1
|
Karmaker S, Rosales PD, Tirumuruhan B, Viravalli A, Boehnke N. More than a delivery system: the evolving role of lipid-based nanoparticles. NANOSCALE 2025; 17:11864-11893. [PMID: 40293317 DOI: 10.1039/d4nr04508d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Lipid-based nanoparticles, including liposomes and lipid nanoparticles (LNPs), make up an important class of drug delivery systems. Their modularity enables encapsulation of a wide range of therapeutic cargoes, their ease of functionalization allows for incorporation of targeting motifs and anti-fouling coatings, and their scalability facilitates rapid translation to the clinic. While the discovery and early understanding of lipid-based nanoparticles is heavily rooted in biology, formulation development has largely focused on materials properties, such as how liposome and lipid nanoparticle composition can be altered to maximize drug loading, stability and circulation. To achieve targeted delivery and enable improved accumulation of therapeutics at target tissues or disease sites, emphasis is typically placed on the use of external modifications, such as peptide, protein, and polymer motifs. However, these approaches can increase the complexity of the nanocarrier and complicate scale up. In this review, we focus on how our understanding of lipid structure and function in biological contexts can be used to design intrinsically functional and targeted nanocarriers. We highlight formulation-based strategies, such as the incorporation of bioactive lipids, that have been used to modulate liposome and lipid nanoparticle properties and improve their functionality while retaining simple nanocarrier designs. We also highlight classes of naturally occurring lipids, their functions, and how they have been incorporated into lipid-based nanoparticles. We will additionally position these approaches into the historical context of both liposome and LNP development.
Collapse
Affiliation(s)
- Senjuti Karmaker
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Plinio D Rosales
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Barath Tirumuruhan
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Amartya Viravalli
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| | - Natalie Boehnke
- Department of Chemical Engineering and Materials Science, University of Minnesota, Twin Cities Minneapolis, MN 55455, USA.
| |
Collapse
|
2
|
Cao Y, Hu X, Wu D, Jiang Y, Yu Y, Wang S, Chen W, Long Y, Xu L, Qu J, Yang B, Chakhabi B, Wang H, Deng Y, Chen L, Chen Z, Li Q. Targeting capacity, safety and efficacy of engineered extracellular vesicles delivered by transdermal microneedles to treat plasmacytoma in mice. Clin Transl Med 2025; 15:e70327. [PMID: 40317915 PMCID: PMC12048306 DOI: 10.1002/ctm2.70327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND Engineered extracellular vesicles (EVs) are emerging as a highly potential platform for targeted drug delivery in cancer therapy. Although intravenous injection is commonly used in EV treatment, there is growing interest in using microneedles (MNs) for transdermal EV delivery; however, comprehensive studies comparing the tissue distribution, safety and antitumour efficacy of these two approaches for delivering engineered EVs remain scarce. METHODS We used EVs derived from umbilical cord mesenchymal stem cells, modified with phospholipid‒polyethylene glycol‒N-hydroxysuccinimide and conjugated with CD38 peptides (CD38-EVs), to target myeloma cells that highly express CD38 antigen, and tested their safety and antitumour efficacy in mice with subcutaneous plasmacytoma, administrated via dissolvable transdermal MNs or intravenous injection. Flow cytometry, immunofluorescence and fluorescence molecular projection imaging analysis were employed to evaluate the distribution of CD38-EVs at the cellular level and within living systems. Additionally, histopathological analysis and biochemical analyses were conducted to assess the antitumour effects and safety of CD38-EVs loaded with doxorubicin (CD38-EVs-Dox). RESULTS Compared to standard EVs, CD38-EVs exhibited enhanced uptake by CD38high tumour cells and reduced uptake by CD38-negative non-tumour cells in vitro. In plasmacytoma NOD/SCID mouse models, CD38-EVs encapsulated within MNs (CD38-EVsMNs) effectively targeted the tumour cells much more than the standard EVs encapsulated within MNs (EVsMNs) and CD38-EVs intravenously administrated (CD38-EVsi.v), with reduced distribution to the lungs and spleen. Additionally, CD38-EVs-Dox induced significantly greater cytotoxicity against the tumour cells than EVs-Dox in vitro, and CD38-EVs-DoxMNs significantly reduced tumour burden compared to both EVs-DoxMNs and CD38-EVs-Doxi.v, while maintaining favourable safety profiles. CONCLUSIONS CD38-EVs-DoxMNs have superior efficacy and safety in treating plasmacytoma mice, compared to CD38-EVs-Doxi.v, providing novel insights into the potential of MNs as a platform for delivering targeted engineered EVs in tumour therapy. HIGHLIGHTS Enhanced tumor targeting: CD38-modified EVs (CD38-EVs) showed increased uptake by CD38high tumor cells while reducing uptake by CD38-negative non-tumor cells. Optimized delivery: MN-loaded CD38-EVs targeted tumors more effectively than MN-loaded EVs and intravenously injected CD38-EVs, with lower lung and spleen accumulation. Superior antitumor efficacy: MN-delivered CD38-EVs-Dox significantly suppressed tumor growth, outperforming intravenous CD38-EVs-Dox and MN-delivered EVs-Dox.
