1
|
Mallareddy V, Daigavane S. Nanoparticle-Mediated Cell Delivery: Advancements in Corneal Endothelial Regeneration. Cureus 2024; 16:e56958. [PMID: 38665717 PMCID: PMC11044897 DOI: 10.7759/cureus.56958] [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: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Corneal endothelial dysfunction poses significant challenges in ophthalmology, leading to corneal edema and vision loss. Traditional treatments, including corneal transplantation, are limited by donor scarcity and potential complications. Nanoparticle-mediated cell delivery emerges as a promising approach for corneal endothelial regeneration, offering targeted and minimally invasive solutions. This comprehensive review provides insights into the role of nanoparticles in enhancing cell survival, integration, and therapeutic efficacy. We discuss the current understanding of corneal endothelial dysfunction, emphasizing the importance of regeneration. Furthermore, we explore the potential implications of nanoparticle-mediated approaches in clinical practice, highlighting opportunities for personalized treatment strategies. Future directions are also discussed, including optimization of nanoparticle design and exploration of combination therapies. Overall, this review elucidates the promising advancements in nanoparticle-mediated cell delivery for corneal endothelial regeneration and underscores the importance of continued research efforts in this evolving field.
Collapse
Affiliation(s)
- Vijaya Mallareddy
- Ophthalmology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sachin Daigavane
- Ophthalmology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
2
|
Ma X, Wu F, Peng C, Chen H, Zhang D, Han T. Exploration of mRNA nanoparticles based on DOTAP through optimization of the helper lipids. Biotechnol J 2023; 18:e2300123. [PMID: 37545293 DOI: 10.1002/biot.202300123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 06/15/2023] [Accepted: 08/02/2023] [Indexed: 08/08/2023]
Abstract
Lipid nanoparticles (LNPs) are one of the most efficient carriers for RNA packaging and delivery, and vaccines based on mRNA-LNPs have received substantial attention since the outbreak of the COVID-19 pandemic. LNPs based on 1,2-dioleoyl-3-trimethylammonium propane (DOTAP) have been widely used in preclinical and clinical settings. A novel non-viral gene delivery system called LNP3 was previously developed, which was composed of DOTAP, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), and cholesterol. One of the helper lipids in this carrier was DOPE, which belongs to phospholipids. Given that substituting DOPE with non-phospholipids as helper lipids can increase the delivery efficiency of some LNPs, this study aimed to examine whether non-phospholipids can be formulated with DOTAP as helper lipids. It was found that monoglycerides with C14:0, C16:0, C18:0, C18:1, and C18:2 mediated mRNA transfection, and the transfection efficiency varied between C18:0, C18:1, and C18:2. Furthermore, substituting of the glycerol with other moieties such as the cholesterol or the ethanolamine similarly mediated mRNA transfection. The introduction of cholesterol can further improve the transfection capacity of some DOTAP-based LNPs. One of the best-performing formulations, LNP3-MO, was used to mediate luciferase-mRNA expression in vivo, and the luminescence signal was found to be mainly enriched in the lung and spleen. In addition, the level of SARS-CoV-2 spike antibody in the serum increased after three doses of LNP3-MO mediated SARS-CoV-2 spike mRNA. Altogether, this study demonstrates that non-phospholipids are promising helper lipids that can be formulated with DOTAP to facilitate efficient delivery of mRNAs in vitro and in vivo with organ-specific targeting.
Collapse
Affiliation(s)
- Xueni Ma
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Fanqi Wu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Respiratory, Lanzhou University Second Hospital, Lanzhou, China
| | - Caihong Peng
- Department of Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Huiling Chen
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Tiyun Han
- Key Laboratory of Digestive Diseases, Lanzhou University Second Hospital, Lanzhou, China
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
3
|
Shannon SR, Ben-Akiva E, Green JJ. Approaches towards biomaterial-mediated gene editing for cancer immunotherapy. Biomater Sci 2022; 10:6675-6687. [PMID: 35858470 PMCID: PMC10112382 DOI: 10.1039/d2bm00806h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gene therapies are transforming treatment modalities for many human diseases and disorders, including those in ophthalmology, oncology, and nephrology. To maximize the clinical efficacy and safety of these treatments, consideration of both delivery materials and cargos is critical. In consideration of the former, a large effort has been placed on transitioning away from potentially immunoreactive and toxic viral delivery mechanisms towards safer and highly tunable nonviral delivery mechanisms, including polymeric, lipid-based, and inorganic carriers. This change of paradigm does not come without obstacles, as efficient non-viral delivery is challenging, particularly to immune cells, and has yet to see clinical translation breakthroughs for gene editing. This mini-review describes notable examples of biomaterial-based gene delivery to immune cells, with emphasis on recent in vivo successes. In consideration of delivery cargos, clustered regularly interspaced palindromic repeat (CRISPR) technology is reviewed and its great promise in the field of immune cell gene editing is described. This mini-review describes how leading non-viral delivery materials and CRISPR technology can be integrated together to advance its clinical potential for therapeutic gene transfer to immune cells to treat cancer.
