151
|
Menichetti A, Mordini D, Montalti M. Penetration of Microplastics and Nanoparticles Through Skin: Effects of Size, Shape, and Surface Chemistry. J Xenobiot 2024; 15:6. [PMID: 39846538 PMCID: PMC11755607 DOI: 10.3390/jox15010006] [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: 10/31/2024] [Revised: 12/23/2024] [Accepted: 12/25/2024] [Indexed: 01/24/2025] Open
Abstract
Skin represents an effective barrier against the penetration of external agents into the human body. Nevertheless, recent research has shown that small particles, especially in the nanosized range, can not only penetrate through the skin but also work as vectors to transport active molecules such as contrast agents or drugs. This knowledge has opened new perspectives on nanomedicine and controlled drug delivery. On the other hand, micro- and nanoplastics represent a form of emerging pollutants, and their concentration in the environment has been reported to drastically increase in the last years. The possible penetration of these particles through the skin has become a major concern for human health. If the actual primary toxicity of these materials is still debated, their possible role in the transport of toxic molecules through the skin, originating as secondary toxicity, is surely alarming. In this review paper, we analyze and critically discuss the most recent scientific publications to underline how these two processes, (i) the controlled delivery of bioactive molecules by micro- and nano-structures and (ii) the unwanted and uncontrolled penetration of toxic species through the skin mediated by micro- and nanoparticles, are deeply related and their efficiency is strongly affected by the nature, size, and shape of the particles.
Collapse
Affiliation(s)
- Arianna Menichetti
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy; (A.M.); (D.M.)
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Tecnopolo di Rimini, Via Dario Campana, 71, 47922 Rimini, Italy
| | - Dario Mordini
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy; (A.M.); (D.M.)
| | - Marco Montalti
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Via Selmi 2, 40126 Bologna, Italy; (A.M.); (D.M.)
- Department of Chemistry “Giacomo Ciamician”, University of Bologna, Tecnopolo di Rimini, Via Dario Campana, 71, 47922 Rimini, Italy
| |
Collapse
|
152
|
Zhang C, Wang Y, Peng J, Wen X, Zhang Y, Li K, Du H, Hu X. Decoding trends in mRNA vaccine research: A comprehensive bibliometric study. Hum Vaccin Immunother 2024; 20:2355037. [PMID: 38813652 PMCID: PMC11141478 DOI: 10.1080/21645515.2024.2355037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/10/2024] [Indexed: 05/31/2024] Open
Abstract
BACKGROUND In recent years, infectious diseases like COVID-19 have had profound global socio-economic impacts. mRNA vaccines have gained prominence due to their rapid development, industrial adaptability, simplicity, and responsiveness to new variants. Notably, the 2023 Nobel Prize in Physiology or Medicine recognized significant contributions to mRNA vaccine research. METHODS Our study employed a comprehensive bibliometric analysis using the Web of Science Core Collection (WoSCC) database, encompassing 5,512 papers on mRNA vaccines from 2003 to 2023. We generated cooperation maps, co-citation analyses, and keyword clustering to evaluate the field's developmental history and achievements. RESULTS The analysis yielded knowledge maps highlighting countries/institutions, influential authors, frequently published and highly cited journals, and seminal references. Ongoing research hotspots encompass immune responses, stability enhancement, applications in cancer prevention and treatment, and combating infectious diseases using mRNA technology. CONCLUSIONS mRNA vaccines represent a transformative development in infectious disease prevention. This study provides insights into the field's growth and identifies key research priorities, facilitating advancements in vaccine technology and addressing future challenges.
Collapse
Affiliation(s)
- Chaobin Zhang
- Southwest Eye Hospital, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuhang Wang
- School of Basic Medicine, Capital Medical University, Beijing, China
| | - Jianding Peng
- School of Basic Medicine, Capital Medical University, Beijing, China
| | - Xiaotian Wen
- School of Basic Medicine, Capital Medical University, Beijing, China
| | - Youwen Zhang
- School of Law, City University of Hongkong, Hong Kong, China
| | - Kejun Li
- Department of Library, Chongqing Vocational Institute of Engineering, Chongqing, China
| | - Hanjian Du
- Department of Neurosurgery, Chongqing Research Center for Glioma Precision Medicine, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Xiaofei Hu
- Department of Nuclear Medicine, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
153
|
Chentoufi AA, Ulmer JB, BenMohamed L. Antigen Delivery Platforms for Next-Generation Coronavirus Vaccines. Vaccines (Basel) 2024; 13:30. [PMID: 39852809 PMCID: PMC11769099 DOI: 10.3390/vaccines13010030] [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: 10/21/2024] [Revised: 12/15/2024] [Accepted: 12/21/2024] [Indexed: 01/26/2025] Open
Abstract
The COVID-19 pandemic, caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), is in its sixth year and is being maintained by the inability of current spike-alone-based COVID-19 vaccines to prevent transmission leading to the continuous emergence of variants and sub-variants of concern (VOCs). This underscores the critical need for next-generation broad-spectrum pan-Coronavirus vaccines (pan-CoV vaccine) to break this cycle and end the pandemic. The development of a pan-CoV vaccine offering protection against a wide array of VOCs requires two key elements: (1) identifying protective antigens that are highly conserved between passed, current, and future VOCs; and (2) developing a safe and efficient antigen delivery system for induction of broad-based and long-lasting B- and T-cell immunity. This review will (1) present the current state of antigen delivery platforms involving a multifaceted approach, including bioinformatics, molecular and structural biology, immunology, and advanced computational methods; (2) discuss the challenges facing the development of safe and effective antigen delivery platforms; and (3) highlight the potential of nucleoside-modified mRNA encapsulated in lipid nanoparticles (LNP) as the platform that is well suited to the needs of a next-generation pan-CoV vaccine, such as the ability to induce broad-based immunity and amenable to large-scale manufacturing to safely provide durable protective immunity against current and future Coronavirus threats.
Collapse
Affiliation(s)
- Aziz A. Chentoufi
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA 92697, USA;
| | - Jeffrey B. Ulmer
- Department of Vaccines and Immunotherapies, TechImmune, LLC, University Lab Partners, Irvine, CA 92660, USA;
| | - Lbachir BenMohamed
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA 92697, USA;
- Department of Vaccines and Immunotherapies, TechImmune, LLC, University Lab Partners, Irvine, CA 92660, USA;
- Institute for Immunology, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
154
|
Agrawal SS, Baliga V, Londhe VY. Liposomal Formulations: A Recent Update. Pharmaceutics 2024; 17:36. [PMID: 39861685 PMCID: PMC11769406 DOI: 10.3390/pharmaceutics17010036] [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: 08/14/2024] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 01/27/2025] Open
Abstract
Liposome-based drug delivery technologies have showed potential in enhancing medication safety and efficacy. Innovative drug loading and release mechanisms highlighted in this review of next-generation liposomal formulations. Due to poor drug release kinetics and loading capacity, conventional liposomes have limited clinical use. Scientists have developed new liposomal carrier medication release control and encapsulation methods to address these limits. Drug encapsulation can be optimized by creating lipid compositions that match a drug's charge and hydrophobicity. By selecting lipids and adding co-solvents or surfactants, scientists have increased drug loading in liposomal formulations while maintaining stability. Nanotechnology has also created multifunctional liposomes with triggered release and personalized drug delivery. Surface modification methods like PEGylation and ligand conjugation can direct liposomes to disease regions, improving therapeutic efficacy and reducing off-target effects. In addition to drug loading, researchers have focused on spatiotemporal modulation of liposomal carrier medication release. Stimuli-responsive liposomes release drugs in response to bodily signals. Liposomes can be pH- or temperature-sensitive. To improve therapeutic efficacy and reduce systemic toxicity, researchers added stimuli-responsive components to liposomal membranes to precisely control drug release kinetics. Advanced drug delivery technologies like magnetic targeting and ultrasound. Pro Drug, RNA Liposomes approach may improve liposomal medication administration. Magnetic targeting helps liposomes aggregate at illness sites and improves drug delivery, whereas ultrasound-mediated drug release facilitates on-demand release of encapsulated medicines. This review also covers recent preclinical and clinical research showing the therapeutic promise of next-generation liposomal formulations for cancer, infectious diseases, neurological disorders and inflammatory disorders. The transfer of these innovative liposomal formulations from lab to clinical practice involves key difficulties such scalability, manufacturing difficulty, and regulatory limits.
Collapse
Affiliation(s)
- Surendra S. Agrawal
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (DU), Sawangi (M), Wardha 442001, Maharashtra, India;
| | - Vrinda Baliga
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, Maharashtra, India
| | - Vaishali Y. Londhe
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM’s NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, Maharashtra, India
| |
Collapse
|
155
|
Meinhard S, Erdmann F, Lucas H, Krabbes M, Krüger S, Wölk C, Mäder K. T14diLys/DOPE Liposomes: An Innovative Option for siRNA-Based Gene Knockdown? Pharmaceutics 2024; 17:25. [PMID: 39861674 PMCID: PMC11769127 DOI: 10.3390/pharmaceutics17010025] [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: 11/23/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Bringing small interfering RNA (siRNA) into the cell cytosol to achieve specific gene silencing is an attractive but also very challenging option for improved therapies. The first step for successful siRNA delivery is the complexation with a permanent cationic or ionizable compound. This protects the negatively charged siRNA and enables transfection through the cell membrane. The current study explores the performance of the innovative, ionizable lipid 2-Tetradecylhexadecanoic acid-(2-bis{[2-(2,6-diamino-1-oxohexyl)amino]ethyl}aminoethyl)-amide (T14diLys), in combination with 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), for siRNA delivery and the impact of the production method (sonication vs. extrusion) on the particle properties. METHODS Liposomes were produced either with sonication or extrusion and characterized. The extruded liposomes were combined with siRNA at different N/P ratios and investigated in terms of size zeta potential, encapsulation efficiency, lipoplex stability against RNase A, and knockdown efficiency using enhanced green fluorescent protein (eGFP)-marked colon adenocarcinoma cells. RESULTS The liposomes prepared by extrusion were smaller and had a narrower size distribution than the sonicated ones. The combination of siRNA and liposomes at a nitrogen-to-phosphate (N/P) ratio of 5 had optimal particle properties, high encapsulation efficiency, and lipoplex stability. Gene knockdown tests confirmed this assumption. CONCLUSIONS Liposomes produced with extrusion were more reproducible and provided enhanced particle properties. The physicochemical characterization and in vitro experiments showed that an N/P ratio of 5 was the most promising ratio for siRNA delivery.
Collapse
Affiliation(s)
- Sophie Meinhard
- Department of Pharmaceutical Technology, Faculty of Natural Sciences I, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle/Saale, Germany; (S.M.); (H.L.)
| | - Frank Erdmann
- Department of Pharmaceutical Pharmacology and Toxicology, Faculty of Natural Sciences I, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle/Saale, Germany;
- Research Center for Drug Therapy Halle, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle/Saale, Germany
| | - Henrike Lucas
- Department of Pharmaceutical Technology, Faculty of Natural Sciences I, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle/Saale, Germany; (S.M.); (H.L.)
- Research Center for Drug Therapy Halle, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle/Saale, Germany
| | - Maria Krabbes
- Pharmaceutical Technology, Medical Faculty, University of Leipzig, Eilenburger Strasse 15A, 04317 Leipzig, Germany; (M.K.); (C.W.)
| | - Stephanie Krüger
- Biocenter, Microscopy Unit, Martin Luther University Halle-Wittenberg, Weinbergweg 22, 06120 Halle/Saale, Germany;
| | - Christian Wölk
- Pharmaceutical Technology, Medical Faculty, University of Leipzig, Eilenburger Strasse 15A, 04317 Leipzig, Germany; (M.K.); (C.W.)
| | - Karsten Mäder
- Department of Pharmaceutical Technology, Faculty of Natural Sciences I, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120 Halle/Saale, Germany; (S.M.); (H.L.)
- Research Center for Drug Therapy Halle, Martin Luther University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle/Saale, Germany
| |
Collapse
|
156
|
Xian J, Xiao F, Zou J, Luo W, Han S, Liu Z, Chen Y, Zhu Q, Li M, Yu C, Saiding Q, Tao W, Kong N, Xie T. Elemene Hydrogel Modulates the Tumor Immune Microenvironment for Enhanced Treatment of Postoperative Cancer Recurrence and Metastases. J Am Chem Soc 2024; 146:35252-35263. [PMID: 39625467 DOI: 10.1021/jacs.4c12531] [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: 12/26/2024]
Abstract
As a representative active ingredient of traditional Chinese medicine (TCM) and a clinically approved anticancer drug, elemene (ELE) exhibits exciting potential in the antitumor field; however, appropriate drug formulations still need to be explored for specific diseases such as postoperative cancer recurrence and metastasis. Herein, we report an ELE hydrogel with controlled drug release kinetics that can allow ELE to maintain effective concentrations at local lesion sites for extended periods to enhance the bioavailability of ELE. Concretely, dopamine-conjugated hyaluronic acid is synthesized and utilized to prepare ELE nanodrug-embedded hydrogels. In a model of postoperative breast cancer recurrence and metastasis, the ELE hydrogel demonstrates a 96% inhibition rate of recurrence; in contrast, the free ELE nanodrug shows only a 65.5% inhibition rate of recurrence. Importantly, the ELE hydrogel markedly stimulates a potent antitumor immune response in the microenvironment of cancer lesions, increasing antitumor immune cells such as CD8+ T cells, CD4+ T cells, and M1-type macrophages, as well as elevating antitumor cytokines including TNF-α, IFN-γ, and IL-6. Overall, this study not only advances the field of TCM but also highlights the transformative impact of controlled-release hydrogels in improving antitumor therapy.
