51
|
Li J, Luo L, He J, Yu J, Li X, Shen X, Zhang J, Li S, Karp JM, Kuai R. A Virus-Inspired Inhalable Liponanogel Induces Potent Antitumor Immunity and Regression in Metastatic Lung Tumors. Cancer Res 2024; 84:2352-2363. [PMID: 38718316 PMCID: PMC11247319 DOI: 10.1158/0008-5472.can-23-3414] [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: 10/31/2023] [Revised: 02/27/2024] [Accepted: 05/02/2024] [Indexed: 07/16/2024]
Abstract
Pulmonary delivery of immunostimulatory agents such as poly(I:C) to activate double-stranded RNA sensors MDA5 and RIG-I within lung-resident antigen-presenting cells is a potential strategy to enhance antitumor immunity by promoting type I interferon secretion. Nevertheless, following pulmonary delivery, poly(I:C) suffers from rapid degradation and poor endosomal escape, thus limiting its potency. Inspired by the structure of a virus that utilizes internal viral proteins to tune the loading and cytosolic delivery of viral nucleic acids, we developed a liponanogel (LNG)-based platform to overcome the delivery challenges of poly(I:C). The LNG comprised an anionic polymer hyaluronic acid-based nanogel core coated by a lipid shell, which served as a protective layer to stabilize the nanogel core in the lungs. The nanogel core was protonated within acidic endosomes to enhance the endosomal membrane permeability and cytosolic delivery of poly(I:C). After pulmonary delivery, LNG-poly(I:C) induced 13.7-fold more IFNβ than poly(I:C) alone and two-fold more than poly(I:C) loaded in the state-of-art lipid nanoparticles [LNP-poly(I:C)]. Additionally, LNG-poly(I:C) induced more potent CD8+ T-cell immunity and stronger therapeutic effects than LNP-poly(I:C). The combination of LNG-poly(I:C) and PD-L1 targeting led to regression of established lung metastases. Due to the ease of manufacturing and the high biocompatibility of LNG, pulmonary delivery of LNG may be broadly applicable to the treatment of different lung tumors and may spur the development of innovative strategies for cancer immunotherapy. Significance: Pulmonary delivery of poly(I:C) with a virus-inspired inhalable liponanogel strongly activates cytosolic MDA5 and RIG-I and stimulates antitumor immunity, representing a promising strategy for safe and effective treatment of metastatic lung tumors.
Collapse
Affiliation(s)
- Junyao Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Lanqing Luo
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Jia He
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Jinchao Yu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Xinyan Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Xueying Shen
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| | - Junxia Zhang
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
- School of Life Sciences, Tsinghua University, Beijing, China.
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing, China.
| | - Sai Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
- School of Life Sciences, Tsinghua University, Beijing, China.
- Frontier Research Center for Biological Structure & State Key Laboratory of Membrane Biology, Beijing, China.
| | - Jeffrey M. Karp
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts.
- Harvard-MIT Program in Health Sciences and Technology, MIT, Cambridge, Massachusetts.
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts.
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts.
| | - Rui Kuai
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
- Tsinghua-Peking Center for Life Sciences, Beijing, China.
| |
Collapse
|
52
|
Duc Nguyen H, Ardeshir A, Fonseca VA, Kim WK. Cluster of differentiation molecules in the metabolic syndrome. Clin Chim Acta 2024; 561:119819. [PMID: 38901629 DOI: 10.1016/j.cca.2024.119819] [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: 05/09/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024]
Abstract
Metabolic syndrome (MetS) represents a significant public health concern due to its association with an increased risk of cardiovascular disease, type 2 diabetes, and other serious health conditions. Despite extensive research, the underlying molecular mechanisms contributing to MetS pathogenesis remain elusive. This review aims to provide a comprehensive overview of the molecular mechanisms linking MetS and cluster of differentiation (CD) markers, which play critical roles in immune regulation and cellular signaling. Through an extensive literature review with a systematic approach, we examine the involvement of various CD markers in MetS development and progression, including their roles in adipose tissue inflammation, insulin resistance, dyslipidemia, and hypertension. Additionally, we discuss potential therapeutic strategies targeting CD markers for the management of MetS. By synthesizing current evidence, this review contributes to a deeper understanding of the complex interplay between immune dysregulation and metabolic dysfunction in MetS, paving the way for the development of novel therapeutic interventions.
