under development, including approaches aimed at augmenting characteristics of viral vectors and others focused on creating secure and efficacious non-viral vectors. This comprehensive review offers an overarching perspective on the present state of gene therapy utilizing both viral and non-viral vectors.
Collapse
Affiliation(s)
- Artyom Kachanov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Anastasiya Kostyusheva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Sergey Brezgin
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Ivan Karandashov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Natalia Ponomareva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Andrey Tikhonov
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Alexander Lukashev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
| | - Vadim Pokrovsky
- Laboratory of Biochemical Fundamentals of Pharmacology and Cancer Models, Blokhin Cancer Research Center, Moscow, Russia
- Department of Biochemistry, People's Friendship University, Russia (RUDN University), Moscow, Russia
| | - Andrey A Zamyatnin
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
- Belozersky Research, Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
| | - Vladimir Chulanov
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Infectious Diseases, Sechenov University, Moscow, Russia
| | - Dmitry Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, Moscow, Russia
- Division of Biotechnology, Scientific Center for Genetics and Life Sciences, Sirius University of Science and Technology, Sochi, Russia
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
5
|
Chauhan S, Nagpal K. Latest Delivery Advancements of Lipid Nanoparticles for Cancer Treatment. Assay Drug Dev Technol 2024; 22:340-360. [PMID: 38968367 DOI: 10.1089/adt.2024.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024] Open
Abstract
As one of the primary causes of illness and death globally, cancer demands novel and potent treatment approaches, which is why lipid nanoparticles (LNPs) have gained attention as a promising delivery system for anticancer drugs with precision and efficacy. The article discusses the salient characteristics of LNPs, such as the lipid components, particle size, polydispersity index, and encapsulation efficiency, followed by strategies that enhance their remarkable drug delivery capabilities. The articles explore LNPs ability to improve the solubility, stability, and bioavailability of various chemotherapeutics, nucleic acids, and immunotherapeutic modalities. It also highlights the recent advancement in surface modification of LNPs, which is essential to improve their effectiveness. Tailored coatings of LNPs improve targeting precision, stability, and biocompatibility; enhancing their transport to boost therapeutic efficacy for cancer targeting. The review summarizes the recent advancements made in using LNPs to treat different forms of cancer and focuses on the most recent clinical studies. Overall, the review highlights that the LNPs can target and treat cancer in a tailored manner through gene therapy, RNA interference, and immunotherapy.
Collapse
Affiliation(s)
- Somia Chauhan
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, India
| | - Kalpana Nagpal
- Amity Institute of Pharmacy, Amity University Uttar Pradesh, Noida, India
| |
Collapse
|
6
|
Li M, Liu L, Li X, Li J, Zhao C, Zhao Y, Zhang X, He P, Wu X, Jiang S, Wang X, Zhang X, Wei L. Lipid Nanoparticles Outperform Electroporation in Delivering Therapeutic HPV DNA Vaccines. Vaccines (Basel) 2024; 12:666. [PMID: 38932395 PMCID: PMC11209142 DOI: 10.3390/vaccines12060666] [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: 05/16/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Therapeutic HPV vaccines that induce potent HPV-specific cellular immunity and eliminate pre-existing infections remain elusive. Among various candidates under development, those based on DNA constructs are considered promising because of their safety profile, stability, and efficacy. However, the use of electroporation (EP) as a main delivery method for such vaccines is notorious for adverse effects like pain and potentially irreversible muscle damage. Moreover, the requirement for specialized equipment adds to the complexity and cost of clinical applications. As an alternative to EP, lipid nanoparticles (LNPs) that are already commercially available for delivering mRNA and siRNA vaccines are likely to be feasible. Here, we have compared three intramuscular delivery systems in a preclinical setting. In terms of HPV-specific cellular immune responses, mice receiving therapeutic HPV DNA vaccines encapsulated with LNP demonstrated superior outcomes when compared to EP administration, while the naked plasmid vaccine showed negligible responses, as expected. In addition, SM-102 LNP M exhibited the most promising results in delivering candidate DNA vaccines. Thus, LNP proves to be a feasible delivery method in vivo, offering improved immunogenicity over traditional approaches.