Collapse
Affiliation(s)
- Sydney R Shannon
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Elana Ben-Akiva
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| | - Jordan J Green
- Department of Biomedical Engineering, Institute for NanoBioTechnology, and the Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
- Departments of Ophthalmology, Oncology, Neurosurgery, Materials Science & Engineering, and Chemical & Biomolecular Engineering, and the Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| |
Collapse
|
4
|
Kiru L, Zlitni A, Tousley AM, Dalton GN, Wu W, Lafortune F, Liu A, Cunanan KM, Nejadnik H, Sulchek T, Moseley ME, Majzner RG, Daldrup-Link HE. In vivo imaging of nanoparticle-labeled CAR T cells. Proc Natl Acad Sci U S A 2022; 119:e2102363119. [PMID: 35101971 PMCID: PMC8832996 DOI: 10.1073/pnas.2102363119] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 12/10/2021] [Indexed: 01/20/2023] Open
Abstract
Metastatic osteosarcoma has a poor prognosis with a 2-y, event-free survival rate of ∼15 to 20%, highlighting the need for the advancement of efficacious therapeutics. Chimeric antigen receptor (CAR) T-cell therapy is a potent strategy for eliminating tumors by harnessing the immune system. However, clinical trials with CAR T cells in solid tumors have encountered significant challenges and have not yet demonstrated convincing evidence of efficacy for a large number of patients. A major bottleneck for the success of CAR T-cell therapy is our inability to monitor the accumulation of the CAR T cells in the tumor with clinical-imaging techniques. To address this, we developed a clinically translatable approach for labeling CAR T cells with iron oxide nanoparticles, which enabled the noninvasive detection of the iron-labeled T cells with magnetic resonance imaging (MRI), photoacoustic imaging (PAT), and magnetic particle imaging (MPI). Using a custom-made microfluidics device for T-cell labeling by mechanoporation, we achieved significant nanoparticle uptake in the CAR T cells, while preserving T-cell proliferation, viability, and function. Multimodal MRI, PAT, and MPI demonstrated homing of the T cells to osteosarcomas and off-target sites in animals administered with T cells labeled with the iron oxide nanoparticles, while T cells were not visualized in animals infused with unlabeled cells. This study details the successful labeling of CAR T cells with ferumoxytol, thereby paving the way for monitoring CAR T cells in solid tumors.
Collapse
Affiliation(s)
- Louise Kiru
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
| | - Aimen Zlitni
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
| | | | | | - Wei Wu
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
| | - Famyrah Lafortune
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
| | - Anna Liu
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| | - Kristen May Cunanan
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
| | - Hossein Nejadnik
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA 19104
| | - Todd Sulchek
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA 30332
| | - Michael Eugene Moseley
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305
| | - Robbie G Majzner
- Department of Pediatrics, Stanford University, Stanford, CA 94305
- Stanford Cancer Institute, Stanford University, Stanford, CA 94305
| | - Heike Elisabeth Daldrup-Link
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305;
- Department of Pediatrics, Stanford University, Stanford, CA 94305
- Stanford Cancer Institute, Stanford University, Stanford, CA 94305
| |
Collapse
|
5
|
Keim D, Gollner K, Gollner U, Jérôme V, Freitag R. Generation of Recombinant Primary Human B Lymphocytes Using Non-Viral Vectors. Int J Mol Sci 2021; 22:8239. [PMID: 34361005 PMCID: PMC8347318 DOI: 10.3390/ijms22158239] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/29/2021] [Accepted: 07/29/2021] [Indexed: 12/26/2022] Open
Abstract
Although the development of gene delivery systems based on non-viral vectors is advancing, it remains a challenge to deliver plasmid DNA into human blood cells. The current "gold standard", namely linear polyethyleneimine (l-PEI 25 kDa), in particular, is unable to produce transgene expression levels >5% in primary human B lymphocytes. Here, it is demonstrated that a well-defined 24-armed poly(2-dimethylamino) ethyl methacrylate (PDMAEMA, 755 kDa) nano-star is able to reproducibly elicit high transgene expression (40%) at sufficient residual viability (69%) in primary human B cells derived from tonsillar tissue. Moreover, our results indicate that the length of the mitogenic stimulation prior to transfection is an important parameter that must be established during the development of the transfection protocol. In our hands, four days of stimulation with rhCD40L post-thawing led to the best transfection results in terms of TE and cell survival. Most importantly, our data argue for an impact of the B cell subsets on the transfection outcomes, underlining that the complexity and heterogeneity of a given B cell population pre- and post-transfection is a critical parameter to consider in the multiparametric approach required for the implementation of the transfection protocol.
