1
|
Mrksich K, Padilla MS, Mitchell MJ. Breaking the final barrier: Evolution of cationic and ionizable lipid structure in lipid nanoparticles to escape the endosome. Adv Drug Deliv Rev 2024; 214:115446. [PMID: 39293650 PMCID: PMC11900896 DOI: 10.1016/j.addr.2024.115446] [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: 07/05/2024] [Revised: 08/18/2024] [Accepted: 09/07/2024] [Indexed: 09/20/2024]
Abstract
In the past decade, nucleic acid therapies have seen a boon in development and clinical translation largely due to advances in nanotechnology that have enabled their safe and targeted delivery. Nanoparticles can protect nucleic acids from degradation by serum enzymes and can facilitate entry into cells. Still, achieving endosomal escape to allow nucleic acids to enter the cytoplasm has remained a significant barrier, where less than 5% of nanoparticles within the endo-lysosomal pathway are able to transfer their cargo to the cytosol. Lipid-based drug delivery vehicles, particularly lipid nanoparticles (LNPs), have been optimized to achieve potent endosomal escape, and thus have been the vector of choice in the clinic as demonstrated by their utilization in the COVID-19 mRNA vaccines. The success of LNPs is in large part due to the rational design of lipids that can specifically overcome endosomal barriers. In this review, we chart the evolution of lipid structure from cationic lipids to ionizable lipids, focusing on structure-function relationships, with a focus on how they relate to endosomal escape. Additionally, we examine recent advancements in ionizable lipid structure as well as discuss the future of lipid design.
Collapse
Affiliation(s)
- Kaitlin Mrksich
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marshall S Padilla
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J Mitchell
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
2
|
Mullin JA, Rahmani E, Kiick KL, Sullivan MO. Growth factors and growth factor gene therapies for treating chronic wounds. Bioeng Transl Med 2024; 9:e10642. [PMID: 38818118 PMCID: PMC11135157 DOI: 10.1002/btm2.10642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 06/01/2024] Open
Abstract
Chronic wounds are an unmet clinical need affecting millions of patients globally, and current standards of care fail to consistently promote complete wound closure and prevent recurrence. Disruptions in growth factor signaling, a hallmark of chronic wounds, have led researchers to pursue growth factor therapies as potential supplements to standards of care. Initial studies delivering growth factors in protein form showed promise, with a few formulations reaching clinical trials and one obtaining clinical approval. However, protein-form growth factors are limited by instability and off-target effects. Gene therapy offers an alternative approach to deliver growth factors to the chronic wound environment, but safety concerns surrounding gene therapy as well as efficacy challenges in the gene delivery process have prevented clinical translation. Current growth factor delivery and gene therapy approaches have primarily used single growth factor formulations, but recent efforts have aimed to develop multi-growth factor approaches that are better suited to address growth factor insufficiencies in the chronic wound environment, and these strategies have demonstrated improved efficacy in preclinical studies. This review provides an overview of chronic wound healing, emphasizing the need and potential for growth factor therapies. It includes a summary of current standards of care, recent advances in growth factor, cell-based, and gene therapy approaches, and future perspectives for multi-growth factor therapeutics.
Collapse
Affiliation(s)
- James A. Mullin
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
| | - Erfan Rahmani
- Department of Biomedical EngineeringUniversity of DelawareNewarkDelawareUSA
| | - Kristi L. Kiick
- Department of Biomedical EngineeringUniversity of DelawareNewarkDelawareUSA
- Department of Materials Science and EngineeringUniversity of DelawareNewarkDelawareUSA
| | - Millicent O. Sullivan
- Department of Chemical and Biomolecular EngineeringUniversity of DelawareNewarkDelawareUSA
- Department of Biomedical EngineeringUniversity of DelawareNewarkDelawareUSA
| |
Collapse
|
3
|
Eş I, Thakur A, Mousavi Khaneghah A, Foged C, de la Torre LG. Engineering aspects of lipid-based delivery systems: In vivo gene delivery, safety criteria, and translation strategies. Biotechnol Adv 2024; 72:108342. [PMID: 38518964 DOI: 10.1016/j.biotechadv.2024.108342] [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: 01/06/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/24/2024]
Abstract
Defects in the genome cause genetic diseases and can be treated with gene therapy. Due to the limitations encountered in gene delivery, lipid-based supramolecular colloidal materials have emerged as promising gene carrier systems. In their non-functionalized form, lipid nanoparticles often demonstrate lower transgene expression efficiency, leading to suboptimal therapeutic outcomes, specifically through reduced percentages of cells expressing the transgene. Due to chemically active substituents, the engineering of delivery systems for genetic drugs with specific chemical ligands steps forward as an innovative strategy to tackle the drawbacks and enhance their therapeutic efficacy. Despite intense investigations into functionalization strategies, the clinical outcome of such therapies still needs to be improved. Here, we highlight and comprehensively review engineering aspects for functionalizing lipid-based delivery systems and their therapeutic efficacy for developing novel genetic cargoes to provide a full snapshot of the translation from the bench to the clinics. We outline existing challenges in the delivery and internalization processes and narrate recent advances in the functionalization of lipid-based delivery systems for nucleic acids to enhance their therapeutic efficacy and safety. Moreover, we address clinical trials using these vectors to expand their clinical use and principal safety concerns.
Collapse
Affiliation(s)
- Ismail Eş
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil; Institute of Biomedical Engineering, Old Road Campus Research Building, University of Oxford, Headington, Oxford OX3 7DQ, UK.
| | - Aneesh Thakur
- Vaccine and Infectious Disease Organization, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| | - Amin Mousavi Khaneghah
- Faculty of Biotechnologies (BioTech), ITMO University 191002, 9 Lomonosova Street, Saint Petersburg, Russia.
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Lucimara Gaziola de la Torre
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
4
|
Gholap AD, Kapare HS, Pagar S, Kamandar P, Bhowmik D, Vishwakarma N, Raikwar S, Garkal A, Mehta TA, Rojekar S, Hatvate N, Mohanto S. Exploring modified chitosan-based gene delivery technologies for therapeutic advancements. Int J Biol Macromol 2024; 260:129581. [PMID: 38266848 DOI: 10.1016/j.ijbiomac.2024.129581] [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: 11/09/2023] [Revised: 12/26/2023] [Accepted: 01/06/2024] [Indexed: 01/26/2024]
Abstract
One of the critical steps in gene therapy is the successful delivery of the genes. Immunogenicity and toxicity are major issues for viral gene delivery systems. Thus, non-viral vectors are explored. A cationic polysaccharide like chitosan could be used as a nonviral gene delivery vector owing to its significant interaction with negatively charged nucleic acid and biomembrane, providing effective cellular uptake. However, the native chitosan has issues of targetability, unpacking ability, and solubility along with poor buffer capability, hence requiring modifications for effective use in gene delivery. Modified chitosan has shown that the "proton sponge effect" involved in buffering the endosomal pH results in osmotic swelling owing to the accumulation of a greater amount of proton and chloride along with water. The major challenges include limited exploration of chitosan as a gene carrier, the availability of high-purity chitosan for toxicity reduction, and its immunogenicity. The genetic drugs are in their infancy phase and require further exploration for effective delivery of nucleic acid molecules as FDA-approved marketed formulations soon.
Collapse
Affiliation(s)
- Amol D Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar 401404, Maharashtra, India
| | - Harshad S Kapare
- Department of Pharmaceutics, Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pune 411018, Maharashtra, India
| | - Sakshi Pagar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai 400019, India
| | - Pallavi Kamandar
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India
| | - Deblina Bhowmik
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India
| | - Nikhar Vishwakarma
- Department of Pharmacy, Gyan Ganga Institute of Technology and Sciences, Jabalpur 482003, Madhya Pradesh, India
| | - Sarjana Raikwar
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Central University, Sagar 470003, Madhya Pradesh, India
| | - Atul Garkal
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Tejal A Mehta
- Department of Pharmaceutics, Institute of Pharmacy, Nirma University, Ahmedabad 382481, Gujrat, India
| | - Satish Rojekar
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| | - Navnath Hatvate
- Institute of Chemical Technology, Mumbai, Marathwada Campus, Jalna 431203, India.
| | - Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangaluru, Karnataka 575018, India
| |
Collapse
|
5
|
Jennings J, Ašćerić D, Semeraro EF, Lohner K, Malanovic N, Pabst G. Combinatorial Screening of Cationic Lipidoids Reveals How Molecular Conformation Affects Membrane-Targeting Antimicrobial Activity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:40178-40190. [PMID: 37602460 PMCID: PMC10472336 DOI: 10.1021/acsami.3c05481] [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: 04/19/2023] [Accepted: 08/04/2023] [Indexed: 08/22/2023]
Abstract
The search for next-generation antibacterial compounds that overcome the development of resistance can be facilitated by identifying how to target the cell membrane of bacteria. Understanding the key molecular features that enable interactions with lipids and lead to membrane disruption is therefore crucial. Here, we employ a library of lipid-like compounds (lipidoids) comprising modular structures with tunable hydrophobic and hydrophilic architecture to shed light on how the chemical functionality and molecular shape of synthetic amphiphilic compounds determine their activity against bacterial membranes. Synthesized from combinations of 8 different polyamines as headgroups and 13 acrylates as tails, 104 different lipidoids are tested for activity against a model Gram-positive bacterial strain (Bacillus subtilis). Results from the combinatorial screening assay show that lipidoids with the most potent antimicrobial properties (down to 2 μM) have intermediate tail hydrophobicity (i.e., c log P values between 3 and 4) and lower headgroup charge density (i.e., longer spacers between charged amines). However, the most important factor appeared to be the ability of a lipidoid to self-assemble into an inverse hexagonal liquid crystalline phase, as observed by small-angle X-ray scattering (SAXS) analysis. The lipidoids active at lowest concentrations, which induced the most significant membrane damage during propidium iodide (PI) permeabilization assays, were those that aggregated into highly curved inverse hexagonal liquid crystal phases. These observations suggest that the introduction of strong curvature stress into the membrane is one way to maximize membrane disruption and lipidoid antimicrobial activity. Lipidoids that demonstrated the ability to furnish this phase consisted of either (i) branched or linear headgroups with shorter linear tails or (ii) cyclic headgroups with 4 bulky nonlinear tails. On the contrary, lipidoids previously observed to adopt disc-like conformations that pack into bicontinuous cubic phases were significantly less effective against B. subtilis. The discovery of these structure-property relationships demonstrates that it is not simply a balance of hydrophobic and hydrophilic moieties that govern membrane-active antibacterial activity, but also their intrinsic curvature and collective behavior.