Collapse
Affiliation(s)
- Mingzhu Li
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, No. 11 Xizhimen South Street, Beijing 100044, China; (M.L.); (J.L.); (C.Z.); (Y.Z.)
| | - Lei Liu
- Aeonvital Institute of Clinical and Translational Immunology (AICTI), Beijing 102600, China; (L.L.); (X.L.); (X.Z.); (P.H.); (X.W.); (S.J.); (X.W.)
| | - Xiaoli Li
- Aeonvital Institute of Clinical and Translational Immunology (AICTI), Beijing 102600, China; (L.L.); (X.L.); (X.Z.); (P.H.); (X.W.); (S.J.); (X.W.)
| | - Jingran Li
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, No. 11 Xizhimen South Street, Beijing 100044, China; (M.L.); (J.L.); (C.Z.); (Y.Z.)
| | - Chao Zhao
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, No. 11 Xizhimen South Street, Beijing 100044, China; (M.L.); (J.L.); (C.Z.); (Y.Z.)
| | - Yun Zhao
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, No. 11 Xizhimen South Street, Beijing 100044, China; (M.L.); (J.L.); (C.Z.); (Y.Z.)
| | - Xiaopeng Zhang
- Aeonvital Institute of Clinical and Translational Immunology (AICTI), Beijing 102600, China; (L.L.); (X.L.); (X.Z.); (P.H.); (X.W.); (S.J.); (X.W.)
| | - Panpan He
- Aeonvital Institute of Clinical and Translational Immunology (AICTI), Beijing 102600, China; (L.L.); (X.L.); (X.Z.); (P.H.); (X.W.); (S.J.); (X.W.)
| | - Xiaoyu Wu
- Aeonvital Institute of Clinical and Translational Immunology (AICTI), Beijing 102600, China; (L.L.); (X.L.); (X.Z.); (P.H.); (X.W.); (S.J.); (X.W.)
| | - Siwen Jiang
- Aeonvital Institute of Clinical and Translational Immunology (AICTI), Beijing 102600, China; (L.L.); (X.L.); (X.Z.); (P.H.); (X.W.); (S.J.); (X.W.)
| | - Xingxing Wang
- Aeonvital Institute of Clinical and Translational Immunology (AICTI), Beijing 102600, China; (L.L.); (X.L.); (X.Z.); (P.H.); (X.W.); (S.J.); (X.W.)
| | - Xiujun Zhang
- Aeonvital Institute of Clinical and Translational Immunology (AICTI), Beijing 102600, China; (L.L.); (X.L.); (X.Z.); (P.H.); (X.W.); (S.J.); (X.W.)
| | - Lihui Wei
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, No. 11 Xizhimen South Street, Beijing 100044, China; (M.L.); (J.L.); (C.Z.); (Y.Z.)
| |
Collapse
|
7
|
Eweje F, Walsh ML, Ahmad K, Ibrahim V, Alrefai A, Chen J, Chaikof EL. Protein-based nanoparticles for therapeutic nucleic acid delivery. Biomaterials 2024; 305:122464. [PMID: 38181574 PMCID: PMC10872380 DOI: 10.1016/j.biomaterials.2023.122464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/25/2023] [Accepted: 12/31/2023] [Indexed: 01/07/2024]
Abstract
To realize the full potential of emerging nucleic acid therapies, there is a need for effective delivery agents to transport cargo to cells of interest. Protein materials exhibit several unique properties, including biodegradability, biocompatibility, ease of functionalization via recombinant and chemical modifications, among other features, which establish a promising basis for therapeutic nucleic acid delivery systems. In this review, we highlight progress made in the use of non-viral protein-based nanoparticles for nucleic acid delivery in vitro and in vivo, while elaborating on key physicochemical properties that have enabled the use of these materials for nanoparticle formulation and drug delivery. To conclude, we comment on the prospects and unresolved challenges associated with the translation of protein-based nucleic acid delivery systems for therapeutic applications.