Collapse
Affiliation(s)
- Yulin Cao
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Xuan Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Di Wu
- Department of HematologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yuxuan Jiang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhanChina
- MoE Key Laboratory for Biomedical Photonics, Department of Biomedical EngineeringHuazhong University of Science and TechnologyWuhanChina
| | - Yali Yu
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Shan Wang
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Wenlan Chen
- Department of HematologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yaoying Long
- Department of HematologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Liuyue Xu
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jiao Qu
- Department of HematologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Bianlei Yang
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Blal Chakhabi
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Hongxiang Wang
- Department of HematologyThe Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yong Deng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for OptoelectronicsHuazhong University of Science and TechnologyWuhanChina
- MoE Key Laboratory for Biomedical Photonics, Department of Biomedical EngineeringHuazhong University of Science and TechnologyWuhanChina
| | - Lei Chen
- Department of HematologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Zhichao Chen
- Department of HematologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Qiubai Li
- Department of Rheumatology and ImmunologyUnion Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Hubei Engineering Research Center for Application of Extracellular VesicleHubei University of Science and TechnologyXianningChina
| |
Collapse
|
3
|
Hofstraat SRJ, Anbergen T, Zwolsman R, Deckers J, van Elsas Y, Trines MM, Versteeg I, Hoorn D, Ros GWB, Bartelet BM, Hendrikx MMA, Darwish YB, Kleuskens T, Borges F, Maas RJF, Verhalle LM, Tielemans W, Vader P, de Jong OG, Tabaglio T, Wee DKB, Teunissen AJP, Brechbühl E, Janssen HM, Fransen PM, de Dreu A, Schrijver DP, Priem B, Toner YC, Beldman TJ, Netea MG, Mulder WJM, Kluza E, van der Meel R. Nature-inspired platform nanotechnology for RNA delivery to myeloid cells and their bone marrow progenitors. NATURE NANOTECHNOLOGY 2025; 20:532-542. [PMID: 39900620 PMCID: PMC12014499 DOI: 10.1038/s41565-024-01847-3] [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: 10/17/2023] [Accepted: 12/06/2024] [Indexed: 02/05/2025]
Abstract
Nucleic acid therapeutics are used for silencing, expressing or editing genes in vivo. However, their systemic stability and targeted delivery to bone marrow resident cells remains a challenge. In this study we present a nanotechnology platform based on natural lipoproteins, designed for delivering small interfering RNA (siRNA), antisense oligonucleotides and messenger RNA to myeloid cells and haematopoietic stem and progenitor cells in the bone marrow. We developed a prototype apolipoprotein nanoparticle (aNP) that stably incorporates siRNA into its core. We then created a comprehensive library of aNP formulations and extensively characterized their physicochemical properties and in vitro performance. From this library, we selected eight representative aNP-siRNA formulations and evaluated their ability to silence lysosomal-associated membrane protein 1 (Lamp1) expression in immune cell subsets in mice after intravenous administration. Using the most effective aNP identified from the screening process, we tested the platform's potential for therapeutic gene silencing in a syngeneic murine tumour model. We also demonstrated the aNP platform's suitability for splice-switching with antisense oligonucleotides and for protein production with messenger RNA by myeloid progenitor cells in the bone marrow. Our data indicate that the aNP platform holds translational potential for delivering various types of nucleic acid therapeutics to myeloid cells and their progenitors.
Collapse
Affiliation(s)
- Stijn R J Hofstraat
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tom Anbergen
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Robby Zwolsman
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Jeroen Deckers
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yuri van Elsas
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mirre M Trines
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Iris Versteeg
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Daniek Hoorn
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Gijs W B Ros
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Branca M Bartelet
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Merel M A Hendrikx
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Biotrip B.V., Eindhoven, the Netherlands
| | - Youssef B Darwish
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Teun Kleuskens
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Francisca Borges
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Rianne J F Maas
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Lars M Verhalle
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Willem Tielemans
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Pieter Vader
- CDL Research & Department of Experimental Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Olivier G de Jong
- Department of Pharmaceutics, Utrecht Institute of Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Tommaso Tabaglio
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Dave Keng Boon Wee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Abraham J P Teunissen
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular, and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eliane Brechbühl
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | | | - Anne de Dreu
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - David P Schrijver
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Bram Priem
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Yohana C Toner
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Thijs J Beldman
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands
- Department of Immunology and Metabolism, Life and Medical Sciences Institute, University of Bonn, Bonn, Germany
| | - Willem J M Mulder
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands.
- Department of Internal Medicine and Radboud Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Ewelina Kluza
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Roy van der Meel
- Laboratory of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
4
|
Tang C, Zhang Y, Li B, Fan X, Wang Z, Su R, Qi W, Wang Y. Modular Design of Lipopeptide-Based Organ-Specific Targeting (POST) Lipid Nanoparticles for Highly Efficient RNA Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415643. [PMID: 39924757 DOI: 10.1002/adma.202415643] [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: 10/14/2024] [Revised: 01/17/2025] [Indexed: 02/11/2025]
Abstract
Lipid nanoparticles (LNPs) with highly efficient and specific extrahepatic targeting abilities are promising in gene delivery, and the lipopeptides (LPs) with excellent designability and functionality are expected to empower the construction of functional LNPs. This study aims to develop highly efficient ionizable components that accurately match different targeting lipid systems through the modular design of LPs. Based on this, a lipopeptide-based organ-specific targeting (POST) LNP screening strategy is constructed, in which lysine-histidine-based lipopeptides (KH-LPs) are designed as highly efficient ionizable components. The optimal KH-LP LNP screened in vitro shows excellent siRNA/mRNA transfecting ability in various hard-to-transfect cell lines. Compared to the classic LNPs, the POST LNPs screened in vivo achieve even higher (or at least comparable) efficiency and specificity in delivering mRNA and siRNA to the lung, liver, and spleen, respectively. The structure-activity relationship (SAR) proves that the modular regulation of LP structures can accurately provide the optimal ionizable components for different targeting lipid systems, demonstrating the potential of this strategy in developing efficient and selective targeting systems, which is expected to open up more possibilities for gene therapy.
Collapse
Affiliation(s)
- Chuanmei Tang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Yexi Zhang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Bowen Li
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Xiangwei Fan
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Zixuan Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Rongxin Su
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Wei Qi
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
- Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin, 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin, 300072, P. R. China
| | - Yuefei Wang
- State Key Laboratory of Chemical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300072, P. R. China
- Tianjin Key Laboratory of Membrane Science and Desalination Technology, Tianjin University, Tianjin, 300072, P. R. China
| |
Collapse
|
5
|
Somu Naidu G, Rampado R, Sharma P, Ezra A, Kundoor GR, Breier D, Peer D. Ionizable Lipids with Optimized Linkers Enable Lung-Specific, Lipid Nanoparticle-Mediated mRNA Delivery for Treatment of Metastatic Lung Tumors. ACS NANO 2025; 19:6571-6587. [PMID: 39912611 PMCID: PMC11841047 DOI: 10.1021/acsnano.4c18636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/31/2025] [Accepted: 01/31/2025] [Indexed: 02/07/2025]
Abstract
Lipid nanoparticles (LNPs) have emerged as a groundbreaking delivery system for vaccines and therapeutic mRNAs. Ionizable lipids are the most pivotal component of LNPs due to their ability to electrostatically interact with mRNA, allowing its encapsulation while concurrently enabling its endosomal escape following cellular internalization. Thus, extensive research has been performed to optimize the ionizable lipid structure and to develop formulations that are well tolerated and allow efficient targeting of different organs that result in a high and sustained mRNA expression. However, one facet of the ionizable lipids' structure has been mostly overlooked: the linker segment between the ionizable headgroup and their tails. Here, we screened a rationally designed library of ionizable lipids with different biodegradable linkers. We extensively characterized LNPs formulated using these ionizable lipids and elucidated how these minor structural changes in the ionizable lipids structure radically influenced the LNPs' biodistribution in vivo. We showed how the use of amide and urea linkers can modulate the LNPs' pKa, resulting in an improved specificity for lung transfection. Finally, we demonstrated how one of these lipids (lipid 35) that form LNPs entrapping a bacterial toxin [pseudomonas exotoxin A (mmPE)] in the form of an mRNA reduced tumor burden and significantly increased the survival of mice with lung metastasis.
