1
|
Hiep Tran T, Thu Phuong Tran T. Current status of nanoparticle-mediated immunogenic cell death in cancer immunotherapy. Int Immunopharmacol 2024; 142:113085. [PMID: 39276455 DOI: 10.1016/j.intimp.2024.113085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/02/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024]
Abstract
Immunogenic cell death (ICD) encompasses various forms of cell death modalities, including apoptosis, necroptosis, ferroptosis, and pyroptosis. It arises from a harmonious interplay of adjuvant (damage-associated molecular patterns-DAMPs and chemokines/cytokines) and antigenicity (tumor-associated antigens-TAA) to induce immune-reaction toward cancer cells. Inducing ICD stands out as a promising approach in cancer immunotherapy, capable of directly eliminating cancer cells and of eliciting enduring antitumor immune responses. Conventional tumor therapies like radiation therapy, photodynamic therapy, and chemotherapy can also induce ICD which could amplify their activities. The development of effective ICD inducers like nano-systems is crucial for ensuring safe and efficacious immunotherapy. Nanoparticles hold considerable promise in cancer therapy, offering enhanced therapeutic outcomes and mitigated side effects. They could be the capacity to adjust systemic biodistribution, augment the accumulation of therapeutic agents at the intended site and protect active agents from the complexity of human biofluid. This review aims to outline the role of nanoparticles in triggering ICD for cancer immunotherapy that potentially pave the way for cancer treatment.
Collapse
Affiliation(s)
- Tuan Hiep Tran
- Faculty of Pharmacy, PHENIKAA University, Hanoi 12116, Vietnam
| | - Thi Thu Phuong Tran
- Department of Life Sciences, University of Science and Technology of Hanoi (USTH), Vietnam Academy of Science and Technology (VAST), 18 Hoang Quoc Viet, Cau Giay, Hanoi, Vietnam.
| |
Collapse
|
2
|
Haghighi E, Abolmaali SS, Dehshahri A, Mousavi Shaegh SA, Azarpira N, Tamaddon AM. Navigating the intricate in-vivo journey of lipid nanoparticles tailored for the targeted delivery of RNA therapeutics: a quality-by-design approach. J Nanobiotechnology 2024; 22:710. [PMID: 39543630 PMCID: PMC11566655 DOI: 10.1186/s12951-024-02972-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 11/03/2024] [Indexed: 11/17/2024] Open
Abstract
RNA therapeutics, such as mRNA, siRNA, and CRISPR-Cas9, present exciting avenues for treating diverse diseases. However, their potential is commonly hindered by vulnerability to degradation and poor cellular uptake, requiring effective delivery systems. Lipid nanoparticles (LNPs) have emerged as a leading choice for in vivo RNA delivery, offering protection against degradation, enhanced cellular uptake, and facilitation of endosomal escape. However, LNPs encounter numerous challenges for targeted RNA delivery in vivo, demanding advanced particle engineering, surface functionalization with targeting ligands, and a profound comprehension of the biological milieu in which they function. This review explores the structural and physicochemical characteristics of LNPs, in-vivo fate, and customization for RNA therapeutics. We highlight the quality-by-design (QbD) approach for targeted delivery beyond the liver, focusing on biodistribution, immunogenicity, and toxicity. In addition, we explored the current challenges and strategies associated with LNPs for in-vivo RNA delivery, such as ensuring repeated-dose efficacy, safety, and tissue-specific gene delivery. Furthermore, we provide insights into the current clinical applications in various classes of diseases and finally prospects of LNPs in RNA therapeutics.
Collapse
Affiliation(s)
- Elahe Haghighi
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ali Dehshahri
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Ali Mousavi Shaegh
- Laboratory of Microfluidics and Medical Microsystems, Research Institute for Medical Sciences, Mashhad University of Medical Sciences, Mashhad, Iran
- Orthopedic Research Center, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
- Clinical Research Development Unit, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Azarpira
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, Shiraz University of Medical Sciences, Shiraz, Iran.
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Pharmaceutics, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Grunberger JW, Newton HS, Donohue D, Dobrovolskaia MA, Ghandehari H. Role of physicochemical properties in silica nanoparticle-mediated immunostimulation. Nanotoxicology 2024; 18:599-617. [PMID: 39460666 DOI: 10.1080/17435390.2024.2418088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/01/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024]
Abstract
Immunostimulation caused by nanoparticles may be beneficial or adverse depending on their intended application. Activation of immune cells is beneficial for indications targeting the immune system for therapeutic purposes, such as tumor microenvironment reprogramming, immunotherapy, and vaccines. When it is unwanted, however, immunostimulation may lead to excessive inflammation, cytokine storm, and hypersensitivity reactions. The increasing use of silica nanoparticles (SiNPs) for the delivery of drugs, imaging agents, and antigens warrants preclinical studies aimed at understanding carrier-mediated effects on the number, activation status, and function of immune cell subsets. Herein, we present an in vitro study utilizing primary human peripheral blood mononuclear cells (PBMC) to investigate the proinflammatory properties of four types of SiNPs varying in size and porosity. Cytokine analysis was performed in resting and LPS-primed PBMC cultures to understand the ability of silica nanoparticles to induce de novo and exaggerate preexisting inflammation, respectively. Changes in the number and activation status of lymphoid and myeloid cells were studied by flow cytometry to gain further insight into SiNP-mediated immunostimulation. Nonporous SiNPs were found to be more proinflammatory than mesoporous SiNPs, and larger-sized particles induced greater cytokine response. LPS-primed PBMC resulted in increased susceptibility to SiNPs. Immunophenotyping analysis of SiNP-treated PBMC resulted in T and B lymphocyte, natural killer cell, and dendritic cell activation. Additionally, a loss of regulatory T cells and an increase in γδ TCR T cell population were observed with all particles. These findings have implications for the utility of SiNPs for the delivery of drugs and imaging agents.
Collapse
Affiliation(s)
- Jason William Grunberger
- Utah Center for Nanomedicine, Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA
| | - Hannah S Newton
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Frederick, MD, USA
| | - Duncan Donohue
- Statistics Department, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Frederick, MD, USA
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Frederick, MD, USA
| | - Hamidreza Ghandehari
- Utah Center for Nanomedicine, Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
4
|
Lee JH, Sergi C, Kast RE, Kanwar BA, Bourbeau J, Oh S, Sohn MG, Lee CJ, Coleman MD. Aggravating mechanisms from COVID-19. Virol J 2024; 21:228. [PMID: 39334442 PMCID: PMC11430051 DOI: 10.1186/s12985-024-02506-8] [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: 08/08/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) induces immune-mediated diseases. The pathophysiology of COVID-19 uses the following three mechanisms: (1) inflammasome activation mechanism; (2) cGAS-STING signaling mechanism; and (3) SAMHD1 tetramerization mechanism, which leads to IFN-I production. Interactions between the host and virus govern induction, resulting in multiorgan impacts. The NLRP3 with cGAS-STING constitutes the primary immune response. The expression of SARS-CoV-2 ORF3a, NSP6, NSP7, and NSP8 blocks innate immune activation and facilitates virus replication by targeting the RIG-I/MDA5, TRIF, and cGAS-STING signaling. SAMHD1 has a target motif for CDK1 to protect virion assembly, threonine 592 to modulate a catalytically active tetramer, and antiviral IFN responses to block retroviral infection. Plastic and allosteric nucleic acid binding of SAMHD1 modulates the antiretroviral activity of SAMHD1. Therefore, inflammasome activation, cGAS-STING signaling, and SAMHD1 tetramerization explain acute kidney injury, hepatic, cardiac, neurological, and gastrointestinal injury of COVID-19. It might be necessary to effectively block the pathological courses of diverse diseases.
Collapse
Affiliation(s)
- Jong Hoon Lee
- Science and Research Center, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Department of Geriatrics, Gyeonggi Medical Center Pocheon Hospital, 1648 Pocheon-ro Sin-eup-dong, Pocheon-si, Gyeonggi-do, 11142, Republic of Korea.
| | - Consolato Sergi
- Division of Anatomical Pathology, Children's Hospital of Eastern Ontario (CHEO), University of Ottawa, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Richard E Kast
- IIAIGC Study Center, 11 Arlington Ct, Burlington, 05408 VT, USA
| | - Badar A Kanwar
- Haider Associates, 1999 Forest Ridge Dr, Bedford, TX, 76021, USA
| | - Jean Bourbeau
- Respiratory Epidemiology and Clinical Research Unit, McGill University Health Centre, Montréal, QC, Canada
| | - Sangsuk Oh
- Department of Food Engineering, Food Safety Laboratory, Memory Unit, Ewha Womans University, Seoul, 03670, Korea
| | - Mun-Gi Sohn
- Department of Food Science, KyungHee University College of Life Science, Seoul, 17104, Republic of Korea
| | - Chul Joong Lee
- Department of Anesthesiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Michael D Coleman
- College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
5
|
López-Espinosa J, Park P, Holcomb M, Godin B, Villapol S. Nanotechnology-driven therapies for neurodegenerative diseases: a comprehensive review. Ther Deliv 2024; 15:997-1024. [PMID: 39297726 PMCID: PMC11583628 DOI: 10.1080/20415990.2024.2401307] [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: 05/03/2024] [Accepted: 09/03/2024] [Indexed: 11/22/2024] Open
Abstract
Neurological diseases, characterized by neuroinflammation and neurodegeneration, impose a significant global burden, contributing to substantial morbidity, disability and mortality. A common feature of these disorders, including stroke, traumatic brain injury and Alzheimer's disease, is the impairment of the blood-brain barrier (BBB), a critical structure for maintaining brain homeostasis. The compromised BBB in neurodegenerative conditions poses a significant challenge for effective treatment, as it allows harmful substances to accumulate in the brain. Nanomedicine offers a promising approach to overcoming this barrier, with nanoparticles (NPs) engineered to deliver therapeutic agents directly to affected brain regions. This review explores the classification and design of NPs, divided into organic and inorganic categories and further categorized based on their chemical and physical properties. These characteristics influence the ability of NPs to carry and release therapeutic agents, target specific tissues and ensure appropriate clearance from the body. The review emphasizes the potential of NPs to enhance the diagnosis and treatment of neurodegenerative diseases through targeted delivery, improved drug bioavailability and real-time therapeutic efficacy monitoring. By addressing the challenges of the compromised BBB and targeting inflammatory biomarkers, NPs represent a cutting-edge strategy in managing neurological disorders, promising better patient outcomes.
Collapse
Affiliation(s)
- Jessica López-Espinosa
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
- School of Medicine and Health Sciences of Tecnológico de Monterrey, Guadalajara, México
| | - Peter Park
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
| | - Morgan Holcomb
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TXUSA
- Department of Obstetrics & Gynecology, Houston Methodist Hospital, Houston, TXUSA
- Department of Obstetrics & Gynecology, Weill Cornell Medicine College, New York, NYUSA
- Department of Biomedical Engineering, Texas A&M University, College Station, TXUSA
| | - Sonia Villapol
- Department of Neurosurgery & Center for Neuroregeneration, Houston, TX USA
- Department of Neuroscience in Neurological Surgery, Weill Cornell Medical College, New York, NY USA
| |
Collapse
|
6
|
Lu RM, Hsu HE, Perez SJLP, Kumari M, Chen GH, Hong MH, Lin YS, Liu CH, Ko SH, Concio CAP, Su YJ, Chang YH, Li WS, Wu HC. Current landscape of mRNA technologies and delivery systems for new modality therapeutics. J Biomed Sci 2024; 31:89. [PMID: 39256822 PMCID: PMC11389359 DOI: 10.1186/s12929-024-01080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Accepted: 08/20/2024] [Indexed: 09/12/2024] Open
Abstract
Realizing the immense clinical potential of mRNA-based drugs will require continued development of methods to safely deliver the bioactive agents with high efficiency and without triggering side effects. In this regard, lipid nanoparticles have been successfully utilized to improve mRNA delivery and protect the cargo from extracellular degradation. Encapsulation in lipid nanoparticles was an essential factor in the successful clinical application of mRNA vaccines, which conclusively demonstrated the technology's potential to yield approved medicines. In this review, we begin by describing current advances in mRNA modifications, design of novel lipids and development of lipid nanoparticle components for mRNA-based drugs. Then, we summarize key points pertaining to preclinical and clinical development of mRNA therapeutics. Finally, we cover topics related to targeted delivery systems, including endosomal escape and targeting of immune cells, tumors and organs for use with mRNA vaccines and new treatment modalities for human diseases.
Collapse
Affiliation(s)
- Ruei-Min Lu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Hsiang-En Hsu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | | | - Monika Kumari
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan
| | - Guan-Hong Chen
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Ming-Hsiang Hong
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Yin-Shiou Lin
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Ching-Hang Liu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Shih-Han Ko
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | | | - Yi-Jen Su
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan
| | - Yi-Han Chang
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan
| | - Wen-Shan Li
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan.
- Institute of Chemistry, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan.
| | - Han-Chung Wu
- Biomedical Translation Research Center, Academia Sinica, Taipei, 11571, Taiwan.
- Institute of Cellular and Organismic Biology, Academia Sinica, No. 128, Academia Road, Section 2, Taipei, 11529, Taiwan.
| |
Collapse
|
7
|
Jang M, Yeom K, Han J, Fagan E, Park JH. Inhalable mRNA Nanoparticle with Enhanced Nebulization Stability and Pulmonary Microenvironment Infiltration. ACS NANO 2024; 18:24204-24218. [PMID: 39174871 DOI: 10.1021/acsnano.4c05653] [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: 08/24/2024]
Abstract
The delivery of mRNA into the lungs is the key to solving infectious and intractable diseases that frequently occur in the lungs. Since inhalation using a nebulizer is the most promising method for mRNA delivery into the lungs, there have been many attempts toward adapting lipid nanoparticles for mRNA inhalation. However, conventional lipid nanoparticles, which have shown great effectiveness for systemic delivery of mRNA and intramuscular vaccination, are not effective for pulmonary delivery due to their structural instability during nebulization and their inability to adapt to the pulmonary microenvironment. To address these issues, we developed an ionizable liposome-mRNA lipocomplex (iLPX). iLPX has a highly ordered lipid bilayer structure, which increases stability during nebulization, and its poly(ethylene glycol)-free composition allows it to infiltrate the low serum environment and the pulmonary surfactant layer in the lungs. We selected an inhalation-optimized iLPX (IH-iLPX) using a multistep screening procedure that mimics the pulmonary delivery process of inhaled nanoparticles. The IH-iLPX showed a higher transfection efficiency in the lungs compared to conventional lipid nanoparticles after inhalation with no observed toxicity in vivo. Furthermore, analysis of lung distribution revealed even protein expression in the deep lungs, with effective delivery to epithelial cells. This study provides insights into the challenges and solutions related to the development of inhaled mRNA pulmonary therapeutics.