Collapse
Affiliation(s)
- Feyisayo Eweje
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Harvard and MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Harvard/MIT MD-PhD Program, Boston, MA, USA, 02115; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA
| | - Michelle L Walsh
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Harvard and MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA; Harvard/MIT MD-PhD Program, Boston, MA, USA, 02115
| | - Kiran Ahmad
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Vanessa Ibrahim
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Assma Alrefai
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Jiaxuan Chen
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| | - Elliot L Chaikof
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA; Wyss Institute of Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.
| |
Collapse
|
8
|
Pozzi D, Caracciolo G. Looking Back, Moving Forward: Lipid Nanoparticles as a Promising Frontier in Gene Delivery. ACS Pharmacol Transl Sci 2023; 6:1561-1573. [PMID: 37974625 PMCID: PMC10644400 DOI: 10.1021/acsptsci.3c00185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Indexed: 11/19/2023]
Abstract
Lipid nanoparticles (LNPs) have shown remarkable success in delivering genetic materials like COVID-19 LNP vaccines, such as mRNA-1273/SpikeVax by Moderna and BNT162b2/Comirnaty by BioNTech/Pfizer, as well as siRNA for rare inherited diseases, such as Onpattro from Alnylam Pharmaceuticals. These LNPs are advantageous since they minimize side effects, target specific cells, and regulate payload delivery. There has been a surge of interest in these particles due to their success stories; however, we still do not know much about how they work. This perspective will recapitulate the evolution of lipid-based gene delivery, starting with Felgner's pioneering 1987 PNAS paper, which introduced the initial DNA-transfection method utilizing a synthetic cationic lipid. Our journey takes us to the early 2020s, a time when advancements in bionano interactions enabled us to create biomimetic lipoplexes characterized by a remarkable ability to evade capture by immune cells in vivo. Through this overview, we propose leveraging previous achievements to assist us in formulating improved research goals when optimizing LNPs for medical conditions such as infectious diseases, cancer, and heritable disorders.
Collapse
Affiliation(s)
- Daniela Pozzi
- NanoDelivery Lab, Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina
Elena 291, 00161 Rome, Italy
| | - Giulio Caracciolo
- NanoDelivery Lab, Department
of Molecular Medicine, Sapienza University
of Rome, Viale Regina
Elena 291, 00161 Rome, Italy
| |
Collapse
|
9
|
Digiacomo L, Renzi S, Quagliarini E, Pozzi D, Amenitsch H, Ferri G, Pesce L, De Lorenzi V, Matteoli G, Cardarelli F, Caracciolo G. Investigating the mechanism of action of DNA-loaded PEGylated lipid nanoparticles. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 53:102697. [PMID: 37507061 DOI: 10.1016/j.nano.2023.102697] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/26/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023]
Abstract
PEGylated lipid nanoparticles (LNPs) are commonly used to deliver bioactive molecules, but the role of PEGylation in DNA-loaded LNP interactions at the cellular and subcellular levels remains poorly understood. In this study, we investigated the mechanism of action of DNA-loaded PEGylated LNPs using gene reporter technologies, dynamic light scattering (DLS), synchrotron small angle X-ray scattering (SAXS), and fluorescence confocal microscopy (FCS). We found that PEG has no significant impact on the size or nanostructure of DNA LNPs but reduces their zeta potential and interaction with anionic cell membranes. PEGylation increases the structural stability of LNPs and results in lower DNA unloading. FCS experiments revealed that PEGylated LNPs are internalized intact inside cells and largely shuttled to lysosomes, while unPEGylated LNPs undergo massive destabilization on the plasma membrane. These findings can inform the design, optimization, and validation of DNA-loaded LNPs for gene delivery and vaccine development.
Collapse
Affiliation(s)
- Luca Digiacomo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Serena Renzi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Erica Quagliarini
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Daniela Pozzi
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy
| | - Heinz Amenitsch
- Institute of Inorganic Chemistry, Graz University of Technology, 8010 Graz, Austria
| | - Gianmarco Ferri
- Laboratorio NEST, Scuola Normale Superiore, 56127 Pisa, Italy
| | - Luca Pesce
- Laboratorio NEST, Scuola Normale Superiore, 56127 Pisa, Italy
| | | | - Giulia Matteoli
- Laboratorio NEST, Scuola Normale Superiore, 56127 Pisa, Italy
| | | | - Giulio Caracciolo
- NanoDelivery Lab, Department of Molecular Medicine, Sapienza University of Rome, 00161 Rome, Italy.