Collapse
Affiliation(s)
- Gonna Somu Naidu
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Riccardo Rampado
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Pharmaceutical Sciences, University of
Padova, Padova 35131, Italy
| | - Preeti Sharma
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Assaf Ezra
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Govinda Reddy Kundoor
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Dor Breier
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| | - Dan Peer
- Laboratory
of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer
Research, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
- Department
of Materials Sciences and Engineering, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Center
for Nanoscience and Nanotechnology, Tel
Aviv University, Tel Aviv-Yafo 69978, Israel
- Cancer
Biology Research Center, Tel Aviv University, Tel Aviv-Yafo 69978, Israel
| |
Collapse
|
6
|
Han EL, Tang S, Kim D, Murray AM, Swingle KL, Hamilton AG, Mrksich K, Padilla MS, Palanki R, Li JJ, Mitchell MJ. Peptide-Functionalized Lipid Nanoparticles for Targeted Systemic mRNA Delivery to the Brain. NANO LETTERS 2025; 25:800-810. [PMID: 39688915 DOI: 10.1021/acs.nanolett.4c05186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Systemic delivery of large nucleic acids, such as mRNA, to the brain remains challenging in part due to the blood-brain barrier (BBB) and the tendency of delivery vehicles to accumulate in the liver. Here, we design a peptide-functionalized lipid nanoparticle (LNP) platform for targeted mRNA delivery to the brain. We utilize click chemistry to functionalize LNPs with peptides that target receptors overexpressed on brain endothelial cells and neurons, namely the RVG29, T7, AP2, and mApoE peptides. We evaluate the effect of LNP targeting on brain endothelial and neuronal cell transfection in vitro, investigating factors such as serum protein adsorption, intracellular trafficking, endothelial transcytosis, and exosome secretion. Finally, we show that LNP peptide functionalization enhances mRNA transfection in the mouse brain and reduces hepatic delivery after systemic administration. Specifically, RVG29 LNPs improved neuronal transfection in vivo, establishing its potential as a nonviral platform for delivering mRNA to the brain.
Collapse
Affiliation(s)
- Emily L Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sophia Tang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Dongyoon Kim
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Amanda M Murray
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Kelsey L Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Alex G Hamilton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Kaitlin Mrksich
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Marshall S Padilla
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Rohan Palanki
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Center for Fetal Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, United States
| | - Jacqueline J Li
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
7
|
Rampado R, Naidu GS, Karpov O, Goldsmith M, Sharma P, Ezra A, Stotsky L, Breier D, Peer D. Lipid Nanoparticles With Fine-Tuned Composition Show Enhanced Colon Targeting as a Platform for mRNA Therapeutics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408744. [PMID: 39585189 PMCID: PMC11744673 DOI: 10.1002/advs.202408744] [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: 07/28/2024] [Revised: 11/11/2024] [Indexed: 11/26/2024]
Abstract
Lipid Nanoparticles (LNPs) recently emerged as an invaluable RNA delivery platform. With many LNP-based therapeutics in the pre-clinical and clinical pipelines, there is extensive research dedicated to improving LNPs. These efforts focus mainly on the tolerability and transfectability of new ionizable lipids and RNAs, or modulating LNPs biodistribution with active targeting strategies. However, most formulations follow the well-established lipid proportions used in clinically approved products. Nevertheless, investigating the effects of LNPs composition on their biodistribution can expand the toolbox for particle design, leading to improved delivery strategies. Herein, a new LNPs (30-n-LNPs) formulation with increasing amounts of phospholipids is investigated as a possible mRNA delivery system for treating Inflammatory Bowel Diseases. Compared to LNPs with benchmark composition (b-LNPs), n-LNPs containing 30% distearoylphosphatidylcholine (DSPC) are well tolerated following intravenous administration and display natural targeting toward the inflamed colon in dextran sodium sulfate (DSS)-colitis bearing mice, while de-targeting clearing organs such as the liver and spleen. Using interleukin-10-encoding mRNA as therapeutic cargo, n-LNPs demonstrated a reduction of pathological burden in colitis-bearing mice. n-LNPs represent a starting point to further investigate the influence of LNPs composition on systemic biodistribution, ultimately opening new therapeutic modalities in different pathologies.
Collapse
Affiliation(s)
- Riccardo Rampado
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Gonna Somu Naidu
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Olga Karpov
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Meir Goldsmith
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Preeti Sharma
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Assaf Ezra
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Lior Stotsky
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Dor Breier
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| | - Dan Peer
- Laboratory of Precision NanomedicineShmunis School of Biomedicine and Cancer ResearchTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Department of Materials Sciences and EngineeringTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Center for Nanoscience and NanotechnologyTel Aviv UniversityTel Aviv‐Yafo69978Israel
- Cancer Biology Research CenterTel Aviv UniversityTel Aviv‐Yafo69978Israel
| |
Collapse
|
8
|
Amabile A, Phelan M, Yu Z, Silva P, Marks A, Morla-Folch J, Sohn M, Mollaoglu G, Falcomata C, Teunissen AJP, Brody JD, Dong Y, Brown BD. Bispecific antibody targeting of lipid nanoparticles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.20.629467. [PMID: 39763831 PMCID: PMC11702604 DOI: 10.1101/2024.12.20.629467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2025]
Abstract
Lipid nanoparticles (LNP) are the most clinically advanced non-viral gene delivery system. While progress has been made for enhancing delivery, cell specific targeting remains a challenge. Targeting moieties such as antibodies can be chemically-conjugated to LNPs however, this approach is complex and has challenges for scaling up. Here, we developed an approach to generate antibody-conjugated LNPs that utilizes a bispecific antibody (bsAb) as the targeting bridge. As a docking site for the bsAb, we generated LNPs with a short epitope, derived from hemagglutinin antigen (HA), embedded in the PEG component of the particle (LNPHA). We generated bsAb in which one domain binds HA and the other binds different cell surface proteins, including PD-L1, CD4, CD5, and SunTag. Non-chemical conjugation of the bsAb and LNP resulted in a major increase in the efficiency and specificity of transfecting cells expressing the cognate target. LNP/bsAb mediated a 4-fold increase in in vivo transfection of PD-L1 expressing cancer cells, and a 26-fold increase in ex vivo transfection of quiescent primary human T cells. Additionally, we created a universal bsAb recognizing HA and anti-rat IgG2, enabling LNP tethering to off-the-shelf antibodies such as CD4, CD8, CD20, CD45, and CD3. By utilizing a molecular dock and bsAb technology, these studies demonstrate a simple and effective strategy to generate antibody-conjugated LNPs, enabling precise and efficient mRNA delivery.