Collapse
Affiliation(s)
- Hai Duc Nguyen
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA
| | - Amir Ardeshir
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Vivian A Fonseca
- Department Endocrinology Metabolism & Diabetes, Tulane University School of Medicine, New Orleans, LA, USA
| | - Woong-Ki Kim
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, LA, USA; Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, LA, USA.
| |
Collapse
|
53
|
Prasad R, Jyothi VGS, Kommineni N, Bulusu RT, Mendes B, Lovell JF, Conde J. Biomimetic Ghost Nanomedicine-Based Optotheranostics for Cancer. NANO LETTERS 2024; 24:8217-8231. [PMID: 38848540 PMCID: PMC11247544 DOI: 10.1021/acs.nanolett.4c01534] [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: 03/31/2024] [Revised: 05/31/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Theranostic medicine combines diagnostics and therapeutics, focusing on solid tumors at minimal doses. Optically activated photosensitizers are significant examples owing to their photophysical and chemical properties. Several optotheranostics have been tested that convert light to imaging signals, therapeutic radicals, and heat. Upon light exposure, conjugated photosensitizers kill tumor cells by producing reactive oxygen species and heat or by releasing cancer antigens. Despite clinical trials, these molecularly conjugated photosensitizers require protection from their surroundings and a localized direction for site-specific delivery during blood circulation. Therefore, cell membrane biomimetic ghosts have been proposed for precise and safe delivery of these optically active large molecules, which are clinically relevant because of their biocompatibility, long circulation time, bypass of immune cell recognition, and targeting ability. This review focuses on the role of biomimetic nanoparticles in the treatment and diagnosis of tumors through light-mediated diagnostics and therapy, providing insights into their preclinical and clinical status.
Collapse
Affiliation(s)
- Rajendra Prasad
- School
of Biochemical Engineering, Indian Institute
of Technology (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Vaskuri G. S. Jyothi
- Department
of Pharmaceutical Sciences, University of
Tennessee Health Science Center (UTHSC), Memphis, Tennessee 38163, United States
| | - Nagavendra Kommineni
- Center
for Biomedical Research, Population Council, New York, New York 10065, United States
| | - Ravi Teja Bulusu
- Department
of Pharmaceutical Sciences, Florida A&M
University, Tallahassee, Florida 32307, United States
| | - Bárbara
B. Mendes
- NOVA
Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon 1169-056, Portugal
- ToxOmics,
NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon, 1169-056, Portugal
| | - Jonathan F. Lovell
- Department
of Biomedical Engineering, University at Buffalo, State University of New York, Buffalo, New York 14260, United States
| | - João Conde
- NOVA
Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon 1169-056, Portugal
- ToxOmics,
NOVA Medical School|Faculdade de Ciências Médicas, NMS|FCM, Universidade NOVA de Lisboa, Lisbon, 1169-056, Portugal
| |
Collapse
|
54
|
Smith CT, Wang Z, Lewis JS. Engineering antigen-presenting cells for immunotherapy of autoimmunity. Adv Drug Deliv Rev 2024; 210:115329. [PMID: 38729265 DOI: 10.1016/j.addr.2024.115329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 03/05/2024] [Accepted: 05/03/2024] [Indexed: 05/12/2024]
Abstract
Autoimmune diseases are burdensome conditions that affect a significant fraction of the global population. The hallmark of autoimmune disease is a host's immune system being licensed to attack its tissues based on specific antigens. There are no cures for autoimmune diseases. The current clinical standard for treating autoimmune diseases is the administration of immunosuppressants, which weaken the immune system and reduce auto-inflammatory responses. However, people living with autoimmune diseases are subject to toxicity, fail to mount a sufficient immune response to protect against pathogens, and are more likely to develop infections. Therefore, there is a concerted effort to develop more effective means of targeting immunomodulatory therapies to antigen-presenting cells, which are involved in modulating the immune responses to specific antigens. In this review, we highlight approaches that are currently in development to target antigen-presenting cells and improve therapeutic outcomes in autoimmune diseases.