| |
Collapse
|
10
|
Chen PY, Qin L, Simons M. TGFβ signaling pathways in human health and disease. Front Mol Biosci 2023; 10:1113061. [PMID: 37325472 PMCID: PMC10267471 DOI: 10.3389/fmolb.2023.1113061] [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: 12/01/2022] [Accepted: 04/27/2023] [Indexed: 06/17/2023] Open
Abstract
Transforming growth factor beta (TGFβ) is named for the function it was originally discovered to perform-transformation of normal cells into aggressively growing malignant cells. It became apparent after more than 30 years of research, however, that TGFβ is a multifaceted molecule with a myriad of different activities. TGFβs are widely expressed with almost every cell in the human body producing one or another TGFβ family member and expressing its receptors. Importantly, specific effects of this growth factor family differ in different cell types and under different physiologic and pathologic conditions. One of the more important and critical TGFβ activities is the regulation of cell fate, especially in the vasculature, that will be the focus of this review.
Collapse
Affiliation(s)
- Pei-Yu Chen
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Lingfeng Qin
- Department of Surgery, Yale University School of Medicine, New Haven, CT, United States
| | - Michael Simons
- Yale Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
11
|
Vogelaar A, Marcotte S, Cheng J, Oluoch B, Zaro J. Use of Microfluidics to Prepare Lipid-Based Nanocarriers. Pharmaceutics 2023; 15:pharmaceutics15041053. [PMID: 37111539 PMCID: PMC10144662 DOI: 10.3390/pharmaceutics15041053] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/18/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Lipid-based nanoparticles (LBNPs) are an important tool for the delivery of a diverse set of drug cargoes, including small molecules, oligonucleotides, and proteins and peptides. Despite their development over the past several decades, this technology is still hindered by issues with the manufacturing processes leading to high polydispersity, batch-to-batch and operator-dependent variability, and limits to the production volumes. To overcome these issues, the use of microfluidic techniques in the production of LBNPs has sharply increased over the past two years. Microfluidics overcomes many of the pitfalls seen with conventional production methods, leading to reproducible LBNPs at lower costs and higher yields. In this review, the use of microfluidics in the preparation of various types of LBNPs, including liposomes, lipid nanoparticles, and solid lipid nanoparticles for the delivery of small molecules, oligonucleotides, and peptide/protein drugs is summarized. Various microfluidic parameters, as well as their effects on the physicochemical properties of LBNPs, are also discussed.
Collapse
Affiliation(s)
- Alicia Vogelaar
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Samantha Marcotte
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Jiaqi Cheng
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Benazir Oluoch
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Jennica Zaro
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
12
|
González-Rioja R, Salazar VA, Bastús NG, Puntes V. The development of highly dense highly protected surfactant ionizable lipid RNA loaded nanoparticles. Front Immunol 2023; 14:1129296. [PMID: 36923400 PMCID: PMC10009161 DOI: 10.3389/fimmu.2023.1129296] [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: 12/21/2022] [Accepted: 02/14/2023] [Indexed: 03/02/2023] Open
Abstract
The long quest for efficient drug administration has been looking for a universal carrier that can precisely transport traditional drugs, new genomic and proteic therapeutic agents. Today, researchers have found conditions to overcome the two main drug delivery dilemmas. On the one side, the versatility of the vehicle to efficiently load, protect and transport the drug and then release it at the target place. On the other hand, the questions related to the degree of PEGylation which are needed to avoid nanoparticle (NP) aggregation and opsonization while preventing cellular uptake. The development of different kinds of lipidic drug delivery vehicles and particles has resulted in the development of ionizable lipid nanoparticles (iLNPs), which can overcome most of the typical drug delivery problems. Proof of their success is the late approval and massive administration as the prophylactic vaccine for SARS-CoV-2. These ILNPs are built by electrostatic aggregation of surfactants, the therapeutic agent, and lipids that self-segregate from an aqueous solution, forming nanoparticles stabilized with lipid polymers, such as PEG. These vehicles overcome previous limitations such as low loading and high toxicity, likely thanks to low charge at the working pH and reduced size, and their entry into the cells via endocytosis rather than membrane perforation or fusion, always associated with higher toxicity. We herein revise their primary features, synthetic methods to prepare and characterize them, pharmacokinetic (administration, distribution, metabolization and excretion) aspects, and biodistribution and fate. Owing to their advantages, iLNPs are potential drug delivery systems to improve the management of various diseases and widely available for clinical use.