Collapse
Affiliation(s)
- Angelo Amabile
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- RNA NanoCore - Lipid Nanoparticle Therapeutics Core, ISMMS, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | - Matthew Phelan
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | - Zhixin Yu
- RNA NanoCore - Lipid Nanoparticle Therapeutics Core, ISMMS, New York, NY, USA
| | - Pedro Silva
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- RNA NanoCore - Lipid Nanoparticle Therapeutics Core, ISMMS, New York, NY, USA
| | - Adam Marks
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | - Judit Morla-Folch
- Biomedical Engineering and Imaging Institute, ISMMS, New Yok, NY, USA
| | - Moah Sohn
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | - Gurkan Mollaoglu
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | - Chiara Falcomata
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | | | - Joshua D Brody
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
- Hematology and Medical Oncology, ISMMS, New York, New York, USA
| | - Yizhou Dong
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- RNA NanoCore - Lipid Nanoparticle Therapeutics Core, ISMMS, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| | - Brian D Brown
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai (ISMMS), New York, NY, USA
- RNA NanoCore - Lipid Nanoparticle Therapeutics Core, ISMMS, New York, NY, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, ISMMS, New York, New York, USA
| |
Collapse
|
9
|
Abbasi Dezfouli S, Michailides ME, Uludag H. Delivery Aspects for Implementing siRNA Therapeutics for Blood Diseases. Biochemistry 2024; 63:3059-3077. [PMID: 39388611 DOI: 10.1021/acs.biochem.4c00327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Hematological disorders result in significant health consequences, and traditional therapies frequently entail adverse reactions without addressing the root cause. A potential solution for hematological disorders characterized by gain-of-function mutations lies in the emergence of small interfering RNA (siRNA) molecules as a therapeutic option. siRNAs are a class of RNA molecules composed of double-stranded RNAs that can degrade specific mRNAs, thereby inhibiting the synthesis of underlying disease proteins. Therapeutic interventions utilizing siRNA can be tailored to selectively target genes implicated in diverse hematological disorders, including sickle cell anemia, β-thalassemia, and malignancies such as lymphoma, myeloma, and leukemia. The development of efficient siRNA silencers necessitates meticulous contemplation of variables such as the RNA backbone, stability, and specificity. Transportation of siRNA to specific cells poses a significant hurdle, prompting investigations of diverse delivery approaches, including chemically modified forms of siRNA and nanoparticle formulations with various biocompatible carriers. This review delves into the crucial role of siRNA technology in targeting and treating hematological malignancies and disorders. It sheds light on the latest research, development, and clinical trials, detailing how various pharmaceutical approaches leverage siRNA against blood disorders, mainly concentrating on cancers. It outlines the preferred molecular targets and physiological barriers to delivery while emphasizing the growing potential of various therapeutic delivery methods. The need for further research is articulated in the context of overcoming the shortcomings of siRNA in order to enrich discussions around siRNA's role in managing blood disorders and aiding the scientific community in advancing more targeted and effective treatments.
Collapse
Affiliation(s)
- Saba Abbasi Dezfouli
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2V2, Canada
| | | | - Hasan Uludag
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta T6G 2V2, Canada
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta T6G 2V2, Canada
| |
Collapse
|
10
|
Pant A, Laliwala A, Holstein SA, Mohs AM. Recent advances in targeted drug delivery systems for multiple myeloma. J Control Release 2024; 376:215-230. [PMID: 39384153 PMCID: PMC11611669 DOI: 10.1016/j.jconrel.2024.10.003] [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: 08/09/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024]
Abstract
Despite significant therapeutic advances, multiple myeloma (MM) remains a challenging, incurable, hematological malignancy. The efficacy of traditional chemotherapy and currently available anti-MM agents is in part limited by their adverse effects, which restrict their therapeutic potential. Nanotherapeutics is an emerging field of cancer therapy that can overcome the biological and chemical barriers of existing anticancer drugs. This review presents an overview of recent advancements in nanoparticle- and immunotherapy-based drug delivery systems for MM treatment. It further delves into the targeting strategies, mechanism of controlled drug release, and challenges associated with the development of drug delivery systems for the treatment of MM.
Collapse
Affiliation(s)
- Ashruti Pant
- Department of Pharmaceutical Sciences, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA.
| | - Aayushi Laliwala
- Department of Pharmaceutical Sciences, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA.
| | - Sarah A Holstein
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Department of Internal Medicine, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA.
| | - Aaron M Mohs
- Department of Pharmaceutical Sciences, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, 505 S 45 St, Omaha, NE 68198, USA; Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, S 45th St, Omaha, NE 68198, USA.
| |
Collapse
|
11
|
Garbayo E, El Moukhtari SH, Rodríguez-Nogales C, Agirre X, Rodriguez-Madoz JR, Rodriguez-Marquez P, Prósper F, Couvreur P, Blanco-Prieto MJ. RNA-loaded nanoparticles for the treatment of hematological cancers. Adv Drug Deliv Rev 2024; 214:115448. [PMID: 39303823 DOI: 10.1016/j.addr.2024.115448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/07/2024] [Accepted: 09/08/2024] [Indexed: 09/22/2024]
Abstract
Hematological cancers encompass a diverse group of malignancies affecting the blood, bone marrow, lymph nodes, and spleen. These disorders present unique challenges due to their complex etiology and varied clinical manifestations. Despite significant advancements in understanding and treating hematological malignancies, innovative therapeutic approaches are continually sought to enhance patient outcomes. This review highlights the application of RNA nanoparticles (RNA-NPs) in the treatment of hematological cancers. We delve into detailed discussions on in vitro and preclinical studies involving RNA-NPs for adult patients, as well as the application of RNA-NPs in pediatric hematological cancer. The review also addresses ongoing clinical trials involving RNA-NPs and explores the emerging field of CAR-T therapy engineered by RNA-NPs. Finally, we discuss the challenges still faced in translating RNA-NP research to clinics.