Collapse
Affiliation(s)
- James Jennings
- Institute
of Molecular Biosciences, University of
Graz, NAWI Graz, 8010 Graz, Austria
- Field
of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Dunja Ašćerić
- Institute
of Molecular Biosciences, University of
Graz, NAWI Graz, 8010 Graz, Austria
- Field
of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Enrico Federico Semeraro
- Institute
of Molecular Biosciences, University of
Graz, NAWI Graz, 8010 Graz, Austria
- Field
of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Karl Lohner
- Institute
of Molecular Biosciences, University of
Graz, NAWI Graz, 8010 Graz, Austria
- Field
of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Nermina Malanovic
- Institute
of Molecular Biosciences, University of
Graz, NAWI Graz, 8010 Graz, Austria
- Field
of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Georg Pabst
- Institute
of Molecular Biosciences, University of
Graz, NAWI Graz, 8010 Graz, Austria
- Field
of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| |
Collapse
|
6
|
Jennings J, Ašćerić D, Malanovic N, Pabst G. Structure-Activity Relationships of Cationic Lipidoids against Escherichia coli. Antibiotics (Basel) 2023; 12:1300. [PMID: 37627720 PMCID: PMC10451255 DOI: 10.3390/antibiotics12081300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/02/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
Membrane-active molecules provide a promising strategy to target and kill pathogenic bacteria. Understanding how specific molecular features drive interactions with membrane components and subsequently cause disruption that leads to antimicrobial activity is a crucial step in designing next-generation treatments. Here, we test a library of lipid-like compounds (lipidoids) against Gram-negative bacteria Escherichia coli to garner in-depth structure-activity relationships using antimicrobial assays. Modular lipidoid molecules were synthesized in high-throughput, such that we could analyze 104 compounds with variable combinations of hydrophobic tails and cationic headgroups. Antibacterial activity was strongly correlated to specific structural features, including tail hydrophobicity and headgroup charge density, and also to the overall molecular shape and propensity for self-assembly into curved liquid crystalline phases. Dye permeabilization assays showed that E. coli membranes were permeabilized by lipidoids, confirming their membrane-active nature. The reduced permeabilization, as compared to Gram-positive Bacillus subtilis, alludes to the challenge of permeabilizing the additional outer membrane layer of E. coli. The effect of headgroup solubility in gemini-type lipidoids was also demonstrated, revealing that a headgroup with a more hydrophilic spacer between amine groups had enhanced activity against B. subtilis but not E. coli. This provides insight into features enabling outer membrane penetration and governing selectivity between bacterial species.
Collapse
Affiliation(s)
- James Jennings
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, 8010 Graz, Austria
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Dunja Ašćerić
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, 8010 Graz, Austria
| | - Nermina Malanovic
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, 8010 Graz, Austria
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| | - Georg Pabst
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, 8010 Graz, Austria
- Field of Excellence BioHealth, University of Graz, 8010 Graz, Austria
| |
Collapse
|
7
|
Jennings J, Pabst G. Multiple Routes to Bicontinuous Cubic Liquid Crystal Phases Discovered by High-Throughput Self-Assembly Screening of Multi-Tail Lipidoids. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023:e2206747. [PMID: 37026678 DOI: 10.1002/smll.202206747] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/16/2023] [Indexed: 06/19/2023]
Abstract
Bicontinuous cubic phases offer advantageous routes to a broad range of applied materials ranging from drug delivery devices to membranes. However, a priori design of molecules that assemble into these phases remains a technological challenge. In this article, a high-throughput synthesis of lipidoids that undergo protonation-driven self-assembly (PrSA) into liquid crystalline (LC) phases is conducted. With this screening approach, 12 different multi-tail lipidoid structures capable of assembling into the bicontinuous double gyroid phase are discovered. The large volume of small-angle X-ray scattering (SAXS) data uncovers unexpected design criteria that enable phase selection as a function of lipidoid headgroup size and architecture, tail length and architecture, and counterion identity. Surprisingly, combining branched headgroups with bulky tails forces lipidoids to adopt unconventional pseudo-disc conformations that pack into double gyroid networks, entirely distinct from other synthetic or biological amphiphiles within bicontinuous cubic phases. From a multitude of possible applications, two examples of functional materials from lipidoid liquid crystals are demonstrated. First, the fabrication of gyroid nanostructured films by interfacial PrSA, which are rapidly responsive to the external medium. Second, it is shown that colloidally-dispersed lipidoid cubosomes, for example, for drug delivery, are easily assembled using top-down solvent evaporation methods.
Collapse
Affiliation(s)
- James Jennings
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz, 8010, Austria
- Field of Excellence BioHealth, University of Graz, Graz, 8010, Austria
| | - Georg Pabst
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz, 8010, Austria
- Field of Excellence BioHealth, University of Graz, Graz, 8010, Austria
| |
Collapse
|
8
|
Li W, Shao L, Liu J, Sheng J, Zheng Q, Wang M. Intracellular delivery of bacterial effectors for cancer therapy using biodegradable lipid nanoparticles. Biomater Sci 2023; 11:3172-3179. [PMID: 36919841 DOI: 10.1039/d3bm00008g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Bacterial effector proteins are virulence factors that are secreted and mediate orthogonal post-translational modifications of proteins that are not found naturally in mammalian systems. They hold great promise for developing biotherapeutics by regulating malignant cell signaling in a specific and targeted manner. However, delivering bacterial effectors into disease cells poses a significant challenge to their therapeutic potential. In this study, we report on the design of a combinatorial library of bioreducible lipid nanoparticles containing disulfide bonds for highly efficient bacterial effector delivery and potential cancer therapy. A leading lipid, PPPDA-O16B, identified from the library, can encapsulate and deliver DNA plasmids into cells. The gene cargo is released in response to the reductive cellular environment that is upregulated in cancer cells, leading to enhanced gene delivery and protein expression efficiency. Furthermore, we demonstrate that PPPDA-O16B can deliver the bacterial effector protein, DUF5, to degrade mutant RAS and inactivate downstream MAPK signaling cascades to suppress cancer cell growth in vitro and in tumor-bearing mouse xenografts. This strategy of delivering bacterial effectors using biodegradable lipid nanoparticles can be expanded for cancer cell signaling regulation and antitumor studies.
Collapse
Affiliation(s)
- Wenting Li
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Leihou Shao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ji Liu
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinhan Sheng
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qizhen Zheng
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ming Wang
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
9
|
Khirallah J, Eimbinder M, Li Y, Xu Q. Clinical progress in genome-editing technology and in vivo delivery techniques. Trends Genet 2023; 39:208-216. [PMID: 36669950 PMCID: PMC9974761 DOI: 10.1016/j.tig.2022.12.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 11/15/2022] [Accepted: 12/15/2022] [Indexed: 01/20/2023]
Abstract
There is wide interest in applying genome-editing tools to prevent, treat, and cure a variety of diseases. Since the discovery of the clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein (Cas) systems, these techniques have been used in combination with different delivery systems to create highly efficacious treatment options. Each delivery system has its own advantages and disadvantages and is being used for various applications. With the large number of gene-editing applications being studied but very few being brought into the clinic, we review current progress in the field, specifically where genome editing has been applied in vivo and in the clinic, and identify current challenges and areas of future growth.
Collapse
Affiliation(s)
- Jennifer Khirallah
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Maximilan Eimbinder
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Yamin Li
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA.
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
10
|
Li Y, Ye Z, Yang H, Xu Q. Tailoring combinatorial lipid nanoparticles for intracellular delivery of nucleic acids, proteins, and drugs. Acta Pharm Sin B 2022; 12:2624-2639. [PMID: 35755280 PMCID: PMC9214058 DOI: 10.1016/j.apsb.2022.04.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 03/17/2022] [Accepted: 04/11/2022] [Indexed: 12/15/2022] Open
Abstract
Lipid nanoparticle (LNP)-based drug delivery systems have become the most clinically advanced non-viral delivery technology. LNPs can encapsulate and deliver a wide variety of bioactive agents, including the small molecule drugs, proteins and peptides, and nucleic acids. However, as the physicochemical properties of small- and macromolecular cargos can vary drastically, every LNP carrier system needs to be carefully tailored in order to deliver the cargo molecules in a safe and efficient manner. Our group applied the combinatorial library synthesis approach and in vitro and in vivo screening strategy for the development of LNP delivery systems for drug delivery. In this Review, we highlight our recent progress in the design, synthesis, characterization, evaluation, and optimization of combinatorial LNPs with novel structures and properties for the delivery of small- and macromolecular therapeutics both in vitro and in vivo. These delivery systems have enormous potentials for cancer therapy, antimicrobial applications, gene silencing, genome editing, and more. We also discuss the key challenges to the mechanistic study and clinical translation of new LNP-enabled therapeutics.
Collapse
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
- Department of Pharmacology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA
| | - Zhongfeng Ye
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Hanyi Yang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| |
Collapse
|
11
|
Bloomer H, Khirallah J, Li Y, Xu Q. CRISPR/Cas9 ribonucleoprotein-mediated genome and epigenome editing in mammalian cells. Adv Drug Deliv Rev 2022; 181:114087. [PMID: 34942274 PMCID: PMC8844242 DOI: 10.1016/j.addr.2021.114087] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/15/2021] [Accepted: 12/16/2021] [Indexed: 02/03/2023]
Abstract
The clustered regularly interspaced short palindromic repeat (CRISPR)/CRISPR-associated protein (Cas) system has revolutionized the ability to edit the mammalian genome, providing a platform for the correction of pathogenic mutations and further investigation into gene function. CRISPR reagents can be delivered into the cell as DNA, RNA, or pre-formed ribonucleoproteins (RNPs). RNPs offer numerous advantages over other delivery approaches due to their ability to rapidly target genomic sites and quickly degrade thereafter. Here, we review the production steps and delivery methods for Cas9 RNPs. Additionally, we discuss how RNPs enhance genome and epigenome editing efficiencies, reduce off-target editing activity, and minimize cellular toxicity in clinically relevant mammalian cell types. We include details on a broad range of editing approaches, including novel base and prime editing techniques. Finally, we summarize key challenges for the use of RNPs, and propose future perspectives on the field.