Collapse
Affiliation(s)
- Ramon González-Rioja
- Institut Català de Nanociència i Nanotecnologia (ICN2), Consejo Superior de Investigaciones Científicas (CSIC), The Barcelona Institute of Science and Technology (BIST), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Vivian A. Salazar
- Institut Català de Nanociència i Nanotecnologia (ICN2), Consejo Superior de Investigaciones Científicas (CSIC), The Barcelona Institute of Science and Technology (BIST), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Neus G. Bastús
- Institut Català de Nanociència i Nanotecnologia (ICN2), Consejo Superior de Investigaciones Científicas (CSIC), The Barcelona Institute of Science and Technology (BIST), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) en Bioingeniería, Biomateriales y Nanomedicina, Centro de Investigación Biomédica en Red en Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
| | - Victor Puntes
- Institut Català de Nanociència i Nanotecnologia (ICN2), Consejo Superior de Investigaciones Científicas (CSIC), The Barcelona Institute of Science and Technology (BIST), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
- Centro de Investigación Biomédica en Red (CIBER) en Bioingeniería, Biomateriales y Nanomedicina, Centro de Investigación Biomédica en Red en Bioingeniería Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain
- Malalties Infeccioses, Nanopartícules farmacocinétiques, Vall d’Hebron Institut de Recerca, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
13
|
Wang M, Tian D, Xu L, Lu M, Yan R, Li X, Song X. Protective efficacy induced by Eimeria maxima rhomboid-like protein 1 against homologous infection. Front Vet Sci 2023; 9:1049551. [PMID: 36686197 PMCID: PMC9845710 DOI: 10.3389/fvets.2022.1049551] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 12/12/2022] [Indexed: 01/06/2023] Open
Abstract
Introduction Avian coccidiosis, caused by apicomplexan protozoa belonging to the Eimeria genus, is considered one of the most important diseases in the intensive poultry industry worldwide. Due to the shortcomings of live anticoccidial vaccines and drugs, the development of novel anticoccidial vaccines is increasingly urgent. Methods Eimeria maxima rhomboid-like protein 1 (EmROM1), an invasion-related molecule, was selected as a candidate antigen to evaluate its protective efficacy against E. maxima in chickens. Firstly, the prokaryotic recombinant plasmid pET-32a-EmROM1 was constructed to prepare EmROM1 recombinant protein (rEmROM1), which was used as a subunit vaccine. The eukaryotic recombinant plasmid pVAX1.0-EmROM1 (pEmROM1) was constructed as a DNA vaccine. Subsequently, 2-week-old chicks were separately vaccinated with the rEmROM1 and pEmROM1 twice every 7 days. One week post the booster vaccination, induced cellular immune responses were determined by evaluating the mRNA level of cytokines including IL-2, IFN-γ, IL-4, IL-10, TGF-β, IL-17, and TNFSF15, as well as the percentages of CD4+ and CD8+ T cells from spleens of vaccinated chickens. Specific serum antibody level in the vaccinated chickens was determined to assess induced humoral immune responses. Finally, the protective efficacy of EmROM1 was evaluated by a vaccination-challenge trial. Results EmROM1 vaccination significantly upregulated the cytokine transcription levels and CD4+/CD8+ T cell percentages in vaccinated chickens compared with control groups, and also significantly increased the levels of serum-specific antibodies in vaccinated chickens. The animal trial showed that EmROM1 vaccination significantly reduced oocyst shedding, enteric lesions, and weight loss of infected birds compared with the controls. The anticoccidial index (ACI) from the rEmROM-vaccination group and pEmROM1-vaccination group were 174.11 and 163.37, respectively, showing moderate protection against E. maxima infection. Discussion EmROM1 is an effective candidate antigen for developing DNA or subunit vaccines against avian coccidiosis.
Collapse
|