Collapse
Affiliation(s)
- Clinton T Smith
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Zhenyu Wang
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Jamal S Lewis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA.
| |
Collapse
|
55
|
Cavaillon JM, Chousterman BG, Skirecki T. Compartmentalization of the inflammatory response during bacterial sepsis and severe COVID-19. JOURNAL OF INTENSIVE MEDICINE 2024; 4:326-340. [PMID: 39035623 PMCID: PMC11258514 DOI: 10.1016/j.jointm.2024.01.001] [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: 08/27/2023] [Revised: 01/04/2024] [Accepted: 01/06/2024] [Indexed: 07/23/2024]
Abstract
Acute infections cause local and systemic disorders which can lead in the most severe forms to multi-organ failure and eventually to death. The host response to infection encompasses a large spectrum of reactions with a concomitant activation of the so-called inflammatory response aimed at fighting the infectious agent and removing damaged tissues or cells, and the anti-inflammatory response aimed at controlling inflammation and initiating the healing process. Fine-tuning at the local and systemic levels is key to preventing local and remote injury due to immune system activation. Thus, during bacterial sepsis and Coronavirus disease 2019 (COVID-19), concomitant systemic and compartmentalized pro-inflammatory and compensatory anti-inflammatory responses are occurring. Immune cells (e.g., macrophages, neutrophils, natural killer cells, and T-lymphocytes), as well as endothelial cells, differ from one compartment to another and contribute to specific organ responses to sterile and microbial insult. Furthermore, tissue-specific microbiota influences the local and systemic response. A better understanding of the tissue-specific immune status, the organ immunity crosstalk, and the role of specific mediators during sepsis and COVID-19 can foster the development of more accurate biomarkers for better diagnosis and prognosis and help to define appropriate host-targeted treatments and vaccines in the context of precision medicine.
Collapse
Affiliation(s)
| | - Benjamin G. Chousterman
- Department of Anesthesia and Critical Care, Lariboisière University Hospital, DMU Parabol, APHP Nord, Paris, France
- Inserm U942, University of Paris, Paris, France
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| |
Collapse
|
56
|
Wang Y, Shi J, Xin M, Kahkoska AR, Wang J, Gu Z. Cell-drug conjugates. Nat Biomed Eng 2024:10.1038/s41551-024-01230-6. [PMID: 38951139 DOI: 10.1038/s41551-024-01230-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 05/01/2024] [Indexed: 07/03/2024]
Abstract
By combining living cells with therapeutics, cell-drug conjugates can potentiate the functions of both components, particularly for applications in drug delivery and therapy. The conjugates can be designed to persist in the bloodstream, undergo chemotaxis, evade surveillance by the immune system, proliferate, or maintain or transform their cellular phenotypes. In this Review, we discuss strategies for the design of cell-drug conjugates with specific functions, the techniques for their preparation, and their applications in the treatment of cancers, autoimmune diseases and other pathologies. We also discuss the translational challenges and opportunities of this class of drug-delivery systems and therapeutics.
Collapse
Affiliation(s)
- Yanfang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Jiaqi Shi
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Jinhua Institute of Zhejiang University, Jinhua, China
| | - Minhang Xin
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Anna R Kahkoska
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jinqiang Wang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhen Gu
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Jinhua Institute of Zhejiang University, Jinhua, China.
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
- Liangzhu Laboratory, Hangzhou, China.