Collapse
Affiliation(s)
- Hanan Bloomer
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, US,School of Medicine and Graduate School of Biomedical Sciences, Tufts University, Boston, MA 02111, US
| | - Jennifer Khirallah
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, US
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, US,Corresponding Authors: (Y. Li) and (Q. Xu)
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, US,Corresponding Authors: (Y. Li) and (Q. Xu)
| |
Collapse
|
12
|
Subhan A, Attia SA, P Torchilin V. Targeted siRNA nanotherapeutics against breast and ovarian metastatic cancer: a comprehensive review of the literature. Nanomedicine (Lond) 2021; 17:41-64. [PMID: 34930021 DOI: 10.2217/nnm-2021-0207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Metastasis is considered the major cause of unsuccessful cancer therapy. The metastatic development requires tumor cells to leave their initial site, circulate in the blood stream, acclimate to new cellular environments at a remote secondary site and endure there. There are several steps in metastasis, including invasion, intravasation, circulation, extravasation, premetastatic niche formation, micrometastasis and metastatic colonization. siRNA therapeutics are appreciated for their usefulness in treatment of cancer metastasis. However, siRNA therapy as a single therapy may not be a sufficient option for control of metastasis. By combining siRNA with targeting, functional agents or small-molecule drugs have shown potential effects that enhance therapeutic effectiveness. This review addresses multidrug resistance and metastasis in breast and ovarian cancers and highlights drug-delivery strategies using siRNA therapeutics.
Collapse
Affiliation(s)
- Abdus Subhan
- Department of Chemistry, ShahJalal University of Science & Technology, Sylhet 3114, Bangladesh
| | - Sara Aly Attia
- Center for Pharmaceutical Biotechnology and Nanomedicine, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA.,Department of Oncology, Radiotherapy & Plastic Surgery, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia
| |
Collapse
|
13
|
Tomé I, Francisco V, Fernandes H, Ferreira L. High-throughput screening of nanoparticles in drug delivery. APL Bioeng 2021; 5:031511. [PMID: 34476328 PMCID: PMC8397474 DOI: 10.1063/5.0057204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/30/2021] [Indexed: 12/19/2022] Open
Abstract
The use of pharmacologically active compounds to manage and treat diseases is of utmost relevance in clinical practice. It is well recognized that spatial-temporal control over the delivery of these biomolecules will greatly impact their pharmacokinetic profile and ultimately their therapeutic effect. Nanoparticles (NPs) prepared from different materials have been tested successfully in the clinic for the delivery of several biomolecules including non-coding RNAs (siRNA and miRNA) and mRNAs. Indeed, the recent success of mRNA vaccines is in part due to progress in the delivery systems (NP based) that have been developed for many years. In most cases, the identification of the best formulation was done by testing a small number of novel formulations or by modification of pre-existing ones. Unfortunately, this is a low throughput and time-consuming process that hinders the identification of formulations with the highest potential. Alternatively, high-throughput combinatorial design of NP libraries may allow the rapid identification of formulations with the required release and cell/tissue targeting profile for a given application. Combinatorial approaches offer several advantages over conventional methods since they allow the incorporation of multiple components with varied chemical properties into materials, such as polymers or lipid-like materials, that will subsequently form NPs by self-assembly or chemical conjugation processes. The current review highlights the impact of high-throughput in the development of more efficient drug delivery systems with enhanced targeting and release kinetics. It also describes the current challenges in this research area as well as future directions.
Collapse
Affiliation(s)
| | - Vitor Francisco
- Biomaterials and Stem-Cell Based Therapeutics Group, Centre of Neuroscience and Cell Biology, University of Coimbra, 3060-197 Cantanhede, Portugal
| | | | - Lino Ferreira
- Authors to whom correspondence should be addressed: and
| |
Collapse
|
14
|
Kim J, Vaughan HJ, Zamboni CG, Sunshine JC, Green JJ. High-throughput evaluation of polymeric nanoparticles for tissue-targeted gene expression using barcoded plasmid DNA. J Control Release 2021; 337:105-116. [PMID: 34097924 DOI: 10.1016/j.jconrel.2021.05.047] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 05/03/2021] [Accepted: 05/31/2021] [Indexed: 12/17/2022]
Abstract
Successful systemic gene delivery requires specific tissue targeting as well as efficient intracellular transfection. Increasingly, research laboratories are fabricating libraries of novel nanoparticles, engineering both new biomaterial structures and composition ratios of multicomponent systems. Yet, methods for screening gene delivery vehicles directly in vivo are often low-throughout, limiting the number of candidate nanoparticles that can be investigated. Here, we report a comprehensive, high-throughput method to evaluate a library of polymeric nanoparticles in vivo for tissue-specific gene delivery. The method involves pairing each nanoparticle formulation with a plasmid DNA (pDNA) that harbors a unique nucleotide sequence serving as the identifying "barcode". Using real time quantitative PCR (qPCR) for detection of the barcoded pDNA and quantitative reverse transcription PCR (RT-qPCR) for transcribed barcoded mRNA, we can quantify accumulation and transfection in tissues of interest. The barcode pDNA and primers were designed with sufficient sensitivity and specificity to evaluate multiple nanoparticle formulations per mouse, improving screening efficiency. Using this platform, we evaluated the biodistribution and transfection of 8 intravenously administered poly(beta-amino ester; PBAE) nanoparticle formulations, each with a PBAE polymer of differential structure. Significant levels of nanoparticle accumulation and gene transfection were observed mainly in organs involved in clearance, including spleen, liver, and kidneys. Interestingly, higher levels of transfection of select organs did not necessarily correlate with higher levels of tissue accumulation, highlighting the importance of directly measuring in vivo transfection efficiency as the key barcoded parameter in gene delivery vector optimization. To validate this method, nanoparticle formulations were used individually for luciferase pDNA delivery in vivo. The distribution of luciferase expression in tissues matched the transfection analysis by the barcode qPCR method, confirming that this platform can be used to accurately evaluate systemic gene delivery.
Collapse
Affiliation(s)
- Jayoung Kim
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Hannah J Vaughan
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Camila G Zamboni
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Joel C Sunshine
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Pathology, Johns Hopkins University, Baltimore, MD, USA
| | - Jordan J Green
- Department of Biomedical Engineering and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, USA; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA; Departments of Neurosurgery, Oncology, and Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
15
|
Osteogenic effects of microRNA-335-5p/lipidoid nanoparticles coated on titanium surface. Arch Oral Biol 2021; 129:105207. [PMID: 34273868 DOI: 10.1016/j.archoralbio.2021.105207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 06/26/2021] [Accepted: 07/03/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE In this study, we aimed to investigate the therapeutic potential of miR-335-5p lipidoid nanocomplexes coated on Titanium (Ti) SLActive surface by lyophilization. DESIGN In our model, we coated miR-335-5p/Lipidoid nanoparticles on titanium implant, seeded GFP-labelled mouse bone marrow stromal cells (BMSCs) onto the functionalized Ti implant surface, and analyzed the transfection efficiency, cell adhesion, proliferation, and osteogenic activity of the bone-implant interface. RESULTS The Ti SLActive surface displayed a suitable hydrophilicity ability and provided a large surface area for miRNA loading, enabling spatial retention of the miRNAs within the nanopores until cellular delivery. We demonstrated a high transfection efficiency of miR-335-5p lipidoid nanoparticles in BMSCs seeded onto the Ti SLActive surface, even after 14 days. Alkaline phosphatase (ALP) activity and cell vitality were significantly increased in BMSCs transfected with miR-335-5p at 7 and 14 days as opposed to cells transfected with negative controls. When miR-335-5p transfected BMSCs were induced to undergo osteogenic differentiation, we detected increased mRNA expression of osteogenic markers including Alkaline phosphatase (ALP), collagen I (COL1), osteocalcin (OCN) and bone sialoprotein (BSP) at 7 and 14 days as compared with negative controls. CONCLUSION MiR-335-5p lipidoid nanoparticles could be used as a new cost-effective methodology to increase the osteogenic capacity of biomedical Ti implants.
Collapse
|
16
|
Zu H, Gao D. Non-viral Vectors in Gene Therapy: Recent Development, Challenges, and Prospects. AAPS JOURNAL 2021; 23:78. [PMID: 34076797 PMCID: PMC8171234 DOI: 10.1208/s12248-021-00608-7] [Citation(s) in RCA: 223] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/07/2021] [Indexed: 12/16/2022]
Abstract
Gene therapy has been experiencing a breakthrough in recent years, targeting various specific cell groups in numerous therapeutic areas. However, most recent clinical studies maintain the use of traditional viral vector systems, which are challenging to manufacture cost-effectively at a commercial scale. Non-viral vectors have been a fast-paced research topic in gene delivery, such as polymers, lipids, inorganic particles, and combinations of different types. Although non-viral vectors are low in their cytotoxicity, immunogenicity, and mutagenesis, attracting more and more researchers to explore the promising delivery system, they do not carry ideal characteristics and have faced critical challenges, including gene transfer efficiency, specificity, gene expression duration, and safety. This review covers the recent advancement in non-viral vectors research and formulation aspects, the challenges, and future perspectives.
Collapse
Affiliation(s)
- Hui Zu
- Abbvie Inc., 1 N. Waukegan Rd, North Chicago, Illinois, 60064, USA
| | - Danchen Gao
- Abbvie Inc., 1 N. Waukegan Rd, North Chicago, Illinois, 60064, USA.
| |
Collapse
|
17
|
Zhao X, Glass Z, Chen J, Yang L, Kaplan DL, Xu Q. mRNA Delivery Using Bioreducible Lipidoid Nanoparticles Facilitates Neural Differentiation of Human Mesenchymal Stem Cells. Adv Healthc Mater 2021; 10:e2000938. [PMID: 32815325 DOI: 10.1002/adhm.202000938] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/12/2020] [Indexed: 12/22/2022]
Abstract
Mesenchymal stem cells (MSCs) are widely used in regenerative medicine and tissue engineering and delivering biological molecules into MSCs has been used to control stem cell behavior. However, the efficient delivery of large biomolecules such as DNA, RNA, and proteins into MSCs using nonviral delivery strategies remains an ongoing challenge. Herein, nanoparticles composed of cationic bioreducible lipid-like materials (lipidoids) are developed to intracellularly deliver mRNA into human mesenchymal stem cells (hMSCs). The delivery efficacy to hMSCs is improved by adding three excipients including cholesterol, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-polyethylene glycol (DSPE-PEG) during lipidoid nanoparticle formulation. Using an optimized lipidoid formulation, Cas9 mRNA and single guide RNA (sgRNA) targeting neuron restrictive silencing factor (NRSF) are delivered to hMSCs, leading to successful neural-like differentiation as demonstrated by the expression of synaptophysin (SYP), brain-derived neurotrophic factor (BDNF), neuron-specific enolase (NSE), and neuron-specific growth-associated protein (SCG10). Overall, a synthetic lipid formulation that can efficiently deliver mRNA to hMSCs is identified, leading to CRISPR-based gene knockdown to facilitate hMSCs transdifferentiation into neural-like lineage.