Collapse
Affiliation(s)
- Mincheol Jang
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Kyunghwan Yeom
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Junhee Han
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Erinn Fagan
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Ji-Ho Park
- Department of Bio and Brain Engineering and KAIST Institute for Health Science and Technology, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| |
Collapse
|
8
|
Shin H, Kang S, Won C, Min D. A Single-Dose mRNA Vaccine Employing Porous Silica Nanoparticles Induces Robust Immune Responses Against the Zika Virus. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2404590. [PMID: 39010673 PMCID: PMC11425238 DOI: 10.1002/advs.202404590] [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: 05/15/2024] [Indexed: 07/17/2024]
Abstract
Recently, lipid nanoparticles (LNPs)-based mRNA delivery has been approved by the FDA for SARS-CoV-2 vaccines. However, there are still considerable points for improvement in LNPs. Especially, local administration of LNPs-formulated mRNA can cause off-target translation of mRNA in distal organs which can induce unintended adverse effects. With the hypothesis that large and rigid nanoparticles can be applied to enhance retention of nanoparticles at the injection site, a polyethyleneimine (PEI)-coated porous silica nanoparticles (PPSNs)-based mRNA delivery platform is designed. PPSNs not only facilitate localized translation of mRNA at the site of injection but also prolonged protein expression. It is further demonstrated that the development of a highly efficacious Zika virus (ZIKV) vaccine using mRNA encoding full-length ZIKV pre-membrane (prM) and envelope (E) protein delivered by PPSNs. The ZIKV prME mRNA-loaded PPSNs vaccine elicits robust immune responses, including high levels of neutralizing antibodies and ZIKV E-specific T cell responses in C57BL/6 mice. Moreover, a single injection of prME-PPSNs vaccine provided complete protection against the ZIKV challenge in mice.
Collapse
Affiliation(s)
- Hojeong Shin
- Department of ChemistrySeoul National UniversitySeoul08826Republic of Korea
| | - Seounghun Kang
- Department of ChemistrySeoul National UniversitySeoul08826Republic of Korea
| | - Cheolhee Won
- Institute of Biotherapeutics Convergence TechnologyLemonex Inc.Seoul06683Republic of Korea
| | - Dal‐Hee Min
- Department of ChemistrySeoul National UniversitySeoul08826Republic of Korea
- Institute of Biotherapeutics Convergence TechnologyLemonex Inc.Seoul06683Republic of Korea
| |
Collapse
|
9
|
Barta BA, Radovits T, Dobos AB, Tibor Kozma G, Mészáros T, Berényi P, Facskó R, Fülöp T, Merkely B, Szebeni J. Comirnaty-induced cardiopulmonary distress and other symptoms of complement-mediated pseudo-anaphylaxis in a hyperimmune pig model: Causal role of anti-PEG antibodies. Vaccine X 2024; 19:100497. [PMID: 38933697 PMCID: PMC11201123 DOI: 10.1016/j.jvacx.2024.100497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 04/27/2024] [Accepted: 05/11/2024] [Indexed: 06/28/2024] Open
Abstract
Background Comirnaty, Pfizer-BioNTech's polyethylene-glycol (PEG)-containing Covid-19 vaccine, can cause hypersensitivity reactions (HSRs), or rarely, life-threatening anaphylaxis in a small fraction of immunized people. A causal role of anti-PEG antibodies (Abs) has been proposed, but causality has not yet proven in an animal model. The aim of this study was to provide such evidence using pigs immunized against PEG, which displayed very high levels of anti-PEG antibodies (Abs). We also aimed to find evidence for a role of complement activation and thromboxane A2 release in blood to explore the mechanism of anaphylaxis. Methods Pigs (n = 6) were immunized with 0.1 mg/kg PEGylated liposome (Doxebo) i.v., and the rise of anti-PEG IgG and IgM were measured in serial blood samples with ELISA. After ∼2-3 weeks the animals were injected i.v. with 1/3 human dose of the PEGylated mRNA vaccine, Comirnaty, and the hemodynamic (PAP, SAP) cardiopulmonary (HR, EtCO2,), hematological (WBC, granulocyte, lymphocyte and platelet counts) parameters and blood immune mediators (anti-PEG IgM and IgG antibodies, thromboxane B2, C3a) were measured as endpoints of HSRs (anaphylaxis). Results The level of anti-PEG IgM and IgG rose 5-10-thousand-fold in all of 6 pigs immunized with Doxebo by day 6, after which time all animals developed anaphylactic shock to i.v. injection of 1/3 human dose of Comirnaty. The reaction, starting within 1 min involved maximal pulmonary hypertension and decreased systemic pulse pressure amplitude, tachycardia, granulo- and thrombocytopenia, and skin reactions (flushing or rash). These physiological changes or their absence were paralleled by C3a and TXB2 rises in blood. Conclusions Consistent with previous studies, these data show a causal role of anti-PEG Abs in the anaphylaxis to Comirnaty, which involves complement activation, and, hence, it represents C activation-related pseudo-anaphylaxis. The setup provides the first large-animal model for mRNA-vaccine-induced anaphylaxis in humans.
Collapse
Affiliation(s)
| | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | | | - Gergely Tibor Kozma
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience LCC, Budapest, Hungary
| | - Tamás Mészáros
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience LCC, Budapest, Hungary
| | - Petra Berényi
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience LCC, Budapest, Hungary
| | - Réka Facskó
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience LCC, Budapest, Hungary
| | | | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - János Szebeni
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, Budapest, Hungary
- SeroScience LCC, Budapest, Hungary
- Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health Sciences, Miskolc University, Miskolc 2880, Hungary
- School of Chemical Engineering and Translational Nanobioscience Research Center, Sungkyunkwan University, Suwon 16419, South Korea
| |
Collapse
|
10
|
Ding T, Wang Y, Meng Y, Wu E, Shao Q, Lin S, Yu Y, Qian J, He Q, Zhang J, Wang J, Kohane DS, Zhan C. Reciprocal Interaction with Neutrophils Facilitates Cutaneous Accumulation of Liposomes. ACS NANO 2024; 18:18769-18784. [PMID: 38950189 DOI: 10.1021/acsnano.4c06638] [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: 07/03/2024]
Abstract
Liposomes are versatile drug delivery systems in clinical use for cancer and many other diseases. Unfortunately, PEGylated liposomal doxorubicin (sLip/DOX) exhibits serious dose-limiting cutaneous toxicities, which are closely related to the extravascular accumulation of sLip/DOX in the dermis. No clinical interventions have been proposed for cutaneous toxicities due to the elusive transport pathways. Herein, we showed that the reciprocal interaction between liposomes and neutrophils played pivotal roles in liposome extravasation into the dermis. Neutrophils captured liposomes via the complement receptor 3 (CD11b/CD18) recognizing the fragment of complement component C3 (iC3b) deposited on the liposomal surface. Uptake of liposomes also activated neutrophils to induce CD11b upregulation and enhanced the ability of neutrophils to migrate outside the capillaries. Furthermore, inhibition of complement activation either by CRIg-L-FH (a C3b/iC3b targeted complement inhibitor) or blocking the phosphate negative charge in mPEG-DSPE could significantly reduce liposome uptake by neutrophils and alleviate the cutaneous accumulation of liposomes. These results validated the liposome extravasation pathway mediated by neutrophils and provided potential solutions to the devastating cutaneous toxicities occurring during sLip/DOX treatment.
Collapse
Affiliation(s)
- Tianhao Ding
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yang Wang
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanchun Meng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
| | - Ercan Wu
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Qianwen Shao
- School of Pharmacy & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P. R. China
| | - Shiqi Lin
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Yifei Yu
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
| | - Jun Qian
- School of Pharmacy & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P. R. China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, West China School of Pharmacy, Sichuan University, Chengdu 610041, P. R. China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P. R. China
| | - Jing Wang
- Shanghai Institute of Immunology, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Changyou Zhan
- Department of Pharmacology, School of Basic Medical Sciences & Department of Pharmacy, Shanghai Pudong Hospital, Fudan University, Shanghai 200032, P. R. China
- School of Pharmacy & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 201203, P. R. China
- Shanghai Engineering Research Center for Synthetic Immunology, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
11
|
Lee JH, Sergi C, Kast RE, Kanwar BA, Bourbeau J, Oh S, Sohn MG, Lee CJ, Coleman MD. Basic implications on three pathways associated with SARS-CoV-2. Biomed J 2024:100766. [PMID: 39004185 DOI: 10.1016/j.bj.2024.100766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/04/2024] [Accepted: 07/10/2024] [Indexed: 07/16/2024] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) interacts between the host and virus and govern induction, resulting in multiorgan impacts. Its pathophysiology involves the followings: 1) the angiotensin-converting enzyme (ACE2) and Toll-like receptor (TLR) pathways: 2) the neuropilin (NRP) pathway: 3) the spike protein pathway. Therefore, it is necessary to block the pathological course with modulating innate lymphoid cells against diverse corona variants in the future.
Collapse
Affiliation(s)
- Jong Hoon Lee
- Science and Research Center, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| | - Consolato Sergi
- Division of Anatomical Pathology, Children's Hospital of Eastern Ontario (CHEO), University of Ottawa, 401 Smyth Road, Ottawa, ON, K1H 8L1, Canada
| | - Richard E Kast
- IIAIGC Study Center, 11 Arlington Ct, Burlington, 05408, VT, USA
| | - Badar A Kanwar
- Haider Associates, 1999 Forest Ridge Dr, Bedford, TX, 76021, USA
| | - Jean Bourbeau
- Respiratory Epidemiology and Clinical Research Unit, McGill University Health Centre, Montréal, QC, Canada
| | - Sangsuk Oh
- Department of Food Engineering, Food Safety Laboratory, Memory Unit, Ewha Womans University, Seoul, 03670, Republic of Korea
| | - Mun-Gi Sohn
- Department of Food Science, KyungHee University College of Life Science, Seoul, 17104, Republic of Korea
| | - Chul Joong Lee
- Department of Anesthesiology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Michael D Coleman
- College of Health and Life Sciences, Aston University, Birmingham, B4 7ET, UK.
| |
Collapse
|
12
|
Lin Y, Chen X, Wang K, Liang L, Zhang H. An Overview of Nanoparticle-Based Delivery Platforms for mRNA Vaccines for Treating Cancer. Vaccines (Basel) 2024; 12:727. [PMID: 39066365 PMCID: PMC11281455 DOI: 10.3390/vaccines12070727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/16/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
With its unique properties and potential applications, nanoparticle-based delivery platforms for messenger RNA (mRNA) vaccines have gained significant attention in recent years. Nanoparticles have the advantages of enhancing immunogenicity, targeting delivery, and improving stability, providing a new solution for drug and vaccine delivery. In some clinical studies, a variety of nanoparticle delivery platforms have been gradually applied to a wide range of vaccine applications. Current research priorities are exploring various types of nanoparticles as vaccine delivery systems to enhance vaccine stability and immunogenicity. Lipid nanoparticles (LNPs) have shown promising potential in preclinical and clinical studies on the efficient delivery of antigens to immune cells. Moreover, lipid nanoparticles and other nanoparticles for nucleic acids, especially for mRNA delivery systems, have shown vast potential for vaccine development. In this review, we present various vaccine platforms with an emphasis on nanoparticles as mRNA vaccine delivery vehicles. We describe several novel nanoparticle delivery platforms for mRNA vaccines, such as lipid-, polymer-, and protein-based nanoparticles. In addition, we provide an overview of the anti-tumor immunity of nanovaccines against different tumors in cancer immunotherapy. Finally, we outline future perspectives and remaining challenges for this promising technology of nanoparticle-based delivery platforms for vaccines.
Collapse
Affiliation(s)
- Yang Lin
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; (Y.L.); (X.C.); (K.W.)
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Xuehua Chen
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; (Y.L.); (X.C.); (K.W.)
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
| | - Ke Wang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; (Y.L.); (X.C.); (K.W.)
| | - Li Liang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; (Y.L.); (X.C.); (K.W.)
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
- Jinfeng Laboratory, Chongqing Science and Technology Innovation Center, Chongqing 401329, China
| | - Hongxia Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; (Y.L.); (X.C.); (K.W.)
- Guangdong Provincial Key Laboratory of Molecular Tumor Pathology, Guangzhou 510515, China
- Department of Pathology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
13
|
Elblová P, Lunova M, Dejneka A, Jirsa M, Lunov O. Impact of mechanical cues on key cell functions and cell-nanoparticle interactions. DISCOVER NANO 2024; 19:106. [PMID: 38907808 PMCID: PMC11193707 DOI: 10.1186/s11671-024-04052-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 06/14/2024] [Indexed: 06/24/2024]
Abstract
In recent years, it has been recognized that mechanical forces play an important regulative role in living organisms and possess a direct impact on crucial cell functions, ranging from cell growth to maintenance of tissue homeostasis. Advancements in mechanobiology have revealed the profound impact of mechanical signals on diverse cellular responses that are cell type specific. Notably, numerous studies have elucidated the pivotal role of different mechanical cues as regulatory factors influencing various cellular processes, including cell spreading, locomotion, differentiation, and proliferation. Given these insights, it is unsurprising that the responses of cells regulated by physical forces are intricately linked to the modulation of nanoparticle uptake kinetics and processing. This complex interplay underscores the significance of understanding the mechanical microenvironment in shaping cellular behaviors and, consequently, influencing how cells interact with and process nanoparticles. Nevertheless, our knowledge on how localized physical forces affect the internalization and processing of nanoparticles by cells remains rather limited. A significant gap exists in the literature concerning a systematic analysis of how mechanical cues might bias the interactions between nanoparticles and cells. Hence, our aim in this review is to provide a comprehensive and critical analysis of the existing knowledge regarding the influence of mechanical cues on the complicated dynamics of cell-nanoparticle interactions. By addressing this gap, we would like to contribute to a detailed understanding of the role that mechanical forces play in shaping the complex interplay between cells and nanoparticles.