Collapse
Affiliation(s)
- Jing Xian
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang 311121, China
| | - Fan Xiao
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang 311121, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jianhua Zou
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Wei Luo
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Shiqi Han
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ziwei Liu
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang 311121, China
| | - Yiquan Chen
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang 311121, China
| | - Qianru Zhu
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Meng Li
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Chuao Yu
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang 311121, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Tian Xie
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| |
Collapse
|
157
|
Mukhopadhyay B, Singh S, Singh A. Utilizing nanomaterials for cancer treatment and diagnosis: an overview. DISCOVER NANO 2024; 19:215. [PMID: 39718700 DOI: 10.1186/s11671-024-04128-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 10/14/2024] [Indexed: 12/25/2024]
Abstract
Cancer is a deadly disease with complex pathophysiological nature and is the leading cause of death worldwide. Traditional diagnosis methods often detect cancer at a considerably critical stage and the conventional methods of treatment like chemotherapy, radiation therapy, targeted therapy, and immunotherapy have several limitations, multidrug resistance, cytotoxicity, and lack of specificity are a few examples. These pose substantial challenge for effective and favourable cancer treatment. The advent of nanotechnology has revolutionized the face of cancer diagnosis and treatment. Nanoparticles, which have a size range of 1-100 nm, are biocompatible and have special optical, magnetic, and electrical capabilities, less toxic, more stable, exhibit permeability and retention effect, and are used for precise targeting. There are several classes of nanoparticles each having their own sets of unique properties. NPs have played an important role in the drug delivery system, overcoming the multi-drug resistance, reducing the side-effects as seen in conventional therapeutic methods and hence able to solve the limitations of conventional methods of diagnosis and treatment. This review discusses the four major classes of nanoparticles (Lipid based NPs, Carbon NPs and Metallic NPs and Polymeric NPs): their discovery and introduction in medical field, unique properties and characteristics, advantages and disadvantages, sub-categories and characteristics of these categories, major area of application in Cancer diagnosis and treatment, and latest methodologies where these are used in cancer treatment.
Collapse
Affiliation(s)
- Bageesha Mukhopadhyay
- Department of Biomedical Engineering, School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab, 144001, India
| | - Sudhakar Singh
- Department of Biomedical Engineering, School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab, 144001, India
| | - Avtar Singh
- School of Electrical Engineering and Computing (SoEEC), Adama Science and Technology University (AS-TU), 1888, Adama, Ethiopia.
| |
Collapse
|
158
|
Flores-Prieto DE, Stabenfeldt SE. Nanoparticle targeting strategies for traumatic brain injury. J Neural Eng 2024; 21:061007. [PMID: 39622184 DOI: 10.1088/1741-2552/ad995b] [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: 05/06/2024] [Accepted: 12/02/2024] [Indexed: 12/21/2024]
Abstract
Nanoparticle (NP)-based drug delivery systems hold immense potential for targeted therapy and diagnosis of neurological disorders, overcoming the limitations of conventional treatment modalities. This review explores the design considerations and functionalization strategies of NPs for precise targeting of the brain and central nervous system. This review discusses the challenges associated with drug delivery to the brain, including the blood-brain barrier and the complex heterogeneity of traumatic brain injury. We also examine the physicochemical properties of NPs, emphasizing the role of size, shape, and surface characteristics in their interactions with biological barriers and cellular uptake mechanisms. The review concludes by exploring the options of targeting ligands designed to augment NP affinity and retention to specific brain regions or cell types. Various targeting ligands are discussed for their ability to mimic receptor-ligand interaction, and brain-specific extracellular matrix components. Strategies to mimic viral mechanisms to increase uptake are discussed. Finally, the emergence of antibody, antibody fragments, and antibody mimicking peptides are discussed as promising targeting strategies. By integrating insights from these scientific fields, this review provides an understanding of NP-based targeting strategies for personalized medicine approaches to neurological disorders. The design considerations discussed here pave the way for the development of NP platforms with enhanced therapeutic efficacy and minimized off-target effects, ultimately advancing the field of neural engineering.
Collapse
Affiliation(s)
- David E Flores-Prieto
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States of America
| | - Sarah E Stabenfeldt
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, United States of America
| |
Collapse
|
159
|
Balaraman AK, Babu MA, Moglad E, Mandaliya V, Rekha MM, Gupta S, Prasad GVS, Kumari M, Chauhan AS, Ali H, Goyal K. Exosome-mediated delivery of CRISPR-Cas9: A revolutionary approach to cancer gene editing. Pathol Res Pract 2024; 266:155785. [PMID: 39708520 DOI: 10.1016/j.prp.2024.155785] [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: 11/13/2024] [Revised: 12/08/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Several molecular strategies based on targeted gene delivery systems have been developed in recent years; however, the CRISPR-Cas9 technology introduced a new era of targeted gene editing, precisely modifying oncogenes, tumor suppressor genes, and other regulatory genes involved in carcinogenesis. However, efficiently and safely delivering CRISPR-Cas9 to cancer cells across the cell membrane and the nucleus is still challenging. Using viral vectors and nanoparticles presents issues of immunogenicity, off-target effects, and low targeting affinity. Naturally, extracellular vesicles called exosomes have garnered the most attention as delivery vehicles in oncology-related CRISPR-Cas9 calls due to their biocompatibility, loading capacity, and inherent targeting features. The following review discusses the current progress in using exosomes to deliver CRISPR-Cas9 components, the approaches to load the CRISPR components into exosomes, and the modification of exosomes to increase stability and tumor-targeted delivery. We discuss the latest strategies in targeting recently accomplished in the exosome field, including modifying the surface of exosomes to enhance their internalization by cancer cells, as well as the measures taken to overcome the impacts of TME on delivery efficiency. Focusing on in vitro and in vivo experimentation, this review shows that exosome-mediated CRISPR-Cas9 can potentially treat cancer types, including pancreatic, lymphoma, and leukemia, for given gene targets. This paper compares exosome-mediated delivery and conventional vectors regarding safety, immune response, and targeting ability. Last but not least, we present the major drawbacks and potential development of the seemingly promising field of exosome engineering in gene editing, with references to CRISPR technologies and applications that may help make the target exosomes therapeutic in oncology.
Collapse
Affiliation(s)
- Ashok Kumar Balaraman
- Research and Enterprise, University of Cyberjaya, Persiaran Bestari, Cyber 11, Cyberjaya, Selangor 63000, Malaysia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA UNIVERSITY, Mathura, UP 281406, India
| | - Ehssan Moglad
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Viralkumar Mandaliya
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat 360003, India
| | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Sofia Gupta
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab 140307, India
| | - G V Siva Prasad
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh 531162, India
| | - Mukesh Kumari
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - Ashish Singh Chauhan
- Uttaranchal Institute of Pharmaceutical Sciences, Division of research and innovation, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India.
| |
Collapse
|
160
|
Pavlov RV, Akimov SA, Dashinimaev EB, Bashkirov PV. Boosting Lipofection Efficiency Through Enhanced Membrane Fusion Mechanisms. Int J Mol Sci 2024; 25:13540. [PMID: 39769303 PMCID: PMC11677079 DOI: 10.3390/ijms252413540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Gene transfection is a fundamental technique in the fields of biological research and therapeutic innovation. Due to their biocompatibility and membrane-mimetic properties, lipid vectors serve as essential tools in transfection. The successful delivery of genetic material into the cytoplasm is contingent upon the fusion of the vector and cellular membranes, which enables hydrophilic polynucleic acids to traverse the hydrophobic barriers of two intervening membranes. This review examines the critical role of membrane fusion in lipofection efficiency, with a particular focus on the molecular mechanisms that govern lipoplex-membrane interactions. This analysis will examine the key challenges inherent to the fusion process, from achieving initial membrane proximity to facilitating final content release through membrane remodeling. In contrast to viral vectors, which utilize specialized fusion proteins, lipid vectors necessitate a strategic formulation and environmental optimization to enhance their fusogenicity. This review discusses recent advances in vector design and fusion-promoting strategies, emphasizing their potential to improve gene delivery yield. It highlights the importance of understanding lipoplex-membrane fusion mechanisms for developing next-generation delivery systems and emphasizes the need for continued fundamental research to advance lipid-mediated transfection technology.
Collapse
Affiliation(s)
- Rais V. Pavlov
- Research Institute for Systems Biology and Medicine, 18 Nauchniy Proezd, Moscow 117246, Russia
| | - Sergey A. Akimov
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, Moscow 119071, Russia;
| | - Erdem B. Dashinimaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia;
| | - Pavel V. Bashkirov
- Research Institute for Systems Biology and Medicine, 18 Nauchniy Proezd, Moscow 117246, Russia
| |
Collapse
|
161
|
Sun X, Tian T, Lian Y, Cui Z. Current Advances in Viral Nanoparticles for Biomedicine. ACS NANO 2024; 18:33827-33863. [PMID: 39648920 DOI: 10.1021/acsnano.4c13146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Viral nanoparticles (VNPs) have emerged as crucial tools in the field of biomedicine. Leveraging their biological and physicochemical properties, VNPs exhibit significant advantages in the prevention, diagnosis, and treatment of human diseases. Through techniques such as chemical bioconjugation, infusion, genetic engineering, and encapsulation, these VNPs have been endowed with multifunctional capabilities, including the display of functional peptides or proteins, encapsulation of therapeutic drugs or inorganic particles, integration with imaging agents, and conjugation with bioactive molecules. This review provides an in-depth analysis of VNPs in biomedicine, elucidating their diverse types, distinctive features, production methods, and complex design principles behind multifunctional VNPs. It highlights recent innovative research and various applications, covering their roles in imaging, drug delivery, therapeutics, gene delivery, vaccines, immunotherapy, and tissue regeneration. Additionally, the review provides an assessment of their safety and biocompatibility and discusses challenges and future opportunities in the field, underscoring the vast potential and evolving nature of VNP research.
Collapse
Affiliation(s)
- Xianxun Sun
- School of Life Sciences, Jianghan University, Wuhan 430056, China
| | - Tao Tian
- School of Life Sciences, Jianghan University, Wuhan 430056, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Yindong Lian
- School of Life Sciences, Jianghan University, Wuhan 430056, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| | - Zongqiang Cui
- State Key Laboratory of Virology, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan 430071, China
| |
Collapse
|
162
|
Cheng J, Jian L, Chen Z, Li Z, Yu Y, Wu Y. In Vivo Delivery Processes and Development Strategies of Lipid Nanoparticles. Chembiochem 2024; 25:e202400481. [PMID: 39101874 DOI: 10.1002/cbic.202400481] [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: 05/31/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 08/06/2024]
Abstract
Lipid nanoparticles (LNPs) represent an advanced and highly efficient delivery system for RNA molecules, demonstrating exceptional biocompatibility and remarkable delivery efficiency. This is evidenced by the clinical authorization of three LNP formulations: Patisiran, BNT162b2, and mRNA-1273. To further maximize the efficacy of RNA-based therapy, it is imperative to develop more potent LNP delivery systems that can effectively protect inherently unstable and negatively charged RNA molecules from degradation by nucleases, while facilitating their cellular uptake into target cells. Therefore, this review presents feasible strategies commonly employed for the development of efficient LNP delivery systems. The strategies encompass combinatorial chemistry for large-scale synthesis of ionizable lipids, rational design strategy of ionizable lipids, functional molecules-derived lipid molecules, the optimization of LNP formulations, and the adjustment of particle size and charge property of LNPs. Prior to introducing these developing strategies, in vivo delivery processes of LNPs, a crucial determinant influencing the clinical translation of LNP formulations, is described to better understand how to develop LNP delivery systems.
Collapse
Affiliation(s)
- Jiashun Cheng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Lina Jian
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhaolin Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhuoyuan Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yaobang Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yihang Wu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
163
|
Khan S, Do CW, Ho EA. Recent updates on drug delivery approaches for improved ocular delivery with an insight into nanostructured drug delivery carriers for anterior and posterior segment disorders. Drug Deliv Transl Res 2024:10.1007/s13346-024-01756-x. [PMID: 39674854 DOI: 10.1007/s13346-024-01756-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 12/16/2024]
Abstract
Ocular diseases have a major impact on patient's vision and quality of life, with approximately 2.2 billion people have visual impairment worldwide according to the findings from the World Health Organization (WHO). The eye is a complex organ with unique morphology and physiology consisting of numerous ocular barriers which hinders the entry of exogenous substances and impedes drug absorption. This in turn has a substantial impact on effective drug delivery to treat ocular diseases, especially intraocular disorders which has consistently presented a challenge to eye care professionals. The most common method of delivering medications to the eye is topical instillation of eye drops. Although this approach is a viable option for treating many ocular diseases remains a major challenge for the effective treatment of posterior ocular conditions. Up till now, incessant efforts have been committed to design innovative drug delivery systems with the hopes of potential clinical application. Modern developments in nanocarrier's technology present a potential chance to overcome these obstacles by enabling targeted delivery of the loaded medication to the eyes with improved solubility, delayed release, higher penetration and increased retention. This review covers the anatomy of eye with associated ocular barriers, ocular diseases and administration routes. In addition it primarily focuses on the latest progress and contemporary applications of ophthalmic formulations providing specific insight on nanostructured drug delivery carriers reported over the past 5 years highlighting their values in achieving efficient ocular drug delivery to both anterior and posterior segments. Most importantly, we outlined in this review the macro and nanotechnology based ophthalmic drug formulations that are being patented or marketed so far for treating ocular diseases. Finally, based on current trends and therapeutic concepts, we highlighted the challenges faced by novel ocular drug delivery systems and provided prospective future developments for further research in these directions. We hope that this review will serve as a source of motivation and ideas for formulation scientists in improving the design of innovative ophthalmic formulations.
Collapse
Affiliation(s)
- Samiullah Khan
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, Hong Kong
| | - Chi-Wai Do
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, Hong Kong.