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
57
|
Desai N, Rana D, Salave S, Benival D, Khunt D, Prajapati BG. Achieving Endo/Lysosomal Escape Using Smart Nanosystems for Efficient Cellular Delivery. Molecules 2024; 29:3131. [PMID: 38999083 PMCID: PMC11243486 DOI: 10.3390/molecules29133131] [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: 06/06/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
The delivery of therapeutic agents faces significant hurdles posed by the endo-lysosomal pathway, a bottleneck that hampers clinical effectiveness. This comprehensive review addresses the urgent need to enhance cellular delivery mechanisms to overcome these obstacles. It focuses on the potential of smart nanomaterials, delving into their unique characteristics and mechanisms in detail. Special attention is given to their ability to strategically evade endosomal entrapment, thereby enhancing therapeutic efficacy. The manuscript thoroughly examines assays crucial for understanding endosomal escape and cellular uptake dynamics. By analyzing various assessment methods, we offer nuanced insights into these investigative approaches' multifaceted aspects. We meticulously analyze the use of smart nanocarriers, exploring diverse mechanisms such as pore formation, proton sponge effects, membrane destabilization, photochemical disruption, and the strategic use of endosomal escape agents. Each mechanism's effectiveness and potential application in mitigating endosomal entrapment are scrutinized. This paper provides a critical overview of the current landscape, emphasizing the need for advanced delivery systems to navigate the complexities of cellular uptake. Importantly, it underscores the transformative role of smart nanomaterials in revolutionizing cellular delivery strategies, leading to a paradigm shift towards improved therapeutic outcomes.
Collapse
Affiliation(s)
- Nimeet Desai
- Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India;
| | - Dhwani Rana
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (D.B.)
| | - Sagar Salave
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (D.B.)
| | - Derajram Benival
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (D.B.)
| | - Dignesh Khunt
- School of Pharmacy, Gujarat Technological University, Gandhinagar 382027, Gujarat, India
| | - Bhupendra G. Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva 384012, Gujarat, India
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| |
Collapse
|
58
|
Smidt JM, Märcher A, Skaanning MK, El-Chami K, Teodori L, Omer M, Kjems J, Gothelf KV. Dual-Targeting of the HER2 Cancer Receptor with an Antibody-Directed Enzyme and a Nanobody-Guided MMAE Prodrug Scaffold. Chembiochem 2024:e202400437. [PMID: 38945824 DOI: 10.1002/cbic.202400437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/02/2024]
Abstract
Antibody-enzyme conjugates have shown potential as tissue-specific prodrug activators by antibody-directed enzyme prodrug therapy (ADEPT), but the approach met challenges clinically due to systemic drug release. Here, we report a novel dual-targeting ADEPT system (DuADEPT) which is based on active cancer receptor targeting of both a trastuzumab-sialidase conjugate (Tz-Sia) and a highly potent sialidase-activated monomethyl auristatin E (MMAE) prodrug scaffold. The scaffold is based on a four-way junction of the artificial nucleic acid analog acyclic (L)-threoninol nucleic acid ((L)-aTNA) which at the ends of its four arms carries one nanobody targeting HER2 and three copies of the prodrug. Dual-targeting of the constructs to two proximal epitopes of HER2 was shown by flow cytometry, and a dual-targeted enzymatic drug release assay revealed cytotoxicity upon prodrug activation specifically for HER2-positive cancer cells. The specific delivery and activation of prodrugs in this way could potentially be used to decrease systemic side effects and increase drug efficacy, and utilization of Tz-Sia provides an opportunity to combine the local chemotherapeutic effect of the DuADEPT with an anticancer immune response.