Collapse
Affiliation(s)
- Petra Elblová
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Faculty of Mathematics and Physics, Charles University, Ke Karlovu 3, 121 16, Prague 2, Czech Republic
| | - Mariia Lunova
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Alexandr Dejneka
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021, Prague, Czech Republic
| | - Oleg Lunov
- Department of Optical and Biophysical Systems, Institute of Physics of the Czech Academy of Sciences, 18200, Prague, Czech Republic.
| |
Collapse
|
14
|
Wang Y, Qu J, Xiong C, Chen B, Xie K, Wang M, Liu Z, Yue Z, Liang Z, Wang F, Zhang T, Zhu G, Kuang YB, Shi P. Transdermal microarrayed electroporation for enhanced cancer immunotherapy based on DNA vaccination. Proc Natl Acad Sci U S A 2024; 121:e2322264121. [PMID: 38865265 PMCID: PMC11194603 DOI: 10.1073/pnas.2322264121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 05/14/2024] [Indexed: 06/14/2024] Open
Abstract
Despite the tremendous clinical potential of nucleic acid-based vaccines, their efficacy to induce therapeutic immune response has been limited by the lack of efficient local gene delivery techniques in the human body. In this study, we develop a hydrogel-based organic electronic device (μEPO) for both transdermal delivery of nucleic acids and in vivo microarrayed cell electroporation, which is specifically oriented toward one-step transfection of DNAs in subcutaneous antigen-presenting cells (APCs) for cancer immunotherapy. The μEPO device contains an array of microneedle-shaped electrodes with pre-encapsulated dry DNAs. Upon a pressurized contact with skin tissue, the electrodes are rehydrated, electrically triggered to release DNAs, and then electroporate nearby cells, which can achieve in vivo transfection of more than 50% of the cells in the epidermal and upper dermal layer. As a proof-of-concept, the μEPO technique is employed to facilitate transdermal delivery of neoantigen genes to activate antigen-specific immune response for enhanced cancer immunotherapy based on a DNA vaccination strategy. In an ovalbumin (OVA) cancer vaccine model, we show that high-efficiency transdermal transfection of APCs with OVA-DNAs induces robust cellular and humoral immune responses, including antigen presentation and generation of IFN-γ+ cytotoxic T lymphocytes with a more than 10-fold dose sparing over existing intramuscular injection (IM) approach, and effectively inhibits tumor growth in rodent animals.
Collapse
Affiliation(s)
- Yuan Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region999077, China
| | - Jin Qu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region999077, China
| | - Chuxiao Xiong
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region999077, China
| | - Bing Chen
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region999077, China
| | - Kai Xie
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region999077, China
| | - Mingxue Wang
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region999077, China
| | - Zhen Liu
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region999077, China
| | - Zhao Yue
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region999077, China
| | - Zhenghua Liang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong Special Administrative Region999077, China
| | - Feng Wang
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region999077, China
| | - Tianlong Zhang
- Department of Mechanical and Aerospace Engineering, The Hong Kong University of Science and Technology, Hong Kong Special Administrative Region999077, China
| | - Guangyu Zhu
- Department of Chemistry, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region999077, China
| | - Yi Becki Kuang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Kowloon, Hong Kong Special Administrative Region999077, China
| | - Peng Shi
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region999077, China
- Center of Super-Diamond and Advanced Films, City University of Hong Kong, Kowloon, Hong Kong Special Administrative Region999077, China
- Hong Kong Centre for Cerebro-Cardiovascular Health Engineering, Hong Kong Special Administrative Region999077, China
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen518000, China
| |
Collapse
|
15
|
Anchordoquy T, Artzi N, Balyasnikova IV, Barenholz Y, La-Beck NM, Brenner JS, Chan WCW, Decuzzi P, Exner AA, Gabizon A, Godin B, Lai SK, Lammers T, Mitchell MJ, Moghimi SM, Muzykantov VR, Peer D, Nguyen J, Popovtzer R, Ricco M, Serkova NJ, Singh R, Schroeder A, Schwendeman AA, Straehla JP, Teesalu T, Tilden S, Simberg D. Mechanisms and Barriers in Nanomedicine: Progress in the Field and Future Directions. ACS NANO 2024; 18:13983-13999. [PMID: 38767983 PMCID: PMC11214758 DOI: 10.1021/acsnano.4c00182] [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] [Indexed: 05/22/2024]
Abstract
In recent years, steady progress has been made in synthesizing and characterizing engineered nanoparticles, resulting in several approved drugs and multiple promising candidates in clinical trials. Regulatory agencies such as the Food and Drug Administration and the European Medicines Agency released important guidance documents facilitating nanoparticle-based drug product development, particularly in the context of liposomes and lipid-based carriers. Even with the progress achieved, it is clear that many barriers must still be overcome to accelerate translation into the clinic. At the recent conference workshop "Mechanisms and Barriers in Nanomedicine" in May 2023 in Colorado, U.S.A., leading experts discussed the formulation, physiological, immunological, regulatory, clinical, and educational barriers. This position paper invites open, unrestricted, nonproprietary discussion among senior faculty, young investigators, and students to trigger ideas and concepts to move the field forward.
Collapse
Affiliation(s)
- Thomas Anchordoquy
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, the University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Natalie Artzi
- Brigham and Woman's Hospital, Department of Medicine, Division of Engineering in Medicine, Harvard Medical School, Boston, Massachusetts 02215, United States
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| | - Irina V Balyasnikova
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University; Northwestern Medicine Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Yechezkel Barenholz
- Membrane and Liposome Research Lab, IMRIC, Hebrew University Hadassah Medical School, Jerusalem 9112102, Israel
| | - Ninh M La-Beck
- Department of Immunotherapeutics and Biotechnology, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center, Abilene, Texas 79601, United States
| | - Jacob S Brenner
- Departments of Medicine and Pharmacology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Warren C W Chan
- Institute of Biomedical Engineering, University of Toronto, Rosebrugh Building, 164 College Street, Toronto, Ontario M5S 3G9, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, Ontario M5S 3E1, Canada
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Italian Institute of Technology, 16163 Genova, Italy
| | - Agata A Exner
- Departments of Radiology and Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Alberto Gabizon
- The Helmsley Cancer Center, Shaare Zedek Medical Center and The Hebrew University of Jerusalem-Faculty of Medicine, Jerusalem, 9103102, Israel
| | - Biana Godin
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas 77030, United States
- Department of Obstetrics and Gynecology, Houston Methodist Hospital, Houston, Texas 77030, United States
- Department of Obstetrics and Gynecology, Weill Cornell Medicine College (WCMC), New York, New York 10065, United States
- Department of Biomedical Engineering, Texas A&M, College Station, Texas 7784,3 United States
| | - Samuel K Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Center for Biohybrid Medical Systems, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Michael J Mitchell
- Department of Bioengineering, 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
| | - S Moein Moghimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne NE1 7RU, U.K
- Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4HH, U.K
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Center, Aurora, Colorado 80045, United States
| | - Vladimir R Muzykantov
- Department of Systems Pharmacology and Translational Therapeutics, The Perelman School of Medicine, The University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Dan Peer
- Laboratory of Precision Nanomedicine, Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, 69978, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, 69978, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Juliane Nguyen
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina-Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Rachela Popovtzer
- Faculty of Engineering and the Institute of Nanotechnology & Advanced Materials, Bar-Ilan University, 5290002 Ramat Gan, Israel
| | - Madison Ricco
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, the University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Natalie J Serkova
- Department of Radiology, University of Colorado Cancer Center, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Ravi Singh
- Department of Cancer Biology, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27101, United States
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, North Carolina 27101, United States
| | - Avi Schroeder
- Department of Chemical Engineering, Technion, Israel Institute of Technology, Haifa 32000, Israel
| | - Anna A Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48108; Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan 48108, United States
| | - Joelle P Straehla
- Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, Massachusetts 02115 United States
- Koch Institute for Integrative Cancer Research at MIT, Cambridge Massachusetts 02139 United States
| | - Tambet Teesalu
- Laboratory of Precision and Nanomedicine, Institute of Biomedicine and Translational Medicine, University of Tartu, 50411 Tartu, Estonia
| | - Scott Tilden
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, the University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Dmitri Simberg
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences, the University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
16
|
Lin W, Li A, Qiu L, Huang H, Cui P, Wang J. Albumin Nanoparticles Increase the Efficacy of Doxorubicin Hydrochloride Liposome Injection Based on Threshold Theory. Mol Pharm 2024; 21:2970-2980. [PMID: 38742943 DOI: 10.1021/acs.molpharmaceut.4c00097] [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: 05/16/2024]
Abstract
One of the most significant reasons hindering the clinical translation of nanomedicines is the rapid clearance of intravenously injected nanoparticles by the mononuclear phagocyte system, particularly by Kupffer cells in the liver, leading to an inefficient delivery of nanomedicines for tumor treatment. The threshold theory suggests that the liver's capacity to clear nanoparticles is limited, and a single high dose of nanoparticles can reduce the hepatic clearance efficiency, allowing more nanomedicines to reach tumor tissues and enhance therapeutic efficacy. Building upon this theory, researchers have conducted numerous validation studies based on the same nanoparticle carrier systems. These studies involve the use of albumin nanoparticles to improve the therapeutic efficacy of albumin nanomedicines as well as polyethylene glycol (PEG)-modified liposomal nanoparticles to enhance the efficacy of PEGylated liposomal nanomedicines. However, there is no research indicating the feasibility of the threshold theory when blank nanoparticles and nanomedicine belong to different nanoparticle carrier systems currently. In this study, we prepared two different sizes of albumin nanoparticles by using bovine serum albumin. We used the marketed nanomedicine liposomal doxorubicin hydrochloride injection (trade name: LIBOD, manufacturer: Shanghai Fudan-zhangjiang Biopharmaceutical Co., Ltd.), as the representative nanomedicine. Through in vivo experiments, we found that using threshold doses of albumin nanoparticles still can reduce the clearance rate of LIBOD, prolong its time in vivo, increase the area under the plasma concentration-time curve (AUC), and also lead to an increased accumulation of the drug at the tumor site. Furthermore, evaluation of in vivo efficacy and safety further indicates that threshold doses of 100 nm albumin nanoparticles can enhance the antitumor effect of LIBOD without causing harm to the animals. During the study, we found that the particle size of albumin nanoparticles influenced the in vivo distribution of the nanomedicine at the same threshold dose. Compared with 200 nm albumin nanoparticles, 100 nm albumin nanoparticles more effectively reduce the clearance efficiency of LIBOD and enhance nanomedicine accumulation at the tumor site, warranting further investigation. This study utilized albumin nanoparticles to reduce hepatic clearance efficiency and enhance the delivery efficiency of nonalbumin nanocarrier liposomal nanomedicine, providing a new avenue to improve the efficacy and clinical translation of nanomedicines with different carrier systems.
Collapse
Affiliation(s)
- Wei Lin
- School of Petroleum and Chemical Engineering, Changzhou University, Changzhou 213164, P. R. China
- School of Pharmacy, Changzhou University, Changzhou 213164, P. R. China
| | - Anyin Li
- School of Pharmacy, Changzhou University, Changzhou 213164, P. R. China
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou 213164, P. R. China
| | - Hai Huang
- School of Petroleum and Chemical Engineering, Changzhou University, Changzhou 213164, P. R. China
| | - Pengfei Cui
- School of Pharmacy, Changzhou University, Changzhou 213164, P. R. China
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, P. R. China
| |
Collapse
|
17
|
Yuan Z, Yan R, Fu Z, Wu T, Ren C. Impact of physicochemical properties on biological effects of lipid nanoparticles: Are they completely safe. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:172240. [PMID: 38582114 DOI: 10.1016/j.scitotenv.2024.172240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/08/2024]
Abstract
Lipid nanoparticles (LNPs) are promising materials and human-use approved excipients, with manifold applications in biomedicine. Researchers have tended to focus on improving the pharmacological efficiency and organ targeting of LNPs, while paid relatively less attention to the negative aspects created by their specific physicochemical properties. Here, we discuss the impacts of LNPs' physicochemical properties (size, surface hydrophobicity, surface charge, surface modification and lipid composition) on the adsorption-transportation-distribution-clearance processes and bio-nano interactions. In addition, since there is a lack of review emphasizing on toxicological profiles of LNPs, this review outlined immunogenicity, inflammation, hemolytic toxicity, cytotoxicity and genotoxicity induced by LNPs and the underlying mechanisms, with the aim to understand the properties that underlie the biological effects of these materials. This provides a basic strategy that increased efficacy of medical application with minimized side-effects can be achieved by modulating the physicochemical properties of LNPs. Therefore, addressing the effects of physicochemical properties on toxicity induced by LNPs is critical for understanding their environmental and health risks and will help clear the way for LNPs-based drugs to eventually fulfill their promise as a highly effective therapeutic agents for diverse diseases in clinic.
Collapse
Affiliation(s)
- Ziyi Yuan
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Ruyu Yan
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Zuyi Fu
- College of Rehabilitation, Captital Medical University, Beijing, China
| | - Tao Wu
- Beijing Key Laboratory of Enze Biomass Fine Chemicals, College of New Materials and Chemical Engineering, Beijing Institute of Petrochemical Technology, Beijing, China.
| | - Chaoxiu Ren
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China.
| |
Collapse
|
18
|
Khalid MB, Zektser E, Chu E, Li M, Utoh J, Ryan P, Loving HS, Harb R, Kattappuram R, Chatman L, Hartono S, Claudio-Etienne E, Sun G, Feener EP, Li Z, Lai SK, Le Q, Schwartz LB, Lyons JJ, Komarow H, Zhou ZH, Raza H, Pao M, Laky K, Holland SM, Brittain E, Frischmeyer-Guerrerio PA. A randomized double-blinded trial to assess recurrence of systemic allergic reactions following COVID-19 mRNA vaccination. J Allergy Clin Immunol 2024; 153:1634-1646. [PMID: 38460680 PMCID: PMC11162316 DOI: 10.1016/j.jaci.2024.03.001] [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: 11/28/2023] [Revised: 02/09/2024] [Accepted: 03/05/2024] [Indexed: 03/11/2024]
Abstract
BACKGROUND Systemic allergic reactions (sARs) following coronavirus disease 2019 (COVID-19) mRNA vaccines were initially reported at a higher rate than after traditional vaccines. OBJECTIVE We aimed to evaluate the safety of revaccination in these individuals and to interrogate mechanisms underlying these reactions. METHODS In this randomized, double-blinded, phase 2 trial, participants aged 16 to 69 years who previously reported a convincing sAR to their first dose of COVID-19 mRNA vaccine were randomly assigned to receive a second dose of BNT162b2 (Comirnaty) vaccine and placebo on consecutive days in a blinded, 1:1 crossover fashion at the National Institutes of Health. An open-label BNT162b2 booster was offered 5 months later if the second dose did not result in severe sAR. None of the participants received the mRNA-1273 (Spikevax) vaccine during the study. The primary end point was recurrence of sAR following second dose and booster vaccination; exploratory end points included biomarker measurements. RESULTS Of 111 screened participants, 18 were randomly assigned to receive study interventions. Eight received BNT162b2 second dose followed by placebo; 8 received placebo followed by BNT162b2 second dose; 2 withdrew before receiving any study intervention. All 16 participants received the booster dose. Following second dose and booster vaccination, sARs recurred in 2 participants (12.5%; 95% CI, 1.6 to 38.3). No sAR occurred after placebo. An anaphylaxis mimic, immunization stress-related response (ISRR), occurred more commonly than sARs following both vaccine and placebo and was associated with higher predose anxiety scores, paresthesias, and distinct vital sign and biomarker changes. CONCLUSIONS Our findings support revaccination of individuals who report sARs to COVID-19 mRNA vaccines. Distinct clinical and laboratory features may distinguish sARs from ISRRs.
Collapse
Affiliation(s)
- Muhammad B Khalid
- Food Allergy Research Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Ellen Zektser
- Food Allergy Research Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Eric Chu
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, Md
| | - Min Li
- Food Allergy Research Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Joanna Utoh
- Food Allergy Research Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Patrick Ryan
- Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, Md
| | - Hanna S Loving
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Md
| | - Roa Harb
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Md
| | - Robbie Kattappuram
- Investigational Drug Management and Research Section, Clinical Center, National Institutes of Health, Bethesda, Md
| | - Lindsay Chatman
- Food Allergy Research Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Stella Hartono
- Food Allergy Research Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Estefania Claudio-Etienne
- Food Allergy Research Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Guangping Sun
- Food Allergy Research Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | | | - Zhongbo Li
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina Chapel Hill, Chapel Hill, NC
| | - Samuel K Lai
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina Chapel Hill, Chapel Hill, NC
| | - Quang Le
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Immunology, Virginia Commonwealth University, Richmond, Va
| | - Lawrence B Schwartz
- Department of Internal Medicine, Division of Rheumatology, Allergy, and Immunology, Virginia Commonwealth University, Richmond, Va
| | - Jonathan J Lyons
- Translational Allergic Immunopathology Unit, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Hirsh Komarow
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Zhao-Hua Zhou
- Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Md
| | - Haniya Raza
- Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, Md
| | - Maryland Pao
- Office of the Clinical Director, National Institute of Mental Health, National Institutes of Health, Bethesda, Md
| | - Karen Laky
- Food Allergy Research Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Erica Brittain
- Biostatistics Research Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Pamela A Frischmeyer-Guerrerio
- Food Allergy Research Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md.
| |
Collapse
|
19
|
Hameedat F, Mendes BB, Conniot J, Di Filippo LD, Chorilli M, Schroeder A, Conde J, Sousa F. Engineering nanomaterials for glioblastoma nanovaccination. NATURE REVIEWS MATERIALS 2024; 9:628-642. [DOI: 10.1038/s41578-024-00684-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 01/03/2025]
|
20
|
Chu R, Wang Y, Kong J, Pan T, Yang Y, He J. Lipid nanoparticles as the drug carrier for targeted therapy of hepatic disorders. J Mater Chem B 2024; 12:4759-4784. [PMID: 38682294 DOI: 10.1039/d3tb02766j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2024]
Abstract
The liver, a complex and vital organ in the human body, is susceptible to various diseases, including metabolic disorders, acute hepatitis, cirrhosis, and hepatocellular carcinoma. In recent decades, these diseases have significantly contributed to global morbidity and mortality. Currently, liver transplantation remains the most effective treatment for hepatic disorders. Nucleic acid therapeutics offer a selective approach to disease treatment through diverse mechanisms, enabling the regulation of relevant genes and providing a novel therapeutic avenue for hepatic disorders. It is expected that nucleic acid drugs will emerge as the third generation of pharmaceuticals, succeeding small molecule drugs and antibody drugs. Lipid nanoparticles (LNPs) represent a crucial technology in the field of drug delivery and constitute a significant advancement in gene therapies. Nucleic acids encapsulated in LNPs are shielded from the degradation of enzymes and effectively delivered to cells, where they are released and regulate specific genes. This paper provides a comprehensive review of the structure, composition, and applications of LNPs in the treatment of hepatic disorders and offers insights into prospects and challenges in the future development of LNPs.
Collapse
Affiliation(s)
- Runxuan Chu
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| | - Yi Wang
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tung, Hong Kong SAR, P. R. China.
| | - Jianglong Kong
- Department of Chemistry, Hong Kong Baptist University, Kowloon Tung, Hong Kong SAR, P. R. China.
| | - Ting Pan
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
- Department of Pharmaceutics School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Yani Yang
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| | - Jun He
- National Advanced Medical Engineering Research Center, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 201203, P. R. China.
| |
Collapse
|
21
|
Yang K, Bai B, Lei J, Yu X, Qi S, Wang Y, Huang F, Tong Z, Yu G. Biodegradable Lipid-Modified Poly(Guanidine Thioctic Acid)s: A Fortifier of Lipid Nanoparticles to Promote the Efficacy and Safety of mRNA Cancer Vaccines. J Am Chem Soc 2024; 146:11679-11693. [PMID: 38482849 DOI: 10.1021/jacs.3c14010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Lipid nanoparticles (LNPs)-based messenger RNA (mRNA) therapeutics have emerged with promising potentials in the fields of infectious diseases, cancer vaccines, and protein replacement therapies; however, their therapeutic efficacy and safety can still be promoted by the optimization of LNPs formulations. Unfortunately, current LNPs suffer from increased production of reactive oxygen species during translation, which leads to a decreased translation efficiency and the onset of inflammation and other side effects. Herein, we synthesize a lipid-modified poly(guanidine thioctic acid) polymer to fabricate novel LNPs for mRNA vaccines. The acquired G-LNPs significantly promote the translation efficiency of loaded mRNA and attenuate inflammation after vaccination through the elimination of reactive oxygen species that are responsible for translational inhibition and inflammatory responses. In vivo studies demonstrate the excellent antitumor efficacy of the G-LNPs@mRNA vaccine, and two-dose vaccination dramatically increases the population and infiltration of cytotoxic T cells due to the intense antitumor immune responses, thus generating superior antitumor outcomes compared with the mRNA vaccine prepared from traditional LNPs. By synergy with immune checkpoint blockade, the tumor inhibition of G-LNPs@mRNA is further boosted, indicating that G-LNPs-based mRNA vaccines will be powerful and versatile platforms to combat cancer.
Collapse
Affiliation(s)
- Kai Yang
- Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, People's Republic of China
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou 310027, People's Republic of China
| | - Bing Bai
- Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, People's Republic of China
| | - Jiaqi Lei
- Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, People's Republic of China
| | - Xinyang Yu
- Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, People's Republic of China
| | - Shaolong Qi
- Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, People's Republic of China
| | - Yangfan Wang
- Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, People's Republic of China
| | - Feihe Huang
- Stoddart Institute of Molecular Science, Department of Chemistry, Zhejiang University, Hangzhou 310027, People's Republic of China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou 311215, People's Republic of China
| | - Zaizai Tong
- College of Materials Science and Engineering, Zhejiang Sci-Tech University, Hangzhou 310018, People's Republic of China
| | - Guocan Yu
- Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, People's Republic of China
| |
Collapse
|
22
|
Tuttle KL, Lynch DM, Marquis K, Besz KM, Matulonis UA, Castells MC. Phenotypes of hypersensitivity reactions to pegylated liposomal doxorubicin: Safety and efficacy of 128 rapid desensitizations. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:1348-1350.e2. [PMID: 38373654 DOI: 10.1016/j.jaip.2024.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 01/30/2024] [Accepted: 02/07/2024] [Indexed: 02/21/2024]
Affiliation(s)
- Katherine L Tuttle
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Golisano Children's Hospital at Strong Memorial Hospital, University of Rochester, Rochester, NY
| | - Donna-Marie Lynch
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Boston, Mass
| | - Kathleen Marquis
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Pharmacy, Brigham and Women's Hospital, Boston, Mass
| | - Kylie Marie Besz
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Pharmacy, Brigham and Women's Hospital, Boston, Mass
| | - Ursula A Matulonis
- Division of Gynecologic Oncology, Susan F. Smith Center for Women's Cancers, Dana Farber Cancer Institute, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass
| | - Mariana C Castells
- Division of Allergy and Clinical Immunology, Department of Medicine, Brigham and Women's Hospital, Boston, Mass; Department of Medicine, Harvard Medical School, Boston, Mass.
| |
Collapse
|
23
|
Hu M, Li X, You Z, Cai R, Chen C. Physiological Barriers and Strategies of Lipid-Based Nanoparticles for Nucleic Acid Drug Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2303266. [PMID: 37792475 DOI: 10.1002/adma.202303266] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/21/2023] [Indexed: 10/06/2023]
Abstract
Lipid-based nanoparticles (LBNPs) are currently the most promising vehicles for nucleic acid drug (NAD) delivery. Although their clinical applications have achieved success, the NAD delivery efficiency and safety are still unsatisfactory, which are, to a large extent, due to the existence of multi-level physiological barriers in vivo. It is important to elucidate the interactions between these barriers and LBNPs, which will guide more rational design of efficient NAD vehicles with low adverse effects and facilitate broader applications of nucleic acid therapeutics. This review describes the obstacles and challenges of biological barriers to NAD delivery at systemic, organ, sub-organ, cellular, and subcellular levels. The strategies to overcome these barriers are comprehensively reviewed, mainly including physically/chemically engineering LBNPs and directly modifying physiological barriers by auxiliary treatments. Then the potentials and challenges for successful translation of these preclinical studies into the clinic are discussed. In the end, a forward look at the strategies on manipulating protein corona (PC) is addressed, which may pull off the trick of overcoming those physiological barriers and significantly improve the efficacy and safety of LBNP-based NADs delivery.
Collapse
Affiliation(s)
- Mingdi Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
- Sino-Danish Center for Education and Research, Beijing, 100049, China
| | - Xiaoyan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Zhen You
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Rong Cai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, 100190, China
- Sino-Danish College, University of Chinese Academy of Sciences, Beijing, 100049, China
- Sino-Danish Center for Education and Research, Beijing, 100049, China
- The GBA National Institute for Nanotechnology Innovation, Guangzhou, 510700, China
| |
Collapse
|
24
|
Forgham H, Zhu J, Zhang T, Huang X, Li X, Shen A, Biggs H, Talbo G, Xu C, Davis TP, Qiao R. Fluorine-modified polymers reduce the adsorption of immune-reactive proteins to PEGylated gold nanoparticles. Nanomedicine (Lond) 2024; 19:995-1012. [PMID: 38593053 PMCID: PMC11221377 DOI: 10.2217/nnm-2023-0357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/23/2024] [Indexed: 04/11/2024] Open
Abstract
Aim: To investigate the influence of fluorine in reducing the adsorption of immune-reactive proteins onto PEGylated gold nanoparticles. Methods: Reversible addition fragmentation chain transfer polymerization, the Turkevich method and ligand exchange were used to prepare polymer-coated gold nanoparticles. Subsequent in vitro physicochemical and biological characterizations and proteomic analysis were performed. Results: Fluorine-modified polymers reduced the adsorption of complement and other immune-reactive proteins while potentially improving circulatory times and modulating liver toxicity by reducing apolipoprotein E adsorption. Fluorine actively discouraged phagocytosis while encouraging the adsorption of therapeutic targets, CD209 and signaling molecule calreticulin. Conclusion: This study suggests that the addition of fluorine in the surface coating of nanoparticles could lead to improved performance in nanomedicine designed for the intravenous delivery of cargos.
Collapse
Affiliation(s)
- Helen Forgham
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Jiayuan Zhu
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Taoran Zhang
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Xumin Huang
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Xiangke Li
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Ao Shen
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Heather Biggs
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Gert Talbo
- Metabolomics Australia (Queensland Node), The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Chun Xu
- School of Dentistry, The University of Queensland, Herston, Queensland, 4006, Australia
| | - Thomas P Davis
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Ruirui Qiao
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland, Brisbane, Queensland, 4072, Australia
| |
Collapse
|
25
|
Misra B, Hughes KA, Pentz WH, Samart P, Geldenhuys WJ, Bobbala S. Flash nanoprecipitation assisted self-assembly of ionizable lipid nanoparticles for nucleic acid delivery. NANOSCALE 2024; 16:6939-6948. [PMID: 38511623 PMCID: PMC11040450 DOI: 10.1039/d4nr00278d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Abstract
Ionizable lipid nanoparticles (LNPs) have emerged as a powerful tool for the intracellular delivery of nucleic acids. Following the recent success of LNP-based siRNA therapeutics and mRNA vaccines, the use of ionizable lipids for nucleic acid delivery has tremendously increased. Here, we introduce a flash nanoprecipitation (FNP) approach using the confined impingement (CIJ) mixer to stably self-assemble ionizable LNPs. To validate this approach, we employed three clinically relevant LNP formulations containing SM102, ALC0315, and DLin-MC3-DMA as ionizable lipids. FNP-assembled LNPs showed >95% encapsulation efficiency of mRNA and siRNA payloads and particle sizes below 150 nm. SM102 or ALC0315 LNPs demonstrated efficient delivery of mRNA into immune cells in vitro and to lymphoid organs in vivo, whereas Dlin-MC3-DMA LNPs allowed effective intracellular siRNA delivery with great functional ability. The FNP technique could economically produce LNPs in smaller volumes that are highly suitable for the discovery phase.