Collapse
Affiliation(s)
- Xuewei Zhao
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - Zachary Glass
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - Jinjin Chen
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - Liu Yang
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - David L. Kaplan
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| | - Qiaobing Xu
- Department of Biomedical Engineering Tufts University 4 Colby Street Medford MA 02155 USA
| |
Collapse
|
18
|
Ponti F, Campolungo M, Melchiori C, Bono N, Candiani G. Cationic lipids for gene delivery: many players, one goal. Chem Phys Lipids 2021; 235:105032. [PMID: 33359210 DOI: 10.1016/j.chemphyslip.2020.105032] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/23/2020] [Accepted: 12/19/2020] [Indexed: 12/28/2022]
Abstract
Lipid-based carriers represent the most widely used alternative to viral vectors for gene expression and gene silencing purposes. This class of non-viral vectors is particularly attractive for their ease of synthesis and chemical modifications to endow them with desirable properties. Despite combinatorial approaches have led to the generation of a large number of cationic lipids displaying different supramolecular structures and improved behavior, additional effort is needed towards the development of more and more effective cationic lipids for transfection purposes. With this review, we seek to highlight the great progress made in the design of each and every constituent domain of cationic lipids, that is, the chemical structure of the headgroup, linker and hydrophobic moieties, and on the specific effect on the assembly with nucleic acids. Since the complexity of such systems is known to affect their performances, the role of formulation, stability and phase behavior on the transfection efficiency of such assemblies will be thoroughly discussed. Our objective is to provide a conceptual framework for the development of ever more performing lipid gene delivery vectors.
Collapse
Affiliation(s)
- Federica Ponti
- GenT LΛB, Dept. of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, 20131, Milan, Italy; Laboratory for Biomaterials and Bioengineering, Canada Research Chair I in Biomaterials and Bioengineering for the Innovation in Surgery, Dept. Min-Met-Materials Engineering, Research Center of CHU de Quebec, Division of Regenerative Medicine, Laval University, Quebec City, QC, Canada
| | - Matilde Campolungo
- GenT LΛB, Dept. of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, 20131, Milan, Italy
| | - Clara Melchiori
- GenT LΛB, Dept. of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, 20131, Milan, Italy
| | - Nina Bono
- GenT LΛB, Dept. of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, 20131, Milan, Italy.
| | - Gabriele Candiani
- GenT LΛB, Dept. of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, 20131, Milan, Italy.
| |
Collapse
|
19
|
Zhao X, Chen J, Qiu M, Li Y, Glass Z, Xu Q. Imidazole‐Based Synthetic Lipidoids for In Vivo mRNA Delivery into Primary T Lymphocytes. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202008082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Xuewei Zhao
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Jinjin Chen
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Min Qiu
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Yamin Li
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Zachary Glass
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Qiaobing Xu
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| |
Collapse
|
20
|
Zhao X, Chen J, Qiu M, Li Y, Glass Z, Xu Q. Imidazole‐Based Synthetic Lipidoids for In Vivo mRNA Delivery into Primary T Lymphocytes. Angew Chem Int Ed Engl 2020; 59:20083-20089. [DOI: 10.1002/anie.202008082] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Indexed: 02/06/2023]
Affiliation(s)
- Xuewei Zhao
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Jinjin Chen
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Min Qiu
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Yamin Li
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Zachary Glass
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| | - Qiaobing Xu
- Department of Biomedical Engineering Tufts University 4 Colby St. Medford MA 02155 USA
| |
Collapse
|
21
|
Jennings J, Carter MCD, Son CY, Cui Q, Lynn DM, Mahanthappa MK. Protonation-Driven Aqueous Lyotropic Self-Assembly of Synthetic Six-Tail Lipidoids. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:8240-8252. [PMID: 32649210 DOI: 10.1021/acs.langmuir.0c01369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
We report the aqueous lyotropic mesophase behaviors of protonated amine-based "lipidoids," a class of synthetic lipid-like molecules that mirrors essential structural features of the multitail bacterial amphiphile lipid A. Small-angle X-ray scattering (SAXS) studies demonstrate that the protonation of the tetra(amine) headgroups of six-tail lipidoids in aqueous HCl, HNO3, H2SO4, and H3PO4 solutions variably drives their self-assembly into lamellar (Lα) and inverse micellar (III) lyotropic liquid crystals (LLCs), depending on acid identity and concentration, amphiphile tail length, and temperature. Lipidoid assemblies formed in H2SO4(aq) exhibit rare inverse body-centered cubic (BCC) and inverse face-centered cubic (FCC) micellar morphologies, the latter of which unexpectedly coexists with zero mean curvature Lα phases. Complementary atomistic molecular dynamics (MD) simulations furnish detailed insights into this unusual self-assembly behavior. The unique aqueous lyotropic mesophase behaviors of ammonium lipidoids originate in their dichotomous ability to adopt both inverse conical and chain-extended molecular conformations depending on the number of counterions and their identity, which lead to coexisting supramolecular assemblies with remarkably different mean interfacial curvatures.
Collapse
Affiliation(s)
| | | | | | | | | | - Mahesh K Mahanthappa
- Department of Chemical Engineering & Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
22
|
Wang Z, Liu A, Zhang H, Wang M, Tang Q, Huang Y, Wang L. Inhibition of retinal neovascularization by VEGF siRNA delivered via bioreducible lipid-like nanoparticles. Graefes Arch Clin Exp Ophthalmol 2020; 258:2407-2418. [PMID: 32620992 DOI: 10.1007/s00417-020-04797-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 06/02/2020] [Accepted: 06/08/2020] [Indexed: 12/20/2022] Open
Abstract
PURPOSE Previously, we have demonstrated the use of lipidoid (lipid-like) nanoparticles (e.g., "1-O16B") for gene delivery to live cells, as an alternative to viral vectors. Here, we encapsulate VEGF siRNA (siVEGF) in bioreducible lipidoid nanoparticles and examine whether these nanocomplexes can reduce intravitreal neovascularization in a rodent model of oxygen-induced retinopathy (OIR). METHODS Firstly, we constructed siVEGF-nanoparticles (NPs) and transfected human umbilical vein endothelial cells, which caused significantly reduced expression of VEGF, compared to exposure to siVEGF in solution. Secondly, we compared the effect of intravitreal siVEGF-NPs and an anti-VEGF drug (ranibizumab) on retinal vascular development and VEGF mRNA/protein expression in the retinas of a rat model of OIR. RESULTS Compared to a non-functional lipid vehicle control group, the level of VEGF mRNA and protein was significantly lower in the siVEGF-NP group (p < 0.01), but the level of VEGF mRNA was not significantly lower in the ranibizumab group. Anatomically, the number of retinal neovascular endothelial nuclei that had protruded through the internal limiting membrane and the number of areas of non-perfusion of the retina were both significantly lower in the siVEGF-NP group and the ranibizumab group than in the OIR group (p < 0.01). CONCLUSION Our results demonstrate that bioreducible lipidoid nanoparticles conveying VEGF siRNA can effectively inhibit retinal neovascularization in a rodent model of OIR, and reduce the expression of VEGF mRNA and protein. This novel treatment modality could have profound implications for treating retinal angiogenic diseases.
Collapse
Affiliation(s)
- Zonghua Wang
- Department of Ophthalmology, Chinese People's Liberation Army General Hospital, Fuxing Road 28, Beijing, 100853, China.,Department of Ophthalmology, The Seventh Medical center of PLA General Hospital, Beijing, 100700, China
| | - Anqi Liu
- Department of Ophthalmology, Chinese People's Liberation Army General Hospital, Fuxing Road 28, Beijing, 100853, China.,The Chinese People's Liberation Army NO.32141, Beijing, China
| | - Han Zhang
- The 2nd Hospital of Shandong University, 247 Beiyuan Road, Jinan, 250031, China
| | - Ming Wang
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100049, China.,Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, the Chinese Academy of Sciences (CAS), Beijing, 100190, China
| | - Qiao Tang
- Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100049, China.,Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, the Chinese Academy of Sciences (CAS), Beijing, 100190, China
| | - Yifei Huang
- Department of Ophthalmology, Chinese People's Liberation Army General Hospital, Fuxing Road 28, Beijing, 100853, China.
| | - Liqiang Wang
- Department of Ophthalmology, Chinese People's Liberation Army General Hospital, Fuxing Road 28, Beijing, 100853, China.
| |
Collapse
|
23
|
Liu F, Yang L, Li Y, Junier A, Ma F, Chen J, Han H, Glass Z, Zhao X, Kumamoto CA, Sang H, Xu Q. In Vitro and In Vivo Study of Amphotericin B Formulation with Quaternized Bioreducible Lipidoids. ACS Biomater Sci Eng 2020; 6:1064-1073. [DOI: 10.1021/acsbiomaterials.9b01722] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Fang Liu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
- Southern Medical University, Jinling Hospital Department of Dermatology, Nanjing, 210002, China
| | - Liu Yang
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Ashlee Junier
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts 02111, United States
| | - Feihe Ma
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Jinjin Chen
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Haobo Han
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, P. R. China
| | - Zachary Glass
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Xuewei Zhao
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| | - Carol A. Kumamoto
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts 02111, United States
| | - Hong Sang
- Southern Medical University, Jinling Hospital Department of Dermatology, Nanjing, 210002, China
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
24
|
Buck J, Grossen P, Cullis PR, Huwyler J, Witzigmann D. Lipid-Based DNA Therapeutics: Hallmarks of Non-Viral Gene Delivery. ACS NANO 2019; 13:3754-3782. [PMID: 30908008 DOI: 10.1021/acsnano.8b07858] [Citation(s) in RCA: 226] [Impact Index Per Article: 37.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Gene therapy is a promising strategy for the treatment of monogenic disorders. Non-viral gene delivery systems including lipid-based DNA therapeutics offer the opportunity to deliver an encoding gene sequence specifically to the target tissue and thus enable the expression of therapeutic proteins in diseased cells. Currently, available gene delivery approaches based on DNA are inefficient and require improvements to achieve clinical utility. In this Review, we discuss state-of-the-art lipid-based DNA delivery systems that have been investigated in a preclinical setting. We emphasize factors influencing the delivery and subsequent gene expression in vitro, ex vivo, and in vivo. In addition, we cover aspects of nanoparticle engineering and optimization for DNA therapeutics. Finally, we highlight achievements of lipid-based DNA therapies in clinical trials.