Collapse
Affiliation(s)
- Jakob Melgaard Smidt
- Interdisciplinary Nanoscience Center (iNANO) and Department of Chemistry, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Anders Märcher
- Interdisciplinary Nanoscience Center (iNANO) and Department of Chemistry, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Mads Koch Skaanning
- Interdisciplinary Nanoscience Center (iNANO) and Department of Chemistry, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Kassem El-Chami
- Interdisciplinary Nanoscience Center (iNANO) and Department of Chemistry, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Laura Teodori
- Interdisciplinary Nanoscience Center (iNANO) and Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Marjan Omer
- Interdisciplinary Nanoscience Center (iNANO) and Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Jørgen Kjems
- Interdisciplinary Nanoscience Center (iNANO) and Department of Molecular Biology and Genetics, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| | - Kurt V Gothelf
- Interdisciplinary Nanoscience Center (iNANO) and Department of Chemistry, Aarhus University, Gustav Wieds Vej 14, 8000, Aarhus, Denmark
| |
Collapse
|
59
|
Baig MS, Ahmad A, Pathan RR, Mishra RK. Precision Nanomedicine with Bio-Inspired Nanosystems: Recent Trends and Challenges in Mesenchymal Stem Cells Membrane-Coated Bioengineered Nanocarriers in Targeted Nanotherapeutics. J Xenobiot 2024; 14:827-872. [PMID: 39051343 PMCID: PMC11270309 DOI: 10.3390/jox14030047] [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: 03/17/2024] [Revised: 05/09/2024] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
In the recent past, the formulation and development of nanocarriers has been elaborated into the broader fields and opened various avenues in their preclinical and clinical applications. In particular, the cellular membrane-based nanoformulations have been formulated to surpass and surmount the limitations and restrictions associated with naïve or free forms of therapeutic compounds and circumvent various physicochemical and immunological barriers including but not limited to systemic barriers, microenvironmental roadblocks, and other cellular or subcellular hinderances-which are quite heterogeneous throughout the diseases and patient cohorts. These limitations in drug delivery have been overcome through mesenchymal cells membrane-based precision therapeutics, where these interventions have led to the significant enhancements in therapeutic efficacies. However, the formulation and development of nanocarriers still focuses on optimization of drug delivery paradigms with a one-size-fits-all resolutions. As mesenchymal stem cell membrane-based nanocarriers have been engineered in highly diversified fashions, these are being optimized for delivering the drug payloads in more and better personalized modes, entering the arena of precision as well as personalized nanomedicine. In this Review, we have included some of the advanced nanocarriers which have been designed and been utilized in both the non-personalized as well as precision applicability which can be employed for the improvements in precision nanotherapeutics. In the present report, authors have focused on various other aspects of the advancements in stem cells membrane-based nanoparticle conceptions which can surmount several roadblocks and barriers in drug delivery and nanomedicine. It has been suggested that well-informed designing of these nanocarriers will lead to appreciable improvements in the therapeutic efficacy in therapeutic payload delivery applications. These approaches will also enable the tailored and customized designs of MSC-based nanocarriers for personalized therapeutic applications, and finally amending the patient outcomes.
Collapse
Affiliation(s)
- Mirza Salman Baig
- Anjuman-I-Islam Kalsekar Technical Campus School of Pharmacy, Sector-16, Near Thana Naka, Khandagao, New Panvel, Navi Mumbai 410206, Maharashtra, India;
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC), Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, Foothills Medical Centre, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | - Rakesh Kumar Mishra
- School of Health Sciences and Technology, University of Petroleum and Energy Studies (UPES), Bidholi, Dehradun 248007, Uttarakhand, India;
| |
Collapse
|
60
|
Dhingra S, Goyal S, Thirumal D, Sharma P, Kaur G, Mittal N. Mesoporous silica nanoparticles: a versatile carrier platform in lung cancer management. Nanomedicine (Lond) 2024; 19:1331-1346. [PMID: 39105754 PMCID: PMC11318747 DOI: 10.1080/17435889.2024.2348438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/24/2024] [Indexed: 08/07/2024] Open
Abstract
Mesoporous silica nanoparticles (MSNPs) are inorganic nanoparticles that have been comprehensively investigated and are intended to deliver therapeutic agents. MSNPs have revolutionized the therapy for various conditions, especially cancer and infectious diseases. In this article, the viability of MSNPs' administration for lung cancer therapy has been reviewed. However, certain challenges lay ahead in the successful translation such as toxicology, immunology, large-scale production, and regulatory matters have made it extremely difficult to translate such discoveries from the bench to the bedside. This review highlights recent developments, characteristics, mechanism of action and customization for targeted delivery. This review also covers the most recent data that sheds light on MSNPs' extraordinary therapeutic potential in fighting lung cancer as well as future hurdles.