Collapse
Affiliation(s)
- Elisa Garbayo
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain
| | - Souhaila H El Moukhtari
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Carlos Rodríguez-Nogales
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain
| | - Xabier Agirre
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain; Hemato-Oncology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pío XII 55, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Juan R Rodriguez-Madoz
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain; Hemato-Oncology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pío XII 55, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Paula Rodriguez-Marquez
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain; Hemato-Oncology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pío XII 55, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Felipe Prósper
- Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain; Hemato-Oncology Program, Center for Applied Medical Research (CIMA), University of Navarra, Avenida Pío XII 55, 31008 Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), 28029 Madrid, Spain; Departmento de Hematología and CCUN, Clínica Universidad de Navarra, University of Navarra, Avenida Pío XII 36, 31008 Pamplona, Spain
| | - Patrick Couvreur
- Institut Galien Paris-Sud, UMR CNRS 8612, Université Paris-Saclay, Orsay Cedex, France.
| | - María J Blanco-Prieto
- Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, Universidad de Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, C/Irunlarrea 3, 31008 Pamplona, Spain; Cancer Center Clinica Universidad de Navarra (CCUN). Avenida Pio XII 36, 31008 Pamplona, Spain.
| |
Collapse
|
12
|
Serpico L, Zhu Y, Maia RF, Sumedha S, Shahbazi MA, Santos HA. Lipid nanoparticles-based RNA therapies for breast cancer treatment. Drug Deliv Transl Res 2024; 14:2823-2844. [PMID: 38831199 PMCID: PMC11384647 DOI: 10.1007/s13346-024-01638-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 06/05/2024]
Abstract
Breast cancer (BC) prevails as a major burden on global healthcare, being the most prevalent form of cancer among women. BC is a complex and heterogeneous disease, and current therapies, such as chemotherapy and radiotherapy, frequently fall short in providing effective solutions. These treatments fail to mitigate the risk of cancer recurrence and cause severe side effects that, in turn, compromise therapeutic responses in patients. Over the last decade, several strategies have been proposed to overcome these limitations. Among them, RNA-based technologies have demonstrated their potential across various clinical applications, notably in cancer therapy. However, RNA therapies are still limited by a series of critical issues like off-target effect and poor stability in circulation. Thus, novel approaches have been investigated to improve the targeting and bioavailability of RNA-based formulations to achieve an appropriate therapeutic outcome. Lipid nanoparticles (LNPs) have been largely proven to be an advantageous carrier for nucleic acids and RNA. This perspective explores the most recent advances on RNA-based technology with an emphasis on LNPs' utilization as effective nanocarriers in BC therapy and most recent progresses in their clinical applications.
Collapse
Affiliation(s)
- Luigia Serpico
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.
| | - Yuewen Zhu
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Renata Faria Maia
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Sumedha Sumedha
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Groningen, The Netherlands.
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
13
|
Masarwy R, Stotsky-Oterin L, Elisha A, Hazan-Halevy I, Peer D. Delivery of nucleic acid based genome editing platforms via lipid nanoparticles: Clinical applications. Adv Drug Deliv Rev 2024; 211:115359. [PMID: 38857763 DOI: 10.1016/j.addr.2024.115359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/17/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024]
Abstract
CRISPR/Cas technology presents a promising approach for treating a wide range of diseases, including cancer and genetic disorders. Despite its potential, the translation of CRISPR/Cas into effective in-vivo gene therapy encounters challenges, primarily due to the need for safe and efficient delivery mechanisms. Lipid nanoparticles (LNPs), FDA-approved for RNA delivery, show potential for delivering also CRISPR/Cas, offering the capability to efficiently encapsulate large mRNA molecules with single guide RNAs. However, achieving precise targeting in-vivo remains a significant obstacle, necessitating further research into optimizing LNP formulations. Strategies to enhance specificity, such as modifying LNP structures and incorporating targeting ligands, are explored to improve organ and cell type targeting. Furthermore, the development of base and prime editing technology presents a potential breakthrough, offering precise modifications without generating double-strand breaks (DSBs). Prime editing, particularly when delivered via targeted LNPs, holds promise for treating diverse diseases safely and precisely. This review assesses both the progress made and the persistent challenges faced in using LNP-encapsulated CRISPR-based technologies for therapeutic purposes, with a particular focus on clinical translation.
Collapse
Affiliation(s)
- Razan Masarwy
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lior Stotsky-Oterin
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
| | - Aviad Elisha
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel; School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Inbal Hazan-Halevy
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.
| | - Dan Peer
- Laboratory of Precision Nanomedicine, The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel; Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
14
|
Park J, Wu Y, Suk Kim J, Byun J, Lee J, Oh YK. Cytoskeleton-modulating nanomaterials and their therapeutic potentials. Adv Drug Deliv Rev 2024; 211:115362. [PMID: 38906478 DOI: 10.1016/j.addr.2024.115362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/25/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
The cytoskeleton, an intricate network of protein fibers within cells, plays a pivotal role in maintaining cell shape, enabling movement, and facilitating intracellular transport. Its involvement in various pathological states, ranging from cancer proliferation and metastasis to the progression of neurodegenerative disorders, underscores its potential as a target for therapeutic intervention. The exploration of nanotechnology in this realm, particularly the use of nanomaterials for cytoskeletal modulation, represents a cutting-edge approach with the promise of novel treatments. Inorganic nanomaterials, including those derived from gold, metal oxides, carbon, and black phosphorus, alongside organic variants such as peptides and proteins, are at the forefront of this research. These materials offer diverse mechanisms of action, either by directly interacting with cytoskeletal components or by influencing cellular signaling pathways that, in turn, modulate the cytoskeleton. Recent advancements have introduced magnetic field-responsive and light-responsive nanomaterials, which allow for targeted and controlled manipulation of the cytoskeleton. Such precision is crucial in minimizing off-target effects and enhancing therapeutic efficacy. This review explores the importance of research into cytoskeleton-targeting nanomaterials for developing therapeutic interventions for a range of diseases. It also addresses the progress made in this field, the challenges encountered, and future directions for using nanomaterials to modulate the cytoskeleton. The continued exploration of nanomaterials for cytoskeleton modulation holds great promise for advancing therapeutic strategies against a broad spectrum of diseases, marking a significant step forward in the intersection of nanotechnology and medicine.
Collapse
Affiliation(s)
- Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Jung Suk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Junho Byun
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | - Jaiwoo Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
15
|
Sanchez AJDS, Loughrey D, Echeverri ES, Huayamares SG, Radmand A, Paunovska K, Hatit M, Tiegreen KE, Santangelo PJ, Dahlman JE. Substituting Poly(ethylene glycol) Lipids with Poly(2-ethyl-2-oxazoline) Lipids Improves Lipid Nanoparticle Repeat Dosing. Adv Healthc Mater 2024; 13:e2304033. [PMID: 38318754 DOI: 10.1002/adhm.202304033] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/22/2024] [Indexed: 02/07/2024]
Abstract
Poly(ethylene glycol) (PEG)-lipids are used in Food-and-Drug-Administration-approved lipid nanoparticle (LNP)-RNA drugs, which are safe and effective. However, it is reported that PEG-lipids may also contribute to accelerated blood clearance and rare cases of hypersensitivity; this highlights the utility of exploring PEG-lipid alternatives. Here, it is shown that LNPs containing poly(2-ethyl-2-oxazoline) (PEOZ)-lipids can deliver messenger RNA (mRNA) to multiple cell types in mice inside and outside the liver. In addition, it is reported that LNPs formulated with PEOZ-lipids show reduced clearance from the bloodstream and lower levels of antistealth lipid immunoglobulin Ms than LNPs formulated with PEG-lipids. These data justify further exploration of PEOZ-lipids as alternatives to PEG-lipids in LNP-RNA formulations.