Collapse
Affiliation(s)
- Daniel Keim
- Process Biotechnology, University of Bayreuth, 95447 Bayreuth, Germany; (D.K.); (V.J.)
| | - Katrin Gollner
- Praxis am Schießgraben, Schießgraben 21, 95326 Kulmbach, Germany; (K.G.); (U.G.)
| | - Ulrich Gollner
- Praxis am Schießgraben, Schießgraben 21, 95326 Kulmbach, Germany; (K.G.); (U.G.)
| | - Valérie Jérôme
- Process Biotechnology, University of Bayreuth, 95447 Bayreuth, Germany; (D.K.); (V.J.)
| | - Ruth Freitag
- Process Biotechnology, University of Bayreuth, 95447 Bayreuth, Germany; (D.K.); (V.J.)
| |
Collapse
|
6
|
Kumar ARK, Shou Y, Chan B, L K, Tay A. Materials for Improving Immune Cell Transfection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007421. [PMID: 33860598 DOI: 10.1002/adma.202007421] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/08/2020] [Indexed: 06/12/2023]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy holds great promise for preventing and treating deadly diseases such as cancer. However, it remains challenging to transfect and engineer primary immune cells for clinical cell manufacturing. Conventional tools using viral vectors and bulk electroporation suffer from low efficiency while posing risks like viral transgene integration and excessive biological perturbations. Emerging techniques using microfluidics, nanoparticles, and high-aspect-ratio nanostructures can overcome these challenges, and on top of that, provide universal and high-throughput cargo delivery. Herein, the strengths and limitations of traditional and emerging materials for immune cell transfection, and commercial development of these tools, are discussed. To enhance the characterization of transfection techniques and uptake by the clinical community, a list of in vitro and in vivo assays to perform, along with relevant protocols, is recommended. The overall aim, herein, is to motivate the development of novel materials to meet rising demand in transfection for clinical CAR-T cell manufacturing.
Collapse
Affiliation(s)
- Arun R K Kumar
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Brian Chan
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Krishaa L
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| |
Collapse
|
7
|
Zhang R, El-Mayta R, Murdoch TJ, Warzecha CC, Billingsley MM, Shepherd SJ, Gong N, Wang L, Wilson JM, Lee D, Mitchell MJ. Helper lipid structure influences protein adsorption and delivery of lipid nanoparticles to spleen and liver. Biomater Sci 2021; 9:1449-1463. [PMID: 33404020 PMCID: PMC8753632 DOI: 10.1039/d0bm01609h] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Nucleic acids, such as messenger RNAs, antisense oligonucleotides, and short interfering RNAs, hold great promise for treating previously 'undruggable' diseases. However, there are numerous biological barriers that hinder nucleic acid delivery to target cells and tissues. While lipid nanoparticles (LNPs) have been developed to protect nucleic acids from degradation and mediate their intracellular delivery, it is challenging to predict how alterations in LNP formulation parameters influence delivery to different organs. In this study, we utilized high-throughput in vivo screening to probe for structure-function relationships of intravenously administered LNPs along with quartz crystal microbalance with dissipation monitoring (QCM-D) to measure the binding affinity of LNPs to apolipoprotein E (ApoE), a protein implicated in the clearance and uptake of lipoproteins by the liver. High-throughput in vivo screening of a library consisting of 96 LNPs identified several formulations containing the helper lipid 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) that preferentially accumulated in the liver, while identical LNPs that substituted DOPE with the helper lipid 1,2-distearoyl-sn-glycero-3-phosphocholine (DSPC) preferentially accumulated in the spleen. Using QCM-D, it was found that one DOPE-containing LNP formulation (LNP 42) had stronger interactions with ApoE than an identical LNP formulation that substituted DOPE with DSPC (LNP 90). In order to further validate our findings, we formulated LNP 42 and LNP 90 to encapsulate Cy3-siRNA or mRNA encoding for firefly luciferase. The DSPC-containing LNP (LNP 90) was found to increase delivery to the spleen for both siRNA (two-fold) and mRNA (five-fold). In terms of liver delivery, the DOPE-containing LNP (LNP 42) enhanced mRNA delivery to the liver by two-fold and improved liver transfection by three-fold. Understanding the role of the helper lipid in LNP biodistribution and ApoE adsorption may aid in the future design of LNPs for nucleic acid therapeutics.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Helfer BM, Bulte JW. Cell Surveillance Using Magnetic Resonance Imaging. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00042-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