- School of Optometry, The Hong Kong Polytechnic University, Hung Hom, Hong Kong.
| | - Emmanuel A Ho
- Centre for Eye and Vision Research (CEVR), 17W Hong Kong Science Park, Hong Kong, Hong Kong.
- School of Pharmacy, University of Waterloo, Waterloo, Canada.
- Waterloo Institute for Nanotechnology, Waterloo, Canada.
| |
Collapse
|
164
|
Liu YL, Liao TY, Ho KW, Liu ES, Huang BC, Hong ST, Hsieh YC, Chang MS, Wu BT, Chen FM, Roffler SR, Chen CY, Yang YC, Cheng TL. Impact of Pre-existing Anti-polyethylene Glycol Antibodies on the Pharmacokinetics and Efficacy of a COVID-19 mRNA Vaccine (Comirnaty) In Vivo. Biomater Res 2024; 28:0112. [PMID: 39665081 PMCID: PMC11633857 DOI: 10.34133/bmr.0112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 10/15/2024] [Accepted: 10/26/2024] [Indexed: 12/13/2024] Open
Abstract
The presence of anti-polyethylene glycol (anti-PEG) antibodies can hinder the therapeutic efficacy of PEGylated drugs. With the widespread use of a PEGylated coronavirus disease 2019 (COVID-19) messenger RNA vaccine (Comirnaty), the impact of pre-existing anti-PEG antibodies on vaccine potency has become a point of debate. To investigate this, we established mouse models with pre-existing anti-PEG antibodies and divided them into 3 groups: group 1 with anti-PEG immunoglobulin G + immunoglobulin M concentrations of 0.76 to 27.41 μg/ml, group 2 with concentrations of 31.27 to 99.52 μg/ml, and a naïve group with no detectable anti-PEG antibodies. Results indicated that anti-spike antibody concentrations significantly decreased in group 1 and group 2 after the 2nd vaccine dose compared to those in the naïve group. Spearman's rank correlation analysis demonstrated a negative relationship between anti-spike antibody production and anti-PEG antibody levels at both the 2nd and 3rd doses (2nd dose: ρ = -0.5296, P = 0.0031; 3rd dose: ρ = -0.387, P = 0.0381). Additionally, spike protein concentrations were 31.4-fold and 46.6-fold lower in group 1 and group 2, respectively, compared to those in the naïve group at 8 h postvaccination. The concentration of complement C3a in group 2 was significantly higher than that in the naïve group after the 3rd dose. These findings confirm that pre-existing anti-PEG antibodies diminish vaccine efficacy, alter pharmacokinetics, and elevate complement activation. Therefore, detecting pre-existing anti-PEG antibodies is crucial for optimizing vaccine efficacy, ensuring patient safety, and developing improved therapeutic strategies.
Collapse
Affiliation(s)
- Yen-Ling Liu
- Graduate Institute of Medicine, College of Medicine,
Kaohsiung Medical University, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center,
Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tzu-Yi Liao
- Graduate Institute of Medicine, College of Medicine,
Kaohsiung Medical University, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center,
Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kai-Wen Ho
- Drug Development and Value Creation Research Center,
Kaohsiung Medical University, Kaohsiung, Taiwan
| | - En-Shuo Liu
- Graduate Institute of Medicine, College of Medicine,
Kaohsiung Medical University, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center,
Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bo-Cheng Huang
- Drug Development and Value Creation Research Center,
Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shih-Ting Hong
- Graduate Institute of Medicine, College of Medicine,
Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuan-Chin Hsieh
- School of Medicine for International Students,
I-Shou University, Kaohsiung, Taiwan
| | - Mu-Shen Chang
- PhD Program in Life Science, College of Life Science,
Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bing-Tsung Wu
- Graduate Institute of Medicine, College of Medicine,
Kaohsiung Medical University, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center,
Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Fang-Ming Chen
- Drug Development and Value Creation Research Center,
Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Radiation Oncology, Faculty of Medicine, College of Medicine,
Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Steve R. Roffler
- Graduate Institute of Medicine, College of Medicine,
Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Biomedical Sciences,
Academia Sinica, Taipei, Taiwan
| | - Chiao-Yun Chen
- Drug Development and Value Creation Research Center,
Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Imaging,
Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yuan-Chieh Yang
- Department of Laboratory Medicine,
Kaohsiung Municipal United Hospital, Kaohsiung, Taiwan
| | - Tian-Lu Cheng
- Graduate Institute of Medicine, College of Medicine,
Kaohsiung Medical University, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center,
Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biomedical Science and Environmental Biology,
Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
165
|
Grigoriev V, Korzun T, Moses AS, Jozic A, Zhu X, Kim J, Newton S, Eygeris Y, Diba P, Sattler AL, Levasseur PR, Olson B, Le N, Singh P, Sharma KS, Goo YT, Mamnoon B, Raitmayr C, Mesquita Souza AP, Taratula OR, Sahay G, Marks DL, Taratula O. Targeting Metastasis in Head and Neck Squamous Cell Carcinoma Using Follistatin mRNA Lipid Nanoparticles. ACS NANO 2024; 18:33330-33347. [PMID: 39569532 PMCID: PMC11916978 DOI: 10.1021/acsnano.4c06930] [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: 11/22/2024]
Abstract
Metastatic progression significantly reduces survival rates and complicates treatment strategies in various cancers. Our study introduces an mRNA therapy for metastasis inhibition by targeting activin A overexpression, a pivotal driver of metastasis and cachexia. Utilizing follistatin mRNA lipid nanoparticles, we effectively downregulated activin A both locally in the tumor environment and systemically. This led to a reduction in tumor burden and suppression of metastatic spread in a murine head and neck squamous cell carcinoma model. Treated mice exhibited minimal metastatic occurrence compared to controls. Additionally, our therapy preserved the cross-sectional area of muscle fibers and adipose tissues, combating the muscle and fat wasting typically observed in cancer-associated cachexia. The therapy also demonstrated a favorable safety profile, underscoring its potential for clinical translation. By integrating metastasis-suppressing and cachexia-alleviating mechanisms, our approach represents a promising advancement in comprehensive cancer management. Considering the widespread upregulation of activin A in many cancer types, our therapy holds considerable potential for application across a broad spectrum of oncologic treatments.
Collapse
Affiliation(s)
- Vladislav Grigoriev
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Tetiana Korzun
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Avenue, Portland, Oregon 97239, United States
- Medical Scientist Training Program, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Abraham S Moses
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Antony Jozic
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Xinxia Zhu
- Papé Family Pediatric Research Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Jeonghwan Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
- College of Pharmacy, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Samuel Newton
- Papé Family Pediatric Research Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Yulia Eygeris
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Parham Diba
- Medical Scientist Training Program, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Ariana L Sattler
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, 2720 S Moody Ave, Portland, Oregon 97201, United States
| | - Peter R Levasseur
- Papé Family Pediatric Research Institute, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
| | - Brennan Olson
- Medical Scientist Training Program, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, United States
- Department of Otolaryngology-Head and Neck Surgery, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, United States
| | - Ngoc Le
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Prem Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Kongbrailatpam Shitaljit Sharma
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Yoon Tae Goo
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Babak Mamnoon
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Constanze Raitmayr
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Ana Paula Mesquita Souza
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Olena R Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
| | - Daniel L Marks
- Endevica Bio, 1935 Techny Road, Northbrook, Illinois 60062, United States
| | - Oleh Taratula
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, 2730 SW Moody Avenue, Portland, Oregon 97201, United States
- Department of Biomedical Engineering, Oregon Health and Science University, 3303 SW Bond Avenue, Portland, Oregon 97239, United States
| |
Collapse
|
166
|
Han Y, Wang M, Chen Y, Ouyang D, Zheng Y, Hu Y. Profiling patent compounds in lipid nanoparticle formulations of siRNA. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102362. [PMID: 39554995 PMCID: PMC11565460 DOI: 10.1016/j.omtn.2024.102362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 10/16/2024] [Indexed: 11/19/2024]
Abstract
Lipid nanoparticles (LNPs) have emerged as a prominent delivery system for nucleic acid drugs, attracting significant attention, especially through the successful development of several commercial products. As a key component in LNPs, cationic lipids have long served as a key technical barrier to block competitors by building up a complex patent thicket. However, there have been few studies as yet that have comprehensively analyzed the patented compounds in LNP formulations, despite a large number of technical reviews and original articles. In this context, this study focuses on analyzing the macroscopic landscapes and microscopic molecular characteristics of LNP patents, aiming to provide a valuable reference for researchers and developers in making informed technological and commercial decisions. By mining 2,994 patents, 265 formulas, 7,674 compounds, and 28,789 fragments, this work sketches the empirical golden ratio of lipid materials in LNP formulation, discloses the advanced technology in the formulation, characterizes high-frequency fragments of heads, linkers and tails in both novel cationic lipids as well as targeting lipids, and establishes a virtual focus library of LNP materials.
Collapse
Affiliation(s)
- Yunfeng Han
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Mengyang Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Yu Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
| | - Defang Ouyang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- Centre for Pharmaceutical Regulatory Sciences, University of Macau, Macao SAR, China
- DPM, Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- DPS, Faculty of Health Sciences, University of Macau, Macao SAR, China
| | - Yuanjia Hu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China
- Centre for Pharmaceutical Regulatory Sciences, University of Macau, Macao SAR, China
- DPM, Faculty of Health Sciences, University of Macau, Macao SAR, China
| |
Collapse
|
167
|
Tang B, Huang R, Ma W. Advances in nanotechnology-based approaches for the treatment of head and neck squamous cell carcinoma. RSC Adv 2024; 14:38668-38688. [PMID: 39654926 PMCID: PMC11626385 DOI: 10.1039/d4ra07193j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC), one of the most common types of cancers occurring in the head and neck region, is often associated with high mortality rates due to its invasiveness and morbidity. The mainstream treatment methods in clinical settings, including surgery, chemotherapy, and radiotherapy, may cause poor overall survival rate and prognosis, with issues such as drug resistance, damage to adjacent healthy tissues, and potential recurrences. Other treatment approaches such as immunotherapy, photodynamic therapy (PDT), and photothermal therapy (PPT) also suffer from inefficient tumor targeting and suboptimal therapeutic outcomes. Early detection is vital for HNSCC patients, but it is always limited by insensitivity and confusing clinical manifestations. Hence, it is highly desirable to develop optimized therapeutic and diagnostic strategies. With the boom in nanomaterials, nanotechnology-conducted HNSCC therapy has attracted widespread attention. Nanoparticles (NPs) are distinguished by their unique morphology and superior physicochemical property, and some can exhibit direct antitumor activity, while others serve as promising candidates for drug delivery. In addition, NPs offer the potential for structural modification for drug delivery and tumor targeting, enabling specific delivery to tumor cells through conjugation with biomarker ligands and improving cargo biocompatibility. This work reviews current therapies and diagnosis methods for HNSCC, highlights the characteristics of the major NPs, surveys their uses and advantages in the treatment of HNSCC, and discusses the obstacles and prospects in clinical applications, aiming to enlighten future research directions for nanotechnology-based therapy for HNSCC.
Collapse
Affiliation(s)
- Bicai Tang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu Sichuan 610041 China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials Chengdu Sichuan 610041 China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| | - Rui Huang
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu Sichuan 610041 China
| | - Wenjuan Ma
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu Sichuan 610041 China
- Sichuan Provincial Engineering Research Center of Oral Biomaterials Chengdu Sichuan 610041 China
- Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University Chengdu 610041 China
| |
Collapse
|
168
|
Büber E, Yaadav R, Schröder T, Franquelim HG, Tinnefeld P. DNA Origami Vesicle Sensors with Triggered Single-Molecule Cargo Transfer. Angew Chem Int Ed Engl 2024; 63:e202408295. [PMID: 39248369 PMCID: PMC11586697 DOI: 10.1002/anie.202408295] [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: 05/01/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/10/2024]
Abstract
Interacting with living systems typically involves the ability to address lipid membranes of cellular systems. The first step of interaction of a nanorobot with a cell will thus be the detection of binding to a lipid membrane. Utilizing DNA origami, we engineered a biosensor with single-molecule Fluorescence Resonance Energy Transfer (smFRET) as transduction mechanism for precise lipid vesicle detection and cargo delivery. The system hinges on a hydrophobic ATTO647N modified single-stranded DNA (ssDNA) leash, protruding from a DNA origami nanostructure. In a vesicle-free environment, the ssDNA coils, ensuring high FRET efficiency. Upon vesicle binding to cholesterol anchors on the DNA origami, hydrophobic ATTO647N induces the ssDNA to stretch towards the lipid bilayer, reducing FRET efficiency. As the next step, the sensing strand serves as molecular cargo that can be transferred to the vesicle through a triggered strand displacement reaction. Depending on the number of cholesterols on the displacer strands, we either induce a diffusive release of the fluorescent load towards neighboring vesicles or a stoichiometric release of a single cargo-unit to the vesicle on the nanosensor. Ultimately, our multi-functional liposome interaction and detection platform opens up pathways for innovative biosensing applications and stoichiometric loading of vesicles with single-molecule control.