Collapse
Affiliation(s)
- Alejandro J Da Silva Sanchez
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Department of Chemical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - David Loughrey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Elisa Schrader Echeverri
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Sebastian G Huayamares
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Afsane Radmand
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Department of Chemical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Marine Hatit
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Karen E Tiegreen
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Philip J Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - James E Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
16
|
Abstract
Carriers for RNA delivery must be dynamic, first stabilizing and protecting therapeutic RNA during delivery to the target tissue and across cellular membrane barriers and then releasing the cargo in bioactive form. The chemical space of carriers ranges from small cationic lipids applied in lipoplexes and lipid nanoparticles, over medium-sized sequence-defined xenopeptides, to macromolecular polycations applied in polyplexes and polymer micelles. This perspective highlights the discovery of distinct virus-inspired dynamic processes that capitalize on mutual nanoparticle-host interactions to achieve potent RNA delivery. From the host side, subtle alterations of pH, ion concentration, redox potential, presence of specific proteins, receptors, or enzymes are cues, which must be recognized by the RNA nanocarrier via dynamic chemical designs including cleavable bonds, alterable physicochemical properties, and supramolecular assembly-disassembly processes to respond to changing biological microenvironment during delivery.
Collapse
Affiliation(s)
- Simone Berger
- Department of Pharmacy, Pharmaceutical Biotechnology, Ludwig-Maximilians-Universität Munich, 81377Munich, Germany
- Center for NanoScience, Ludwig-Maximilians-Universität Munich, 80799Munich, Germany
| | - Ulrich Lächelt
- Center for NanoScience, Ludwig-Maximilians-Universität Munich, 80799Munich, Germany
- Department of Pharmaceutical Sciences, University of Vienna, Vienna1090, Austria
| | - Ernst Wagner
- Department of Pharmacy, Pharmaceutical Biotechnology, Ludwig-Maximilians-Universität Munich, 81377Munich, Germany
- Center for NanoScience, Ludwig-Maximilians-Universität Munich, 80799Munich, Germany
| |
Collapse
|
17
|
Radmand A, Kim H, Beyersdorf J, Dobrowolski CN, Zenhausern R, Paunovska K, Huayamares SG, Hua X, Han K, Loughrey D, Hatit MZC, Del Cid A, Ni H, Shajii A, Li A, Muralidharan A, Peck HE, Tiegreen KE, Jia S, Santangelo PJ, Dahlman JE. Cationic cholesterol-dependent LNP delivery to lung stem cells, the liver, and heart. Proc Natl Acad Sci U S A 2024; 121:e2307801120. [PMID: 38437539 PMCID: PMC10945827 DOI: 10.1073/pnas.2307801120] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 09/22/2023] [Indexed: 03/06/2024] Open
Abstract
Adding a cationic helper lipid to a lipid nanoparticle (LNP) can increase lung delivery and decrease liver delivery. However, it remains unclear whether charge-dependent tropism is universal or, alternatively, whether it depends on the component that is charged. Here, we report evidence that cationic cholesterol-dependent tropism can differ from cationic helper lipid-dependent tropism. By testing how 196 LNPs delivered mRNA to 22 cell types, we found that charged cholesterols led to a different lung:liver delivery ratio than charged helper lipids. We also found that combining cationic cholesterol with a cationic helper lipid led to mRNA delivery in the heart as well as several lung cell types, including stem cell-like populations. These data highlight the utility of exploring charge-dependent LNP tropism.
Collapse
Affiliation(s)
- Afsane Radmand
- Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA30332
- Department of Chemical Engineering, Georgia Institute of Technology, Atlanta, GA30332
| | - Hyejin Kim
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Jared Beyersdorf
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Curtis N. Dobrowolski
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Ryan Zenhausern
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Kalina Paunovska
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Sebastian G. Huayamares
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Xuanwen Hua
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Keyi Han
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - David Loughrey
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Marine Z. C. Hatit
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Ada Del Cid
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Huanzhen Ni
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Aram Shajii
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Andrea Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Abinaya Muralidharan
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA30332
- Institute for Electronics and Nanotechnology, Georgia Institute of Technology, Atlanta, GA30332
| | - Hannah E. Peck
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Karen E. Tiegreen
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Shu Jia
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - Philip J. Santangelo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| | - James E. Dahlman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA30332
| |
Collapse
|
18
|
Jia J, Yang J, Qian L, Zhou B, Tang X, Liu S, Wu L, Chen J, Kuang Y. Controlled siRNA Release of Nanopolyplex for Effective Targeted Anticancer Therapy in Animal Model. Int J Nanomedicine 2024; 19:1145-1161. [PMID: 38344438 PMCID: PMC10859097 DOI: 10.2147/ijn.s443636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 01/16/2024] [Indexed: 02/15/2024] Open
Abstract
Introduction Spatiotemporally controlled release of siRNA for anti-tumor therapy poses significant challenges. Near-infrared (NIR) light, known for its exceptional tissue penetration and minimal tissue invasiveness, holds promise as a viable exogenous stimulus for inducing controlled siRNA release in vivo. However, the majority of light-responsive chemical bonds exhibit absorption wavelengths in the ultraviolet (UV) or short-wavelength visible light range. Methods To achieve NIR-controlled siRNA release, the study synthesized a UV-sensitive triblock copolymer cRGD-poly(ethylene glycol)-b-poly(aspartic acid ester-5-(2'-(dimethylamino)ethoxy)-2-nitrobenzyl alcohol)-b-polyphenylalanine, abbreviated as cRGD-PEG-PAsp(EDONB)-PPHE. This copolymer is composed of a cRGD-capped PEG block (cRGD-PEG), a poly(aspartate) block modified with cationic moieties through UV-cleavable 2-nitrobenzyl ester bonds [PAsp(EDONB)], and a hydrophobic polyphenylalanine block (PPHE). The cationic amphiphilic polymer cRGD-PEG-PAsp(EDONB)-PPHE can assemble with hydrophobic upconversion nanoparticles (UCNPs) to form a cationic micelle designated as T-UCNP, which subsequently complexes with siRNA to create the final nanopolyplex T-si/UCNP. siRNA-PLK1 was employed to prepare T-PLK1/UCNP nanopolyplex for anti-tumor therapy. Results T-PLK1/UCNP not only exhibited outstanding tumor cell targeting through cRGD modification but also achieved 980 nm NIR-controlled PLK1 gene silencing. This was achieved by utilizing the encapsulated UCNPs to convert NIR into UV light, facilitating the cleavage of 2-nitrobenzyl ester bonds. As a result, there was a significant suppression of tumor growth. Conclusion The UCNPs-encapsulated nanopolyplex T-si/UCNP, capable of co-delivering siRNA and UCNPs, enables precise NIR-controlled release of siRNA at the tumor site for cancer RNAi therapy. This nanopolyplex can enhance the controllability and safety of RNAi therapy for tumors, and it also holds the potential to serve as a platform for achieving controlled release and activation of other drugs, such as mRNA and DNA.