9
|
Chen Y, Wang J, Li X, Hu N, Voelcker NH, Xie X, Elnathan R. Emerging Roles of 1D Vertical Nanostructures in Orchestrating Immune Cell Functions. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2001668. [PMID: 32844502 PMCID: PMC7461044 DOI: 10.1002/adma.202001668] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 05/16/2020] [Indexed: 05/07/2023]
Abstract
Engineered nano-bio cellular interfaces driven by 1D vertical nanostructures (1D-VNS) are set to prompt radical progress in modulating cellular processes at the nanoscale. Here, tuneable cell-VNS interfacial interactions are probed and assessed, highlighting the use of 1D-VNS in immunomodulation, and intracellular delivery into immune cells-both crucial in fundamental and translational biomedical research. With programmable topography and adaptable surface functionalization, 1D-VNS provide unique biophysical and biochemical cues to orchestrate innate and adaptive immunity, both ex vivo and in vivo. The intimate nanoscale cell-VNS interface leads to membrane penetration and cellular deformation, facilitating efficient intracellular delivery of diverse bioactive cargoes into hard-to-transfect immune cells. The unsettled interfacial mechanisms reported to be involved in VNS-mediated intracellular delivery are discussed. By identifying up-to-date progress and fundamental challenges of current 1D-VNS technology in immune-cell manipulation, it is hoped that this report gives timely insights for further advances in developing 1D-VNS as a safe, universal, and highly scalable platform for cell engineering and enrichment in advanced cancer immunotherapy such as chimeric antigen receptor-T therapy.
Collapse
Affiliation(s)
- Yaping Chen
- Monash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClayton3168Australia
| | - Ji Wang
- The First Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityGuangzhou510006China
| | - Xiangling Li
- State Key Laboratory of Optoelectronic Materials and TechnologiesSchool of Electronics and Information TechnologySun Yat‐sen UniversityGuangzhou510006China
| | - Ning Hu
- State Key Laboratory of Optoelectronic Materials and TechnologiesSchool of Electronics and Information TechnologySun Yat‐sen UniversityGuangzhou510006China
| | - Nicolas H. Voelcker
- Monash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClayton3168Australia
- Department of Materials Science and EngineeringMonash University22 Alliance LaneClaytonVIC3168Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO)ClaytonVIC3168Australia
- INM‐Leibniz Institute for New MaterialsCampus D2 2Saarbrücken66123Germany
| | - Xi Xie
- The First Affiliated Hospital of Sun Yat‐sen UniversitySun Yat‐sen UniversityGuangzhou510006China
- State Key Laboratory of Optoelectronic Materials and TechnologiesSchool of Electronics and Information TechnologySun Yat‐sen UniversityGuangzhou510006China
| | - Roey Elnathan
- Monash Institute of Pharmaceutical SciencesMonash University381 Royal ParadeParkvilleVIC3052Australia
- Melbourne Centre for NanofabricationVictorian Node of the Australian National Fabrication Facility151 Wellington RoadClayton3168Australia
- Department of Materials Science and EngineeringMonash University22 Alliance LaneClaytonVIC3168Australia
| |
Collapse
|
10
|
Wilk AJ, Weidenbacher NLB, Vergara R, Haabeth OAW, Levy R, Waymouth RM, Wender PA, Blish CA. Charge-altering releasable transporters enable phenotypic manipulation of natural killer cells for cancer immunotherapy. Blood Adv 2020; 4:4244-4255. [PMID: 32898247 PMCID: PMC7479957 DOI: 10.1182/bloodadvances.2020002355] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022] Open
Abstract
Chimeric antigen receptor (CAR) natural killer (NK) cells are an emerging cell therapy with promising results in oncology trials. However, primary human NK cells are difficult to transfect, hampering both mechanistic studies and clinical applications of NK cells. Currently, NK cell CAR modification relies on viral vectors or cell activation. The former raises cost and tolerability issues, while the latter alters NK cell biology. Here, we report that readily synthesized and inexpensive nonviral charge-altering releasable transporters (CARTs) efficiently transfect primary human NK cells with messenger RNA without relying on NK cell activation. Compared with electroporation, CARTs transfect NK cells more efficiently, better preserve cell viability, and cause minimal reconfiguration of NK cell phenotype and function. We use CARTs to generate cytotoxic primary anti-CD19 CAR NK cells, demonstrating this technology can drive clinical applications of NK cells. To our knowledge, CARTs represent the first efficacious transfection technique for resting primary human NK cells that preserves NK cell phenotype and can enable new biological discoveries and therapeutic applications of this understudied lymphocyte subset.