Collapse
Affiliation(s)
- Smriti Dhingra
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Shuchi Goyal
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Divya Thirumal
- Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, 576104,India
| | - Preety Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| | - Gurpreet Kaur
- Department of Pharmaceutical Sciences & Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Neeraj Mittal
- Chitkara College of Pharmacy, Chitkara University, Punjab, 140401, India
| |
Collapse
|
61
|
Fang T, Chen G. Non-viral vector-based genome editing for cancer immunotherapy. Biomater Sci 2024; 12:3068-3085. [PMID: 38716572 DOI: 10.1039/d4bm00286e] [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: 06/12/2024]
Abstract
Despite the exciting promise of cancer immunotherapy in the clinic, immune checkpoint blockade therapy and T cell-based therapies are often associated with low response rates, intrinsic and adaptive immune resistance, and systemic side effects. CRISPR-Cas-based genome editing appears to be an effective strategy to overcome these unmet clinical needs. As a safer delivery platform for the CRISPR-Cas system, non-viral nanoformulations have been recently explored to target tumor cells and immune cells, aiming to improve cancer immunotherapy on a gene level. In this review, we summarized the efforts of non-viral vector-based CRISPR-Cas-mediated genome editing in tumor cells and immune cells for cancer immunotherapy. Their design rationale and specific applications were highlighted.
Collapse
Affiliation(s)
- Tianxu Fang
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, Montreal, QC, H3G 0B1, Canada.
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, H3G 0B1, Canada
| |
Collapse
|
62
|
S. V. S, Augustine D, Mushtaq S, Baeshen HA, Ashi H, Hassan RN, Alshahrani M, Patil S. Revitalizing oral cancer research: Crispr-Cas9 technology the promise of genetic editing. Front Oncol 2024; 14:1383062. [PMID: 38915370 PMCID: PMC11194394 DOI: 10.3389/fonc.2024.1383062] [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: 02/06/2024] [Accepted: 05/08/2024] [Indexed: 06/26/2024] Open
Abstract
This review presents an in-depth analysis of the immense potential of CRISPR-Cas9 technology in revolutionizing oral cancer research. It underscores the inherent limitations of conventional treatments while emphasizing the pressing need for groundbreaking approaches. The unparalleled capability of CRISPR-Cas9 to precisely target and modify specific genes involved in cancer progression heralds a new era in therapeutic intervention. Employing genome-wide CRISPR screens, vulnerabilities in oral cancer cells can be identified, thereby unravelling promising targets for therapeutic interventions. In the realm of oral cancer, the disruptive power of CRISPR-Cas9 manifests through its capacity to perturb genes that are intricately associated with drug resistance, consequently augmenting the efficacy of chemotherapy. To address the challenges that arise, this review diligently examines pertinent issues such as off-target effects, efficient delivery mechanisms, and the ethical considerations surrounding germline editing. Through precise gene editing, facilitated by CRISPR/Cas9, it becomes possible to overcome drug resistance by rectifying mutations, thereby enhancing the efficacy of personalized treatment strategies. This review delves into the prospects of CRISPR-Cas9, illuminating its potential applications in the domains of medicine, agriculture, and biotechnology. It is paramount to emphasize the necessity of ongoing research endeavors and the imperative to develop targeted therapies tailored specifically for oral cancer. By embracing this comprehensive overview, we can pave the way for ground-breaking treatments that instill renewed hope for enhanced outcomes in individuals afflicted by oral cancer.
Collapse
Affiliation(s)
- Sowmya S. V.
- Department of Oral Pathology and Microbiology, Faculty of Dental Sciences, MS Ramaiah University of Applied Sciences, Bengaluru, Karnataka, India
| | - Dominic Augustine
- Department of Oral Pathology and Microbiology, Faculty of Dental Sciences, MS Ramaiah University of Applied Sciences, Bengaluru, Karnataka, India
| | - Shazia Mushtaq
- College of Applied Medical Sciences, Dental Health Department, King Saud University, Riyadh, Saudi Arabia
| | - