1
|
Chrysant SG. Better blood pressure control with the nanoformulation of antihypertensive drugs. Expert Rev Cardiovasc Ther 2024:1-9. [PMID: 39635781 DOI: 10.1080/14779072.2024.2438813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 11/12/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION Hypertension is very common and a major risk factor for cardiovascular disease, heart failure, chronic kidney disease, strokes, and death. However, at present only 14% of patients of developing countries have their blood pressure (BP) well controlled. The causes for the failure to control the BP are multiple and one of them could be the formulation of antihypertensive drugs. AREAS COVERED The recent development of nanotechnology by incorporating the drugs into nanoparticles is a new promising field of nanomedicine and preliminary studies have shown this nanoformulation to be more effective in the treatment of hypertension than the existing drug formulations. Another recent development is the nanoformulation of genes used for the treatment of hypertension and cardiovascular diseases. For current information, a Medline search was conducted between 2017 and 2024 and 36 pertinent papers were selected. EXPERT OPINION The nanoformulations of drugs help achieve better drug concentrations, improve drug stability, low solubility, short half life, oral bioavailability, narrow therapeutic index, and poor pharmacokinetic and pharmacodynamic profiles, and decrease the adverse effects of antihypertensive drugs. Also, the nanoformulation of genes for the treatment of hypertension has been shown in preliminary studies to be effective, but more research is needed.
Collapse
Affiliation(s)
- Steven G Chrysant
- Department of Cardiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
2
|
Fu T, Zhou B, Li Y, Liu W, Xie Y, Mo Z, Yin F, Wang Y, Fang K, Fang Y, Xiong Z, Yu K, Le A. Innovative Dual mRNA-Lipid Nanoparticle Therapy Targeting CRHBP and CFHR3 for Enhanced Treatment of Hepatocellular Carcinoma. Int J Nanomedicine 2024; 19:13183-13199. [PMID: 39664759 PMCID: PMC11633302 DOI: 10.2147/ijn.s498065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/26/2024] [Indexed: 12/13/2024] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) is a deadly disease requiring the identification of new therapeutic targets and strategies. Methods This study identified genes linked to HCC progression via differential analysis. Key genes were identified through univariate and multivariate Cox regression analysis. The biological effects of co-expressed CRHBP and CFHR3 were evaluated in vitro. mRNAs encoding CRHBP and CFHR3 were encapsulated in lipid nanoparticles (LNPs), with the addition of SP94 peptide on the LNPs surface to enhance targeting. The therapeutic efficacy of dual-mRNA LNPs was evaluated in HCC cells and mouse models. Results CRHBP and CFHR3 were closely associated with HCC progression. Low expression of CRHBP (P < 0.01, HR = 1.931 [1.174-3.175]) and CFHR3 (P < 0.05, HR = 1.755 [1.066-2.890]) was identified as a poor prognostic factor for HCC. The risk score model combining CRHBP and CFHR3 demonstrated superior predictive power (P < 0.001, HR = 2.935 [1.768-4.872]). Co-expression of CRHBP and CFHR3 significantly inhibited the malignant biological functions of HCC cells. Treatment with SP94 peptide-modified dual-mRNA LNPs markedly suppressed HCC tumor growth and exhibited excellent biocompatibility and safety. Conclusion Our study proposes a dual-targeted therapeutic strategy for HCC, which may represent a promising treatment approach.
Collapse
Affiliation(s)
- Tianmei Fu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Boxuan Zhou
- Department of Breast Disease Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Yingliang Li
- Department of Breast Disease Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Wei Liu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Yuankang Xie
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, People’s Republic of China
| | - Zhaohong Mo
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanchang, People’s Republic of China
| | - Fang Yin
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Yu Wang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Kang Fang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Yangyang Fang
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Ziqing Xiong
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Kuai Yu
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| | - Aiping Le
- Department of Transfusion Medicine, Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, People’s Republic of China
| |
Collapse
|
3
|
Repkova M, Mazurkov O, Filippova E, Protsenko M, Mazurkova N, Meschaninova M, Levina A, Zarytova V. Effect of modification of siRNA molecules delivered with aminopropylsilanol nanoparticles on suppression of A/H5N1 virus in cell culture. Biochim Biophys Acta Gen Subj 2024; 1868:130727. [PMID: 39437973 DOI: 10.1016/j.bbagen.2024.130727] [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: 07/02/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
The application of siRNAs as antiviral agents is limited by several obstacles including their poor penetration into cells and instability in biological media. To overcome these problems, we used non-agglomerated aminopropylsilanol nanoparticles (NP) to deliver siRNA into cells. All studied siRNAs had identical nucleoside sequences comprising phosphodiester or phosphorothioate (PS) internucleotide groups and the 2'-OMe and/or 2'-F groups in nucleoside units at different positions of RNA. The siRNA molecules were attached to NP, thus forming the NP-siRNA nanocomplexes. We studied the effect of siRNA modification in the nanocomplexes on suppressing the highly pathogenic influenza A/H5N1 virus replication. The results demonstrated that all siRNA-containing nanocomplexes inhibited the replication of the A/H5N1 virus by 1-3 orders of magnitude. The nanocomplexes containing partially modified siRNAs exhibited the most pronounced inhibition with an efficacy of 900-fold. This result was achieved by using siRNA consisting of the canonical 19-bp RNA duplex with the 3'-dTdT dangling ends, with the antisense strand in this duplex being protected from endonucleases (one UMeA site within the strand). The additional modifications of siRNA reduce their antiviral activity. Promising sense strands for loading into the RISC complex are likely to be phosphodiester sequences that contain dTdT at the 3' end (such as S4) to be protected against exonucleases. The sense strands of this type can probably be the most suitable for designing siRNAs as therapeutic agents. The proposed NP-siRNA nanocomplexes that consisted of low toxic and non-agglomerated aminopropylsilanol nanoparticles and siRNA molecules could be hopeful agents for gene silencing.
Collapse
Affiliation(s)
- Marina Repkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of RAS, pr. Lavrent'eva 8, Novosibirsk 630090, Russia
| | - Oleg Mazurkov
- FBRI State Research Center of Virology and Biotechnology "Vector" Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Ekaterina Filippova
- FBRI State Research Center of Virology and Biotechnology "Vector" Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Maria Protsenko
- FBRI State Research Center of Virology and Biotechnology "Vector" Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Natalia Mazurkova
- FBRI State Research Center of Virology and Biotechnology "Vector" Rospotrebnadzor, Koltsovo, Novosibirsk 630559, Russia
| | - Maria Meschaninova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of RAS, pr. Lavrent'eva 8, Novosibirsk 630090, Russia
| | - Asya Levina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of RAS, pr. Lavrent'eva 8, Novosibirsk 630090, Russia
| | - Valentina Zarytova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of RAS, pr. Lavrent'eva 8, Novosibirsk 630090, Russia.
| |
Collapse
|
4
|
Su Z, Boucetta H, Shao J, Huang J, Wang R, Shen A, He W, Xu ZP, Zhang L. Next-generation aluminum adjuvants: Immunomodulatory layered double hydroxide NanoAlum reengineered from first-line drugs. Acta Pharm Sin B 2024; 14:4665-4682. [PMID: 39664431 PMCID: PMC11628803 DOI: 10.1016/j.apsb.2024.09.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 12/13/2024] Open
Abstract
Aluminum adjuvants (Alum), approved by the US Food and Drug Administration, have been extensively used in vaccines containing recombinant antigens, subunits of pathogens, or toxins for almost a century. While Alums typically elicit strong humoral immune responses, their ability to induce cellular and mucosal immunity is limited. As an alternative, layered double hydroxide (LDH), a widely used antacid, has emerged as a novel class of potent nano-aluminum adjuvants (NanoAlum), demonstrating advantageous physicochemical properties, biocompatibility and adjuvanticity in both humoral and cellular immune responses. In this review, we summarize and compare the advantages and disadvantages of Alum and NanoAlum in these properties and their performance as adjuvants. Moreover, we propose the key features for ideal adjuvants and demonstrate that LDH NanoAlum is a promising candidate by summarizing its current progress in immunotherapeutic cancer treatments. Finally, we conclude the review by offering our integrated perspectives about the remaining challenges and future directions for NanoAlum's application in preclinical/clinical settings.
Collapse
Affiliation(s)
- Zhenwei Su
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Hamza Boucetta
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Jiahui Shao
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Jinling Huang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Ran Wang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Aining Shen
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Zhi Ping Xu
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518107, China
- School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Lingxiao Zhang
- Interdisciplinary Nanoscience Center (INANO), Aarhus University, Aarhus 8000, Denmark
| |
Collapse
|
5
|
Lu C, Rohilla P, Felner EI, Byagathvalli G, Azizoglu E, Bhamla MS, Prausnitz MR. Tolerability of a piezoelectric microneedle electroporator in human subjects. Bioeng Transl Med 2024; 9:e10662. [PMID: 39036075 PMCID: PMC11256137 DOI: 10.1002/btm2.10662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 02/08/2024] [Accepted: 02/27/2024] [Indexed: 07/23/2024] Open
Abstract
Electroporation, or the use of electric pulses to facilitate the intracellular delivery of DNA, RNA, and other molecules, is a well-established technique, that has been demonstrated to significantly augment the immunogenicity of DNA/mRNA vaccines and therapeutics. However, the clinical translation of traditional electroporators has been limited due to high costs, large size, complex user operation, and poor tolerability in humans due to nerve stimulation. In prior work, we introduced ePatch: an ultra-low-cost, handheld, battery-free electroporator employing a piezoelectric pulser coupled with a microneedle electrode array that showed enhanced immunogenic responses to an intradermal SARS-CoV-2 DNA vaccine in mice. The current study shifts focus from efficacy to tolerability, hypothesizing that ePatch's microneedle array, which localizes the electric field to the superficial skin strata, will minimize nerve stimulation and improve patient comfort. We tested this hypothesis in 14 healthy adults, monitoring pain and other potential adverse effects associated with electroporation. Compared to the insertion of a traditional hypodermic needle, the ePatch was less painful. Adverse effects such as pain, tenderness, erythema and swelling at the application sites were minimal, transient, and statistically indistinguishable between the experimental and placebo ePatch application, suggesting excellent tolerability towards electroporation. In summary, ePatch has a favorable tolerability profile in humans and offers the potential for the safe use of electroporation in a variety of clinical settings, including DNA and mRNA vaccination.
Collapse
Affiliation(s)
- Chao‐Yi Lu
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Pankaj Rohilla
- School of Chemical and Biomolecular Engineering, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Eric I. Felner
- School of Chemical and Biomolecular Engineering, Georgia Institute of TechnologyAtlantaGeorgiaUSA
- Department of Pediatrics, Division of EndocrinologyEmory University School of MedicineAtlantaGeorgiaUSA
| | - Gaurav Byagathvalli
- School of Chemical and Biomolecular Engineering, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Erkan Azizoglu
- School of Chemical and Biomolecular Engineering, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| | - M. Saad Bhamla
- School of Chemical and Biomolecular Engineering, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| | - Mark R. Prausnitz
- Wallace H. Coulter Department of Biomedical Engineering at Georgia Tech and Emory University, Georgia Institute of TechnologyAtlantaGeorgiaUSA
- School of Chemical and Biomolecular Engineering, Georgia Institute of TechnologyAtlantaGeorgiaUSA
| |
Collapse
|
6
|
Schober GB, Story S, Arya DP. A careful look at lipid nanoparticle characterization: analysis of benchmark formulations for encapsulation of RNA cargo size gradient. Sci Rep 2024; 14:2403. [PMID: 38287070 PMCID: PMC10824725 DOI: 10.1038/s41598-024-52685-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 01/19/2024] [Indexed: 01/31/2024] Open
Abstract
With the recent success of lipid nanoparticle (LNP) based SARS-CoV-2 mRNA vaccines, the potential for RNA therapeutics has gained widespread attention. LNPs are promising non-viral delivery vectors to protect and deliver delicate RNA therapeutics, which are ineffective and susceptible to degradation alone. While food and drug administration (FDA) approved formulations have shown significant promise, benchmark lipid formulations still require optimization and improvement. In addition, the translatability of these formulations for several different RNA cargo sizes has not been compared under the same conditions. Herein we analyze "gold standard" lipid formulations for encapsulation efficiency of various non-specific RNA cargo lengths representing antisense oligonucleotides (ASO), small interfering RNA (siRNA), RNA aptamers, and messenger RNA (mRNA), with lengths of 10 bases, 21 base pairs, 96 bases, 996 bases, and 1929 bases, respectively. We evaluate encapsulation efficiency as the percentage of input RNA encapsulated in the final LNP product (EEinput%), which shows discrepancy with the traditional calculation of encapsulation efficiency (EE%). EEinput% is shown to be < 50% for all formulations tested, when EE% is consistently > 85%. We also compared formulations for LNP size (Z-average) and polydispersity index (PDI). LNP size does not appear to be strongly influenced by cargo size, which is a counterintuitive finding. Thoughtful characterization of LNPs, in parallel with consideration of in vitro or in vivo behavior, will guide design and optimization for better understanding and improvement of future RNA therapeutics.
Collapse
Affiliation(s)
| | | | - Dev P Arya
- NUBAD LLC, Greer, 29650, USA.
- Department of Chemistry, Clemson University, Clemson, 29631, USA.
| |
Collapse
|
7
|
Sanati M, Afshari AR, Ahmadi SS, Jamialahmadi T, Sahebkar A. Application of RNA-based therapeutics in glioma: A review. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 204:133-161. [PMID: 38458736 DOI: 10.1016/bs.pmbts.2023.12.001] [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: 03/10/2024]
Abstract
Despite the extensive advancements made in the field of cancer therapy, the outlook of individuals suffering from glioblastoma multiforme remains highly detrimental. The absence of specific treatments for cancerous cells significantly hinders the effectiveness of conventional anticancer techniques. Multiple research studies have demonstrated that the suppression of specific genes or the augmentation of therapeutic proteins through RNA-based therapeutics may represent a valuable approach when combined with chemotherapy or immunotherapy. In recent years, there has been a significant increase in the application of RNA therapeutics in conjunction with chemotherapy and immunotherapy. This emerging field has become a prominent area of research for advancing various types of cancer treatments. The present investigation provides an in-depth overview of the classification and application of RNA therapy, focusing on the mechanisms of RNA antitumor treatment and the current status of clinical studies on RNA drugs.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Amir R Afshari
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran; Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyed Sajad Ahmadi
- Department of Ophthalmology, Khatam-Ol-Anbia Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Zhang J, Chen B, Gan C, Sun H, Zhang J, Feng L. A Comprehensive Review of Small Interfering RNAs (siRNAs): Mechanism, Therapeutic Targets, and Delivery Strategies for Cancer Therapy. Int J Nanomedicine 2023; 18:7605-7635. [PMID: 38106451 PMCID: PMC10725753 DOI: 10.2147/ijn.s436038] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/29/2023] [Indexed: 12/19/2023] Open
Abstract
Small interfering RNA (siRNA) delivery by nanocarriers has been identified as a promising strategy in the study and treatment of cancer. Short nucleotide sequences are synthesized exogenously to create siRNA, which triggers RNA interference (RNAi) in cells and silences target gene expression in a sequence-specific way. As a nucleic acid-based medicine that has gained popularity recently, siRNA exhibits novel potential for the treatment of cancer. However, there are still many obstacles to overcome before clinical siRNA delivery devices can be developed. In this review, we discuss prospective targets for siRNA drug design, explain siRNA drug properties and benefits, and give an overview of the current clinical siRNA therapeutics for the treatment of cancer. Additionally, we introduce the siRNA chemical modifications and delivery systems that are clinically sophisticated and classify bioresponsive materials for siRNA release in a methodical manner. This review will serve as a reference for researchers in developing more precise and efficient targeted delivery systems, promoting ongoing advances in clinical applications.