Collapse
Affiliation(s)
- Ece Büber
- Department of ChemistryCenter for NanoScienceLudwig-Maximilians-UniversityButenandtstraße 5–1381377MunichGermany
| | - Renukka Yaadav
- Department of ChemistryCenter for NanoScienceLudwig-Maximilians-UniversityButenandtstraße 5–1381377MunichGermany
| | - Tim Schröder
- Department of ChemistryCenter for NanoScienceLudwig-Maximilians-UniversityButenandtstraße 5–1381377MunichGermany
| | - Henri G. Franquelim
- Interfaculty Centre for Bioactive MatterLeipzig UniversityDeutscher Platz 5 (BBZ)04103LeipzigGermany
| | - Philip Tinnefeld
- Department of ChemistryCenter for NanoScienceLudwig-Maximilians-UniversityButenandtstraße 5–1381377MunichGermany
| |
Collapse
|
169
|
Li X, Lai Y, Wan G, Zou J, He W, Yang P. Approved natural products-derived nanomedicines for disease treatment. Chin J Nat Med 2024; 22:1100-1116. [PMID: 39725511 DOI: 10.1016/s1875-5364(24)60726-0] [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: 08/12/2024] [Indexed: 12/28/2024]
Abstract
In recent years, there has been an increasing emphasis on exploring innovative drug delivery approaches due to the limitations of conventional therapeutic strategies, such as inadequate drug targeting, insufficient therapeutic efficacy, and significant adverse effects. Nanomedicines have emerged as a promising solution with notable advantages, including extended drug circulation, targeted delivery, and improved bioavailability, potentially enhancing the clinical treatment of various diseases. Natural products/materials-derived nanomedicines, characterized by their natural therapeutic efficacy, superior biocompatibility, and safety profile, play a crucial role in nanomedicine-based treatments. This review provides a comprehensive overview of currently approved natural products-derived nanomedicines, emphasizing the essential properties of natural products-derived drug carriers, their applications in clinical diagnosis and treatment, and the current therapeutic potential and challenges. The aim is to offer guidance for the application and further development of these innovative therapeutic approaches.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Yaoyao Lai
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Guanghan Wan
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China.
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 2111198, China.
| |
Collapse
|
170
|
Liu Y, Xu Q, Liu Y, Cao S, Luo J, Zheng Z, Zhou J, Lu X, Zhang L, Tan Y, Chen Q, Zuo D. Hepatocyte-Targeted Lipid Nanoparticle Delivery of HERC2 Plasmid Controls Drug-Induced Hepatotoxicity by Limiting β-Catenin-Regulated CYP2E1 Expression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401633. [PMID: 39440550 PMCID: PMC11633468 DOI: 10.1002/advs.202401633] [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: 02/15/2024] [Revised: 10/06/2024] [Indexed: 10/25/2024]
Abstract
Understanding the molecular mechanisms that bridge hepatic inflammation and liver injury is crucial for developing effective therapeutic strategies for drug-induced liver injury (DILI) management. HECT domain and RCC1-like domain 2 (HERC2) belongs to the large Herc family of ubiquitin E3 ligases, which are implicated in tissue development and inflammation. The observation reveals a pronounced HERC2 expression in specific hepatocyte subsets that proliferate in response to DILI in humans, prompting an investigation into the role of HERC2 in distinct DILI progression. Under the APAP challenge, liver-specific HERC2-deficient mice suffer more severe liver damage. Integrated single-cell RNA sequencing analysis unveils a negative correlation between HERC2 and CYP2E1, a vital metabolic enzyme for xenobiotics, in hepatocytes from APAP-challenged mice. Mechanistically, HERC2 interacts with β-catenin to promote its ubiquitination, thereby governing CYP2E1 transcriptional regulation. Targeted hepatic delivery of lipid nanoparticle-encapsulated HERC2-overexpressing plasmid markedly reduces liver damage caused by APAP overdose. Collectively, these findings elucidate a previously unrecognized protective role of HERC2 in protecting against acute liver injury associated with drug metabolism disorders, highlighting its potential as a therapeutic target in treating DILI.
Collapse
Affiliation(s)
- Yunzhi Liu
- Institute of Molecular ImmunologySchool of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouGuangdong510515China
- Clinical Oncology CenterShenzhen Key Laboratory for cancer metastasis and personalized therapyThe University of Hong Kong‐Shenzhen HospitalShenzhenGuangdong518053China
- Shenzhen Institute of Advanced TechnologyChinese Academy of SciencesShenzhen518055China
| | - Qishan Xu
- Institute of Molecular ImmunologySchool of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouGuangdong510515China
- Department of Precision LaboratoryAffiliated Hospital of Guangdong Medical UniversityZhanjiangGuangdong510180China
| | - Yan Liu
- Institute of Molecular ImmunologySchool of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouGuangdong510515China
| | - Sihang Cao
- Institute of Molecular ImmunologySchool of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouGuangdong510515China
- Department of MicrobiologyLi Ka Shing Faculty of MedicineThe University of Hong KongPokfulamHong Kong SAR999077China
| | - Jialiang Luo
- Institute of Molecular ImmunologySchool of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Province Key Laboratory of ProteomicsDepartment of ImmunologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Zhuojun Zheng
- Institute of Molecular ImmunologySchool of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouGuangdong510515China
- Medical Research InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510080China
- Guangdong Province Key Laboratory of Immune Regulation and ImmunotherapySchool of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouGuangdong510515China
| | - Jia Zhou
- Guangdong Province Key Laboratory of ProteomicsDepartment of ImmunologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Xiao Lu
- Guangdong Province Key Laboratory of ProteomicsDepartment of ImmunologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Liyun Zhang
- Guangdong Province Key Laboratory of ProteomicsDepartment of ImmunologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Yanan Tan
- Clinical Oncology CenterShenzhen Key Laboratory for cancer metastasis and personalized therapyThe University of Hong Kong‐Shenzhen HospitalShenzhenGuangdong518053China
- Advanced Energy Science and Technology Guangdong LaboratoryHuizhouGuangdong516001China
| | - Qingyun Chen
- Medical Research InstituteGuangdong Provincial People's HospitalGuangdong Academy of Medical SciencesSouthern Medical UniversityGuangzhouGuangdong510080China
| | - Daming Zuo
- Institute of Molecular ImmunologySchool of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouGuangdong510515China
- Guangdong Province Key Laboratory of Immune Regulation and ImmunotherapySchool of Laboratory Medicine and BiotechnologySouthern Medical UniversityGuangzhouGuangdong510515China
| |
Collapse
|
171
|
Wang Y, Sun C, Liu Z, Zhang S, Gao K, Yi F, Zhou W, Liu H. Nanoengineered Endocytic Biomaterials for Stem Cell Therapy. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202410714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Indexed: 01/05/2025]
Abstract
AbstractStem cells, ideal for the tissue repair and regeneration, possess extraordinary capabilities of multidirectional differentiation and self‐renewal. However, the limited spontaneous differentiation potential makes it challenging to harness them for tissue repair without external intervention. Although conventional approaches using biomolecules, small organic molecules, and ions have shown specific and effective functions, they face challenges such as in vivo diffusion and degradation, poor internalization, and side effects on adjacent cells. Nanoengineered biomaterials offer a solution by solidifying and nanosizing these soluble regulating molecules and ions, facilitating their uptake by stem cells. Once inside lysosomes, these nanoparticles release their contents in a controlled “molecule or ion storm,” efficiently altering the intracellular biological and chemical microenvironment to tune the differentiation of stem cells. This newly emerged approach for regulating stem cell fate has attracted much attention in recent years. This method has shown promising results and is poised to enhance clinical stem cell therapy. This review provides an overview of the design principles for nanoengineered biomaterials, discusses the categories and characteristics of nanoparticles, summarizes the application of nanoparticles in tissue repair and regeneration, and discusses the direction of nanoparticle‐enhanced stem cell therapy and prospects for its clinical application in regenerative medicine.
Collapse
Affiliation(s)
- Yingxue Wang
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Chunhui Sun
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Zhaoying Liu
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Shengmin Zhang
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Ke Gao
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
| | - Fan Yi
- School of Basic Medical Sciences Shandong University Jinan 250012 P. R. China
| | - Wenjuan Zhou
- School of Basic Medical Sciences Shandong University Jinan 250012 P. R. China
| | - Hong Liu
- Institute for Advanced Interdisciplinary Research (iAIR) University of Jinan Jinan 250022 P. R. China
- State Key Laboratory of Crystal Materials Shandong University Jinan 250100 P. R. China
| |
Collapse
|
172
|
Qi L, Hong S, Zhao T, Yan J, Ge W, Wang J, Fang X, Jiang W, Shen SG, Zhang L. DNA Tetrahedron Delivering miR-21-5p Promotes Senescent Bone Defects Repair through Synergistic Regulation of Osteogenesis and Angiogenesis. Adv Healthc Mater 2024; 13:e2401275. [PMID: 38979868 DOI: 10.1002/adhm.202401275] [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: 04/07/2024] [Revised: 06/08/2024] [Indexed: 07/10/2024]
Abstract
Compromised osteogenesis and angiogenesis is the character of stem cell senescence, which brought difficulties for bone defects repairing in senescent microenvironment. As the most abundant bone-related miRNA, miRNA-21-5p plays a crucial role in inducing osteogenic and angiogenic differentiation. However, highly efficient miR-21-5p delivery still confronts challenges including poor cellular uptake and easy degradation. Herein, TDN-miR-21-5p nanocomplex is constructed based on DNA tetrahedral (TDN) and has great potential in promoting osteogenesis and alleviating senescence of senescent bone marrow stem cells (O-BMSCs), simultaneously enhancing angiogenic capacity of senescent endothelial progenitor cells (O-EPCs). Of note, the activation of AKT and Erk signaling pathway may direct regulatory mechanism of TDN-miR-21-5p mediated osteogenesis and senescence of O-BMSCs. Also, TDN-miR-21-5p can indirectly mediate osteogenesis and senescence of O-BMSCs through pro-angiogenic growth factors secreted from O-EPCs. In addition, gelatin methacryloyl (GelMA) hydrogels are mixed with TDN and TDN-miR-21-5p to fabricate delivery scaffolds. TDN-miR-21-5p@GelMA scaffold exhibits greater bone repair with increased expression of osteogenic- and angiogenic-related markers in senescent critical-size cranial defects in vivo. Collectively, TDN-miR-21-5p can alleviate senescence and induce osteogenesis and angiogenesis in senescent microenvironment, which provides a novel candidate strategy for senescent bone repair and widen clinical application of TDNs-based gene therapy.
Collapse
Affiliation(s)
- Lei Qi
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Shebin Hong
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Tong Zhao
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Jinge Yan
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Weiwen Ge
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Jing Wang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Xin Fang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Weidong Jiang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Steve Gf Shen
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| | - Lei Zhang
- Department of Oral & Cranio-Maxillofacial Surgery, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai, 200011, P. R. China
| |
Collapse
|
173
|
Alfutaimani AS, Alharbi NK, S. Alahmari A, A. Alqabbani A, Aldayel AM. Exploring the landscape of Lipid Nanoparticles (LNPs): A comprehensive review of LNPs types and biological sources of lipids. Int J Pharm X 2024; 8:100305. [PMID: 39669003 PMCID: PMC11635012 DOI: 10.1016/j.ijpx.2024.100305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 12/14/2024] Open
Abstract
Lipid nanoparticles (LNPs) have emerged as promising carriers for delivering therapeutic agents, including mRNA-based immunotherapies, in various biomedical applications. The use of LNPs allows for efficient delivery of drugs, resulting in enhanced targeted delivery to specific tissues or cells. These LNPs can be categorized into several types, including liposomes, solid lipid nanoparticles, nanostructured lipid carriers, and lipid-polymer hybrid nanoparticles. The preparation of LNPs involves the manipulation of their structural, dimensional, compositional, and physical characteristics via the use of different methods in the industry. Lipids used to construct LNPs can also be derived from various biological sources, such as natural lipids extracted from plants, animals, or microorganisms. This review dives into the different types of LNPs and their preparation methods. More importantly, it discusses all possible biological sources that are known to supply lipids for the creation of LNPs. Natural lipid reservoirs have surfaced as promising sources for generating LNPs. The use of LNPs in drug delivery is expected to increase significantly in the coming years. Herein, we suggest some environmentally friendly and biocompatible sources that can produce lipids for future LNPs production.
Collapse
Affiliation(s)
- Alanood S. Alfutaimani
- Nanomedicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University (PNU), P.O Box 84428, Riyadh 11671, Saudi Arabia
| | - Nouf K. Alharbi
- Nanomedicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
| | - Amirah S. Alahmari
- Department of Biology, College of Science, Princess Nourah Bint Abdulrahman University (PNU), P.O Box 84428, Riyadh 11671, Saudi Arabia
| | - Almaha A. Alqabbani
- The Ear, Nose, and Throat (ENT) Department at King Salman Hospital, Riyadh 12769, Saudi Arabia
| | - Abdulaziz M. Aldayel
- Nanomedicine Department, King Abdullah International Medical Research Center, King Abdulaziz Medical City, Riyadh 11426, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences (KSAU-HS), King Abdulaziz Medical City (KAMC), Riyadh 11426, Saudi Arabia
| |
Collapse
|
174
|
Bai L, Chen X, Li C, Zhou H, Li Y, Xiao J, Zhang F, Cheng H, Zhou M. Mannose/stearyl chloride doubly functionalized polyethylenimine as a nucleic acid vaccine carrier to promote macrophage uptake. Drug Deliv 2024; 31:2427138. [PMID: 39540234 PMCID: PMC11565675 DOI: 10.1080/10717544.2024.2427138] [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: 07/23/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Transmembrane transport remains a significant challenge for nucleic acid vaccine vectors. Promoting the ability of immune cells, such as macrophages, to capture foreign stimuli is also an effective approach to improving cross-presentation. In addition, polyethyleneimine (PEI) has gained attention in the field of nucleic acid vaccine carriers due to its excellent gene transfection efficiency and unique proton buffering effect. However, although high molecular weight PEI exhibits high efficiency, its high-density positive charges make it highly toxic, which limits its application. In this study, mannose/stearyl chloride functionalized polyethylenimine (SA-Man-PEI) was prepared by functionalizing PEI (molecular weight of 25 kDa) with mannose with immunomodulatory and phagocyte targeting effects, and an alkyl hydrophobic chain segment, which could easily promote cell uptake. Moreover, the functionalized-PEI retains a strong proton buffering effect, which helps the carrier escape from the lysosome. The particle sizes of the composite particles formed by SA-Man-PEI and ovalbumin (OVA) were below 200 nm, with good storage stability at both 4 °C and 37 °C. At a drug concentration of 2 μg/mL, the cell survival rate of functionalized-PEI was 19.2% higher than that of unfunctionalized PEI. In vitro macrophage endocytosis experiments showed that SA-Man-PEI could significantly enhance the macrophage uptake of composite particles, compared to unfunctionalized PEI or single-functionalized PEI. This study offers a new approach for developing PEI as a nucleic acid vaccine carrier, which could simultaneously enhance cell targeting and promote cell uptake.