Collapse
Affiliation(s)
- Aaron J Wilk
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
- Program in Immunology
- Medical Scientist Training Program
| | | | - Rosemary Vergara
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
| | | | - Ronald Levy
- Division of Oncology, Department of Medicine
| | | | - Paul A Wender
- Department of Chemistry
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA; and
| | - Catherine A Blish
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine
- Chan Zuckerberg Biohub, San Francisco, CA
| |
Collapse
|
11
|
AbouAitah K, Swiderska-Sroda A, Kandeil A, Salman AMM, Wojnarowicz J, Ali MA, Opalinska A, Gierlotka S, Ciach T, Lojkowski W. Virucidal Action Against Avian Influenza H5N1 Virus and Immunomodulatory Effects of Nanoformulations Consisting of Mesoporous Silica Nanoparticles Loaded with Natural Prodrugs. Int J Nanomedicine 2020; 15:5181-5202. [PMID: 32801685 PMCID: PMC7398888 DOI: 10.2147/ijn.s247692] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Background Combating infectious diseases caused by influenza virus is a major challenge due to its resistance to available drugs and vaccines, side effects, and cost of treatment. Nanomedicines are being developed to allow targeted delivery of drugs to attack specific cells or viruses. Materials and Methods In this study, mesoporous silica nanoparticles (MSNs) functionalized with amino groups and loaded with natural prodrugs of shikimic acid (SH), quercetin (QR) or both were explored as a novel antiviral nanoformulations targeting the highly pathogenic avian influenza H5N1 virus. Also, the immunomodulatory effects were investigated in vitro tests and anti-inflammatory activity was determined in vivo using the acute carrageenan-induced paw edema rat model. Results Prodrugs alone or the MSNs displayed weaker antiviral effects as evidenced by virus titers and plaque formation compared to nanoformulations. The MSNs-NH2-SH and MSNs-NH2-SH-QR2 nanoformulations displayed a strong virucidal by inactivating the H5N1 virus. They induced also strong immunomodulatory effects: they inhibited cytokines (TNF-α, IL-1β) and nitric oxide production by approximately 50% for MSNs-NH2-SH-QR2 (containing both SH and QR). Remarkable anti-inflammatory effects were observed during in vivo tests in an acute carrageenan-induced rat model. Conclusion Our preliminary findings show the potential of nanotechnology for the application of natural prodrug substances to produce a novel safe, effective, and affordable antiviral drug.
Collapse
Affiliation(s)
- Khaled AbouAitah
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland.,Medicinal and Aromatic Plants Research Department, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), P.C.12622, Dokki, Giza, Egypt
| | - Anna Swiderska-Sroda
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Ahmed Kandeil
- Center of Scientific Excellence for Influenza Viruses, Water Pollution Research Department, Environmental Research Division, National Research Centre (NRC) P.C.12622, Dokki, Giza, Egypt
| | - Asmaa M M Salman
- Medicinal and Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), P.C. 12622, Dokki, Giza, Egypt
| | - Jacek Wojnarowicz
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Mohamed A Ali
- Center of Scientific Excellence for Influenza Viruses, Water Pollution Research Department, Environmental Research Division, National Research Centre (NRC) P.C.12622, Dokki, Giza, Egypt
| | - Agnieszka Opalinska
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Stanislaw Gierlotka
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| | - Tomasz Ciach
- Biomedical Engineering Laboratory, Faculty of Chemical and Process Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Witold Lojkowski
- Laboratory of Nanostructures and Nanomedicine, Institute of High Pressure Physics, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
12
|
Tay A, Melosh N. Transfection with Nanostructure Electro‐Injection is Minimally Perturbative. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900133] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Andy Tay
- Department of Materials Science and Engineering Stanford University Stanford CA 94305 USA
- Department of Biomedical Engineering National University of Singapore Singapore 117583 Singapore
| | - Nicholas Melosh
- Department of Materials Science and Engineering Stanford University Stanford CA 94305 USA
| |
Collapse
|
13
|
Chen Z, Lee WG. A switching role of hard-uptake nanoparticles in microalgae cell electroporation. Analyst 2019; 144:3581-3589. [PMID: 31065636 DOI: 10.1039/c9an00314b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The microalgal cell wall is a natural barrier that limits the efficiency of gene delivery in algae genetic engineering. Here, we report the role of hard-uptake nanoparticles (huNPs) in microalgae cell electroporation to enhance the delivery of genes in Chlamydomonas reinhardtii. This role can be divided into two categories: (i) a 'transient state' for short-term behavior under confocal visualization and (ii) a 'steady state' for long-term behavior in cell culture. First, the 'transient' role of gene-huNP complexes was investigated after washing for clear confocal imaging to observe the location of huNPs after electroporation. Second, the 'steady-state' role of the gene-huNP complexes was examined after electroporation by transferring cells to a fresh, medium-rich culture environment without washing to obtain a stable cell culture. For selection of the huNPs, we used two types of nanoparticles (NPs, 250 nm and 530 nm) larger than the threshold size of electroporation uptake to avoid unwanted endocytic uptake of NPs. In the transient state, the visualization results indicate that gene-NP (250 nm) complexes were positioned on the cells and helped to deliver more genes than did the 530 nm NPs. In the steady state, the gene-NP (530 nm) complexes helped stably deliver more genes to the cells by precipitation of NPs due to gravity. We believe that these findings illustrate how gene-NP complexes function in microalgae cell electroporation and could help set up a protocol for enhanced microalgae applications associated with NPs such as environmental waste removal and biofuel production.
Collapse
Affiliation(s)
- Zhenzhong Chen
- Department of Mechanical Engineering, Kyung Hee University, Yongin 17104, Republic of Korea.
| | - Won Gu Lee
- Department of Mechanical Engineering, Kyung Hee University, Yongin 17104, Republic of Korea.
| |
Collapse
|
14
|
Saini S, Korf H, Liang S, Verbeke R, Manshian B, Raemdonck K, Lentacker I, Gysemans C, De Smedt SC, Himmelreich U. Challenges for labeling and longitudinal tracking of adoptively transferred autoreactive T lymphocytes in an experimental type-1 diabetes model. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2019; 32:295-305. [PMID: 30648196 DOI: 10.1007/s10334-018-0720-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 11/20/2018] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Tracking the autoreactive T-cell migration in the pancreatic region after labeling with fluorinated nanoparticles (1,2-dioleoyl-sn-glycero-3-phosphoethanolamine-N-[3-(2-pyridyldithio)propionate]-perfluoro-15-crown-5-ether nanoparticles, PDP-PFCE NPs) in a diabetic murine model using 19F MRI. MATERIALS AND METHODS Synthesis of novel PDP-PFCE fluorine tracer was performed for in vitro labeling of T cells. Labeling conditions were optimized using different PDP-PFCE NPs concentrations. For in vivo 19F MRI, mice were longitudinally followed after adoptive transfer of activated, autoreactive, labeled T cells in NOD.SCID mice. RESULTS Established MR protocols were used for challenging T cell labeling to track inflammation in a model of diabetes after successful labeling of CD4+ and CD8+ T cells with PDP-PFCE NPs. However, T cells were difficult to be detected in vivo after their engraftment in animals. DISCUSSION We showed successful in vitro labeling of T cells using novel fluorinated liposomal nanoparticles. However, insufficient and slow accumulation of labeled T cells and subsequent T cell proliferation in the pancreatic region remains as limitations of in vivo cell imaging by 19F MRI.