Collapse
Affiliation(s)
- Jiaying Zhang
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, People’s Republic of China
| | - Bo Chen
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, People’s Republic of China
| | - Chunyuan Gan
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, People’s Republic of China
| | - Hongyan Sun
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, People’s Republic of China
| | - Jiaxin Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
- Institute of Liver Diseases, Beijing University of Chinese Medicine, Beijing, People’s Republic of China
| | - Lin Feng
- School of Mechanical Engineering and Automation, Beihang University, Beijing, 100191, People’s Republic of China
- Beijing Advanced Innovation Center for Biomedical Engineering, Beihang University, Beijing, 100191, People’s Republic of China
| |
Collapse
|
9
|
Saiding Q, Zhang Z, Chen S, Xiao F, Chen Y, Li Y, Zhen X, Khan MM, Chen W, Koo S, Kong N, Tao W. Nano-bio interactions in mRNA nanomedicine: Challenges and opportunities for targeted mRNA delivery. Adv Drug Deliv Rev 2023; 203:115116. [PMID: 37871748 DOI: 10.1016/j.addr.2023.115116] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/17/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023]
Abstract
Upon entering the biological milieu, nanomedicines swiftly interact with the surrounding tissue fluid, subsequently being enveloped by a dynamic interplay of biomacromolecules, such as carbohydrates, nucleic acids, and cellular metabolites, but with predominant serum proteins within the biological corona. A notable consequence of the protein corona phenomenon is the unintentional loss of targeting ligands initially designed to direct nanomedicines toward particular cells or organs within the in vivo environment. mRNA nanomedicine displays high demand for specific cell and tissue-targeted delivery to effectively transport mRNA molecules into target cells, where they can exert their therapeutic effects with utmost efficacy. In this review, focusing on the delivery systems and tissue-specific applications, we aim to update the nanomedicine population with the prevailing and still enigmatic paradigm of nano-bio interactions, a formidable hurdle in the pursuit of targeted mRNA delivery. We also elucidate the current impediments faced in mRNA therapeutics and, by contemplating prospective avenues-either to modulate the corona or to adopt an 'ally from adversary' approach-aim to chart a course for advancing mRNA nanomedicine.
Collapse
Affiliation(s)
- Qimanguli Saiding
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Zhongyang Zhang
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States; The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Shuying Chen
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Fan Xiao
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang 311121, China; Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Yumeng Chen
- The Danish National Research Foundation and Villum Foundation's Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, 2800 Kgs. Lyngby, Denmark
| | - Yongjiang Li
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Xueyan Zhen
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Muhammad Muzamil Khan
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Wei Chen
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Seyoung Koo
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States.
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang 311121, China; Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States.
| | - Wei Tao
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States.
| |
Collapse
|
10
|
Story D, Aminoroaya A, Skelton Z, Kumari M, Zhang Y, Smith BR. Nanoparticle-Based Therapies in Hypertension. Hypertension 2023; 80:2506-2514. [PMID: 37767725 PMCID: PMC10651274 DOI: 10.1161/hypertensionaha.123.19523] [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: 09/29/2023]
Abstract
Nearly 1.4 billion people worldwide suffer from arterial hypertension, a significant risk factor for cardiovascular disease which is now the leading cause of death. Despite numerous drugs designed to treat hypertension, only ≈14% of hypertensive individuals have their blood pressure under control. A critical factor negatively impacting the efficacy of available treatments is their poor bioavailability. This leads to increased dosing requirements which can result in more side effects, resulting in patient noncompliance. A recent solution to improve dosing and bioavailability issues has been to incorporate drugs into nanoparticle carriers, with over 50 nanodrugs currently on the market across all diseases, and another 51 currently in clinical trials. Given their ability to improve solubility and bioavailability, nanoparticles may offer significant advantages in the formulation of antihypertensives to overcome pharmacokinetic shortcomings. To date, however, no antihypertensive nanoformulations have been clinically approved. This review assesses in vivo study data from preclinical antihypertensive nanoformulation development and testing. Combined, the results of these studies suggest nanoformulation of antihypertensive drugs may be a promising solution to overcome the poor efficacy of currently available antihypertensives, and with further advances has the potential to open paths for new substances that have heretofore been clinically unrealistic due to poor bioavailability.
Collapse
Affiliation(s)
- Darren Story
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering (D.S., M.K., Y.Z., B.R.S.), Michigan State University, East Lansing, MI
| | - Alireza Aminoroaya
- Department of Chemical Engineering and Materials Science (A.A., B.R.S.), Michigan State University, East Lansing, MI
| | - Zak Skelton
- College of Osteopathic Medicine (Z.S.), Michigan State University, East Lansing, MI
| | - Manisha Kumari
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering (D.S., M.K., Y.Z., B.R.S.), Michigan State University, East Lansing, MI
| | - Yapei Zhang
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering (D.S., M.K., Y.Z., B.R.S.), Michigan State University, East Lansing, MI
| | - Bryan Ronain Smith
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering (D.S., M.K., Y.Z., B.R.S.), Michigan State University, East Lansing, MI
- Department of Chemical Engineering and Materials Science (A.A., B.R.S.), Michigan State University, East Lansing, MI
| |
Collapse
|
11
|
Saraswat A, Patel K. Delineating effect of headgroup and preparation method on transfection versus toxicity of DNA-loaded lipid nanocarriers. Nanomedicine (Lond) 2023; 18:1921-1940. [PMID: 38078422 DOI: 10.2217/nnm-2023-0219] [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: 12/19/2023] Open
Abstract
Aim: To perform a parallel comparison of key parameters affecting the safety and efficiency of lipid-based nanovectors (i.e., complexing headgroups, composition and preparation method). Materials & methods: Various cationic and ionizable headgroups were screened for formulating lipoplexes with GFP-plasmid DNA. Ethanol injection and microfluidics were used to prepare nanoparticles with GFP-plasmid DNA complexed on the surface or within the interior of lipid bilayers. Results: Lipoplexes composed of sphingomyelin 102 exhibited the highest transfection efficiency given their higher cellular uptake in BRAF inhibitor-resistant melanoma cells. Lipid nanoparticles demonstrated acceptable transfection efficiency and high spheroid penetration while protecting plasmid DNA under simulated physiological conditions. Conclusion: Selecting the right complexing lipid and preparation method is critical for developing lipid nanocarriers to treat intractable diseases.
Collapse
Affiliation(s)
- Aishwarya Saraswat
- College of Pharmacy & Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Ketan Patel
- College of Pharmacy & Health Sciences, St. John's University, Queens, NY 11439, USA
| |
Collapse
|
12
|
Abbo LM, Vasiliu-Feltes I. Disrupting the infectious disease ecosystem in the digital precision health era innovations and converging emerging technologies. Antimicrob Agents Chemother 2023; 67:e0075123. [PMID: 37724872 PMCID: PMC10583659 DOI: 10.1128/aac.00751-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023] Open
Abstract
This commentary explores the convergence of precision health and evolving technologies, including the critical role of artificial intelligence (AI) and emerging technologies in infectious diseases (ID) and microbiology. We discuss their disruptive impact on the ID ecosystem and examine the transformative potential of frontier technologies in precision health, public health, and global health when deployed with robust ethical and data governance guardrails in place.
Collapse
Affiliation(s)
- Lilian M. Abbo
- Jackson Health System, Miami, Florida, USA
- Division of Infectious Diseases, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | | |
Collapse
|
13
|
Ribeiro J, Lopes I, Gomes AC. A New Perspective for the Treatment of Alzheimer's Disease: Exosome-like Liposomes to Deliver Natural Compounds and RNA Therapies. Molecules 2023; 28:6015. [PMID: 37630268 PMCID: PMC10458935 DOI: 10.3390/molecules28166015] [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: 06/15/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
With the increment of the aging population in recent years, neurodegenerative diseases exert a major global disease burden, essentially as a result of the lack of treatments that stop the disease progression. Alzheimer's Disease (AD) is an example of a neurodegenerative disease that affects millions of people globally, with no effective treatment. Natural compounds have emerged as a viable therapy to fill a huge gap in AD management, and in recent years, mostly fueled by the COVID-19 pandemic, RNA-based therapeutics have become a hot topic in the treatment of several diseases. Treatments of AD face significant limitations due to the complex and interconnected pathways that lead to their hallmarks and also due to the necessity to cross the blood-brain barrier. Nanotechnology has contributed to surpassing this bottleneck in the treatment of AD by promoting safe and enhanced drug delivery to the brain. In particular, exosome-like nanoparticles, a hybrid delivery system combining exosomes and liposomes' advantageous features, are demonstrating great potential in the treatment of central nervous system diseases.
Collapse
Affiliation(s)
- Joana Ribeiro
- Centre of Molecular and Environmental Biology (CBMA)/Aquatic Research Network (ARNET) Associate Laboratory, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (J.R.); (I.L.)
- Institute of Science and Innovation for Sustainability (IB-S), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Ivo Lopes
- Centre of Molecular and Environmental Biology (CBMA)/Aquatic Research Network (ARNET) Associate Laboratory, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (J.R.); (I.L.)
| | - Andreia Castro Gomes
- Centre of Molecular and Environmental Biology (CBMA)/Aquatic Research Network (ARNET) Associate Laboratory, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (J.R.); (I.L.)
- Institute of Science and Innovation for Sustainability (IB-S), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| |
Collapse
|
14
|
Subhan MA, Filipczak N, Torchilin VP. Advances with Lipid-Based Nanosystems for siRNA Delivery to Breast Cancers. Pharmaceuticals (Basel) 2023; 16:970. [PMID: 37513882 PMCID: PMC10386415 DOI: 10.3390/ph16070970] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
Breast cancer is the most frequently diagnosed cancer among women. Breast cancer is also the key reason for worldwide cancer-related deaths among women. The application of small interfering RNA (siRNA)-based drugs to combat breast cancer requires effective gene silencing in tumor cells. To overcome the challenges of drug delivery to tumors, various nanosystems for siRNA delivery, including lipid-based nanoparticles that protect siRNA from degradation for delivery to cancer cells have been developed. These nanosystems have shown great potential for efficient and targeted siRNA delivery to breast cancer cells. Lipid-based nanosystems remain promising as siRNA drug delivery carriers for effective and safe cancer therapy including breast cancer. Lipid nanoparticles (LNPs) encapsulating siRNA enable efficient and specific silencing of oncogenes in breast tumors. This review discusses a variety of lipid-based nanosystems including cationic lipids, sterols, phospholipids, PEG-lipid conjugates, ionizable liposomes, exosomes for effective siRNA drug delivery to breast tumors, and the clinical translation of lipid-based siRNA nanosystems for solid tumors.