Collapse
Affiliation(s)
- Lu Bai
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Xiaoqi Chen
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Chengyu Li
- School of Materials Science and Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei Province, China
| | - Haijun Zhou
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Yantao Li
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Jijun Xiao
- School of Materials Science and Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei Province, China
| | - Fen Zhang
- Institute of Energy Resources, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Hua Cheng
- Institute of Biology, Hebei Academy of Sciences, Shijiazhuang, Hebei Province, China
| | - Mengmeng Zhou
- Shijiazhuang Polymer Composite Technological Innovation Center; Shijiazhuang Key Laboratory of Low Carbon Energy Materials, College of Chemical Engineering, Shijiazhuang University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
175
|
Nagpal S, Palaniappan T, Wang JW, Wacker MG. Revisiting nanomedicine design strategies for follow-on products: A model-informed approach to optimize performance. J Control Release 2024; 376:1251-1270. [PMID: 39510258 DOI: 10.1016/j.jconrel.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 10/27/2024] [Accepted: 11/03/2024] [Indexed: 11/15/2024]
Abstract
The field of nanomedicine is undergoing a seismic transformations with the rise of nanosimilars, reshaping the pharmaceutical landscape and expanding beyond traditional innovators and generic manufacturers. Nanodrugs are increasingly replacing conventional therapies, offering improved efficacy and safety, while the demand for follow-on products drives market diversification. However, the transition from preclinical to clinical stages presents challenges due to the complex biopharmaceutical behavior of nanodrugs. This review highlights the integration of Quality-by-Design (QbD), in vitro-in vivo correlations (IVIVCs), machine learning, and Model-Informed Drug Development (MIDD) as key strategies to address these complexities. Additionally, it discusses the role of high-throughput processes in the optimization of the nanodrug development pipelines. Covering generations of delivery systems from liposomes to RNA-loaded nanoparticles, this review underscores the evolving market dynamics driven by recent advances in nanomedicine.
Collapse
Affiliation(s)
- Shakti Nagpal
- National University of Singapore, Faculty of Science, Department of Pharmacy and Pharmaceutical Sciences, Singapore
| | | | - Jiong-Wei Wang
- National University of Singapore, Department of Surgery, Yong Loo Lin School of Medicine, Singapore 119228, Singapore; Cardiovascular Research Institute, National University Heart Centre Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Matthias G Wacker
- National University of Singapore, Faculty of Science, Department of Pharmacy and Pharmaceutical Sciences, Singapore.
| |
Collapse
|
176
|
Hajimolaali M, Dorkoosh FA, Antimisiaris SG. Review of recent preclinical and clinical research on ligand-targeted liposomes as delivery systems in triple negative breast cancer therapy. J Liposome Res 2024; 34:671-696. [PMID: 38520185 DOI: 10.1080/08982104.2024.2325963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/06/2024] [Accepted: 02/27/2024] [Indexed: 03/25/2024]
Abstract
Triple-negative breast Cancer (TNBC) is one of the deadliest types, making up about 20% of all breast cancers. Chemotherapy is the traditional manner of progressed TNBC treatment; however, it has a short-term result with a high reversibility pace. The lack of targeted treatment limited and person-dependent treatment options for those suffering from TNBC cautions to be the worst type of cancer among breast cancer patients. Consequently, appropriate treatment for this disease is considered a major clinical challenge. Therefore, various treatment methods have been developed to treat TNBC, among which chemotherapy is the most common and well-known approach recently studied. Although effective methods are chemotherapies, they are often accompanied by critical limitations, especially the lack of specific functionality. These methods lead to systematic toxicity and, ultimately, the expansion of multidrug-resistant (MDR) cancer cells. Therefore, finding novel and efficient techniques to enhance the targeting of TNBC treatment is an essential requirement. Liposomes have demonstrated that they are an effective method for drug delivery; however, among a large number of liposome-based drug delivery systems annually developed, a small number have just received authorization for clinical application. The new approaches to using liposomes target their structure with various ligands to increase therapeutic efficiency and diminish undesired side effects on various body tissues. The current study describes the most recent strategies and research associated with functionalizing the liposomes' structure with different ligands as targeted drug carriers in treating TNBCs in preclinical and clinical stages.
Collapse
Affiliation(s)
- Mohammad Hajimolaali
- Department of Pharmacy, Laboratory of Pharmaceutical Technology, University of Patras, Patras, Greece
| | - Farid Abedin Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Sophia G Antimisiaris
- Department of Pharmacy, Laboratory of Pharmaceutical Technology, University of Patras, Patras, Greece
- Institute of Chemical Engineering, Foundation for Research and Technology Hellas, FORTH/ICEHT, Patras, Greece
| |
Collapse
|
177
|
Huang Z, Meng H, Xu L, Pei X, Xiong J, Wang Y, Zhan X, Li S, He Y. Liposomes in the cosmetics: present and outlook. J Liposome Res 2024; 34:715-727. [PMID: 38712581 DOI: 10.1080/08982104.2024.2341139] [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: 08/03/2023] [Revised: 02/29/2024] [Accepted: 04/04/2024] [Indexed: 05/08/2024]
Abstract
Liposomes are small spherical vesicles composed of phospholipid bilayers capable of encapsulating a variety of ingredients, including water- and oil-soluble compound, which are one of the most commonly used piggybacking and delivery techniques for many active ingredients and different compounds in biology, medicine and cosmetics. With the increasing number of active cosmetic ingredients, the concomitant challenge is to effectively protect, transport, and utilize these substances in a judicious manner. Many cosmetic ingredients are ineffective both topically and systemically when applied to the skin, thus changing the method of delivery and interaction with the skin of the active ingredients is a crucial step toward improving their effectiveness. Liposomes can improve the delivery of active ingredients to the skin, enhance their stability, and ultimately, improve the efficacy of cosmetics and and pharmaceuticals. In this review, we summarized the basic properties of liposomes and their recent advances of functionalities in cosmetics and and pharmaceuticals. Also, the current state of the art in the field is discussed and the prospects for future research areas are highlighted. We hope that this review will provide ideas and inspiration on the application and development of cosmetics and pharmaceuticals.
Collapse
Affiliation(s)
- Zhaohe Huang
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Hong Meng
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Li Xu
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Xiaojing Pei
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Jie Xiong
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Yanan Wang
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Xin Zhan
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Shujing Li
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Yifan He
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| |
Collapse
|
178
|
Yang H, Fei C, Wang S, Shen X, Yang L, Yang H, Li G. Validation of an HPLC-CAD method for measuring the lipid content of novel LNP-encapsulated COVID-19 mRNA vaccines. J Virol Methods 2024; 330:115040. [PMID: 39384157 DOI: 10.1016/j.jviromet.2024.115040] [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: 06/16/2024] [Revised: 09/29/2024] [Accepted: 09/29/2024] [Indexed: 10/11/2024]
Abstract
Lipid nanoparticles (LNPs) are frequently employed as mRNA vaccine delivery vehicles. LNPs are made up of four types of lipids: cationic lipid, PEG-lipid conjugate, zwitterionic helper phospholipid, and cholesterol. LNP distribution efficiency is significantly impacted by lipid composition, which also controls LNP stability and bilayer fluidity. The various lipids used in the formulation system have distinct properties and contents. To aid in the development of new drugs and vaccines, we developed and validated an HPLC-CAD method for identifying and determining the amounts of four lipids in Yuxi Watson Biotechnology Co., Ltd.'s LNP-encapsulated COVID-19 mRNA vaccines (OmicronXBB.1.5).
Collapse
Affiliation(s)
- Huan Yang
- Yunnan Institute of Supervision and Inspection for Food and Drug, Kunming, Yunnan 650500, PR China; Public Service Platform for Industrial Technology Foundation of the Ministry of Industry and Information Technology, Kunming, Yunnan 650500, PR China
| | - Chengrui Fei
- Yunnan Institute of Supervision and Inspection for Food and Drug, Kunming, Yunnan 650500, PR China; Public Service Platform for Industrial Technology Foundation of the Ministry of Industry and Information Technology, Kunming, Yunnan 650500, PR China
| | - Sijie Wang
- Yunnan Institute of Supervision and Inspection for Food and Drug, Kunming, Yunnan 650500, PR China; Public Service Platform for Industrial Technology Foundation of the Ministry of Industry and Information Technology, Kunming, Yunnan 650500, PR China
| | - Xue Shen
- Yunnan Institute of Supervision and Inspection for Food and Drug, Kunming, Yunnan 650500, PR China; Public Service Platform for Industrial Technology Foundation of the Ministry of Industry and Information Technology, Kunming, Yunnan 650500, PR China
| | - Li Yang
- Yunnan Institute of Supervision and Inspection for Food and Drug, Kunming, Yunnan 650500, PR China; Public Service Platform for Industrial Technology Foundation of the Ministry of Industry and Information Technology, Kunming, Yunnan 650500, PR China
| | - Hefeng Yang
- Yunnan Key Laboratory of Stomatology, Kunming Medical University, Kunming, Yunnan 650500, PR China; Department of Dental Research, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan 650500, PR China.
| | - Guiding Li
- Yunnan Key Laboratory of Stomatology, Kunming Medical University, Kunming, Yunnan 650500, PR China; Department of Dental Research, The Affiliated Stomatology Hospital of Kunming Medical University, Kunming, Yunnan 650500, PR China.
| |
Collapse
|
179
|
Hameed H, Faheem S, Younas K, Jamshaid M, Ereej N, Hameed A, Munir R, Khokhar R. A comprehensive review on lipid-based nanoparticles via nose to brain targeting as a novel approach. J Microencapsul 2024; 41:681-714. [PMID: 39286884 DOI: 10.1080/02652048.2024.2404414] [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: 06/23/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
The central nervous system (CNS) has been a chief concern for millions of people worldwide, and many therapeutic medications are unable to penetrate the blood-brain barrier. Advancements in nanotechnology have enabled safe, effective, and precise delivery of medications towards specific brain regions by utilising a nose-to-brain targeting route. This method reduces adverse effects, increases medication bioavailability, and facilitates mucociliary clearance while promoting accumulation of drug in the targeted brain region. Recent developments in lipid-based nanoparticles, for instance solid lipid nanoparticles (SLNs), liposomes, nanoemulsions, and nano-structured lipid carriers have been explored. SLNs are currently the most promising drug carrier system because of their capability of transporting drugs across the blood-brain barrier at the intended brain site. This approach offers higher efficacy, controlled drug delivery, target specificity, longer circulation time, and a reduction in toxicity through a biomimetic mechanism.
Collapse
Affiliation(s)
- Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Saleha Faheem
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Komel Younas
- Faculty of Pharmacy, University Paris Saclay, Orsay, France
| | - Muhammad Jamshaid
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Nelofer Ereej
- Faculty of Pharmaceutical Sciences, University of Central Punjab, Lahore, Pakistan
| | - Anam Hameed
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Lahore, Pakistan
| | - Rabia Munir
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad, Pakistan
| | - Rabia Khokhar
- Institute of Pharmaceutical Sciences, University of Veterinary and Animal Sciences, Lahore, Pakistan
| |
Collapse
|
180
|
Kana Veedu A, Panthalattu Parambil A, Manheri MK. Sequential Release of Ibuprofen and the Gasotransmitter Hydrogen sulfide using Oxanorbornane-Based Synthetic Lipids as Carriers. Chempluschem 2024; 89:e202400323. [PMID: 39235160 DOI: 10.1002/cplu.202400323] [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: 05/06/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 09/06/2024]
Abstract
After understanding the biological signaling roles of hydrogen sulfide and its involvement in various physiological processes, there has been enormous interest in exploring its therapeutic utility in areas such as cancer, inflammation, cardiovascular diseases, etc. There is also growing interest in using suitable H2S donors in combination with other drugs to improve the treatment outcome through the modulation of multiple pathways. The premature release of H2S from small molecule donors and the difficulty in controlling its spatio-temporal distribution are the major challenges during these efforts. Hence the development of appropriate carriers that can release this gasotransmitter along with the therapeutic entity of interest in a controlled manner has high significance. In this regard, this report presents a novel drug delivery system from oxanorbornane-based synthetic lipids that carries a H2S-releasing 1,2-dithiole-3-thione moiety as part of the head group. Nanoaggregates of the resulting conjugate are not only capable of efficiently entrapping a non-steroidal anti-inflammatory drug such as ibuprofen, but also release this drug and H2S in a controlled and sequential manner.