Collapse
Affiliation(s)
- Bishal Misra
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA.
| | - Krystal A Hughes
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA.
| | - William H Pentz
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA.
- School of Medicine, West Virginia University, Morgantown, WV 26506, USA
| | - Parinya Samart
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA.
| | - Werner J Geldenhuys
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA.
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
26
|
Bitounis D, Jacquinet E, Rogers MA, Amiji MM. Strategies to reduce the risks of mRNA drug and vaccine toxicity. Nat Rev Drug Discov 2024; 23:281-300. [PMID: 38263456 DOI: 10.1038/s41573-023-00859-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/30/2023] [Indexed: 01/25/2024]
Abstract
mRNA formulated with lipid nanoparticles is a transformative technology that has enabled the rapid development and administration of billions of coronavirus disease 2019 (COVID-19) vaccine doses worldwide. However, avoiding unacceptable toxicity with mRNA drugs and vaccines presents challenges. Lipid nanoparticle structural components, production methods, route of administration and proteins produced from complexed mRNAs all present toxicity concerns. Here, we discuss these concerns, specifically how cell tropism and tissue distribution of mRNA and lipid nanoparticles can lead to toxicity, and their possible reactogenicity. We focus on adverse events from mRNA applications for protein replacement and gene editing therapies as well as vaccines, tracing common biochemical and cellular pathways. The potential and limitations of existing models and tools used to screen for on-target efficacy and de-risk off-target toxicity, including in vivo and next-generation in vitro models, are also discussed.
Collapse
Affiliation(s)
- Dimitrios Bitounis
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA, USA
- Moderna, Inc., Cambridge, MA, USA
| | | | | | - Mansoor M Amiji
- Departments of Pharmaceutical Sciences and Chemical Engineering, Northeastern University, Boston, MA, USA.
| |
Collapse
|
27
|
Llovet JM, Pinyol R, Yarchoan M, Singal AG, Marron TU, Schwartz M, Pikarsky E, Kudo M, Finn RS. Adjuvant and neoadjuvant immunotherapies in hepatocellular carcinoma. Nat Rev Clin Oncol 2024; 21:294-311. [PMID: 38424197 DOI: 10.1038/s41571-024-00868-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2024] [Indexed: 03/02/2024]
Abstract
Liver cancer, specifically hepatocellular carcinoma (HCC), is the sixth most common cancer and the third leading cause of cancer mortality worldwide. The development of effective systemic therapies, particularly those involving immune-checkpoint inhibitors (ICIs), has substantially improved the outcomes of patients with advanced-stage HCC. Approximately 30% of patients are diagnosed with early stage disease and currently receive potentially curative therapies, such as resection, liver transplantation or local ablation, which result in median overall survival durations beyond 60 months. Nonetheless, up to 70% of these patients will have disease recurrence within 5 years of resection or local ablation. To date, the results of randomized clinical trials testing adjuvant therapy in patients with HCC have been negative. This major unmet need has been addressed with the IMbrave 050 trial, demonstrating a recurrence-free survival benefit in patients with a high risk of relapse after resection or local ablation who received adjuvant atezolizumab plus bevacizumab. In parallel, studies testing neoadjuvant ICIs alone or in combination in patients with early stage disease have also reported efficacy. In this Review, we provide a comprehensive overview of the current approaches to manage patients with early stage HCC. We also describe the tumour immune microenvironment and the mechanisms of action of ICIs and cancer vaccines in this setting. Finally, we summarize the available evidence from phase II/III trials of neoadjuvant and adjuvant approaches and discuss emerging clinical trials, identification of biomarkers and clinical trial design considerations for future studies.
Collapse
Affiliation(s)
- Josep M Llovet
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain.
- Mount Sinai Liver Cancer Program, Divisions of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain.
| | - Roser Pinyol
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Mark Yarchoan
- Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amit G Singal
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Thomas U Marron
- Mount Sinai Liver Cancer Program, Divisions of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Myron Schwartz
- Department of Liver Surgery, Recanati/Miller Transplantation Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eli Pikarsky
- The Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Richard S Finn
- Department of Medicine, Division of Hematology/Oncology, Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
28
|
Bakos T, Mészáros T, Kozma GT, Berényi P, Facskó R, Farkas H, Dézsi L, Heirman C, de Koker S, Schiffelers R, Glatter KA, Radovits T, Szénási G, Szebeni J. mRNA-LNP COVID-19 Vaccine Lipids Induce Complement Activation and Production of Proinflammatory Cytokines: Mechanisms, Effects of Complement Inhibitors, and Relevance to Adverse Reactions. Int J Mol Sci 2024; 25:3595. [PMID: 38612407 PMCID: PMC11012056 DOI: 10.3390/ijms25073595] [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: 02/08/2024] [Revised: 03/16/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
A small fraction of people vaccinated with mRNA-lipid nanoparticle (mRNA-LNP)-based COVID-19 vaccines display acute or subacute inflammatory symptoms whose mechanism has not been clarified to date. To better understand the molecular mechanism of these adverse events (AEs), here, we analyzed in vitro the vaccine-induced induction and interrelations of the following two major inflammatory processes: complement (C) activation and release of proinflammatory cytokines. Incubation of Pfizer-BioNTech's Comirnaty and Moderna's Spikevax with 75% human serum led to significant increases in C5a, sC5b-9, and Bb but not C4d, indicating C activation mainly via the alternative pathway. Control PEGylated liposomes (Doxebo) also induced C activation, but, on a weight basis, it was ~5 times less effective than that of Comirnaty. Viral or synthetic naked mRNAs had no C-activating effects. In peripheral blood mononuclear cell (PBMC) cultures supplemented with 20% autologous serum, besides C activation, Comirnaty induced the secretion of proinflammatory cytokines in the following order: IL-1α < IFN-γ < IL-1β < TNF-α < IL-6 < IL-8. Heat-inactivation of C in serum prevented a rise in IL-1α, IL-1β, and TNF-α, suggesting C-dependence of these cytokines' induction, although the C5 blocker Soliris and C1 inhibitor Berinert, which effectively inhibited C activation in both systems, did not suppress the release of any cytokines. These findings suggest that the inflammatory AEs of mRNA-LNP vaccines are due, at least in part, to stimulation of both arms of the innate immune system, whereupon C activation may be causally involved in the induction of some, but not all, inflammatory cytokines. Thus, the pharmacological attenuation of inflammatory AEs may not be achieved via monotherapy with the tested C inhibitors; efficacy may require combination therapy with different C inhibitors and/or other anti-inflammatory agents.
Collapse
Affiliation(s)
- Tamás Bakos
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (T.B.); (T.M.); (G.T.K.); (P.B.); (R.F.); (L.D.); (G.S.)
| | - Tamás Mészáros
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (T.B.); (T.M.); (G.T.K.); (P.B.); (R.F.); (L.D.); (G.S.)
- SeroScience LCC., 1089 Budapest, Hungary
- Department of Cardiology, Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary;
- Department of Surgical Research and Techniques, Heart and Vascular Center, Semmelweis University, 1089 Budapest, Hungary
| | - Gergely Tibor Kozma
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (T.B.); (T.M.); (G.T.K.); (P.B.); (R.F.); (L.D.); (G.S.)
- SeroScience LCC., 1089 Budapest, Hungary
| | - Petra Berényi
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (T.B.); (T.M.); (G.T.K.); (P.B.); (R.F.); (L.D.); (G.S.)
- SeroScience LCC., 1089 Budapest, Hungary
| | - Réka Facskó
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (T.B.); (T.M.); (G.T.K.); (P.B.); (R.F.); (L.D.); (G.S.)
- SeroScience LCC., 1089 Budapest, Hungary
- Department of Cardiology, Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary;
- Department of Surgical Research and Techniques, Heart and Vascular Center, Semmelweis University, 1089 Budapest, Hungary
| | - Henriette Farkas
- Hungarian Center of Reference and Excellence, Department of Internal Medicine and Hematology, Semmelweis University, 1088 Budapest, Hungary;
| | - László Dézsi
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (T.B.); (T.M.); (G.T.K.); (P.B.); (R.F.); (L.D.); (G.S.)
| | - Carlo Heirman
- Etherna Biopharmaceuticals, 2845 Niel, Belgium; (C.H.); (S.d.K.)
| | - Stefaan de Koker
- Etherna Biopharmaceuticals, 2845 Niel, Belgium; (C.H.); (S.d.K.)
| | - Raymond Schiffelers
- Division of Laboratories and Pharmacy, University Medical Center, 3584 CX Utrecht, The Netherlands;
| | | | - Tamás Radovits
- Department of Cardiology, Heart and Vascular Center, Semmelweis University, 1122 Budapest, Hungary;
- Department of Surgical Research and Techniques, Heart and Vascular Center, Semmelweis University, 1089 Budapest, Hungary
| | - Gábor Szénási
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (T.B.); (T.M.); (G.T.K.); (P.B.); (R.F.); (L.D.); (G.S.)
| | - János Szebeni
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, 1085 Budapest, Hungary; (T.B.); (T.M.); (G.T.K.); (P.B.); (R.F.); (L.D.); (G.S.)
- SeroScience LCC., 1089 Budapest, Hungary
- Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health Sciences, Miskolc University, 3530 Miskolc, Hungary
- Translational Nanobioscience Research Center, Sungkyunkwan University, Suwon 06351, Republic of Korea
| |
Collapse
|
29
|
Estapé Senti M, García Del Valle L, Schiffelers RM. mRNA delivery systems for cancer immunotherapy: Lipid nanoparticles and beyond. Adv Drug Deliv Rev 2024; 206:115190. [PMID: 38307296 DOI: 10.1016/j.addr.2024.115190] [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: 10/31/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 02/04/2024]
Abstract
mRNA-based vaccines are emerging as a promising alternative to standard cancer treatments and the conventional vaccines. Moreover, the FDA-approval of three nucleic acid based therapeutics (Onpattro, BNT162b2 and mRNA-1273) has further increased the interest and trust on this type of therapeutics. In order to achieve a significant therapeutic efficacy, the mRNA needs from a drug delivery system. In the last years, several delivery platforms have been explored, being the lipid nanoparticles (LNPs) the most well characterized and studied. A better understanding on how mRNA-based therapeutics operate (both the mRNA itself and the drug delivery system) will help to further improve their efficacy and safety. In this review, we will provide an overview of what mRNA cancer vaccines are and their mode of action and we will highlight the advantages and challenges of the different delivery platforms that are under investigation.
Collapse
Affiliation(s)
- Mariona Estapé Senti
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Lucía García Del Valle
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands
| | - Raymond M Schiffelers
- CDL Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX, Utrecht, the Netherlands.
| |
Collapse
|
30
|
Wu L, Li X, Qian X, Wang S, Liu J, Yan J. Lipid Nanoparticle (LNP) Delivery Carrier-Assisted Targeted Controlled Release mRNA Vaccines in Tumor Immunity. Vaccines (Basel) 2024; 12:186. [PMID: 38400169 PMCID: PMC10891594 DOI: 10.3390/vaccines12020186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
In recent years, lipid nanoparticles (LNPs) have attracted extensive attention in tumor immunotherapy. Targeting immune cells in cancer therapy has become a strategy of great research interest. mRNA vaccines are a potential choice for tumor immunotherapy, due to their ability to directly encode antigen proteins and stimulate a strong immune response. However, the mode of delivery and lack of stability of mRNA are key issues limiting its application. LNPs are an excellent mRNA delivery carrier, and their structural stability and biocompatibility make them an effective means for delivering mRNA to specific targets. This study summarizes the research progress in LNP delivery carrier-assisted targeted controlled release mRNA vaccines in tumor immunity. The role of LNPs in improving mRNA stability, immunogenicity, and targeting is discussed. This review aims to systematically summarize the latest research progress in LNP delivery carrier-assisted targeted controlled release mRNA vaccines in tumor immunity to provide new ideas and strategies for tumor immunotherapy, as well as to provide more effective treatment plans for patients.
Collapse
Affiliation(s)
- Liusheng Wu
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China; (L.W.); (X.Q.); (S.W.)
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Xiaoqiang Li
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China;
| | - Xinye Qian
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China; (L.W.); (X.Q.); (S.W.)
| | - Shuang Wang
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China; (L.W.); (X.Q.); (S.W.)
| | - Jixian Liu
- Department of Thoracic Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China;
| | - Jun Yan
- Center of Hepatobiliary Pancreatic Disease, Beijing Tsinghua Changgung Hospital, School of Medicine, Tsinghua University, Beijing 100084, China; (L.W.); (X.Q.); (S.W.)
| |
Collapse
|
31
|
Igyártó BZ, Qin Z. The mRNA-LNP vaccines - the good, the bad and the ugly? Front Immunol 2024; 15:1336906. [PMID: 38390323 PMCID: PMC10883065 DOI: 10.3389/fimmu.2024.1336906] [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: 11/11/2023] [Accepted: 01/23/2024] [Indexed: 02/24/2024] Open
Abstract
The mRNA-LNP vaccine has received much attention during the COVID-19 pandemic since it served as the basis of the most widely used SARS-CoV-2 vaccines in Western countries. Based on early clinical trial data, these vaccines were deemed safe and effective for all demographics. However, the latest data raise serious concerns about the safety and effectiveness of these vaccines. Here, we review some of the safety and efficacy concerns identified to date. We also discuss the potential mechanism of observed adverse events related to the use of these vaccines and whether they can be mitigated by alterations of this vaccine mechanism approach.