Collapse
Affiliation(s)
- Shweta Saini
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Hannelie Korf
- Laboratory of Hepatology, CHROMETA Department, KU Leuven, Leuven, Belgium
| | | | - Rein Verbeke
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium
| | - Bella Manshian
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Koen Raemdonck
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Ghent University, Ghent, Belgium
| | - Conny Gysemans
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | | | - Uwe Himmelreich
- Biomedical MRI/MoSAIC, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
15
|
Fornaguera C, Castells-Sala C, Borrós S. Unraveling Polymeric Nanoparticles Cell Uptake Pathways: Two Decades Working to Understand Nanoparticles Journey to Improve Gene Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1288:117-138. [PMID: 31916235 DOI: 10.1007/5584_2019_467] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Polymeric nanoparticles have aroused an increasing interest in the last decades as novel advanced delivery systems to improve the treatment of many diseases. Hard work has been performed worldwide designing and developing polymeric nanoparticles using different building blocks, which target specific cell types, trying to avoid bioaccumulation and degradation pathways. The main handicap of the design is to understand the final fate and the journey that the nanoparticle will follow, which is intimately ligated with the chemical and physical properties of the nanoparticles themselves and specific factors of the targeted cells. Although the huge number of published scientific articles regarding polymeric nanoparticles for biomedical applications, their use in clinics is still limited. This fact could be explained by the limited data reporting the interaction of the huge diversity of polymeric nanoparticles with cells. This knowledge is essential to understand nanoparticle uptake and trafficking inside cells to the subcellular target structure.In this chapter, we aim to contribute to this field of knowledge by: (1) summarizing the polymeric nanoparticles properties and cellular factors that influence nanoparticle endocytosis and (2) reviewing the endocytic pathways classified as a function of nanoparticle size and as a function of the receptor playing a role. The revision of previously reported endocytic pathways for particular polymeric nanoparticles could facilitate scientist involved in this field to easily delineate efficient delivery systems based on polymeric nanoparticles.
Collapse
Affiliation(s)
- C Fornaguera
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain.
| | - C Castells-Sala
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain
| | - S Borrós
- Grup d'Enginyeria de Materials (Gemat), Institut Químic de Sarrià (IQS), Universitat Ramon Llull (URL), Barcelona, Spain
| |
Collapse
|
16
|
Viral and Nonviral Engineering of Natural Killer Cells as Emerging Adoptive Cancer Immunotherapies. J Immunol Res 2018; 2018:4054815. [PMID: 30306093 PMCID: PMC6166361 DOI: 10.1155/2018/4054815] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/26/2018] [Accepted: 08/01/2018] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells are powerful immune effectors whose antitumor activity is regulated through a sophisticated network of activating and inhibitory receptors. As effectors of cancer immunotherapy, NK cells are attractive as they do not attack healthy self-tissues nor do they induce T cell-driven inflammatory cytokine storm, enabling their use as allogeneic adoptive cellular therapies. Clinical responses to adoptive NK-based immunotherapy have been thwarted, however, by the profound immunosuppression induced by the tumor microenvironment, particularly severe in the context of solid tumors. In addition, the short postinfusion persistence of NK cells in vivo has limited their clinical efficacy. Enhancing the antitumor immunity of NK cells through genetic engineering has been fueled by the promise that impaired cytotoxic functionality can be restored or augmented with the use of synthetic genetic approaches. Alongside expressing chimeric antigen receptors to overcome immune escape by cancer cells, enhance their recognition, and mediate their killing, NK cells have been genetically modified to enhance their persistence in vivo by the expression of cytokines such as IL-15, avoid functional and metabolic tumor microenvironment suppression, or improve their homing ability, enabling enhanced targeting of solid tumors. However, NK cells are notoriously adverse to endogenous gene uptake, resulting in low gene uptake and transgene expression with many vector systems. Though viral vectors have achieved the highest gene transfer efficiencies with NK cells, nonviral vectors and gene transfer approaches—electroporation, lipofection, nanoparticles, and trogocytosis—are emerging. And while the use of NK cell lines has achieved improved gene transfer efficiencies particularly with viral vectors, challenges with primary NK cells remain. Here, we discuss the genetic engineering of NK cells as they relate to NK immunobiology within the context of cancer immunotherapy, highlighting the most recent breakthroughs in viral vectors and nonviral approaches aimed at genetic reprogramming of NK cells for improved adoptive immunotherapy of cancer, and, finally, address their clinical status.
Collapse
|