Collapse
Affiliation(s)
- Akshaya Kana Veedu
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, 600036, Tamil Nadu, India
| | | | - Muraleedharan K Manheri
- Department of Chemistry, Indian Institute of Technology Madras, Chennai, 600036, Tamil Nadu, India
| |
Collapse
|
181
|
Qi Y, Wang C, Lang H, Wang Y, Wang X, Zheng H, Lu Y. Liposome-based RNAi delivery in honeybee for inhibiting parasite Nosema ceranae. Synth Syst Biotechnol 2024; 9:853-860. [PMID: 39139857 PMCID: PMC11320372 DOI: 10.1016/j.synbio.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 06/14/2024] [Accepted: 07/17/2024] [Indexed: 08/15/2024] Open
Abstract
Nosema ceranae, a parasite that parasitizes and reproduces in the gut of honeybees, has become a serious threat to the global apiculture industry. RNA interference (RNAi) technology can be used to inhibit N. ceranae growth by targeting silencing the thioredoxin reductase (TrxR) in N. ceranae. However, suitable carriers are one of the reasons limiting the application of RNAi due to the easy degradation of dsRNA in honeybees. As a vesicle composed of a lipid bilayer, liposomes are a good carrier for nucleic acid delivery, but studies in honeybees are lacking. In this study, liposomes were used for double-stranded RNA (dsRNA) dsTrxR delivery triggering RNAi to inhibit the N. ceranae growth in honeybees. Compared to naked dsTrxR, liposome-dsTrxR reduced N. ceranae numbers in the midgut and partially restored midgut morphology without affecting bee survival and gut microbial composition. The results of this study confirmed that liposomes could effectively protect dsRNA from entering the honeybee gut and provide a reference for using RNAi technology to suppress honeybee pests and diseases.
Collapse
Affiliation(s)
- Yue Qi
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Chen Wang
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Haoyu Lang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yueyi Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| | - Xiaofei Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Hao Zheng
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, 100083, China
| | - Yuan Lu
- Department of Chemical Engineering, Tsinghua University, Beijing, 100084, China
- Key Laboratory of Industrial Biocatalysis, Ministry of Education, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
182
|
Yuan Y, Li J, Chen M, Zhao Y, Zhang B, Chen X, Zhao J, Liang H, Chen Q. Nano-encapsulation of drugs to target hepatic stellate cells: Toward precision treatments of liver fibrosis. J Control Release 2024; 376:318-336. [PMID: 39413846 DOI: 10.1016/j.jconrel.2024.10.012] [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: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Liver fibrosis is characterized by excessive extracellular matrix (ECM) deposition triggered by hepatic stellate cells (HSCs). As central players in fibrosis progression, HSCs are the most important therapeutic targets for antifibrotic therapy. However, owing to the limitations of systemic drug administration, there is still no suitable and effective clinical treatment. In recent years, nanosystems have demonstrated expansive therapeutic potential and evolved into a clinical modality. In liver fibrosis, nanosystems have undergone a paradigm shift from targeting the whole liver to locally targeted modifying processes. Nanomedicine delivered to HSCs has significant potential in managing liver fibrosis, where optimal management would benefit from targeted delivery, personalized therapy based on the specific site of interest, and minor side effects. In this review, we present a brief overview of the role of HSCs in the pathogenesis of liver fibrosis, summarize the different types of nanocarriers and their specific delivery applications in liver fibrosis, and highlight the biological barriers associated with the use of nanosystems to target HSCs and approaches available to solve this issue. We further discuss in-depth all the molecular target receptors overexpressed during HSC activation in liver fibrosis and their corresponding ligands that have been used for drug or gene delivery targeting HSCs.
Collapse
Affiliation(s)
- Yue Yuan
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Jiaxuan Li
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Min Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Ying Zhao
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Jianping Zhao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China.
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China.
| | - Qian Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China.
| |
Collapse
|
183
|
Nasr SS, Paul P, Loretz B, Lehr CM. Realizing time-staggered expression of nucleic acid-encoded proteins by co-delivery of messenger RNA and plasmid DNA on a single nanocarrier. Drug Deliv Transl Res 2024; 14:3339-3353. [PMID: 39009932 DOI: 10.1007/s13346-024-01668-w] [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] [Accepted: 07/04/2024] [Indexed: 07/17/2024]
Abstract
Co-delivery of different protein-encoding polynucleotide species with varying expression kinetics of their therapeutic product will become a prominent requirement in the realm of combined nucleic acid(NA)-based therapies in the upcoming years. The current study explores the capacity for time-staggered expression of encoded proteins by simultaneous delivery of plasmid DNA (pDNA) in the core and mRNA on the shell of the same nanocarrier. The core is based on a Gelatin Type A-pDNA coacervate, thermally stabilized to form an irreversible nanogel stable enough for the deposition of cationic coats namely, protamine sulfate or LNP-related lipid mixtures. Only the protamine-coated nanocarriers remained colloidally stable following mRNA loading and could successfully co-transfect murine dendritic cell line DC2.4 with fluorescent reporter mRNA(mCherry) and pDNA (pAmCyan1). Further investigation of the protamine-coated nanosystem only, the transfection efficiency (percentage of transfected cells) and level of protein expression (mean fluorescence intensity, MFI) of mRNA and pDNA, simultaneously delivered by the same nanocarrier, were compared and kinetically assessed over 48 h in DC2.4 using flow cytometry. The onset of transfection for both nucleotides was initially delayed, with levels < 5% at 6 h. Thereafter, mRNA transfection reached 90% after 24 h and continued to slightly increase until 48 h. In contrast, pDNA transfection was clearly slower, reaching approximately 40% after 24 h, but continuing to increase to reach 94% at 48 h. The time course of protein expression (represented by MFI) for both NAs essentially followed that of transfection. Model-independent as well as model-dependent kinetic parameters applied to the data further confirmed such time-staggered expression of the two NA's where mRNA's rate of transfection and protein expression initially exceeded those of pDNA in the first 24 h of the experiment whereas the opposite was true during the second 24 h of the experiment where pDNA displayed the higher response rates. We expect that innovative nanocarriers capable of time-staggered co-delivery of different nucleotides could open new perspectives for multi-dosing, pulsatile or sustained expression of nucleic acid-based therapeutics in protein replacement, vaccination, and CRISPR-mediated gene editing scenarios.
Collapse
Affiliation(s)
- Sarah S Nasr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123, Saarbrücken, Germany.
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany.
- Fischell Department of Bioengineering, University of Maryland, College Park, USA.
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, 21521, Egypt.
| | - Pascal Paul
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123, Saarbrücken, Germany
| | - Brigitta Loretz
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123, Saarbrücken, Germany
| | - Claus-Michael Lehr
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University, Campus E8 1, 66123, Saarbrücken, Germany.
- Department of Pharmacy, Saarland University, 66123, Saarbrücken, Germany.
| |
Collapse
|
184
|
Zou B, Long Y, Gao R, Liu Q, Tian X, Liu B, Zhou Q. Nanodelivery system of traditional Chinese medicine bioactive compounds: Application in the treatment of prostate cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:155554. [PMID: 39341127 DOI: 10.1016/j.phymed.2024.155554] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND The long history of clinical experience in China have confirmed the effectiveness of traditional Chinese medicine (TCM) in treating prostate cancer (PCa). Until now, several bioactive compounds with anti-PCa potential, such as curcumin, gallic acid, and quercetin, have been extracted from TCM. Recent studies have shown that encapsulating these TCM bioactive compounds into nano-delivery system enhanced their bioavailability and improved their ability to target PCa tumors. PURPOSE This review aims to summarize the anti-PCa effects and molecular mechanisms of TCM bioactive compounds and discuss the clinical application prospects and future research trends of nano-delivery system based on these compounds. METHODS Literatures focusing on the treatment of PCa using traditional Chinese medicine compounds via nano-drug delivery system were searched from Electronic databases, including PubMed, Web of Science, and Scopus until December 2023. RESULTS Polyphenols, alkaloids, terpenes, and quinones exhibit anti-PCa effects through various pathways. Notably, compounds like curcumin, gallic acid, quercetin, and tanshinone have been extensively studied in nano-delivery systems for anti-PCa purpose. Nano-delivery systems enhance the biological activity of free compounds and reduce toxic side effects, as well. Commonly used nanomaterials for delivering TCM compounds include polymer nanomaterials, liposomes, solid lipid nanoparticles, nanostructured lipid carriers, and niosomes. CONCLUSION Research on nano-delivery systems for TCM bioactive compounds holds promising prospects for anti-PCa therapy. However, extensive clinical trials are necessary to evaluate the effectiveness and safety of these nanodrugs.
Collapse
Affiliation(s)
- Bo Zou
- The First Hospital of Hunan University of Chinese Medicine, 95, Changsha 410007, Hunan, China
| | - Yan Long
- The First Hospital of Hunan University of Chinese Medicine, 95, Changsha 410007, Hunan, China
| | - Ruisong Gao
- The First Hospital of Hunan University of Chinese Medicine, 95, Changsha 410007, Hunan, China
| | - Qizhi Liu
- Hunan University of Chinese Medicine, 300, Changsha 410208, Hunan, China
| | - Xuefei Tian
- Hunan University of Chinese Medicine, 300, Changsha 410208, Hunan, China
| | - Bin Liu
- College of Biology of Hunan University, Changsha 410208, Hunan, China.
| | - Qing Zhou
- The First Hospital of Hunan University of Chinese Medicine, 95, Changsha 410007, Hunan, China.
| |
Collapse
|
185
|
Cai J, Chen S, Liu Z, Li H, Wang P, Yang F, Li Y, Chen K, Sun M, Qiu M. RNA technology and nanocarriers empowering in vivo chimeric antigen receptor therapy. Immunology 2024; 173:634-653. [PMID: 39340367 DOI: 10.1111/imm.13861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
The remarkable success of mRNA-based coronavirus 2019 (COVID-19) vaccines has propelled the advancement of nanomedicine, specifically in the realm of RNA technology and nanomaterial delivery systems. Notably, significant strides have been made in the development of RNA-based in vivo chimeric antigen receptor (CAR) therapy. In comparison to the conventional ex vivo CAR therapy, in vivo CAR therapy offers several benefits including simplified preparation, reduced costs, broad applicability and decreased potential for carcinogenic effects. This review summarises the RNA-based CAR constructs in in vivo CAR therapy, discusses the current applications of in vivo delivery vectors and outlines the immune cells edited with CAR molecules. We aim for the conveyed messages to contribute towards the advancement of in vivo CAR application.
Collapse
Affiliation(s)
- Jingsheng Cai
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, People's Republic of China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, People's Republic of China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, People's Republic of China
| | - Shaoyi Chen
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, People's Republic of China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, People's Republic of China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, People's Republic of China
| | - Zheng Liu
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, People's Republic of China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, People's Republic of China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, People's Republic of China
| | - Haoran Li
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, People's Republic of China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, People's Republic of China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, People's Republic of China
| | - Peiyu Wang
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, People's Republic of China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, People's Republic of China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, People's Republic of China
| | - Fan Yang
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, People's Republic of China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Yun Li
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, People's Republic of China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, People's Republic of China
| | - Kezhong Chen
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, People's Republic of China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, People's Republic of China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, People's Republic of China
| | - Ming Sun
- Department of Oncology Center, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, People's Republic of China
| | - Mantang Qiu
- Thoracic Oncology Institute, Peking University People's Hospital, Beijing, People's Republic of China
- Department of Thoracic Surgery, Peking University People's Hospital, Beijing, People's Republic of China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, People's Republic of China
| |
Collapse
|
186
|
Liu J, Xi Z, Fan C, Mei Y, Zhao J, Jiang Y, Zhao M, Xu L. Hydrogels for Nucleic Acid Drugs Delivery. Adv Healthc Mater 2024; 13:e2401895. [PMID: 39152918 DOI: 10.1002/adhm.202401895] [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: 05/22/2024] [Revised: 07/05/2024] [Indexed: 08/19/2024]
Abstract
Nucleic acid drugs are one of the hot spots in the field of biomedicine in recent years, and play a crucial role in the treatment of many diseases. However, its low stability and difficulty in target drug delivery are the bottlenecks restricting its application. Hydrogels are proven to be promising for improving the stability of nucleic acid drugs, reducing the adverse effects of rapid degradation, sudden release, and unnecessary diffusion of nucleic acid drugs. In this review, the strategies of loading nucleic acid drugs in hydrogels are summarized for various biomedical research, and classify the mechanism principles of these strategies, including electrostatic binding, hydrogen bond based binding, hydrophobic binding, covalent bond based binding and indirect binding using various carriers. In addition, this review also describes the release strategies of nucleic acid drugs, including photostimulation-based release, enzyme-responsive release, pH-responsive release, and temperature-responsive release. Finally, the applications and future research directions of hydrogels for delivering nucleic acid drugs in the field of medicine are discussed.
Collapse
Affiliation(s)
- Jiaping Liu
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Ziyue Xi
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Chuanyong Fan
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Yihua Mei
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Jiale Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Yingying Jiang
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Ming Zhao
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| | - Lu Xu
- School of Pharmacy, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang, 110016, P. R. China
| |
Collapse
|
187
|
Nguyen L, Nguyen TT, Kim JY, Jeong JH. Advanced siRNA delivery in combating hepatitis B virus: mechanistic insights and recent updates. J Nanobiotechnology 2024; 22:745. [PMID: 39616384 PMCID: PMC11608496 DOI: 10.1186/s12951-024-03004-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/09/2024] [Indexed: 12/06/2024] Open
Abstract
Hepatitis B virus (HBV) infection is a major health problem, causing thousands of deaths each year worldwide. Although current medications can often inhibit viral replication and reduce the risk of liver carcinoma, several obstacles still hinder their effectiveness. These include viral resistance, prolonged treatment duration, and low efficacy in clearing viral antigens. To address these challenges in current HBV treatment, numerous approaches have been developed with remarkable success. Among these strategies, small-interfering RNA (siRNA) stands out as one of the most promising therapies for hepatitis B. However, naked siRNAs are vulnerable to enzymatic digestion, easily eliminated by renal filtration, and unable to cross the cell membrane due to their large, anionic structure. Therefore, effective delivery systems are required to protect siRNAs and maintain their functionality. In this review, we have discussed the promises of siRNA therapy in treating HBV, milestones in their delivery systems, and products that have entered clinical trials. Finally, we have outlined the future perspectives of siRNA-based therapy for HBV treatment.