Collapse
Affiliation(s)
- Botond Z. Igyártó
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, United States
| | | |
Collapse
|
32
|
Shi Q, Zhang Z, Liu S. Precision Sequence-Defined Polymers: From Sequencing to Biological Functions. Angew Chem Int Ed Engl 2024; 63:e202313370. [PMID: 37875462 DOI: 10.1002/anie.202313370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 10/26/2023]
Abstract
Precise sequence-defined polymers (SDPs) with uniform chain-to-chain structure including chain length, unit sequence, and end functionalities represent the pinnacle of sophistication in the realm of polymer science. For example, the absolute control over the unit sequence of SDPs allows for the bottom-up design of polymers with hierarchical microstructures and functions. Accompanied with the development of synthetic techniques towards precision SDPs, the decoding of SDP sequences and construction of advanced functions irreplaceable by other synthetic materials is of central importance. In this Minireview, we focus on recent advances in SDP sequencing techniques including tandem mass spectrometry (MS), chemically assisted primary MS, as well as other non-destructive sequencing methods such as nuclear magnetic resonance (NMR) spectroscopy, circular dichroism (CD), and nanopore sequencing. Additionally, we delve into the promising prospects of SDP functions in the area of cutting-edge biological research. Topics of exploration include gene delivery systems, the development of hybrid materials combining SDPs and nucleic acids, protein recognition and regulation, as well as the interplay between chirality and biological functions. A brief outlook towards the future directions of SDPs is also presented.
Collapse
Affiliation(s)
- Qiangqiang Shi
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, China
| | - Zhengbiao Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China
| | - Shiyong Liu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and Key Laboratory of Precision and Intelligent Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, China
| |
Collapse
|
33
|
Sharma P, Hoorn D, Aitha A, Breier D, Peer D. The immunostimulatory nature of mRNA lipid nanoparticles. Adv Drug Deliv Rev 2024; 205:115175. [PMID: 38218350 DOI: 10.1016/j.addr.2023.115175] [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: 10/25/2023] [Revised: 12/25/2023] [Accepted: 12/28/2023] [Indexed: 01/15/2024]
Abstract
mRNA-Lipid nanoparticles (LNPs) are at the forefront of global medical research. With the development of mRNA-LNP vaccines to combat the COVID-19 pandemic, the clinical potential of this platform was unleashed. Upon administering 16 billion doses that protected billions of people, it became clear that a fraction of them witnessed mild and in some cases even severe adverse effects. Therefore, it is paramount to define the safety along with the therapeutic efficacy of the mRNA-LNP platform for the successful translation of new genetic medicines based on this technology. While mRNA was the effector molecule of this platform, the ionizable lipid component of the LNPs played an indispensable role in its success. However, both of these components possess the ability to induce undesired immunostimulation, which is an area that needs to be addressed systematically. The immune cell agitation caused by this platform is a two-edged sword as it may prove beneficial for vaccination but detrimental to other applications. Therefore, a key challenge in advancing the mRNA-LNP drug delivery platform from bench to bedside is understanding the immunostimulatory behavior of these components. Herein, we provide a detailed overview of the structural modifications and immunogenicity of synthetic mRNA. We discuss the effect of ionizable lipid structure on LNP functionality and offer a mechanistic overview of the ability of LNPs to elicit an immune response. Finally, we shed some light on the current status of this technology in clinical trials and discuss a few challenges to be addressed to advance the field.
Collapse
Affiliation(s)
- Preeti Sharma
- Laboratory of Precision Nanomedicine, 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; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Daniek Hoorn
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Anjaiah Aitha
- Laboratory of Precision Nanomedicine, 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; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dor Breier
- Laboratory of Precision Nanomedicine, 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; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Dan Peer
- Laboratory of Precision Nanomedicine, 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; Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
34
|
Rajora MA, Dhaliwal A, Zheng M, Choi V, Overchuk M, Lou JWH, Pellow C, Goertz D, Chen J, Zheng G. Quantitative Pharmacokinetics Reveal Impact of Lipid Composition on Microbubble and Nanoprogeny Shell Fate. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304453. [PMID: 38032129 PMCID: PMC10811482 DOI: 10.1002/advs.202304453] [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: 07/03/2023] [Revised: 10/19/2023] [Indexed: 12/01/2023]
Abstract
Microbubble-enabled focused ultrasound (MB-FUS) has revolutionized nano and molecular drug delivery capabilities. Yet, the absence of longitudinal, systematic, quantitative studies of microbubble shell pharmacokinetics hinders progress within the MB-FUS field. Microbubble radiolabeling challenges contribute to this void. This barrier is overcome by developing a one-pot, purification-free copper chelation protocol able to stably radiolabel diverse porphyrin-lipid-containing Definity® analogues (pDefs) with >95% efficiency while maintaining microbubble physicochemical properties. Five tri-modal (ultrasound-, positron emission tomography (PET)-, and fluorescent-active) [64 Cu]Cu-pDefs are created with varying lipid acyl chain length and charge, representing the most prevalently studied microbubble compositions. In vitro, C16 chain length microbubbles yield 2-3x smaller nanoprogeny than C18 microbubbles post FUS. In vivo, [64 Cu]Cu-pDefs are tracked in healthy and 4T1 tumor-bearing mice ± FUS over 48 h qualitatively through fluorescence imaging (to characterize particle disruption) and quantitatively through PET and γ-counting. These studies reveal the impact of microbubble composition and FUS on microbubble dissolution rates, shell circulation, off-target tissue retention (predominantly the liver and spleen), and FUS enhancement of tumor delivery. These findings yield pharmacokinetic microbubble structure-activity relationships that disrupt conventional knowledge, the implications of which on MB-FUS platform design, safety, and nanomedicine delivery are discussed.
Collapse
Affiliation(s)
- Maneesha A. Rajora
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Alexander Dhaliwal
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Mark Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Victor Choi
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Marta Overchuk
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Joint Department of Biomedical EngineeringUniversity of North Carolina at Chapel Hill and North Carolina State UniversityChapel HillNC27599USA
| | - Jenny W. H. Lou
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| | - Carly Pellow
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - David Goertz
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Sunnybrook Research InstituteTorontoOntarioM4N 3M5Canada
| | - Juan Chen
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
| | - Gang Zheng
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoOntarioM5G 1L7Canada
- Institute of Biomedical EngineeringUniversity of TorontoTorontoOntarioM5G 1L7Canada
- Department of Medical BiophysicsUniversity of TorontoTorontoOntarioM5G 1L7Canada
| |
Collapse
|
35
|
Neun BW, Dobrovolskaia MA. Detection of Pre-Existing Antibodies to Polyethylene Glycol and PEGylated Liposomes in Human Serum. Methods Mol Biol 2024; 2789:185-192. [PMID: 38507004 DOI: 10.1007/978-1-0716-3786-9_19] [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: 03/22/2024]
Abstract
Polyethylene glycol, or PEG, is common in consumer products, over-the-counter medications, food, and pharmaceutical products. Concerns about PEG immunogenicity and the subsequent negative impact of pre-existing and product-induced antibodies often shadow the benefits of using PEG in nanotechnology-based products. Such anti-PEG antibodies contribute to the accelerated blood clearance of PEGylated nanomedicines and result in premature drug release and antibody-mediated toxicities. Recent data demonstrated that using PEG in COVID-19 lipid nanoparticle-mRNA vaccines is associated with an induction of anti-PEG antibodies in healthy individuals, further contributing to the development or boosting of pre-existing antibodies and increasing the risks of antibody-mediated toxicities to other products containing PEG. Therefore, monitoring the levels of pre-existing and product-induced anti-PEG antibodies provides mechanistic insights for pharmacology, toxicology, and immunological studies of PEGylated drug products.
Collapse
Affiliation(s)
- Barry W Neun
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| |
Collapse
|
36
|
Taharabaru T, Kihara T, Obata A, Onodera R, Wen Y, Li J, Motoyama K, Higashi T. Cyclodextrin-based tailored polyrotaxanes for highly efficient delivery of the genome-editing molecule. Carbohydr Polym 2024; 323:121443. [PMID: 37940259 DOI: 10.1016/j.carbpol.2023.121443] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/10/2023] [Accepted: 09/27/2023] [Indexed: 11/10/2023]
Abstract
Direct cytosolic delivery of the Cas9 ribonucleoprotein is the most promising method for inducing CRISPR-Cas9 genome editing in mammalian cells. Recently, we focused the movable properties of cyclodextrin-based polyrotaxanes (PRXs), which consist of numerous cyclodextrins threaded onto the axile molecule with bulky endcaps at both ends of the axile molecule, and developed aminated PRXs as multistep transformable carriers for Cas9 ribonucleoprotein, ensuring efficient complexation, cellular internalization, endosomal escape, release, and nuclear localization. This study reports the structural fine-tuning and structure-property relationship of multistep transformable PRXs for more efficient Cas9 ribonucleoprotein delivery. Among various PRXs, PRX derivatives with a longer molecular length (35 kDa polyethylene glycol as the axile molecule) and a low total degree of substitution (1.5 amino groups/α-cyclodextrins), as well as the modified ratio of two modified amines (cystamine and diethylenetriamine) = ≈1:1, exhibited the highest genome-editing efficacy and intracellular dynamics control. These structural properties are important for efficient endosomal escape and Cas9 RNP release. Furthermore, ligand-modified-β-CD, which can endow the ligand through complexation with PRX termini, improved the cellular uptake and genome-editing effects of the optimized PRX/Cas9 RNP in target cells. Thus, structural fine-tuning and the addition of ligand-modified-β-cyclodextrin enabled efficient genome editing by the Cas9 RNP.
Collapse
Affiliation(s)
- Toru Taharabaru
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Takuya Kihara
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Airi Obata
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Risako Onodera
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Yuting Wen
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Jun Li
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore 119276, Singapore
| | - Keiichi Motoyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Taishi Higashi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Priority Organization for Innovation and Excellence, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto 860-8555, Japan.
| |
Collapse
|
37
|
Ilinskaya A, Shah A, Van Dusen A, Dobrovolskaia MA. Detection of Intracellular Complement Activation by Nanoparticles in Human T Lymphocytes. Methods Mol Biol 2024; 2789:109-120. [PMID: 38506996 DOI: 10.1007/978-1-0716-3786-9_11] [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: 03/22/2024]
Abstract
The complement system is complex and includes two main components: the systemic or plasma complement and the so-called intracellular complement or complosome. The complement proteins expressed by the liver and secreted into blood plasma compose the plasma complement system, whereas complement proteins expressed by and functioning inside the cell represent the intracellular complement. The complement system plays an essential role in host defense; however, complement activation may lead to pathologies when uncontrolled. When such undesirable activation of the plasma complement occurs in response to a drug product, it leads to immediate-type hypersensitivity reactions independent of immunoglobulin E. These reactions are often called complement activation-related pseudoallergy (CARPA). In addition to the blood plasma, the complement protein C3 is found in many cells, including lymphocytes, monocytes, endothelial, and even cancer cells. The activation of the intracellular complement generates split products, which are exported from the cell onto the membrane. Since the activation of the intracellular complement in T lymphocytes was found to correlate with autoimmune disorders, and growing evidence is available for the involvement of T lymphocytes in the development of drug-induced hypersensitivity reactions, understanding the ability of nanomaterials to activate intracellular complement may aid in establishing a long-term safety profile for these materials. This chapter describes a flow cytometry-based protocol for detecting intracellular complement activation by engineered nanomaterials.
Collapse
Affiliation(s)
- Anna Ilinskaya
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Ankit Shah
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Marina A Dobrovolskaia
- Nanotechnology Characterization Laboratory, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA.
| |
Collapse
|
38
|
Wang WD, Guo YY, Yang ZL, Su GL, Sun ZJ. Sniping Cancer Stem Cells with Nanomaterials. ACS NANO 2023; 17:23262-23298. [PMID: 38010076 DOI: 10.1021/acsnano.3c07828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Cancer stem cells (CSCs) drive tumor initiation, progression, and therapeutic resistance due to their self-renewal and differentiation capabilities. Despite encouraging progress in cancer treatment, conventional approaches often fail to eliminate CSCs, necessitating the development of precise targeted strategies. Recent advances in materials science and nanotechnology have enabled promising CSC-targeted approaches, harnessing the power of tailoring nanomaterials in diverse therapeutic applications. This review provides an update on the current landscape of nanobased precision targeting approaches against CSCs. We elucidate the nuanced application of organic, inorganic, and bioinspired nanomaterials across a spectrum of therapeutic paradigms, encompassing targeted therapy, immunotherapy, and multimodal synergistic therapies. By examining the accomplishments and challenges in this potential field, we aim to inform future efforts to advance nanomaterial-based therapies toward more effective "sniping" of CSCs and tumor clearance.
Collapse
Affiliation(s)
- Wen-Da Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Yan-Yu Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Zhong-Lu Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Guang-Liang Su
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, China
- Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
39
|
Ma Y, Sun L, Zhang J, Chiang C, Pan J, Wang X, Kwak KJ, Li H, Zhao R, Rima XY, Zhang C, Zhang A, Liu Y, He Z, Hansford D, Reategui E, Liu C, Lee AS, Yuan Y, Lee LJ. Exosomal mRNAs for Angiogenic-Osteogenic Coupled Bone Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302622. [PMID: 37847907 PMCID: PMC10667797 DOI: 10.1002/advs.202302622] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 08/25/2023] [Indexed: 10/19/2023]
Abstract
Regenerative medicine in tissue engineering often relies on stem cells and specific growth factors at a supraphysiological dose. These approaches are costly and may cause severe side effects. Herein, therapeutic small extracellular vesicles (t-sEVs) endogenously loaded with a cocktail of human vascular endothelial growth factor A (VEGF-A) and human bone morphogenetic protein 2 (BMP-2) mRNAs within a customized injectable PEGylated poly (glycerol sebacate) acrylate (PEGS-A) hydrogel for bone regeneration in rats with challenging femur critical-size defects are introduced. Abundant t-sEVs are produced by a facile cellular nanoelectroporation system based on a commercially available track-etched membrane (TM-nanoEP) to deliver plasmid DNAs to human adipose-derived mesenchymal stem cells (hAdMSCs). Upregulated microRNAs associated with the therapeutic mRNAs are enriched in t-sEVs for enhanced angiogenic-osteogenic regeneration. Localized and controlled release of t-sEVs within the PEGS-A hydrogel leads to the retention of therapeutics in the defect site for highly efficient bone regeneration with minimal low accumulation in other organs.