Collapse
Affiliation(s)
- Jonas Buck
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences , University of Basel , Klingelbergstrasse 50 , 4056 Basel , Switzerland
| | - Philip Grossen
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences , University of Basel , Klingelbergstrasse 50 , 4056 Basel , Switzerland
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology , University of British Columbia , 2350 Health Sciences Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences , University of Basel , Klingelbergstrasse 50 , 4056 Basel , Switzerland
| | - Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences , University of Basel , Klingelbergstrasse 50 , 4056 Basel , Switzerland
- Department of Biochemistry and Molecular Biology , University of British Columbia , 2350 Health Sciences Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| |
Collapse
|
25
|
Carboni V, Maaliki C, Alyami M, Alsaiari S, Khashab N. Synthetic Vehicles for Encapsulation and Delivery of CRISPR/Cas9 Gene Editing Machinery. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201800085] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Valentina Carboni
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials CenterKing Abdullah University of Science and Technology (KAUST) Thuwal 23955‐6900 Saudi Arabia
| | - Carine Maaliki
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials CenterKing Abdullah University of Science and Technology (KAUST) Thuwal 23955‐6900 Saudi Arabia
| | - Mram Alyami
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials CenterKing Abdullah University of Science and Technology (KAUST) Thuwal 23955‐6900 Saudi Arabia
| | - Shahad Alsaiari
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials CenterKing Abdullah University of Science and Technology (KAUST) Thuwal 23955‐6900 Saudi Arabia
| | - Niveen Khashab
- Smart Hybrid Materials Laboratory (SHMs), Advanced Membranes and Porous Materials CenterKing Abdullah University of Science and Technology (KAUST) Thuwal 23955‐6900 Saudi Arabia
| |
Collapse
|
26
|
Lin Z, Bao M, Yu Z, Xue L, Ju C, Zhang C. The development of tertiary amine cationic lipids for safe and efficient siRNA delivery. Biomater Sci 2019; 7:2777-2792. [DOI: 10.1039/c9bm00494g] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tertiary amine-derived cationic lipid serves as the primary lipid of cationic liposomes, which can balance the effectiveness and safety of siRNA vectors.
Collapse
Affiliation(s)
- Ziming Lin
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of New Drug Discovery
- China Pharmaceutical University
- Nanjing
- China
| | - Moxyel Bao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of New Drug Discovery
- China Pharmaceutical University
- Nanjing
- China
| | - Zexuan Yu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of New Drug Discovery
- China Pharmaceutical University
- Nanjing
- China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of New Drug Discovery
- China Pharmaceutical University
- Nanjing
- China
| | - Caoyun Ju
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of New Drug Discovery
- China Pharmaceutical University
- Nanjing
- China
| | - Can Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases
- Center of New Drug Discovery
- China Pharmaceutical University
- Nanjing
- China
| |
Collapse
|
27
|
Li Y, Yang T, Yu Y, Shi N, Yang L, Glass Z, Bolinger J, Finkel IJ, Li W, Xu Q. Combinatorial library of chalcogen-containing lipidoids for intracellular delivery of genome-editing proteins. Biomaterials 2018; 178:652-662. [PMID: 29549971 DOI: 10.1016/j.biomaterials.2018.03.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/19/2022]
Abstract
Protein based therapeutics with high specificities and low off-target effects are used for transient and accurate manipulation of cell functions. However, developing safe and efficient carriers for intracellular delivery of active therapeutic proteins is a long-standing challenge. Here we report a combinatorial library of chalcogen (O, S, Se) containing lipidoid nanoparticles (LNPs) as efficient nanocarriers for intracellular delivery of negatively supercharged Cre recombinase ((-30)GFP-Cre) and anionic Cas9:single-guide RNA (Cas9:sgRNA) ribonucleoprotein (RNP) for genome editing. The structure-activity relationship between the lipidoids and intracellular protein delivery efficiencies was explored and it was demonstrated that the newly developed LNPs are effective for gene recombination in vivo.
Collapse
Affiliation(s)
- Yamin Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Tao Yang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610065, PR China
| | - Yingjie Yu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Nicola Shi
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Liu Yang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Zachary Glass
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Justin Bolinger
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Isaac James Finkel
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Wenhan Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
28
|
Damen M, Groenen AJJ, van Dongen SFM, Nolte RJM, Scholte BJ, Feiters MC. Transfection by cationic gemini lipids and surfactants. MEDCHEMCOMM 2018; 9:1404-1425. [PMID: 30288217 PMCID: PMC6148748 DOI: 10.1039/c8md00249e] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/11/2018] [Indexed: 12/13/2022]
Abstract
Diseases that are linked to defective genes or mutations can in principle be cured by gene therapy, in which damaged or absent genes are either repaired or replaced by new DNA in the nucleus of the cell. Related to this, disorders associated with elevated protein expression levels can be treated by RNA interference via the delivery of siRNA to the cytoplasm of cells. Polynucleotides can be brought into cells by viruses, but this is not without risk for the patient. Alternatively, DNA and RNA can be delivered by transfection, i.e. by non-viral vector systems such as cationic surfactants, which are also referred to as cationic lipids. In this review, recent progress on cationic lipids as transfection vectors will be discussed, with special emphasis on geminis, surfactants with 2 head groups and 2 tails connected by a spacer.
Collapse
Affiliation(s)
- M Damen
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| | - A J J Groenen
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| | - S F M van Dongen
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| | - R J M Nolte
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| | - B J Scholte
- Departments of Pediatric pulmonology and Cell Biology , Erasmus MC, P. O. Box 2040 , 3000 CA Rotterdam , The Netherlands
| | - M C Feiters
- Institute for Molecules and Materials , Faculty of Science , Radboud University , Heyendaalseweg 135 , 6525 AJ Nijmegen , The Netherlands .
| |
Collapse
|
29
|
Altınoğlu SA, Wang M, Li KQ, Li Y, Xu Q. Intracellular delivery of the PTEN protein using cationic lipidoids for cancer therapy. Biomater Sci 2018; 4:1773-1780. [PMID: 27748775 DOI: 10.1039/c6bm00580b] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a tumor suppressor, mutated or inactive in a large percentage of human cancers. Restoring PTEN activity in cancer cells through gene therapy has shown to inhibit cell growth and induce apoptosis, particularly in cells with a PTEN deficiency. Gene therapy, however, comes with some inherent risks such as triggering an immune response and permanent off target effects. Nanoparticle assisted protein delivery could mitigate these liabilities while maintaining therapeutic integrity. In this report, we evaluated the use of cationic lipid-like (lipidoid) materials to intracellularly deliver the PTEN protein. We synthesized a small library of cationic lipidoid materials and screened for the delivery of PTEN based on cell viability. The lipidoid material EC16-80 was selected for high efficacy and the subsequent lipidoid-protein complex was characterized using DLS, zeta potential, and TEM. Intracellular delivery of PTEN with EC16-80 to the PTEN deficient prostate cancer cell line PC-3 resulted in a significant decrease in activated AKT and induced apoptosis. Interestingly, delivery of PTEN to PTEN deficient prostate cancer cell lines PC-3 and LNCaP compared to the breast cancer cell line, MCF-7 with endogenous PTEN, resulted in significantly lower IC50 values in PC-3 and LNCaP cells indicating that the treatment is predominantly specific to PTEN-deficient cells. Altogether, these results demonstrate the first intracellular delivery of recombinant PTEN using a synthetic delivery vehicle and highlight the potential of intracellular PTEN protein delivery as a potential targeted cancer therapy.
Collapse
Affiliation(s)
- Sarah A Altınoğlu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| | - Ming Wang
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| | - Kathleen Q Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| | - Yuyang Li
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| | - Qiaobing Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA.
| |
Collapse
|
30
|
de Groot AM, Thanki K, Gangloff M, Falkenberg E, Zeng X, van Bijnen DCJ, van Eden W, Franzyk H, Nielsen HM, Broere F, Gay NJ, Foged C, Sijts AJAM. Immunogenicity Testing of Lipidoids In Vitro and In Silico: Modulating Lipidoid-Mediated TLR4 Activation by Nanoparticle Design. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:159-169. [PMID: 29858051 PMCID: PMC5992342 DOI: 10.1016/j.omtn.2018.02.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 02/06/2018] [Accepted: 02/06/2018] [Indexed: 11/28/2022]
Abstract
Therapeutics based on small interfering RNA (siRNA) have promising potential as antiviral and anti-inflammatory agents. To deliver siRNA across cell membranes to reach the RNAi pathway in the cytosol of target cells, non-viral nanoparticulate delivery approaches are explored. Recently, we showed that encapsulation of siRNA in lipid-polymer hybrid nanoparticles (LPNs), based on poly(DL-lactic-co-glycolic acid) (PLGA) and cationic lipid-like materials (lipidoids), remarkably enhances intracellular delivery of siRNA as compared to siRNA delivery with LPNs modified with dioleoyltrimethylammoniumpropane (DOTAP) as the lipid component. However, the potential immune modulation by these cationic lipids remains unexplored. By testing lipidoids and DOTAP for innate immune-receptor-activating properties in vitro, we found that neither lipidoids nor DOTAP activate human Toll-like receptor (TLR) 2, 3, 7, and 9. However, in contrast to DOTAP, lipidoids are strong agonists for TLR4 and activate murine antigen-presenting cells in vitro. This agonistic effect was further confirmed in silico using a prediction model based on crystal structures. Also, lipidoids formulated as lipoplexes or as stable nucleic acid lipid particles, which was the reference formulation for siRNA delivery, proved to activate TLR4. However, by combining lipidoids with PLGA into LPNs, TLR4 activation was abrogated. Thus, lipidoid-mediated TLR4 activation during siRNA delivery may be modulated via optimization of the formulation design.