Collapse
Affiliation(s)
- Md Abdus Subhan
- Department of Chemistry, ShahJalal University of Science and Technology, Sylhet 3114, Bangladesh
- Division of Nephrology, University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave, Box 675, Rochester, NY 14642, USA
| | - Nina Filipczak
- Center for Pharmaceutical Biotechnology and Nanomedicine, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
| | - Vladimir P Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
15
|
Golubovic A, Tsai S, Li B. Bioinspired Lipid Nanocarriers for RNA Delivery. ACS BIO & MED CHEM AU 2023; 3:114-136. [PMID: 37101812 PMCID: PMC10125326 DOI: 10.1021/acsbiomedchemau.2c00073] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 04/28/2023]
Abstract
RNA therapy is a disruptive technology comprising a rapidly expanding category of drugs. Further translation of RNA therapies to the clinic will improve the treatment of many diseases and help enable personalized medicine. However, in vivo delivery of RNA remains challenging due to the lack of appropriate delivery tools. Current state-of-the-art carriers such as ionizable lipid nanoparticles still face significant challenges, including frequent localization to clearance-associated organs and limited (1-2%) endosomal escape. Thus, delivery vehicles must be improved to further unlock the full potential of RNA therapeutics. An emerging strategy is to modify existing or new lipid nanocarriers by incorporating bioinspired design principles. This method generally aims to improve tissue targeting, cellular uptake, and endosomal escape, addressing some of the critical issues facing the field. In this review, we introduce the different strategies for creating bioinspired lipid-based RNA carriers and discuss the potential implications of each strategy based on reported findings. These strategies include incorporating naturally derived lipids into existing nanocarriers and mimicking bioderived molecules, viruses, and exosomes. We evaluate each strategy based on the critical factors required for delivery vehicles to succeed. Finally, we point to areas of research that should be furthered to enable the more successful rational design of lipid nanocarriers for RNA delivery.
Collapse
Affiliation(s)
- Alex Golubovic
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Shannon Tsai
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Bowen Li
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
- Institute
of Biomedical Engineering, University of
Toronto, Toronto, Ontario M5S 3G9, Canada
| |
Collapse
|
16
|
Zhong Y, Du S, Dong Y. mRNA delivery in cancer immunotherapy. Acta Pharm Sin B 2023; 13:1348-1357. [PMID: 37139419 PMCID: PMC10150179 DOI: 10.1016/j.apsb.2023.03.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/28/2023] [Accepted: 03/01/2023] [Indexed: 03/07/2023] Open
Abstract
Messenger RNA (mRNA) has drawn much attention in the medical field. Through various treatment approaches including protein replacement therapies, gene editing, and cell engineering, mRNA is becoming a potential therapeutic strategy for cancers. However, delivery of mRNA into targeted organs and cells can be challenging due to the unstable nature of its naked form and the low cellular uptake. Therefore, in addition to mRNA modification, efforts have been devoted to developing nanoparticles for mRNA delivery. In this review, we introduce four categories of nanoparticle platform systems: lipid, polymer, lipid-polymer hybrid, and protein/peptide-mediated nanoparticles, together with their roles in facilitating mRNA-based cancer immunotherapies. We also highlight promising treatment regimens and their clinical translation.
Collapse
Affiliation(s)
- Yichen Zhong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
- Icahn Genomics Institute, Precision Immunology Institute, Department of Oncological Sciences, Tisch Cancer Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shi Du
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
- Icahn Genomics Institute, Precision Immunology Institute, Department of Oncological Sciences, Tisch Cancer Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
- Department of Radiation Oncology, Department of Biomedical Engineering, Center for Clinical and Translational Science, Comprehensive Cancer Center, Dorothy M. Davis Heart & Lung Research Institute, Center for Cancer Engineering, Center for Cancer Metabolism, Pelotonia Institute for Immune-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Icahn Genomics Institute, Precision Immunology Institute, Department of Oncological Sciences, Tisch Cancer Institute, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Corresponding author.
| |
Collapse
|
17
|
Zeng M, Guo D, Fernández-Varo G, Zhang X, Fu S, Ju S, Yang H, Liu X, Wang YC, Zeng Y, Casals G, Casals E. The Integration of Nanomedicine with Traditional Chinese Medicine: Drug Delivery of Natural Products and Other Opportunities. Mol Pharm 2023; 20:886-904. [PMID: 36563052 DOI: 10.1021/acs.molpharmaceut.2c00882] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The integration of progressive technologies such as nanomedicine with the use of natural products from traditional medicine (TM) provides a unique opportunity for the longed-for harmonization between traditional and modern medicine. Although several actions have been initiated decades ago, a disparity of reasons including some misunderstandings between each other limits the possibilities of a truly complementation. Herein, we analyze some common challenges between nanomedicine and traditional Chinese medicine (TCM). These challenges, if solved in a consensual way, can give a boost to such harmonization. Nanomedicine is a recently born technology, while TCM has been used by the Chinese people for thousands of years. However, for these disciplines, the regulation and standardization of many of the protocols, especially related to the toxicity and safety, regulatory aspects, and manufacturing procedures, are under discussion. Besides, both TCM and nanomedicine still need to achieve a wider social acceptance. Herein, we first briefly discuss the strengths and weaknesses of TCM. This analysis serves to focus afterward on the aspects where TCM and nanomedicine can mutually help to bridge the existing gaps between TCM and Western modern medicine. As discussed, many of these challenges can be applied to TM in general. Finally, recent successful cases in scientific literature that merge TCM and nanomedicine are reviewed as examples of the benefits of this harmonization.
Collapse
Affiliation(s)
- Muling Zeng
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Dongdong Guo
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Guillermo Fernández-Varo
- Service of Biochemistry and Molecular Genetics, Hospital Clinic Universitari, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Carrer de Villarroel, 170, 08036 Barcelona, Spain.,Department of Biomedicine, University of Barcelona, 08007 Barcelona, Spain
| | - Xu Zhang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Siqi Fu
- Department of Dermatology, Second Xiangya Hospital Central South University, Changsha 410011, China
| | - Shijie Ju
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Huiling Yang
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Xingfei Liu
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Ya-Chao Wang
- The Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China
| | - Yu Zeng
- Department of Neurosurgery, Xiangya Hospital Central South University, Changsha 410008, China.,First Clinical Department of Changsha Medical University, Changsha 410219, China
| | - Gregori Casals
- Service of Biochemistry and Molecular Genetics, Hospital Clinic Universitari, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Carrer de Villarroel, 170, 08036 Barcelona, Spain
| | - Eudald Casals
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| |
Collapse
|
18
|
Ye X, Holland R, Wood M, Pasetka C, Palmer L, Samaridou E, McClintock K, Borisevich V, Geisbert TW, Cross RW, Heyes J. Combination treatment of mannose and GalNAc conjugated small interfering RNA protects against lethal Marburg virus infection. Mol Ther 2023; 31:269-281. [PMID: 36114672 PMCID: PMC9840110 DOI: 10.1016/j.ymthe.2022.09.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/28/2022] [Accepted: 09/12/2022] [Indexed: 02/02/2023] Open
Abstract
Marburg virus (MARV) infection results in severe viral hemorrhagic fever with mortalities up to 90%, and there is a pressing need for effective therapies. Here, we established a small interfering RNA (siRNA) conjugate platform that enabled successful subcutaneous delivery of siRNAs targeting the MARV nucleoprotein. We identified a hexavalent mannose ligand with high affinity to macrophages and dendritic cells, which are key cellular targets of MARV infection. This ligand enabled successful siRNA conjugate delivery to macrophages both in vitro and in vivo. The delivered hexa-mannose-siRNA conjugates rendered substantial target gene silencing in macrophages when supported by a mannose functionalized endosome release polymer. This hexa-mannose-siRNA conjugate was further evaluated alongside our hepatocyte-targeting GalNAc-siRNA conjugate, to expand targeting of infected liver cells. In MARV-Angola-infected guinea pigs, these platforms offered limited survival benefit when used as individual agents. However, in combination, they achieved up to 100% protection when dosed 24 h post infection. This novel approach, using two different ligands to simultaneously deliver siRNA to multiple cell types relevant to infection, provides a convenient subcutaneous route of administration for treating infection by these dangerous pathogens. The mannose conjugate platform has potential application to other diseases involving macrophages and dendritic cells.
Collapse
Affiliation(s)
- Xin Ye
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | - Richard Holland
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | - Mark Wood
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | - Chris Pasetka
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | - Lorne Palmer
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | - Eleni Samaridou
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | | | - Viktoriya Borisevich
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Thomas W Geisbert
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Robert W Cross
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - James Heyes
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada.
| |
Collapse
|
19
|
Keyvani V, Mahmoudian RA, Mollazadeh S, Kheradmand N, Ghorbani E, Khazaei M, Saeed Al-Hayawi I, Hassanian SM, Ferns GA, Avan A, Anvari K. Insight into RNA-based Therapies for Ovarian Cancer. Curr Pharm Des 2023; 29:2692-2701. [PMID: 37916491 DOI: 10.2174/0113816128270476231023052228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/14/2023] [Indexed: 11/03/2023]
Abstract
Ovarian cancer (OC) is one of the most common malignancies in women and is associated with poor outcomes. The treatment for OC is often associated with resistance to therapies and hence this has stimulated the search for alternative therapeutic approaches, including RNA-based therapeutics. However, this approach has some challenges that include RNA degradation. To solve this critical issue, some novel delivery systems have been proposed. In current years, there has been growing interest in the improvement of RNAbased therapeutics as a promising approach to target ovarian cancer and improve patient outcomes. This paper provides a practical insight into the use of RNA-based therapeutics in ovarian cancers, highlighting their potential benefits, challenges, and current research progress. RNA-based therapeutics offer a novel and targeted approach to treat ovarian cancer by exploiting the unique characteristics of RNA molecules. By targeting key oncogenes or genes responsible for drug resistance, siRNAs can effectively inhibit tumor growth and sensitize cancer cells to conventional therapies. Furthermore, messenger RNA (mRNA) vaccines have emerged as a revolutionary tool in cancer immunotherapy. MRNA vaccines can be designed to encode tumor-specific antigens, stimulating the immune system to distinguish and eliminate ovarian cancer cells. A nano-based delivery platform improves the release of loaded RNAs to the target location and reduces the off-target effects. Additionally, off-target effects and immune responses triggered by RNA molecules necessitate careful design and optimization of these therapeutics. Several preclinical and clinical researches have shown promising results in the field of RNA-based therapeutics for ovarian cancer. In a preclinical study, siRNA-mediated silencing of the poly (ADP-ribose) polymerase 1 (PARP1) gene, involved in DNA repair, sensitized ovarian cancer cells to PARP inhibitors, leading to enhanced therapeutic efficacy. In clinical trials, mRNA-based vaccines targeting tumor-associated antigens have demonstrated safety and efficacy in stimulating immune responses in ovarian cancer patients. In aggregate, RNA-based therapeutics represent a promising avenue for the therapy of ovarian cancers. The ability to specifically target oncogenes or stimulate immune responses against tumor cells holds great potential for improving patient outcomes. However, further research is needed to address challenges related to delivery, permanence, and off-target effects. Clinical trials assessing the care and effectiveness of RNAbased therapeutics in larger patient cohorts are warranted. With continued advancements in the field, RNAbased therapeutics have the potential to develop the management of ovarian cancer and provide new hope for patients.
Collapse
Affiliation(s)
- Vahideh Keyvani
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reihaneh Alsadat Mahmoudian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Nahid Kheradmand
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Ghorbani
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Seyed Mahdi Hassanian
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane 4059, Australia
| | - Kazem Anvari
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
20
|
Vinciguerra D, Gelb MB, Maynard HD. Synthesis and Application of Trehalose Materials. JACS AU 2022; 2:1561-1587. [PMID: 35911465 PMCID: PMC9327084 DOI: 10.1021/jacsau.2c00309] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Trehalose is a naturally occurring, nonreducing disaccharide that is widely used in the biopharmaceutical, food, and cosmetic industries due to its stabilizing and cryoprotective properties. Over the years, scientists have developed methodologies to synthesize linear polymers with trehalose units either in the polymer backbone or as pendant groups. These macromolecules provide unique properties and characteristics, which often outperform trehalose itself. Additionally, numerous reports have focused on the synthesis and formulation of materials based on trehalose, such as nanoparticles, hydrogels, and thermoset networks. Among many applications, these polymers and materials have been used as protein stabilizers, as gene delivery systems, and to prevent amyloid aggregate formation. In this Perspective, recent developments in the synthesis and application of trehalose-based linear polymers, hydrogels, and nanomaterials are discussed, with a focus on utilization in the biomedical field.
Collapse
Affiliation(s)
- Daniele Vinciguerra
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood
Plaza, Los Angeles, California 90095-1569, United States
| | - Madeline B. Gelb
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood
Plaza, Los Angeles, California 90095-1569, United States
| | - Heather D. Maynard
- Department
of Chemistry and Biochemistry, University
of California, Los Angeles, 607 Charles E. Young Drive East, Los Angeles, California 90095-1569, United States
- California
NanoSystems Institute, University of California,
Los Angeles, 570 Westwood
Plaza, Los Angeles, California 90095-1569, United States
| |
Collapse
|
21
|
Arshad R, Fatima I, Sargazi S, Rahdar A, Karamzadeh-Jahromi M, Pandey S, Díez-Pascual AM, Bilal M. Novel Perspectives towards RNA-Based Nano-Theranostic Approaches for Cancer Management. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:3330. [PMID: 34947679 PMCID: PMC8708502 DOI: 10.3390/nano11123330] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 11/24/2021] [Accepted: 12/05/2021] [Indexed: 12/19/2022]
Abstract
In the fight against cancer, early diagnosis is critical for effective treatment. Traditional cancer diagnostic technologies, on the other hand, have limitations that make early detection difficult. Therefore, multi-functionalized nanoparticles (NPs) and nano-biosensors have revolutionized the era of cancer diagnosis and treatment for targeted action via attaching specified and biocompatible ligands to target the tissues, which are highly over-expressed in certain types of cancers. Advancements in multi-functionalized NPs can be achieved via modifying molecular genetics to develop personalized and targeted treatments based on RNA interference. Modification in RNA therapies utilized small RNA subunits in the form of small interfering RNAs (siRNA) for overexpressing the specific genes of, most commonly, breast, colon, gastric, cervical, and hepatocellular cancer. RNA-conjugated nanomaterials appear to be the gold standard for preventing various malignant tumors through focused diagnosis and delivering to a specific tissue, resulting in cancer cells going into programmed death. The latest advances in RNA nanotechnology applications for cancer diagnosis and treatment are summarized in this review.
Collapse
Affiliation(s)
- Rabia Arshad
- Faculty of Pharmacy, University of Lahore, Lahore 45320, Pakistan;
| | - Iqra Fatima
- Department of Pharmacy, Quaid-i-Azam University, Islamabad 45320, Pakistan;
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 98167-43463, Iran;
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran
| | | | - Sadanand Pandey
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Korea;
| | - Ana M. Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Km. 33.6, 28805 Alcalá de Henares, Madrid, Spain
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huai’an 223003, China;
| |
Collapse
|