Collapse
Affiliation(s)
- Yifan Ma
- Department of Biomedical EngineeringThe Ohio State UniversityColumbusOH43210USA
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOH43210USA
| | - Lili Sun
- Key Laboratory for Ultrafine Materials of Ministry of Education and Frontiers Science Center for Materiobiology and Dynamic ChemistryEast China University of Science and Technology200237ShanghaiP. R. China
| | - Jingjing Zhang
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOH43210USA
| | - Chi‐ling Chiang
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOH43210USA
| | - Junjie Pan
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOH43210USA
| | - Xinyu Wang
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOH43210USA
| | | | - Hong Li
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOH43210USA
| | - Renliang Zhao
- Department of Orthopedic Surgery and Shanghai Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's Hospital200233ShanghaiChina
| | - Xilal Y. Rima
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOH43210USA
| | - Chi Zhang
- College of PharmacyThe Ohio State UniversityColumbusOH43210USA
| | - Anan Zhang
- Key Laboratory for Ultrafine Materials of Ministry of Education and Frontiers Science Center for Materiobiology and Dynamic ChemistryEast China University of Science and Technology200237ShanghaiP. R. China
| | - Yutong Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education and Frontiers Science Center for Materiobiology and Dynamic ChemistryEast China University of Science and Technology200237ShanghaiP. R. China
| | - Zirui He
- Key Laboratory for Ultrafine Materials of Ministry of Education and Frontiers Science Center for Materiobiology and Dynamic ChemistryEast China University of Science and Technology200237ShanghaiP. R. China
| | - Derek Hansford
- Department of Biomedical EngineeringThe Ohio State UniversityColumbusOH43210USA
| | - Eduardo Reategui
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOH43210USA
| | - Changsheng Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education and Frontiers Science Center for Materiobiology and Dynamic ChemistryEast China University of Science and Technology200237ShanghaiP. R. China
| | - Andrew S. Lee
- School of Chemical Biology and BiotechnologyPeking University Shenzhen Graduate School518055ShenzhenChina
- Institute for Cancer ResearchShenzhen Bay Laboratory518055ShenzhenChina
| | - Yuan Yuan
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOH43210USA
- Key Laboratory for Ultrafine Materials of Ministry of Education and Frontiers Science Center for Materiobiology and Dynamic ChemistryEast China University of Science and Technology200237ShanghaiP. R. China
| | - Ly James Lee
- Department of Biomedical EngineeringThe Ohio State UniversityColumbusOH43210USA
- William G. Lowrie Department of Chemical and Biomolecular EngineeringThe Ohio State UniversityColumbusOH43210USA
| |
Collapse
|
40
|
Chase LS, Zaleski MH, Morrell LJ, Brenner JS. Automated measurement of distance-walked as a "sixth vital sign" for detecting infusion reactions during preclinical testing. Int J Pharm 2023; 645:123369. [PMID: 37696344 PMCID: PMC10718583 DOI: 10.1016/j.ijpharm.2023.123369] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/13/2023]
Abstract
Infusion reactions are a major risk for advanced therapeutics (e.g., engineered proteins nanoparticles, etc.), which can trigger the complement cascade, anaphylaxis, and other life-threatening immune responses. However, during the early phases of development, it is uncommon to assess for infusion reactions, given the labor involved in measuring multiple physiological parameters in rodents. Therefore, we sought to develop an automated quantification of rodent locomotion to serve as a sensitive screening tool for infusion reactions, with minimal added labor-time for each experiment. Here we present the detailed methods for building a motion tracking cage for mice, requiring ∼$100 of materials, ∼2 h to build and set up completely, and employing freely available software (DeepLabCut). The distance-walked after injection was first shown to have the predicted effects for stimulants (caffeine), sedatives (ketamine), and toxins (lipopolysaccharide). Additionally, the distance-walked more sensitively detected the effects of these compounds than did pulse oximetry-based measurements of the classical vital signs of heart rate, respiratory rate, and blood oxygen saturation. Finally, we examined a nanomedicine formulation that has been in preclinical development, liposomes targeted to the cell adhesion molecule ICAM. While this formulation has been studied across dozens of publications, it has not previously been noted to produce an infusion reaction. However, the automated motion tracking cage showed that ICAM-liposomes markedly reduce the distance-walked, which we confirmed by measuring the other vital signs. Importantly, the motion tracking cage added < 5 min of labor time per 5-mouse condition, while pulse oximetry with a neck cuff (by far the most stable oximetry signal in mice) required ∼ 100 min of labor time. Thus, automated measurement of distance-walked can indeed serve as a "sixth vital sign" for detecting infusion reactions during preclinical testing. Additionally, the device to measure distance-walked is easy and cheap to build and requires negligible labor time for each experiment, enabling distance-walked to be recorded in nearly every infusion experiment.
Collapse
Affiliation(s)
- Liam S Chase
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| | - Michael H Zaleski
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lianne J Morrell
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob S Brenner
- Department of Pharmacology, The Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
41
|
Krauson AJ, Casimero FVC, Siddiquee Z, Stone JR. Duration of SARS-CoV-2 mRNA vaccine persistence and factors associated with cardiac involvement in recently vaccinated patients. NPJ Vaccines 2023; 8:141. [PMID: 37758751 PMCID: PMC10533894 DOI: 10.1038/s41541-023-00742-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
At the start of the COVID-19 pandemic, the BNT162b2 (BioNTech-Pfizer) and mRNA-1273 (Moderna) mRNA vaccines were expediently designed and mass produced. Both vaccines produce the full-length SARS-CoV-2 spike protein for gain of immunity and have greatly reduced mortality and morbidity from SARS-CoV-2 infection. The distribution and duration of SARS-CoV-2 mRNA vaccine persistence in human tissues is unclear. Here, we developed specific RT-qPCR-based assays to detect each mRNA vaccine and screened lymph nodes, liver, spleen, and myocardium from recently vaccinated deceased patients. Vaccine was detected in the axillary lymph nodes in the majority of patients dying within 30 days of vaccination, but not in patients dying more than 30 days from vaccination. Vaccine was not detected in the mediastinal lymph nodes, spleen, or liver. Vaccine was detected in the myocardium in a subset of patients vaccinated within 30 days of death. Cardiac ventricles in which vaccine was detected had healing myocardial injury at the time of vaccination and had more myocardial macrophages than the cardiac ventricles in which vaccine was not detected. These results suggest that SARS-CoV-2 mRNA vaccines routinely persist up to 30 days from vaccination and can be detected in the heart.
Collapse
Affiliation(s)
- Aram J Krauson
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Faye Victoria C Casimero
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Zakir Siddiquee
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - James R Stone
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA.
- Department of Pathology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
42
|
Hınçer A, Ahan RE, Aras E, Şeker UÖŞ. Making the Next Generation of Therapeutics: mRNA Meets Synthetic Biology. ACS Synth Biol 2023; 12:2505-2515. [PMID: 37672348 PMCID: PMC10510722 DOI: 10.1021/acssynbio.3c00253] [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: 05/26/2023] [Indexed: 09/08/2023]
Abstract
The development of mRNA-based therapeutics centers around the natural functioning of mRNA molecules to provide the genetic information required for protein translation. To improve the efficacy of these therapeutics and minimize side effects, researchers can focus on the features of mRNA itself or the properties of the delivery agent to achieve the desired response. The tools considered for mRNA manipulation can be improved in terms of targetability, tunability, and translatability to medicine. While ongoing studies are dedicated to improving conventional approaches, innovative approaches can also be considered to unleash the full potential of mRNA-based therapeutics. Here, we discuss the opportunities that emerged from introducing synthetic biology to mRNA therapeutics. It includes a discussion of modular self-assembled mRNA nanoparticles, logic gates on a single mRNA molecule, and other possibilities.
Collapse
Affiliation(s)
- Ahmet Hınçer
- UNAM
− Institute of Materials Science and Nanotechnology, National
Nanotechnology Research Center, Bilkent
University, Ankara 06800, Turkey
| | - Recep Erdem Ahan
- UNAM
− Institute of Materials Science and Nanotechnology, National
Nanotechnology Research Center, Bilkent
University, Ankara 06800, Turkey
| | - Ebru Aras
- UNAM
− Institute of Materials Science and Nanotechnology, National
Nanotechnology Research Center, Bilkent
University, Ankara 06800, Turkey
| | - Urartu Özgür Şafak Şeker
- UNAM
− Institute of Materials Science and Nanotechnology, National
Nanotechnology Research Center, Bilkent
University, Ankara 06800, Turkey
| |
Collapse
|
43
|
Palanki R, Bose SK, Dave A, White BM, Berkowitz C, Luks V, Yaqoob F, Han E, Swingle KL, Menon P, Hodgson E, Biswas A, Billingsley MM, Li L, Yiping F, Carpenter M, Trokhan A, Yeo J, Johana N, Wan TY, Alameh MG, Bennett FC, Storm PB, Jain R, Chan J, Weissman D, Mitchell MJ, Peranteau WH. Ionizable Lipid Nanoparticles for Therapeutic Base Editing of Congenital Brain Disease. ACS NANO 2023; 17:13594-13610. [PMID: 37458484 PMCID: PMC11025390 DOI: 10.1021/acsnano.3c02268] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
Delivery of mRNA-based therapeutics to the perinatal brain holds great potential in treating congenital brain diseases. However, nonviral delivery platforms that facilitate nucleic acid delivery in this environment have yet to be rigorously studied. Here, we screen a diverse library of ionizable lipid nanoparticles (LNPs) via intracerebroventricular (ICV) injection in both fetal and neonatal mice and identify an LNP formulation with greater functional mRNA delivery in the perinatal brain than an FDA-approved industry standard LNP. Following in vitro optimization of the top-performing LNP (C3 LNP) for codelivery of an adenine base editing platform, we improve the biochemical phenotype of a lysosomal storage disease in the neonatal mouse brain, exhibit proof-of-principle mRNA brain transfection in vivo in a fetal nonhuman primate model, and demonstrate the translational potential of C3 LNPs ex vivo in human patient-derived brain tissues. These LNPs may provide a clinically translatable platform for in utero and postnatal mRNA therapies including gene editing in the brain.
Collapse
Affiliation(s)
- Rohan Palanki
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sourav K Bose
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Apeksha Dave
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Brandon M. White
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Cara Berkowitz
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Valerie Luks
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Fazeela Yaqoob
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emily Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kelsey L Swingle
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pallavi Menon
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Emily Hodgson
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Arijit Biswas
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
| | | | - Li Li
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fan Yiping
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
| | - Marco Carpenter
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexandra Trokhan
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Julie Yeo
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
| | | | - Tan Yi Wan
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
| | - Mohamad-Gabriel Alameh
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Frederick Chris Bennett
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Phillip B. Storm
- Division of Neurosurgery, Children’s Hospital of Philadelphia, PA 19104, USA
| | - Rajan Jain
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jerry Chan
- Duke-NUS Graduate Medical School, Singapore, 169547, SG
- Department of Reproductive Medicine, KK Women’s and Children’s Hospital, Singapore, 229899, SG
| | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael J. Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Regenerative Medicine, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - William H. Peranteau
- Center for Fetal Research, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of General, Thoracic, and Fetal Surgery, Children’s Hospital of Philadelphia, PA, USA
| |
Collapse
|
44
|
Kozma GT, Mészáros T, Berényi P, Facskó R, Patkó Z, Oláh CZ, Nagy A, Fülöp TG, Glatter KA, Radovits T, Merkely B, Szebeni J. Role of anti-polyethylene glycol (PEG) antibodies in the allergic reactions to PEG-containing Covid-19 vaccines: Evidence for immunogenicity of PEG. Vaccine 2023; 41:4561-4570. [PMID: 37330369 PMCID: PMC10239905 DOI: 10.1016/j.vaccine.2023.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 05/26/2023] [Accepted: 06/02/2023] [Indexed: 06/19/2023]
Abstract
A small fraction of recipients who receive polyethylene-glycol (PEG)-containing COVID-19 mRNA-LNP vaccines (Comirnaty and Spikevax) develop hypersensitivity reactions (HSRs) or anaphylaxis. A causal role of anti-PEG antibodies (Abs) has been proposed, but not yet been proven in humans.We used ELISA for serial measurements of SARS-CoV-2 neutralizing Ab (anti-S) and anti-PEG IgG/IgM Ab levels before and after the first and subsequent booster vaccinations with mRNA-LNP vaccines in a total of 291 blood donors. The HSRs in 15 subjects were graded and correlated with anti-PEG IgG/IgM, just as the anti-S and anti-PEG Ab levels with each other. The impacts of gender, allergy, mastocytosis and use of cosmetics were also analyzed. Serial testing of two or more plasma samples showed substantial individual variation of anti-S Ab levels after repeated vaccinations, just as the levels of anti-PEG IgG and IgM, which were over baseline in 98-99 % of unvaccinated individuals. About 3-4 % of subjects in the strongly left-skewed distribution had 15-45-fold higher values than the median, referred to as anti-PEG Ab supercarriers. Both vaccines caused significant rises of anti-PEG IgG/IgM with >10-fold rises in about ∼10 % of Comirnaty, and all Spikevax recipients. The anti-PEG IgG and/or IgM levels in the 15 vaccine reactors (3 anaphylaxis) were significantly higher compared to nonreactors. Serial testing of plasma showed significant correlation between the booster injection-induced rises of anti-S and anti-PEG IgGs, suggesting coupled anti-S and anti-PEG immunogenicity.Conclusions: The small percentage of people who have extremelevels of anti-PEG Ab in their blood may be at increased risk for HSRs/anaphylaxis to PEGylated vaccines and other PEGylated injectables. This risk might be further increased by the anti-PEG immunogenicity of these vaccines. Screening for anti-PEG Ab "supercarriers" may help predicting reactors and thus preventing these adverse phenomena.
Collapse
Affiliation(s)
- Gergely Tibor Kozma
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, Budapest, Hungary; SeroScience LLC, Budapest, Hungary
| | - Tamás Mészáros
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, Budapest, Hungary; SeroScience LLC, Budapest, Hungary
| | - Petra Berényi
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, Budapest, Hungary; SeroScience LLC, Budapest, Hungary
| | - Réka Facskó
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, Budapest, Hungary; SeroScience LLC, Budapest, Hungary
| | - Zsófia Patkó
- Department of Radiology, BAZ County Central Hospital and Borsod County University Teaching Hospital and Miskolc University, Miskolc, Hungary
| | - Csaba Zs Oláh
- Department of Neurosurgery, BAZ County Central Hospital and Borsod County University Teaching Hospital, Miskolc, Hungary
| | - Adrienne Nagy
- Department of Allergy, Heim Pál Children's Hospital, Budapest, Hungary
| | | | | | - Tamás Radovits
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Béla Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - János Szebeni
- Nanomedicine Research and Education Center, Department of Translational Medicine, Semmelweis University, Budapest, Hungary; SeroScience LLC, Budapest, Hungary; Department of Nanobiotechnology and Regenerative Medicine, Faculty of Health Sciences, Miskolc University, Miskolc, Hungary; Translational Nanobioscience Research Center, Sungkyunkwan University, Suwon 16419, South Korea.
| |
Collapse
|
45
|
Liu Y, Fei Y, Wang X, Yang B, Li M, Luo Z. Biomaterial-enabled therapeutic modulation of cGAS-STING signaling for enhancing antitumor immunity. Mol Ther 2023; 31:1938-1959. [PMID: 37002605 PMCID: PMC10362396 DOI: 10.1016/j.ymthe.2023.03.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/07/2023] [Accepted: 03/23/2023] [Indexed: 04/03/2023] Open
Abstract
cGAS-STING signaling is a central component in the therapeutic action of most existing cancer therapies. The accumulated knowledge of tumor immunoregulatory network in recent years has spurred the development of cGAS-STING agonists for tumor treatment as an effective immunotherapeutic strategy. However, the clinical translation of these agonists is thus far unsatisfactory because of the low immunostimulatory efficacy and unrestricted side effects under clinically relevant conditions. Interestingly, the rational integration of biomaterial technology offers a promising approach to overcome these limitations for more effective and safer cGAS-STING-mediated tumor therapy. Herein, we first outline the cGAS-STING signaling axis and generally discuss its association with tumors. We then symmetrically summarize the recent progress in those biomaterial-based cGAS-STING agonism strategies to generate robust antitumor immunity, categorized by the chemical nature of those cGAS-STING stimulants and carrier substrates. Finally, a perspective is provided to discuss the existing challenges and potential opportunities in cGAS-STING modulation for tumor therapy.
Collapse
Affiliation(s)
- Yingqi Liu
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Yang Fei
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Xuan Wang
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Bingbing Yang
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China.
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing 400044, P. R. China.
| |
Collapse
|
46
|
Pignatti P, Ramirez GA, Russo M, Marraccini P, Nannipieri S, Asperti C, Torre FD, Tiri A, Gatti BM, Gurrado A, Meriggi A, Benanti G, Cilona MB, Pigatto P, Burastero SE, Dagna L, Yacoub MR. Hypersensitivity reactions to anti-SARS-CoV-2 vaccines: Basophil reactivity to excipients. Vaccine 2023:S0264-410X(23)00714-4. [PMID: 37349223 DOI: 10.1016/j.vaccine.2023.06.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 06/09/2023] [Accepted: 06/11/2023] [Indexed: 06/24/2023]
Abstract
Basophil activation test (BAT) can tackle multiple mechanisms underlying acute and delayed hypersensitivity to drugs and vaccines and might complement conventional allergy diagnostics but its role in anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine-related hypersensitivity is ill-defined. Therefore, 89 patients with possible hypersensitivity (56 % with delayed mucocutaneous manifestations) to anti-SARS-CoV-2 vaccines were tested with BAT for Macrogol 3350, DMG-PEG 2000, PEG 20000, polysorbate-80 and trometamol and compared to 156 subjects undergoing pre-vaccine BAT. A positive BAT was associated with delayed reaction onset (p = 0.010) and resolution (p = 0.011). BAT was more frequently positive to DMG-PEG 2000 than to other excipients in both groups (p < 0.001). DMG-PEG 2000 reactivity was less frequent in vaccine-naïve (6 %) than vaccinated subjects (35 %, p < 0.001) and associated with mRNA-1273 vaccination. DMG-PEG 2000 BAT might therefore have a diagnostic role in subjects with delayed hypersensitivity reactions. Natural immunity might be a key player in basophil activation.
Collapse
Affiliation(s)
- Patrizia Pignatti
- Allergy and Immunology Unit, Istituti Clinici Scientifici Maugeri IRCCS Pavia, 27100 Pavia, Italy
| | - Giuseppe Alvise Ramirez
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| | - Marco Russo
- Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Marraccini
- Preventive and Occupational Medicine Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Foundation Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Serena Nannipieri
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Chiara Asperti
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | | | | | | | - Antonella Gurrado
- Allergy and Immunology Unit, Istituti Clinici Scientifici Maugeri IRCCS Pavia, 27100 Pavia, Italy
| | | | - Giovanni Benanti
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Maria Bernadette Cilona
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Paolo Pigatto
- Section of Clinical Dermatology, Department of Biomedical, Surgical and Dental Sciences, IRCCS Galeazzi Orthopedic Institute, University of Milan, Milan, Italy
| | - Samuele E Burastero
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Mona-Rita Yacoub
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| |
Collapse
|
47
|
Liu C, Shi Q, Huang X, Koo S, Kong N, Tao W. mRNA-based cancer therapeutics. Nat Rev Cancer 2023:10.1038/s41568-023-00586-2. [PMID: 37311817 DOI: 10.1038/s41568-023-00586-2] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/26/2023] [Indexed: 06/15/2023]
Abstract
Due to the fact that mRNA technology allows the production of diverse vaccines and treatments in a shorter time frame and with reduced expense compared to conventional approaches, there has been a surge in the use of mRNA-based therapeutics in recent years. With the aim of encoding tumour antigens for cancer vaccines, cytokines for immunotherapy, tumour suppressors to inhibit tumour development, chimeric antigen receptors for engineered T cell therapy or genome-editing proteins for gene therapy, many of these therapeutics have shown promising efficacy in preclinical studies, and some have even entered clinical trials. Given the evidence supporting the effectiveness and safety of clinically approved mRNA vaccines, coupled with growing interest in mRNA-based therapeutics, mRNA technology is poised to become one of the major pillars in cancer drug development. In this Review, we present in vitro transcribed mRNA-based therapeutics for cancer treatment, including the characteristics of the various types of synthetic mRNA, the packaging systems for efficient mRNA delivery, preclinical and clinical studies, current challenges and future prospects in the field. We anticipate the translation of promising mRNA-based treatments into clinical applications, to ultimately benefit patients.
Collapse
Affiliation(s)
- Chuang Liu
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Qiangqiang Shi
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| | - Xiangang Huang
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China.
| | - Wei Tao
- Center for Nanomedicine and Department of Anaesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
48
|
Münter R, Christensen E, Andresen TL, Larsen JB. Studying how administration route and dose regulates antibody generation against LNPs for mRNA delivery with single-particle resolution. Mol Ther Methods Clin Dev 2023; 29:450-459. [PMID: 37251983 PMCID: PMC10220314 DOI: 10.1016/j.omtm.2023.05.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 05/08/2023] [Indexed: 05/31/2023]
Abstract
Following the recent approval of both siRNA- and mRNA-based therapeutics, nucleic acid therapies are considered a game changer in medicine. Their envisioned widespread use for many therapeutic applications with an array of cellular target sites means that various administration routes will be employed. Concerns exist regarding adverse reactions against the lipid nanoparticles (LNPs) used for mRNA delivery, as PEG coatings on nanoparticles can induce severe antibody-mediated immune reactions, potentially being boosted by the inherently immunogenic nucleic acid cargo. While exhaustive information is available on how physicochemical features of nanoparticles affects immunogenicity, it remains unexplored how the fundamental choice of administration route regulates anti-particle immunity. Here, we directly compared antibody generation against PEGylated mRNA-carrying LNPs administered by the intravenous, intramuscular, or subcutaneous route, using a novel sophisticated assay capable of measuring antibody binding to authentic LNP surfaces with single-particle resolution. Intramuscular injections in mice were found to generate overall low and dose-independent levels of anti-LNP antibodies, while both intravenous and subcutaneous LNP injections generated substantial and highly dose-dependent levels. These findings demonstrate that before LNP-based mRNA medicines can be safely applied to new therapeutic applications, it will be crucial to carefully consider the choice of administration route.
Collapse
Affiliation(s)
- Rasmus Münter
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), 2800 Kongens Lyngby, Denmark
| | - Esben Christensen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), 2800 Kongens Lyngby, Denmark
| | - Thomas L. Andresen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), 2800 Kongens Lyngby, Denmark
| | - Jannik B. Larsen
- Biotherapeutic Engineering and Drug Targeting, Department of Health Technology, Technical University of Denmark (DTU), 2800 Kongens Lyngby, Denmark
| |
Collapse
|
49
|
Taharabaru T, Kihara T, Onodera R, Kogo T, Wen Y, Li J, Motoyama K, Higashi T. Versatile delivery platform for nucleic acids, negatively charged protein drugs, and genome-editing ribonucleoproteins using a multi-step transformable polyrotaxane. Mater Today Bio 2023; 20:100690. [PMID: 37441133 PMCID: PMC10333717 DOI: 10.1016/j.mtbio.2023.100690] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 05/07/2023] [Accepted: 06/02/2023] [Indexed: 07/15/2023] Open
Abstract
Various biopharmaceuticals, such as nucleic acids, proteins, and genome-editing molecules, have been developed. Generally, carriers are prepared for each biopharmaceutical to deliver it intracellularly; thus, the applications of individual carriers are limited. Moreover, the development of carriers is laborious and expensive. Therefore, in the present study, versatile and universal delivery carriers were developed for various biopharmaceuticals using aminated polyrotaxane libraries. Step-by-step and logical screening revealed that aminated polyrotaxane, including the carbamate bond between the axile molecule and endcap, is suitable as a backbone polymer. Movable and flexible properties of the amino groups modified on polyrotaxane facilitated efficient complexation with various biopharmaceuticals, such as small interfering RNA, antisense oligonucleotides, messenger RNA, β-galactosidase, and genome-editing ribonucleoproteins. Diethylenetriamine and cystamine modifications of polyrotaxane provided endosomal-escape abilities and drug-release properties in the cytosol, allowing higher delivery efficacies than commercially available high-standard carriers without cytotoxicity. Thus, the resulting polyrotaxane might serve as a versatile and universal delivery platform for various biopharmaceuticals.
Collapse
Affiliation(s)
- Toru Taharabaru
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore, 119276, Singapore
| | - Takuya Kihara
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Risako Onodera
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Tetsuya Kogo
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Yuting Wen
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore, 119276, Singapore
| | - Jun Li
- Department of Biomedical Engineering, National University of Singapore, 15 Kent Ridge Crescent, Singapore, 119276, Singapore
| | - Keiichi Motoyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
| | - Taishi Higashi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan
- Priority Organization for Innovation and Excellence, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, 860-8555, Japan
| |
Collapse
|
50
|
Tenchov R, Sasso JM, Zhou QA. PEGylated Lipid Nanoparticle Formulations: Immunological Safety and Efficiency Perspective. Bioconjug Chem 2023. [PMID: 37162501 DOI: 10.1021/acs.bioconjchem.3c00174] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Lipid nanoparticles (LNPs) have been recognized as efficient vehicles to transport a large variety of therapeutics. Currently in the spotlight as important constituents of the COVID-19 mRNA vaccines, LNPs play a significant role in protecting and transporting mRNA to cells. As one of their key constituents, polyethylene glycol (PEG)-lipid conjugates are important in defining LNP physicochemical characteristics and biological activity. PEGylation has proven particularly efficient in conferring longer systemic circulation of LNPs, thus greatly improving their pharmacokinetics and efficiency. Along with revealing the benefits of PEG conjugates, studies have revealed unexpected immune reactions against PEGylated nanocarriers such as accelerated blood clearance (ABC), involving the production of anti-PEG antibodies at initial injection, which initiates accelerated blood clearance upon subsequent injections, as well as a hypersensitivity reaction referred to as complement activation-related pseudoallergy (CARPA). Further, data have been accumulated indicating consistent yet sometimes controversial correlations between various structural parameters of the PEG-lipids, the properties of the PEGylated LNPs, and the magnitude of the observed adverse effects. Detailed knowledge and comprehension of such correlations are of foremost importance in the efforts to diminish and eliminate the undesirable immune reactions and improve the safety and efficiency of the PEGylated medicines. Here, we present an overview based on analysis of data from the CAS Content Collection regarding the PEGylated LNP immunogenicity and overall safety concerns. A comprehensive summary has been compiled outlining how various structural parameters of the PEG-lipids affect the immune responses and activities of the LNPs, with regards to their efficiency in drug delivery. This Review is thus intended to serve as a helpful resource in understanding the current knowledge in the field, in an effort to further solve the remaining challenges and to achieve full potential.
Collapse
Affiliation(s)
- Rumiana Tenchov
- CAS, a division of the American Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Janet M Sasso
- CAS, a division of the American Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| | - Qiongqiong Angela Zhou
- CAS, a division of the American Chemical Society, 2540 Olentangy River Road, Columbus, Ohio 43202, United States
| |
Collapse
|