Collapse
Affiliation(s)
- Anne Marit de Groot
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands
| | - Kaushik Thanki
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Monique Gangloff
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Emily Falkenberg
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Xianghui Zeng
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Djai C J van Bijnen
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands
| | - Willem van Eden
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Hanne M Nielsen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Femke Broere
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands
| | - Nick J Gay
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark.
| | - Alice J A M Sijts
- Department of Infectious Diseases and Immunology, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
31
|
A novel Lipidoid-MicroRNA formulation promotes calvarial bone regeneration. Biomaterials 2018; 177:88-97. [PMID: 29886386 DOI: 10.1016/j.biomaterials.2018.05.038] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/20/2018] [Accepted: 05/22/2018] [Indexed: 11/23/2022]
Abstract
Specific microRNAs (miRs) and the Wnt signaling pathway play critical roles in regulating bone development and homeostasis. Our previous studies revealed the ability of miR-335-5p to promote osteogenic differentiation by downregulating Wnt antagonist Dickkopf-1 (DKK1). The purpose of this study was to use nano-materials to efficiently deliver miR-335-5p into osteogenic cells for tissue engineering applications. We synthesized and screened a library of 12 candidate nano-lipidoids,of which L8 was identified as the preferred biodegradable lipidoid for miRNA molecule delivery into cells. We then investigated whether a lipidoid-miRNA formulation of miR-335-5-p (LMF-335) could successfully deliver miR-335-5-p into cells to promote osteogenesis in vitro and calvarial bone healing in vivo. Transfection of C3H10T1/2 cells and bone marrow stromal cells (BMSCs) with LMF-335 led to decreased expression of DKK1 and increased expression of the key osteogenic genes. LMF-335 and LMF-335-transfected BMSCs were then used in combination with silk scaffolds to evaluate healing of critical-size calvarial bone defects in mice. The results revealed significant new bone formation in the defects in LMF-335 groups as compared with control groups. In conclusion, this first report supports the notion that lipidoid delivery of miRNA can be used to induce osteogenic differentiation of stem cells and bone regeneration.
Collapse
|
32
|
Krivitsky A, Polyak D, Scomparin A, Eliyahu S, Ofek P, Tiram G, Kalinski H, Avkin-Nachum S, Feiner Gracia N, Albertazzi L, Satchi-Fainaro R. Amphiphilic poly(α)glutamate polymeric micelles for systemic administration of siRNA to tumors. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2018; 14:303-315. [PMID: 29127036 DOI: 10.1016/j.nano.2017.10.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 10/08/2017] [Accepted: 10/30/2017] [Indexed: 01/27/2023]
Abstract
RNAi therapeutics carried a great promise to the area of personalized medicine: the ability to target "undruggable" oncogenic pathways. Nevertheless, their efficient tumor targeting via systemic administration had not been resolved yet. Amphiphilic alkylated poly(α)glutamate amine (APA) can serve as a cationic carrier to the negatively-charged oligonucleotides. APA polymers complexed with siRNA to form round-shaped, homogenous and reproducible nano-sized polyplexes bearing ~50 nm size and slightly negative charge. In addition, APA:siRNA polyplexes were shown to be potent gene regulators in vitro. In light of these preferred physico-chemical characteristics, their performance as systemically-administered siRNA nanocarriers was investigated. Intravenously-injected APA:siRNA polyplexes accumulated selectively in tumors and did not accumulate in the lungs, heart, liver or spleen. Nevertheless, the polyplexes failed to induce specific mRNA degradation, hence neither reduction in tumor volume nor prolonged mice survival was seen.
Collapse
Affiliation(s)
- Adva Krivitsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel
| | - Dina Polyak
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel
| | - Anna Scomparin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel
| | - Shay Eliyahu
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel
| | - Paula Ofek
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel
| | - Galia Tiram
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Room 607, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
33
|
Wang X, Li Y, Li Q, Neufeld CI, Pouli D, Sun S, Yang L, Deng P, Wang M, Georgakoudi I, Tang S, Xu Q. Hyaluronic acid modification of RNase A and its intracellular delivery using lipid-like nanoparticles. J Control Release 2017; 263:39-45. [DOI: 10.1016/j.jconrel.2017.01.037] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/19/2017] [Accepted: 01/27/2017] [Indexed: 11/28/2022]
|
34
|
Thanki K, Zeng X, Justesen S, Tejlmann S, Falkenberg E, Van Driessche E, Mørck Nielsen H, Franzyk H, Foged C. Engineering of small interfering RNA-loaded lipidoid-poly(DL-lactic-co-glycolic acid) hybrid nanoparticles for highly efficient and safe gene silencing: A quality by design-based approach. Eur J Pharm Biopharm 2017; 120:22-33. [PMID: 28756280 DOI: 10.1016/j.ejpb.2017.07.014] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 07/19/2017] [Accepted: 07/25/2017] [Indexed: 01/29/2023]
Abstract
Safety and efficacy of therapeutics based on RNA interference, e.g., small interfering RNA (siRNA), are dependent on the optimal engineering of the delivery technology, which is used for intracellular delivery of siRNA to the cytosol of target cells. We investigated the hypothesis that commonly used and poorly tolerated cationic lipids might be replaced with more efficacious and safe lipidoids as the lipid component of siRNA-loaded lipid-polymer hybrid nanoparticles (LPNs) for achieving more efficient gene silencing at lower and safer doses. However, formulation design of such a complex formulation is highly challenging due to a strong interplay between several contributing factors. Hence, critical formulation variables, i.e. the lipidoid content and siRNA:lipidoid ratio, were initially identified, followed by a systematic quality-by-design approach to define the optimal operating space (OOS), eventually resulting in the identification of a robust, highly efficacious and safe formulation. A 17-run design of experiment with an I-optimal approach was performed to systematically assess the effect of selected variables on critical quality attributes (CQAs), i.e. physicochemical properties (hydrodynamic size, zeta potential, siRNA encapsulation/loading) and the biological performance (in vitro gene silencing and cell viability). Model fitting of the obtained data to construct predictive models revealed non-linear relationships for all CQAs, which can be readily overlooked in one-factor-at-a-time optimization approaches. The response surface methodology further enabled the identification of an OOS that met the desired quality target product profile. The optimized lipidoid-modified LPNs revealed more than 50-fold higher in vitro gene silencing at well-tolerated doses and approx. a twofold increase in siRNA loading as compared to reference LPNs modified with the commonly used cationic lipid dioleyltrimethylammonium propane (DOTAP). Thus, lipidoid-modified LPNs show highly promising prospects for efficient and safe intracellular delivery of siRNA.
Collapse
Affiliation(s)
- Kaushik Thanki
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Xianghui Zeng
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Sarah Justesen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen Ø, Denmark
| | - Sarah Tejlmann
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Emily Falkenberg
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Elize Van Driessche
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark; Department of Pharmaceutics, Laboratory of General Biochemistry and Physical Pharmacy, Ghent University Campus Heymans, Ottergemsesteenweg 460, 9000 Gent, Belgium
| | - Hanne Mørck Nielsen
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark
| | - Henrik Franzyk
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Jagtvej 162, DK-2100 Copenhagen Ø, Denmark
| | - Camilla Foged
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, DK-2100 Copenhagen Ø, Denmark.
| |
Collapse
|
35
|
Wu Y, Smith AE, Reineke TM. Lipophilic Polycation Vehicles Display High Plasmid DNA Delivery to Multiple Cell Types. Bioconjug Chem 2017; 28:2035-2040. [PMID: 28731685 DOI: 10.1021/acs.bioconjchem.7b00306] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
A class of cationic poly(alkylamidoamine)s (PAAAs) containing lipophilic methylene linkers were designed and examined as in vitro plasmid DNA (pDNA) delivery agents. The PAAAs were synthesized via step-growth polymerization between a diamine monomer and each of four different diacid chloride monomers with varying methylene linker lengths, including glutaryl chloride, adipoyl chloride, pimeloyl chloride, and suberoyl chloride, which served to systematically increase the lipophilicity of the polymers. The synthesized polymers successfully complexed with pDNA in reduced serum medium at N/P ratios of 5 and greater, resulting in polyplexes with hydrodynamic diameters of approximately 1 μm. These polyplexes were tested for in vitro transgene expression and cytotoxicity using HDFa (human dermal fibroblast), HeLa (human cervical carcinoma), HMEC (human mammary epithelial), and HUVEC (human umbilical vein endothelial) cells. Interestingly, select PAAA polyplex formulations were found to be more effective than Lipofectamine 2000 at promoting transgene expression (GFP) while maintaining comparable or higher cell viability. Transgene expression was highest in HeLa cells (∼90% for most formulations) and lowest in HDFa cells (up to ∼20%) as measured by GFP fluorescence. In addition, the cytotoxicity of PAAA polyplex formulations was significantly increased as the molecular weight, N/P ratio, and methylene linker length were increased. The PAAA vehicles developed herein provide a new delivery vehicle design strategy of displaying attributes of both polycations and lipids, which show promise as a tunable scaffold for refining the structure-activity-toxicity profiles for future genome editing studies.
Collapse
Affiliation(s)
- Yaoying Wu
- Department of Chemistry, University of Minnesota , 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| | - Adam E Smith
- Department of Chemistry, University of Minnesota , 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States.,Department of Chemical Engineering, University of Mississippi , 134 Anderson, University, Mississippi 38677, United States
| | - Theresa M Reineke
- Department of Chemistry, University of Minnesota , 207 Pleasant Street SE, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
36
|
Wang LL, Burdick JA. Engineered Hydrogels for Local and Sustained Delivery of RNA-Interference Therapies. Adv Healthc Mater 2017; 6:10.1002/adhm.201601041. [PMID: 27976524 PMCID: PMC5226889 DOI: 10.1002/adhm.201601041] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 10/21/2016] [Indexed: 12/20/2022]
Abstract
It has been nearly two decades since RNA-interference (RNAi) was first reported. While there are no approved clinical uses, several phase II and III clinical trials suggest the great promise of RNAi therapeutics. One challenge for RNAi therapies is the controlled localization and sustained presentation to target tissues, to both overcome systemic toxicity concerns and to enhance in vivo efficacy. One approach that is emerging to address these limitations is the entrapment of RNAi molecules within hydrogels for local and sustained release. In these systems, nucleic acids are either delivered as siRNA conjugates or within nanoparticles. A plethora of hydrogels has been implemented using these approaches, including both traditional hydrogels that have already been developed for other applications and new hydrogels developed specifically for RNAi delivery. These hydrogels have been applied to various applications in vivo, including cancer, bone regeneration, inflammation and cardiac repair. This review will examine the design and implementation of such hydrogel RNAi systems and will cover the most recent applications of these systems.
Collapse
Affiliation(s)
- Leo L. Wang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| | - Jason A. Burdick
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
37
|
Li B, Dong Y. Preparation and Optimization of Lipid-Like Nanoparticles for mRNA Delivery. Methods Mol Biol 2017; 1632:207-217. [PMID: 28730441 DOI: 10.1007/978-1-4939-7138-1_13] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lipid-like nanoparticles (LLNs) have shown great promise for nucleic acid delivery. Recently, we have developed N 1,N 3,N 5-tris(2-aminoethyl)benzene-1,3,5-tricarboxamide (TT) derived lipid-like compounds, formulated them into TT LLNs for mRNA delivery, and applied an orthogonal array design to facilitate formulation optimization. This chapter focuses on the following contents relevant to lipid-like nanoparticles: formulation method, particle characterization, orthogonal array design, and in vitro assays.
Collapse
Affiliation(s)
- Bin Li
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, 496 W. 12th Ave., Columbus, OH, 43210, USA
| | - Yizhou Dong
- Division of Pharmaceutics and Pharmaceutical Chemistry, College of Pharmacy, The Ohio State University, 496 W. 12th Ave., Columbus, OH, 43210, USA.
| |
Collapse
|
38
|
Wang M, Glass ZA, Xu Q. Non-viral delivery of genome-editing nucleases for gene therapy. Gene Ther 2016; 24:144-150. [PMID: 27797355 DOI: 10.1038/gt.2016.72] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 09/22/2016] [Accepted: 10/13/2016] [Indexed: 01/02/2023]
Abstract
Manipulating the genetic makeup of mammalian cells using programmable nuclease-based genome-editing technology has recently evolved into a powerful avenue that holds great potential for treating genetic disorders. There are four types of genome-editing nucleases, including meganucleases, zinc finger nucleases, transcription activator-like effector nucleases and clustered, regularly interspaced, short palindromic repeat-associated nucleases such as Cas9. These nucleases have been harnessed to introduce precise and specific changes of the genome sequence at virtually any genome locus of interest. The therapeutic relevance of these genome-editing technologies, however, is challenged by the safe and efficient delivery of nuclease into targeted cells. Herein, we summarize recent advances that have been made on non-viral delivery of genome-editing nucleases. In particular, we focus on non-viral delivery of Cas9/sgRNA ribonucleoproteins for genome editing. In addition, the future direction for developing non-viral delivery of programmable nucleases for genome editing is discussed.
Collapse
Affiliation(s)
- M Wang
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA.,Beijing National Laboratory for Molecular Sciences, Key Laboratory of Analytical Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences, Beijing, China
| | - Z A Glass
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| | - Q Xu
- Department of Biomedical Engineering, Tufts University, Medford, MA, USA
| |
Collapse
|
39
|
Ueda E, Feng W, Levkin PA. Superhydrophilic-Superhydrophobic Patterned Surfaces as High-Density Cell Microarrays: Optimization of Reverse Transfection. Adv Healthc Mater 2016; 5:2646-2654. [PMID: 27568500 DOI: 10.1002/adhm.201600518] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 06/30/2016] [Indexed: 01/07/2023]
Abstract
High-density microarrays can screen thousands of genetic and chemical probes at once in a miniaturized and parallelized manner, and thus are a cost-effective alternative to microwell plates. Here, high-density cell microarrays are fabricated by creating superhydrophilic-superhydrophobic micropatterns in thin, nanoporous polymer substrates such that the superhydrophobic barriers confine both aqueous solutions and adherent cells within each superhydrophilic microspot. The superhydrophobic barriers confine and prevent the mixing of larger droplet volumes, and also control the spreading of droplets independent of the volume, minimizing the variability that arises due to different liquid and surface properties. Using a novel liposomal transfection reagent, ScreenFect A, the method of reverse cell transfection is optimized on the patterned substrates and several factors that affect transfection efficiency and cytotoxicity are identified. Higher levels of transfection are achieved on HOOC- versus NH2 -functionalized superhydrophilic spots, as well as when gelatin and fibronectin are added to the transfection mixture, while minimizing the amount of transfection reagent improves cell viability. Almost no diffusion of the printed transfection mixtures to the neighboring microspots is detected. Thus, superhydrophilic-superhydrophobic patterned surfaces can be used as cell microarrays and for optimizing reverse cell transfection conditions before performing further cell screenings.
Collapse
Affiliation(s)
- Erica Ueda
- Institute of Toxicology and Genetics; Karlsruhe Institute of Technology; 76021 Karlsruhe Germany
| | - Wenqian Feng
- Institute of Toxicology and Genetics; Karlsruhe Institute of Technology; 76021 Karlsruhe Germany
| | - Pavel A. Levkin
- Institute of Toxicology and Genetics; Karlsruhe Institute of Technology; 76021 Karlsruhe Germany
| |
Collapse
|
40
|
siRNA targeting the κ light chain constant region: preclinical testing of an approach to nonfibrillar and fibrillar light chain deposition diseases. Gene Ther 2016; 23:727-733. [DOI: 10.1038/gt.2016.50] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 05/30/2016] [Accepted: 06/08/2016] [Indexed: 01/27/2023]
|
41
|
Knapp CM, He J, Lister J, Whitehead KA. Lipidoid nanoparticle mediated silencing of Mcl-1 induces apoptosis in mantle cell lymphoma. Exp Biol Med (Maywood) 2016; 241:1007-13. [PMID: 27022142 DOI: 10.1177/1535370216640944] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Conventional chemo-immunotherapy fails to cure the majority of mantle cell lymphoma patients and causes substantial toxicity. Resistant mantle cell lymphoma cells commonly overexpress and are dependent on the anti-apoptotic protein, Mcl-1, for survival. In this study, we use potent lipidoid nanoparticles to deliver siRNA to silence Mcl-1 expression. Studies were conducted using two different mantle cell lymphoma cell lines, a normal (JeKo-1) and an aggressive (MAVER-1) line, to assess the ability of lipidoid nanoparticles to be used broadly in the treatment of mantle cell lymphoma. Mcl-1 mRNA silencing and protein knockdown was observed as early as one day after treatment and the lipidoid nanoparticles achieved sustained silencing of Mcl-1 mRNA for at least four days in both JeKo-1 and MAVER-1 cells. Eighty percent silencing was achieved at three days post-transfection in JeKo-1 cells while 50% silencing was achieved in MAVER-1 cells, which are more resistant to transfection. Interestingly, silencing of Mcl-1 induced apoptosis in nearly 30% of both JeKo-1 and MAVER-1 cells three days post-transfection. Additionally, Mcl-1 silencing and the resultant apoptosis in mantle cell lymphoma cells were dose dependent. These data suggest that lipidoid nanoparticles siRNA therapy targeting Mcl-1 has potential as a new treatment modality for mantle cell lymphoma and many other cancers that overexpress Mcl-1. The combination of anti-Mcl-1 lipidoid nanoparticles with other forms of targeted therapy offers hope for reducing or replacing cytotoxic chemotherapy as standard treatment for mantle cell lymphoma.
Collapse
Affiliation(s)
- Christopher M Knapp
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jia He
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - John Lister
- Division of Hematology and Cellular Therapy, Allegheny Health Network Cancer Institute, Pittsburgh, PA 15224, USA
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
42
|
Efficient delivery of genome-editing proteins using bioreducible lipid nanoparticles. Proc Natl Acad Sci U S A 2016; 113:2868-73. [PMID: 26929348 DOI: 10.1073/pnas.1520244113] [Citation(s) in RCA: 480] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
A central challenge to the development of protein-based therapeutics is the inefficiency of delivery of protein cargo across the mammalian cell membrane, including escape from endosomes. Here we report that combining bioreducible lipid nanoparticles with negatively supercharged Cre recombinase or anionic Cas9:single-guide (sg)RNA complexes drives the electrostatic assembly of nanoparticles that mediate potent protein delivery and genome editing. These bioreducible lipids efficiently deliver protein cargo into cells, facilitate the escape of protein from endosomes in response to the reductive intracellular environment, and direct protein to its intracellular target sites. The delivery of supercharged Cre protein and Cas9:sgRNA complexed with bioreducible lipids into cultured human cells enables gene recombination and genome editing with efficiencies greater than 70%. In addition, we demonstrate that these lipids are effective for functional protein delivery into mouse brain for gene recombination in vivo. Therefore, the integration of this bioreducible lipid platform with protein engineering has the potential to advance the therapeutic relevance of protein-based genome editing.
Collapse
|
43
|
Molla MR, Levkin PA. Combinatorial Approach to Nanoarchitectonics for Nonviral Delivery of Nucleic Acids. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:1159-1175. [PMID: 26608939 DOI: 10.1002/adma.201502888] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/01/2015] [Indexed: 06/05/2023]
Abstract
Nanoparticles based on cationic polymers, lipids or lipidoids are of great interest in the field of gene delivery applications. The research on these nanosystems is rapidly growing as they hold promise to treat wide variety of human diseases ranging from viral infections to genetic disorders and cancer. Recently, combinatorial design principles have been adopted for rapid generation of large numbers of chemically diverse polymers and lipids capable of forming multifunctional nanocarriers for the use in gene delivery applications. At the same time, current high-throughput screening systems as well as convenient cell assays and readout techniques allow for fast evaluation of cell transfection efficiencies and toxicities of libraries of novel gene delivery agents. This allows for a rapid evaluation of structure-function relationship as well as identification of novel efficient nanocarriers for cell transfection and gene therapy. Here, the recent contribution of high-throughput synthesis to the development of novel nanocarriers for gene delivery applications is described.
Collapse
Affiliation(s)
- Mijanur Rahaman Molla
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Pavel A Levkin
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
- University of Heidelberg, Department of Applied Physical Chemistry, 69120, Heidelberg, Germany
| |
Collapse
|
44
|
He D, Müller K, Krhac Levacic A, Kos P, Lächelt U, Wagner E. Combinatorial Optimization of Sequence-Defined Oligo(ethanamino)amides for Folate Receptor-Targeted pDNA and siRNA Delivery. Bioconjug Chem 2016; 27:647-59. [DOI: 10.1021/acs.bioconjchem.5b00649] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Dongsheng He
- Pharmaceutical
Biotechnology, Center for System-based Drug Research and Center for
NanoScience (CeNS), Ludwig-Maximilians-University, 81377 Munich, Germany
- Nanosystems Initiative Munich, Schellingstrasse
4, 80799 Munich, Germany
| | - Katharina Müller
- Pharmaceutical
Biotechnology, Center for System-based Drug Research and Center for
NanoScience (CeNS), Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Ana Krhac Levacic
- Pharmaceutical
Biotechnology, Center for System-based Drug Research and Center for
NanoScience (CeNS), Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Petra Kos
- Pharmaceutical
Biotechnology, Center for System-based Drug Research and Center for
NanoScience (CeNS), Ludwig-Maximilians-University, 81377 Munich, Germany
| | - Ulrich Lächelt
- Pharmaceutical
Biotechnology, Center for System-based Drug Research and Center for
NanoScience (CeNS), Ludwig-Maximilians-University, 81377 Munich, Germany
- Nanosystems Initiative Munich, Schellingstrasse
4, 80799 Munich, Germany
| | - Ernst Wagner
- Pharmaceutical
Biotechnology, Center for System-based Drug Research and Center for
NanoScience (CeNS), Ludwig-Maximilians-University, 81377 Munich, Germany
- Nanosystems Initiative Munich, Schellingstrasse
4, 80799 Munich, Germany
| |
Collapse
|
45
|
Wu Y, Li L, Chen Q, Su Y, Levkin PA, Davidson G. Single-Tailed Lipidoids Enhance the Transfection Activity of Their Double-Tailed Counterparts. ACS COMBINATORIAL SCIENCE 2016; 18:43-50. [PMID: 26651853 DOI: 10.1021/acscombsci.5b00117] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Cationic lipid-like molecules (lipidoids) are widely used for in vitro and in vivo gene delivery. Nearly all lipidoids developed to date employ double-tail or multiple-tail structures for transfection. Single-tail lipidoids are seldom considered for transfection as they have low efficiency in gene delivery. So far, there is no detailed study on the contribution to transfection efficiency of single-tail lipidoids when combined with standard double-tail lipidoids. Here, we use combinatorial chemistry to synthesize 17 double-tail and 17 single-tail lipidoids using thiol-yne and thiol-ene click chemistry, respectively. HEK 293T cells were used to analyze transfection efficiency by fluorescence microscopy and calculated based on the percentage of cells transfected. The size and zeta potential of liposomes and lipoplexes were characterized by dynamic light scattering (DLS). Intracellular DNA delivery and trafficking was further examined using confocal microscopy. Our study shows that combining single with double-tail lipidoids increases uptake of lipoplexes, as well as cellular transfection efficiency.
Collapse
Affiliation(s)
- Yihang Wu
- Institute of Toxicology and
Genetics, Karlsruhe Institute of Technology, 76344 Karlsruhe, Germany
| | - Linxian Li
- Institute of Toxicology and
Genetics, Karlsruhe Institute of Technology, 76344 Karlsruhe, Germany
| | - Qing Chen
- Institute of Toxicology and
Genetics, Karlsruhe Institute of Technology, 76344 Karlsruhe, Germany
| | - Yi Su
- Institute of Toxicology and
Genetics, Karlsruhe Institute of Technology, 76344 Karlsruhe, Germany
| | - Pavel A. Levkin
- Institute of Toxicology and
Genetics, Karlsruhe Institute of Technology, 76344 Karlsruhe, Germany
| | - Gary Davidson
- Institute of Toxicology and
Genetics, Karlsruhe Institute of Technology, 76344 Karlsruhe, Germany
| |
Collapse
|
46
|
Carter MD, Miller DS, Jennings J, Wang X, Mahanthappa MK, Abbott NL, Lynn DM. Synthetic Mimics of Bacterial Lipid A Trigger Optical Transitions in Liquid Crystal Microdroplets at Ultralow Picogram-per-Milliliter Concentrations. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2015; 31:12850-12855. [PMID: 26562069 PMCID: PMC4671391 DOI: 10.1021/acs.langmuir.5b03557] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/11/2015] [Indexed: 05/29/2023]
Abstract
We report synthetic six-tailed mimics of the bacterial glycolipid Lipid A that trigger changes in the internal ordering of water-dispersed liquid crystal (LC) microdroplets at ultralow (picogram-per-milliliter) concentrations. These molecules represent the first class of synthetic amphiphiles to mimic the ability of Lipid A and bacterial endotoxins to trigger optical responses in LC droplets at these ultralow concentrations. This behavior stands in contrast to all previously reported synthetic surfactants and lipids, which require near-complete monolayer coverage at the LC droplet surface to trigger ordering transitions. Surface-pressure measurements and SAXS experiments reveal these six-tailed synthetic amphiphiles to mimic key aspects of the self-assembly of Lipid A at aqueous interfaces and in solution. These and other results suggest that these amphiphiles trigger orientational transitions at ultralow concentrations through a unique mechanism that is similar to that of Lipid A and involves formation of inverted self-associated nanostructures at topological defects in the LC droplets.
Collapse
Affiliation(s)
- Matthew
C. D. Carter
- Department
of Chemistry, and Department of Chemical and Biological Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Daniel S. Miller
- Department
of Chemistry, and Department of Chemical and Biological Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - James Jennings
- Department
of Chemistry, and Department of Chemical and Biological Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Xiaoguang Wang
- Department
of Chemistry, and Department of Chemical and Biological Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Mahesh K. Mahanthappa
- Department
of Chemistry, and Department of Chemical and Biological Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - Nicholas L. Abbott
- Department
of Chemistry, and Department of Chemical and Biological Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| | - David M. Lynn
- Department
of Chemistry, and Department of Chemical and Biological Engineering, University of Wisconsin—Madison, Madison, Wisconsin 53706, United States
| |
Collapse
|
47
|
Altınoglu S, Wang M, Xu Q. Combinatorial library strategies for synthesis of cationic lipid-like nanoparticles and their potential medical applications. Nanomedicine (Lond) 2015; 10:643-57. [PMID: 25723096 DOI: 10.2217/nnm.14.192] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The past two decades have witnessed the high efficiency and efficacy of cationic lipids and liposomal formations for drug delivery. The tedious synthesis of conventional lipids and the inefficiency in studying structure-activity relationships, however, have hindered the clinical translation of lipid nanoparticle delivery systems. Combinatorial synthesis of lipid-like nanoparticles ('lipidoids') has recently emerged as an approach to accelerate the development of these delivery platforms. Utilizing a high-throughput screening strategy, the libraries of lipidoids are sorted and prime candidates for the delivery in the intended application can be identified and optimized for the next generation. In this review, we outline methods used for combinatorial lipidoid synthesis, the application of high-throughput screening, and the current medical applications of candidate lipidoids.
Collapse
Affiliation(s)
- Sarah Altınoglu
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | | | | |
Collapse
|
48
|
Singh Y, Tomar S, Khan S, Meher JG, Pawar VK, Raval K, Sharma K, Singh PK, Chaurasia M, Surendar Reddy B, Chourasia MK. Bridging small interfering RNA with giant therapeutic outcomes using nanometric liposomes. J Control Release 2015; 220:368-387. [PMID: 26528900 DOI: 10.1016/j.jconrel.2015.10.050] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 10/26/2015] [Accepted: 10/27/2015] [Indexed: 01/04/2023]
Abstract
The scope of RNAi based therapeutics is unquestionable. However, if we dissect the current trend of clinical trials for afore mentioned drug class, some stark trends appear: 1) naked siRNA only exerts influence in topical mode whilst systemic delivery requires a carrier and 2) even after two decades of extensive efforts, not even a single siRNA containing product is commercially available. It was therefore felt that a perspective simplifying the unique intricacies of working with a merger of siRNA and liposomes from a pharmaceutical viewpoint could draw the attention of a wider array of interested researchers. We begin from the beginning and attempt to conduit the gap between theoretical logic and experimental/actual constraints. This, in turn could stimulate the next generation of investigators, gearing them to tackle the conundrum, which is siRNA delivery.
Collapse
Affiliation(s)
- Yuvraj Singh
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Sandeep Tomar
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Shariq Khan
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Jaya Gopal Meher
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Vivek K Pawar
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Kavit Raval
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Komal Sharma
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Pankaj K Singh
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Mohini Chaurasia
- Amity Institute of Pharmacy, Amity University, Lucknow, UP 226028, India
| | - B Surendar Reddy
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Manish K Chourasia
- Pharmaceutics Division, CSIR-Central Drug Research Institute, Lucknow 226031, India.
| |
Collapse
|
49
|
Wu N, Zhang X, Li F, Zhang T, Gan Y, Li J. Spray-dried powders enhance vaginal siRNA delivery by potentially modulating the mucus molecular sieve structure. Int J Nanomedicine 2015; 10:5383-96. [PMID: 26347257 PMCID: PMC4554414 DOI: 10.2147/ijn.s87978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Vaginal small interfering RNA (siRNA) delivery provides a promising strategy for the prevention and treatment of vaginal diseases. However, the densely cross-linked mucus layer on the vaginal wall severely restricts nanoparticle-mediated siRNA delivery to the vaginal epithelium. In order to overcome this barrier and enhance vaginal mucus penetration, we prepared spray-dried powders containing siRNA-loaded nanoparticles. Powders with Pluronic F127 (F127), hydroxypropyl methyl cellulose (HPMC), and mannitol as carriers were obtained using an ultrasound-assisted spray-drying technique. Highly dispersed dry powders with diameters of 5-15 μm were produced. These powders showed effective siRNA protection and sustained release. The mucus-penetrating properties of the powders differed depending on their compositions. They exhibited different potential of opening mesh size of molecular sieve in simulated vaginal mucus system. A powder formulation with 0.6% F127 and 0.1% HPMC produced the maximum increase in the pore size of the model gel used to simulate vaginal mucus by rapidly extracting water from the gel and interacting with the gel; the resulting modulation of the molecular sieve effect achieved a 17.8-fold improvement of siRNA delivery in vaginal tract and effective siRNA delivery to the epithelium. This study suggests that powder formulations with optimized compositions have the potential to alter the steric barrier posed by mucus and hold promise for effective vaginal siRNA delivery.
Collapse
Affiliation(s)
- Na Wu
- School of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China ; Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Xinxin Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Feifei Li
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Tao Zhang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Yong Gan
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Juan Li
- School of Pharmacy, China Pharmaceutical University, Nanjing, People's Republic of China
| |
Collapse
|
50
|
Draghici B, Ilies MA. Synthetic Nucleic Acid Delivery Systems: Present and Perspectives. J Med Chem 2015; 58:4091-130. [PMID: 25658858 DOI: 10.1021/jm500330k] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Bogdan Draghici
- Department
of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, Pennsylvania 19140, United States
| | - Marc A. Ilies
- Department
of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, 3307 North Broad Street, Philadelphia, Pennsylvania 19140, United States
- Temple Materials Institute, 1803 North Broad Street, Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|