Collapse
Affiliation(s)
- Jingchao Jia
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
- Department of General Surgery, Jiangyin Hospital Affiliated to Nantong University, Wuxi, People’s Republic of China
| | - Jing Yang
- Jiangnan University Medical Center, Wuxi, People’s Republic of China
| | - Leimin Qian
- Department of General Surgery, Jiangyin Hospital Affiliated to Nantong University, Wuxi, People’s Republic of China
| | - Biao Zhou
- Department of General Surgery, Jiangyin Hospital Affiliated to Nantong University, Wuxi, People’s Republic of China
| | - Xiaodong Tang
- Department of General Surgery, Jiangyin Hospital Affiliated to Nantong University, Wuxi, People’s Republic of China
| | - Shuanghai Liu
- Department of General Surgery, Jiangyin Hospital Affiliated to Nantong University, Wuxi, People’s Republic of China
| | - Li Wu
- Department of Pharmaceutics, People’s Hospital of Shanggao, Yichun, People’s Republic of China
| | - Jifeng Chen
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Jinan University, Guangzhou, People’s Republic of China
| | - Yuting Kuang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, People’s Republic of China
| |
Collapse
|
19
|
Hu Z, Zhao X, Wu Z, Qu B, Yuan M, Xing Y, Song Y, Wang Z. Lymphatic vessel: origin, heterogeneity, biological functions, and therapeutic targets. Signal Transduct Target Ther 2024; 9:9. [PMID: 38172098 PMCID: PMC10764842 DOI: 10.1038/s41392-023-01723-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 11/03/2023] [Accepted: 11/23/2023] [Indexed: 01/05/2024] Open
Abstract
Lymphatic vessels, comprising the secondary circulatory system in human body, play a multifaceted role in maintaining homeostasis among various tissues and organs. They are tasked with a serious of responsibilities, including the regulation of lymph absorption and transport, the orchestration of immune surveillance and responses. Lymphatic vessel development undergoes a series of sophisticated regulatory signaling pathways governing heterogeneous-origin cell populations stepwise to assemble into the highly specialized lymphatic vessel networks. Lymphangiogenesis, as defined by new lymphatic vessels sprouting from preexisting lymphatic vessels/embryonic veins, is the main developmental mechanism underlying the formation and expansion of lymphatic vessel networks in an embryo. However, abnormal lymphangiogenesis could be observed in many pathological conditions and has a close relationship with the development and progression of various diseases. Mechanistic studies have revealed a set of lymphangiogenic factors and cascades that may serve as the potential targets for regulating abnormal lymphangiogenesis, to further modulate the progression of diseases. Actually, an increasing number of clinical trials have demonstrated the promising interventions and showed the feasibility of currently available treatments for future clinical translation. Targeting lymphangiogenic promoters or inhibitors not only directly regulates abnormal lymphangiogenesis, but improves the efficacy of diverse treatments. In conclusion, we present a comprehensive overview of lymphatic vessel development and physiological functions, and describe the critical involvement of abnormal lymphangiogenesis in multiple diseases. Moreover, we summarize the targeting therapeutic values of abnormal lymphangiogenesis, providing novel perspectives for treatment strategy of multiple human diseases.
Collapse
Affiliation(s)
- Zhaoliang Hu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Xushi Zhao
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Zhonghua Wu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Bicheng Qu
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Minxian Yuan
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China
| | - Yanan Xing
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
| | - Yongxi Song
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
| | - Zhenning Wang
- Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors (China Medical University), Ministry of Education, 155 North Nanjing Street, Heping District, Shenyang, 110001, China.
| |
Collapse
|
20
|
Tafech B, Mohabatpour F, Hedtrich S. Surface modification of lipid nanoparticles for gene therapy. J Gene Med 2024; 26:e3642. [PMID: 38043928 DOI: 10.1002/jgm.3642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 10/30/2023] [Accepted: 11/05/2023] [Indexed: 12/05/2023] Open
Abstract
Gene therapies have the potential to target and effectively treat a variety of diseases including cancer as well as genetic, neurological, and autoimmune disorders. Although we have made significant advances in identifying non-viral strategies to deliver genetic cargo, certain limitations remain. In general, gene delivery is challenging for several reasons including the instabilities of nucleic acids to enzymatic and chemical degradation and the presence of restrictive biological barriers such as cell, endosomal and nuclear membranes. The emergence of lipid nanoparticles (LNPs) helped overcome many of these challenges. Despite its success, further optimization is required for LNPs to yield efficient gene delivery to extrahepatic tissues, as LNPs favor accumulation in the liver after systemic administration. In this mini-review, we provide an overview of current preclinical approaches in that LNP surface modification was leveraged for cell and tissue targeting by conjugating aptamers, antibodies, and peptides among others. In addition to their cell uptake and efficiency-enhancing effects, we outline the (dis-)advantages of the different targeting moieties and commonly used conjugation strategies.