Collapse
Affiliation(s)
- Linh Nguyen
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea
| | - Tiep Tien Nguyen
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea.
| | - Ju-Yeon Kim
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea.
| | - Jee-Heon Jeong
- Department of Precision Medicine, School of Medicine, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea.
| |
Collapse
|
188
|
Hsiung KC, Chiang HJ, Reinig S, Shih SR. Vaccine Strategies Against RNA Viruses: Current Advances and Future Directions. Vaccines (Basel) 2024; 12:1345. [PMID: 39772007 PMCID: PMC11679499 DOI: 10.3390/vaccines12121345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 01/11/2025] Open
Abstract
The development of vaccines against RNA viruses has undergone a rapid evolution in recent years, particularly driven by the COVID-19 pandemic. This review examines the key roles that RNA viruses, with their high mutation rates and zoonotic potential, play in fostering vaccine innovation. We also discuss both traditional and modern vaccine platforms and the impact of new technologies, such as artificial intelligence, on optimizing immunization strategies. This review evaluates various vaccine platforms, ranging from traditional approaches (inactivated and live-attenuated vaccines) to modern technologies (subunit vaccines, viral and bacterial vectors, nucleic acid vaccines such as mRNA and DNA, and phage-like particle vaccines). To illustrate these platforms' practical applications, we present case studies of vaccines developed for RNA viruses such as SARS-CoV-2, influenza, Zika, and dengue. Additionally, we assess the role of artificial intelligence in predicting viral mutations and enhancing vaccine design. The case studies underscore the successful application of RNA-based vaccines, particularly in the fight against COVID-19, which has saved millions of lives. Current clinical trials for influenza, Zika, and dengue vaccines continue to show promise, highlighting the growing efficacy and adaptability of these platforms. Furthermore, artificial intelligence is driving improvements in vaccine candidate optimization and providing predictive models for viral evolution, enhancing our ability to respond to future outbreaks. Advances in vaccine technology, such as the success of mRNA vaccines against SARS-CoV-2, highlight the potential of nucleic acid platforms in combating RNA viruses. Ongoing trials for influenza, Zika, and dengue demonstrate platform adaptability, while artificial intelligence enhances vaccine design by predicting viral mutations. Integrating these innovations with the One Health approach, which unites human, animal, and environmental health, is essential for strengthening global preparedness against future RNA virus threats.
Collapse
Affiliation(s)
- Kuei-Ching Hsiung
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
| | - Huan-Jung Chiang
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
- Graduate Institute of Biomedical Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Sebastian Reinig
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
| | - Shin-Ru Shih
- Research Center for Emerging Viral Infections, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; (K.-C.H.); (H.-J.C.); (S.R.)
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
- Department of Medical Biotechnology & Laboratory Science, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Research Center for Chinese Herbal Medicine, Research Center for Food & Cosmetic Safety, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science & Technology, Taoyuan 33303, Taiwan
| |
Collapse
|
189
|
Ochoa-Sánchez C, Rodríguez-León E, Iñiguez-Palomares R, Rodríguez-Beas C. Brief Comparison of the Efficacy of Cationic and Anionic Liposomes as Nonviral Delivery Systems. ACS OMEGA 2024; 9:46664-46678. [PMID: 39619565 PMCID: PMC11603276 DOI: 10.1021/acsomega.4c06714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 10/28/2024] [Accepted: 11/05/2024] [Indexed: 01/05/2025]
Abstract
In recent decades, the development and application of nonviral vectors, such as liposomes and lipidic nanoparticles, for gene therapy and drug delivery have seen substantial progress. The interest in the physicochemical properties and structures of the complexes liposome/DNA and liposome/RNA is due to their potential to substitute viruses as carriers of drugs or genetic material into cells with minimal cytotoxicity, which could lead to their use in gene therapy. Initially, cationic liposomes were utilized as nonviral DNA delivery vectors; subsequently, different molecules, such as polymers, were incorporated to enhance transfection efficiency. Additionally, liposome/protein complexes have been developed as nonviral vectors for the treatment of diseases. The most relevant internalization pathways of these vectors and the few transfection results obtained using targeted and nontargeted liposomes are discussed below. The high cytotoxicity of cationic liposomes represents a significant challenge for the development of gene therapy and drug delivery. Anionic liposomes offer a promising alternative to address the limitations of conventional cationic liposomes, including immune response, short circulation time, and low toxicity. This review will discuss the advantages of cationic liposomes and the novel anionic liposome-based systems that have emerged as a result. The advent of novel designs and manufacturing techniques has facilitated the development of innovative systems, designated as lipid nanoparticles (LNPs), which serve as highly efficacious regulators of the immune system.
Collapse
Affiliation(s)
- Carlos Ochoa-Sánchez
- Physics Department, Universidad de Sonora. Rosales and Luis Encinas 8300, Hermosillo, Sonora 83000, México
| | - Ericka Rodríguez-León
- Physics Department, Universidad de Sonora. Rosales and Luis Encinas 8300, Hermosillo, Sonora 83000, México
| | - Ramón Iñiguez-Palomares
- Physics Department, Universidad de Sonora. Rosales and Luis Encinas 8300, Hermosillo, Sonora 83000, México
| | - César Rodríguez-Beas
- Physics Department, Universidad de Sonora. Rosales and Luis Encinas 8300, Hermosillo, Sonora 83000, México
| |
Collapse
|
190
|
Pals MJ, Lindberg A, Velema WA. Chemical strategies for antisense antibiotics. Chem Soc Rev 2024; 53:11303-11320. [PMID: 39436264 PMCID: PMC11495246 DOI: 10.1039/d4cs00238e] [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] [Received: 03/13/2024] [Indexed: 10/23/2024]
Abstract
Antibacterial resistance is a severe threat to modern medicine and human health. To stay ahead of constantly-evolving bacteria we need to expand our arsenal of effective antibiotics. As such, antisense therapy is an attractive approach. The programmability allows to in principle target any RNA sequence within bacteria, enabling tremendous selectivity. In this Tutorial Review we provide guidelines for devising effective antibacterial antisense agents and offer a concise perspective for future research. We will review the chemical architectures of antibacterial antisense agents with a special focus on the delivery and target selection for successful antisense design. This Tutorial Review will strive to serve as an essential guide for antibacterial antisense technology development.
Collapse
Affiliation(s)
- Mathijs J Pals
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| | - Alexander Lindberg
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| | - Willem A Velema
- Institute for Molecules and Materials, Radboud University Nijmegen, the Netherlands. Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands.
| |
Collapse
|
191
|
Laila UE, An W, Xu ZX. Emerging prospects of mRNA cancer vaccines: mechanisms, formulations, and challenges in cancer immunotherapy. Front Immunol 2024; 15:1448489. [PMID: 39654897 PMCID: PMC11625737 DOI: 10.3389/fimmu.2024.1448489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 10/18/2024] [Indexed: 12/12/2024] Open
Abstract
Cancer continues to pose an alarming threat to global health, necessitating the need for the development of efficient therapeutic solutions despite massive advances in the treatment. mRNA cancer vaccines have emerged as a hopeful avenue, propelled by the victory of mRNA technology in COVID-19 vaccines. The article delves into the intricate mechanisms and formulations of cancer vaccines, highlighting the ongoing efforts to strengthen mRNA stability and ensure successful translation inside target cells. Moreover, it discusses the design and mechanism of action of mRNA, showcasing its potential as a useful benchmark for developing efficacious cancer vaccines. The significance of mRNA therapy and selecting appropriate tumor antigens for the personalized development of mRNA vaccines are emphasized, providing insights into the immune mechanism. Additionally, the review explores the integration of mRNA vaccines with other immunotherapies and the utilization of progressive delivery platforms, such as lipid nanoparticles, to improve immune responses and address challenges related to immune evasion and tumor heterogeneity. While underscoring the advantages of mRNA vaccines, the review also addresses the challenges associated with the susceptibility of RNA to degradation and the difficulty in identifying optimum tumor-specific antigens, along with the potential solutions. Furthermore, it provides a comprehensive overview of the ongoing research efforts aimed at addressing these hurdles and enhancing the effectiveness of mRNA-based cancer vaccines. Overall, this review is a focused and inclusive impression of the present state of mRNA cancer vaccines, outlining their possibilities, challenges, and future predictions in the fight against cancer, ultimately aiding in the development of more targeted therapies against cancer.
Collapse
Affiliation(s)
| | | | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| |
Collapse
|
192
|
Favas R, Almeida H, Peixoto AF, Ferreira D, Silva AC. Advances in Encapsulating Marine Bioactive Compounds Using Nanostructured Lipid Carriers (NLCs) and Solid Lipid Nanoparticles (SLNs) for Health Applications. Pharmaceutics 2024; 16:1517. [PMID: 39771497 PMCID: PMC11728729 DOI: 10.3390/pharmaceutics16121517] [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: 10/18/2024] [Revised: 11/13/2024] [Accepted: 11/20/2024] [Indexed: 01/16/2025] Open
Abstract
As life expectancy rises and modern lifestyles improve, there is an increasing focus on health, disease prevention, and enhancing physical appearance. Consumers are more aware of the benefits of natural ingredients in healthcare products while also being mindful of sustainability challenges. Consequently, marine bioactive compounds have gained popularity as ingredients in cosmetics and food supplements due to their diverse beneficial properties. Nonetheless, the use of some of these compounds is restricted by their low stability and poor aqueous solubility, necessitating solutions to overcome these limitations. In this context, lipid nanoparticles, such as solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs), have been investigated for their potential to protect and improve the absorption of molecules through various routes, including oral and cutaneous. Numerous studies have shown that nanoencapsulating these compounds and incorporating them into cosmetics and food supplements can be effective. However, this application remains unregulated at the global level and is not currently addressed by existing legislation. Additional in vivo studies in both animals and humans are necessary to fully assess safety concerns.
Collapse
Affiliation(s)
- Rita Favas
- UCIBIO (Applied Molecular Biosciences Unit), Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Hugo Almeida
- UCIBIO (Applied Molecular Biosciences Unit), Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Mesosystem Investigação & Investimentos by Spinpark, 4805-017 Guimarães, Portugal
| | - Andreia F. Peixoto
- LAQV-REQUIMTE (Associated Laboratory for Green Chemistry of the Network of Chemistry and Technology), Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, 4169-007 Porto, Portugal
| | - Domingos Ferreira
- UCIBIO (Applied Molecular Biosciences Unit), Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Ana C. Silva
- UCIBIO (Applied Molecular Biosciences Unit), Laboratory of Pharmaceutical Technology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- Associate Laboratory i4HB Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- FP-BHS (Biomedical and Health Sciences Research Unit), FP-I3ID (Instituto de Investigação, Inovação e Desenvolvimento), Faculty of Health Sciences, University Fernando Pessoa, 4249-004 Porto, Portugal
| |
Collapse
|
193
|
Anders AG, Ruotolo BT. Ion Mobility-Mass Spectrometry Captures the Structural Consequences of Lipid Nanoparticle Encapsulation on Ribonucleic Acid Cargo. J Am Chem Soc 2024; 146:31885-31891. [PMID: 39508132 PMCID: PMC11910746 DOI: 10.1021/jacs.4c11066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Ribonucleic acids (RNAs) are becoming increasingly significant in our search for improved biotherapeutics. RNA-based treatments offer high specificity, targeted delivery, and potentially lower-cost options for various debilitating human diseases. Despite these benefits, there are still relatively few FDA-approved RNA-based therapies, with the notable exceptions being the mRNA (mRNA) COVID-19 vaccines, which are delivered using lipid nanoparticle (LNP) systems. LNPs are distinctive drug delivery systems (DDSs) because of their ability to target specific cells, their biocompatibility, and their efficiency in merging with cellular membranes to enhance treatment effectiveness. While the biophysical landscapes of RNA structures in solution are relatively well understood, the impact of the LNP environment on RNA remains less clear. This study uses native ion mobility-mass spectrometry (IM-MS) and collision-induced unfolding (CIU) techniques to investigate how LNP encapsulation affects RNA structure and stability. We examine how various factors, such as ionization polarity, cofactor binding, lipid types, and lipid ratios, influence LNP-released RNA cargo. Our findings reveal that LNP DDSs induce significant changes in the structures and stabilities of their RNA cargo. However, the extent of these changes strongly depends on the type and composition of the lipids used. We conclude by discussing how IM-MS and CIU can aid in the continued development of more efficient LNP DDSs and improve DDS selection methodologies overall.
Collapse
Affiliation(s)
- Anna G Anders
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Brandon T Ruotolo
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
194
|
van Staden D, Gerber M, Lemmer HJR. The Application of Nano Drug Delivery Systems in Female Upper Genital Tract Disorders. Pharmaceutics 2024; 16:1475. [PMID: 39598598 PMCID: PMC11597179 DOI: 10.3390/pharmaceutics16111475] [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: 10/07/2024] [Revised: 11/11/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
The prevalence of female reproductive system disorders is increasing, especially among women of reproductive age, significantly impacting their quality of life and overall health. Managing these diseases effectively is challenging due to the complex nature of the female reproductive system, characterized by dynamic physiological environments and intricate anatomical structures. Innovative drug delivery approaches are necessary to facilitate the precise regulation and manipulation of biological tissues. Nanotechnology is increasingly considered to manage reproductive system disorders, for example, nanomaterial imaging allows for early detection and enhances diagnostic precision to determine disease severity and progression. Additionally, nano drug delivery systems are gaining attention for their ability to target the reproductive system successfully, thereby increasing therapeutic efficacy and decreasing side effects. This comprehensive review outlines the anatomy of the female upper genital tract by highlighting the complex mucosal barriers and their impact on systemic and local drug delivery. Advances in nano drug delivery are described for their sustainable therapeutic action and increased biocompatibility to highlight the potential of nano drug delivery strategies in managing female upper genital tract disorders.