Collapse
Affiliation(s)
- Belal Tafech
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Fatemeh Mohabatpour
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sarah Hedtrich
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
- Center of Biological Design, Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Berlin, Germany
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
- Max-Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| |
Collapse
|
21
|
Elsafy S, Metselaar J, Lammers T. Nanomedicine - Immune System Interactions: Limitations and Opportunities for the Treatment of Cancer. Handb Exp Pharmacol 2024; 284:231-265. [PMID: 37578622 DOI: 10.1007/164_2023_685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Nanoparticles interact with immune cells in many different ways. These interactions are crucially important for determining nanoparticles' ability to be used for cancer therapy. Traditionally, strategies such as PEGylation have been employed to reduce (the kinetics of) nanoparticle uptake by immune cells, to endow them with long circulation properties, and to enable them to exploit the Enhanced Permeability and Retention (EPR) effect to accumulate in tumors. More recently, with immunotherapy becoming an increasingly important cornerstone in the clinical management of cancer, ever more research efforts in academia and industry are focusing on specifically targeting immune cells with nanoparticles. In this chapter, we describe the barriers and opportunities of immune cell targeting with nanoparticles, and we discuss how nanoparticle-based drug delivery to specific immune cell populations in tumors as well as in secondary myeloid and lymphoid organs (such as bone marrow, lymph nodes, and spleen) can be leveraged to boost the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Sara Elsafy
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), Center for Biohybrid Medical Systems (CBMS), University Hospital RWTH Aachen, Aachen, Germany
| | - Josbert Metselaar
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), Center for Biohybrid Medical Systems (CBMS), University Hospital RWTH Aachen, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), Center for Biohybrid Medical Systems (CBMS), University Hospital RWTH Aachen, Aachen, Germany.
| |
Collapse
|
22
|
Sela M, Poley M, Mora-Raimundo P, Kagan S, Avital A, Kaduri M, Chen G, Adir O, Rozencweig A, Weiss Y, Sade O, Leichtmann-Bardoogo Y, Simchi L, Aga-Mizrachi S, Bell B, Yeretz-Peretz Y, Zaid Or A, Choudhary A, Rosh I, Cordeiro D, Cohen-Adiv S, Berdichevsky Y, Odeh A, Shklover J, Shainsky-Roitman J, Schroeder JE, Hershkovitz D, Hasson P, Ashkenazi A, Stern S, Laviv T, Ben-Zvi A, Avital A, Ashery U, Maoz BM, Schroeder A. Brain-Targeted Liposomes Loaded with Monoclonal Antibodies Reduce Alpha-Synuclein Aggregation and Improve Behavioral Symptoms in Parkinson's Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304654. [PMID: 37753928 PMCID: PMC7615408 DOI: 10.1002/adma.202304654] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 09/04/2023] [Indexed: 09/28/2023]
Abstract
Monoclonal antibodies (mAbs) hold promise in treating Parkinson's disease (PD), although poor delivery to the brain hinders their therapeutic application. In the current study, it is demonstrated that brain-targeted liposomes (BTL) enhance the delivery of mAbs across the blood-brain-barrier (BBB) and into neurons, thereby allowing the intracellular and extracellular treatment of the PD brain. BTL are decorated with transferrin to improve brain targeting through overexpressed transferrin-receptors on the BBB during PD. BTL are loaded with SynO4, a mAb that inhibits alpha-synuclein (AS) aggregation, a pathological hallmark of PD. It is shown that 100-nm BTL cross human BBB models intact and are taken up by primary neurons. Within neurons, SynO4 is released from the nanoparticles and bound to its target, thereby reducing AS aggregation, and enhancing neuronal viability. In vivo, intravenous BTL administration results in a sevenfold increase in mAbs in brain cells, decreasing AS aggregation and neuroinflammation. Treatment with BTL also improve behavioral motor function and learning ability in mice, with a favorable safety profile. Accordingly, targeted nanotechnologies offer a valuable platform for drug delivery to treat brain neurodegeneration.
Collapse
Affiliation(s)
- Mor Sela
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Maria Poley
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Patricia Mora-Raimundo
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Shaked Kagan
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Aviram Avital
- The Norman Seiden Multidisciplinary Program for Nanoscience and Nanotechnology, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Maya Kaduri
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Gal Chen
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
- The Interdisciplinary Program for Biotechnology, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Omer Adir
- The Norman Seiden Multidisciplinary Program for Nanoscience and Nanotechnology, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Adi Rozencweig
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Yfat Weiss
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ofir Sade
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | | | - Lilach Simchi
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa 3498838, Israel
| | - Shlomit Aga-Mizrachi
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa 3498838, Israel
| | - Batia Bell
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9190500, Israel
| | - Yoel Yeretz-Peretz
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9190500, Israel
| | - Aviv Zaid Or
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ashwani Choudhary
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Idan Rosh
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Diogo Cordeiro
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Stav Cohen-Adiv
- The Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Yevgeny Berdichevsky
- The Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Anas Odeh
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Jeny Shklover
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Janna Shainsky-Roitman
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Joshua E. Schroeder
- Spine Unit, Orthopedic Complex, Hadassah Hebrew University Medical Center, Kiryat Hadassah, POB 12000, Jerusalem 9190500, Israel
| | - Dov Hershkovitz
- Department of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv 6997801, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Peleg Hasson
- Department of Genetics and Developmental Biology, The Rappaport Faculty of Medicine and Research Institute, Technion – Israel Institute of Technology, Haifa 32000, Israel
| | - Avraham Ashkenazi
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- The Department of Cell and Developmental Biology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa 3498838, Israel
| | - Tal Laviv
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ayal Ben-Zvi
- Department of Developmental Biology and Cancer Research, The Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9190500, Israel
| | - Avi Avital
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa 3498838, Israel
| | - Uri Ashery
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ben M. Maoz
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol Center for Regenerative Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Avi Schroeder
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion – Israel Institute of Technology, Haifa 32000, Israel
| |
Collapse
|
23
|
Lin Y, Cheng Q, Wei T. Surface engineering of lipid nanoparticles: targeted nucleic acid delivery and beyond. BIOPHYSICS REPORTS 2023; 9:255-278. [PMID: 38516300 PMCID: PMC10951480 DOI: 10.52601/bpr.2023.230022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 11/28/2023] [Indexed: 03/23/2024] Open
Abstract
Harnessing surface engineering strategies to functionalize nucleic acid-lipid nanoparticles (LNPs) for improved performance has been a hot research topic since the approval of the first siRNA drug, patisiran, and two mRNA-based COVID-19 vaccines, BNT162b2 and mRNA-1273. Currently, efforts have been mainly made to construct targeted LNPs for organ- or cell-type-specific delivery of nucleic acid drugs by conjugation with various types of ligands. In this review, we describe the surface engineering strategies for nucleic acid-LNPs, considering ligand types, conjugation chemistries, and incorporation methods. We then outline the general purification and characterization techniques that are frequently used following the engineering step and emphasize the specific techniques for certain types of ligands. Next, we comprehensively summarize the currently accessible organs and cell types, as well as the other applications of the engineered LNPs. Finally, we provide considerations for formulating targeted LNPs and discuss the challenges of successfully translating the "proof of concept" from the laboratory into the clinic. We believe that addressing these challenges could accelerate the development of surface-engineered LNPs for targeted nucleic acid delivery and beyond.
Collapse
Affiliation(s)
- Yi Lin
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Qiang Cheng
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Tuo Wei
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|