Collapse
Affiliation(s)
| | | | - Hendrik J. R. Lemmer
- Centre of Excellence for Pharmaceutical Sciences (PharmacenTM), North-West University, Potchefstroom 2531, South Africa; (D.v.S.); (M.G.)
| |
Collapse
|
195
|
Milanesi L, Tomas S. The interaction of a self-assembled nanoparticle and a lipid membrane: Binding, disassembly and re-distribution. Heliyon 2024; 10:e39681. [PMID: 39524779 PMCID: PMC11550047 DOI: 10.1016/j.heliyon.2024.e39681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Here we report a detailed study of the interactions of nanoparticles, formed by the self-assembly of cholesterol-containing porphyrins, with lipid membranes. We show that the interaction is a two-step process: first, the docking and fusion, then, the redistribution of the building blocks of the self-assembled nanoparticles (SANs henceforth). Analysis of the binding and structural data is consistent with the docking step being driven by a multivalence cooperative effect and with the formation of SAN aggregates on the membrane, whilst the solubility of the cholesterol anchor in the membrane is key to both the fusion and redistribution of the SANs building blocks. The tendency of the SAN to aggregate in the membrane helps explain the photosensitizer properties of the SANs, essential to their anti-microbial activity. The solubility of the cholesteryl anchors drives fusion to the membrane and de-assembly of the SAN, explaining the capability of the SANs to deliver therapeutic cargos at the lipid interface. The subsequent redistribution of the SANs building blocks offer a plausible pathway to body clearance that is not immediately available to hard nanoparticles. These properties, and the modularity of the synthesis, point to the SANs being an excellent platform for the development of nanomedicines. An unexpected consequence of unraveling the mechanism of membrane interaction of these SANs is that it allows us to derive a value of the free energy of binding of cholesterol (the membrane anchor of the SAN building blocks) to a lipid membrane, that is consistent with the literature values. This is an additional property that can be exploited to determine the affinity of a variety of membrane anchors to membranes of various compositions.
Collapse
Affiliation(s)
- Lilia Milanesi
- Department of Chemistry, University of the Balearic Islands, Ctra. Valldemossa, Km 7.5. 07122, Palma de Mallorca, Spain
| | - Salvador Tomas
- Department of Chemistry, University of the Balearic Islands, Ctra. Valldemossa, Km 7.5. 07122, Palma de Mallorca, Spain
| |
Collapse
|
196
|
Wu K, Xu F, Dai Y, Jin S, Zheng A, Zhang N, Xu Y. Characterization of mRNA-LNP structural features and mechanisms for enhanced mRNA vaccine immunogenicity. J Control Release 2024; 376:1288-1299. [PMID: 39522562 DOI: 10.1016/j.jconrel.2024.11.007] [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: 07/21/2024] [Revised: 10/30/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024]
Abstract
Lipid nanoparticles (LNPs) used for nonviral gene delivery have achieved significant success, particularly in COVID-19 mRNA vaccines. LNPs are routinely characterized by their particle size, polydispersity, and mRNA loading efficiency. However, the internal structure of these particles has not been specified, despite evidence showing that LNPs can be highly heterogeneous, with variations in lipid composition and preparation methods. How these structural features contributed to mRNA LNP vaccine activities is also unclear. In this study, we prepared LNPs with distinctly different internal structures. They were named the emulsion-like LNPs (eLNPs) and membrane-rich LNPs (mLNPs) respectively and compared with the classic "bleb" structure LNPs (cLNPs). The eLNPs contained higher molar percent of the ionizable lipid and lower molar percent of DSPC and cholesterol. The different lipid organization structures lead to varying mRNA delivery activities in vitro and in vivo. After intramuscular injection, eLNPs remained at the injection site and expressed antigens locally. The resulted immune responses had a very fast onset (higher titer at week 2) and lasted longer and stronger (higher titers at week 8) than other LNPs (cLNPs and mLNPs). We hypothesize that the rapid onset and local expression of antigens by muscle cells in the eLNP groups may be favored by the antigen recognition and presentation process, despite the overall mRNA expression activities was not as high especially in liver and other organ. Our data support that eLNPs are potentially the more suitable delivery system for mRNA vaccine due to their high immunogenicity and low systemic toxicity.
Collapse
Affiliation(s)
- Kangzeng Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China; Zhejiang-California International Nanosystems Institute, Zhejiang University, Hangzhou 310058, PR China
| | - Fengwei Xu
- HighField Biopharmaceuticals Inc., Hangzhou 310000, PR China
| | - Yongchao Dai
- HighField Biopharmaceuticals Inc., Hangzhou 310000, PR China
| | - Shanshan Jin
- HighField Biopharmaceuticals Inc., Hangzhou 310000, PR China
| | - Anjie Zheng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, PR China
| | - Ning Zhang
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, PR China.
| | - Yuhong Xu
- College of Pharmacy, Dali University, Dali 671003, PR China; HighField Biopharmaceuticals Inc., Hangzhou 310000, PR China.
| |
Collapse
|
197
|
Daram A, Sawant SS, Mehta DA, Sanhueza CA, Kunda NK. Inhalable Anti-EGFR Antibody-Conjugated Osimertinib Liposomes for Non-Small Cell Lung Cancer. Pharmaceutics 2024; 16:1444. [PMID: 39598567 PMCID: PMC11597056 DOI: 10.3390/pharmaceutics16111444] [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: 09/05/2024] [Revised: 11/04/2024] [Accepted: 11/10/2024] [Indexed: 11/29/2024] Open
Abstract
Background: Non-small cell lung cancer (NSCLC) is a leading cause of cancer deaths globally. The most extensive treatment is Tyrosine Kinase Inhibitors (TKIs) that target epidermal growth factor receptor (EGFR) overexpression. Osimertinib, a third-generation TKI is approved to target EGFR exon 19 deletions or exon 21 L858R mutations. However, resistance is inevitable due to emergence of triple mutations (sensitizing mutations, T790M and C797S). To overcome this challenge, a combinatorial approach was used wherein Osimertinib liposomes were conjugated with cetuximab (CTX), an anti-EGFR monoclonal antibody, to improve drug efficacy and delivery. Additionally, pulmonary administration was employed to minimize systemic toxicity and achieve high lung concentrations. Methods: Osimertinib liposomes (OB-LPs) were prepared using thin film hydration method and immunoliposomes (CTX-OB-LPs) were prepared by conjugating the OB-LPs surface with CTX. Liposomes were characterized for particle size, zeta-potential, drug loading, antibody conjugation efficiency, in vitro drug release, and aerosolization performance. Further, the in vitro efficacy of immunoliposomes was evaluated in H1975 cell line. Results: Immunoliposomes exhibited a particle size of 150 nm, high antibody conjugation efficiency (87%), efficient drug release, and excellent aerosolization properties with an aerodynamic diameter of 3 μm and fine particle fraction of 88%. Furthermore, in vitro studies in H1975 cells showed enhanced cytotoxicity with CTX-OB-LPs displaying 1.7-fold reduction and 1.2-fold reduction in IC50 compared to Osimertinib and OB-LPs, respectively. The CTX-OB-LPs also significantly reduced tumor cell migration and colonization compared to Osimertinib and OB-LPs. Conclusions: These successful results for EGFR-targeting inhalable immunoliposomes exhibited potential for contributing to greater anti-tumor efficacy for the treatment of non-small cell lung cancer.
Collapse
Affiliation(s)
| | | | | | | | - Nitesh K. Kunda
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York City, NY 11439, USA
| |
Collapse
|
198
|
Tang S, Levy ES, Zang N, Goyon A, Chang D, Cebrero A, Tang Y, Pellett JD. Scaling laws for nanoparticles - Online shape heterogeneity analysis by size exclusion chromatography coupled with multi-angle light scattering. J Chromatogr A 2024; 1736:465386. [PMID: 39341170 DOI: 10.1016/j.chroma.2024.465386] [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: 07/25/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Nanoparticle-based drug delivery systems are rising technologies to access challenging therapeutic targets. Following commercial success of lipid-based nanoparticles (LBNP), accruing understandings of nanoparticle structures and critical quality attributes through advanced analytics are beneficial to future clinical development and generalization of this delivery platform. The morphological attributes of nanoparticles, such as shape, can affect uptake, cell-interaction, drug release, circulation, and flow. Gaining an understanding of these structure-activity relationships in early-stage formulation development is important because mix morphologies can affect quality and potency but often exist before process control strategies are fully implemented. In this study, we used shape heterogeneous nanoparticle mixtures, containing various populations of liposomes and lipodisks, as a model system and developed an online semi-quantitative method to characterize the nanoparticle shape heterogeneity by size exclusion chromatography (SEC) coupled with multi-angle light scattering (MALS). The liposomes and lipodisks were separated in SEC when their sizes were ∼3 fold different. When the particles of different shapes were in similar sizes, size-based separation was not always feasible. Instead, light scattering data distinguished liposomes and lipodisks by the scaling law linking radius of gyration and molecular weight of the nanoparticles, enabling morphological identification. A semi-quantitative model was built based on the exponential correlation between the scaling law exponents and the ratios of liposomes and lipodisks. The model was applied to test 6 random formulations made with different compositions and manufacturing processes, and the predicted liposome percentage for 5 formulations was within 25 % absolute difference from the percentage determined by cryogenic electron microscopy (cryo-EM). We envision this method being routinely used to characterize liposome and lipodisk shape heterogeneity during formulation screening as well as on stability studies. Potentially, the method can be converted to in-process control method and extended to other categories of nanoparticles beyond liposomes.
Collapse
Affiliation(s)
- Shijia Tang
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, CA 94080, USA.
| | - Elizabeth S Levy
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Nanzhi Zang
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Alexandre Goyon
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Debby Chang
- Pharma Technical Development, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Abigail Cebrero
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Yijing Tang
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, CA 94080, USA
| | - Jackson D Pellett
- Synthetic Molecule Pharmaceutical Sciences, Genentech, Inc., South San Francisco, CA 94080, USA
| |
Collapse
|
199
|
Epanchintseva AV, Baranova SV, Poletaeva JE, Bakhno IA, Ryabchikova EI, Dovydenko IS. Study of Hard Protein Corona on Lipid Surface of Composite Nanoconstruction. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1767. [PMID: 39513847 PMCID: PMC11547845 DOI: 10.3390/nano14211767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/12/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
The composition of the protein corona covering any nanoparticle (NP) when it enters a biological fluid determines the parameters of the NP's interaction with the body. To "control" these parameters, it is important to know the composition of the protein corona, the determination of which is a complex task associated with the two-layer organization of the corona (hard and soft coronas). In a previous publication, we reported obtaining lipid-coated NPs with a full protein corona, isolating them, and proving the presence of the corona on the surface of the NPs. This work reports on the preparation, isolation, and purification of lipid-coated NPs bearing a hard corona. The protein corona composition was determined by using the LC-MS/MS method. Thirty-seven serum proteins were identified with a high degree of reliability. The hard corona contained various apolipoproteins, including apolipoprotein E, which can potentially affect the penetration of NPs into the cell.
Collapse
Affiliation(s)
| | | | | | | | - Elena I. Ryabchikova
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (A.V.E.); (S.V.B.); (J.E.P.); (I.A.B.)
| | - Ilya S. Dovydenko
- Institute of Chemical Biology and Fundamental Medicine SB RAS, 630090 Novosibirsk, Russia; (A.V.E.); (S.V.B.); (J.E.P.); (I.A.B.)
| |
Collapse
|
200
|
Li M, Jiang S, Wang X, Xu W, Du C, Wei Y, Deng X, Liu M. Chirality and Curvature on Protein Adsorption and Osteogenesis. Angew Chem Int Ed Engl 2024; 63:e202411733. [PMID: 39115949 DOI: 10.1002/anie.202411733] [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: 06/22/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 08/10/2024]
Abstract
Here we designed enantiomeric lipid-mimetic glutamic acid derivatives (L/D-UG) and investigated their self-assembled chiral nanostructures' interaction with the protein adsorption as well as the osteogenesis. It was found that L or D-UG molecules can self-assemble into vesicle bilayers and two-dimensional (2D) nanocrystals via a kinetic and thermodynamic control, respectively. These chiral vesicles and 2D crystals showed differentiated adsorption of proteins, determined by their curvature and chirality. Specifically, fibronectin constituted by L-amino acids adsorbed preferentially on L-UG 2D crystal in a semi-random pattern and L-2D nanocrystal show as the most effective structures to promote bone regeneration. The controlled vesicle and 2D crystal assemblies with different chirality and curvature helps to clarify their determine roles in protein adsorption and osteogenesis.
Collapse
Affiliation(s)
- Meijun Li
- Beijing National Laboratory for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, No.2, 1st North Street, Zhongguancun, Beijing, 100190, P.R.China
| | - Shengjie Jiang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, No.22 Zhongguancun South Street, Zhongguancun, Beijing, 100081, P.R.China
| | - Xiaowei Wang
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, No.22 Zhongguancun South Street, Zhongguancun, Beijing, 100081, P.R.China
| | - Wei Xu
- Beijing National Laboratory for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, No.2, 1st North Street, Zhongguancun, Beijing, 100190, P.R.China
| | - Cong Du
- Beijing National Laboratory for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, No.2, 1st North Street, Zhongguancun, Beijing, 100190, P.R.China
| | - Yan Wei
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, No.22 Zhongguancun South Street, Zhongguancun, Beijing, 100081, P.R.China
| | - Xuliang Deng
- Department of Geriatric Dentistry, Peking University School and Hospital of Stomatology, No.22 Zhongguancun South Street, Zhongguancun, Beijing, 100081, P.R.China
| | - Minghua Liu
- Beijing National Laboratory for Molecular Science, Institute of Chemistry, Chinese Academy of Sciences, No.2, 1st North Street, Zhongguancun, Beijing, 100190, P.R.China
| |
Collapse
|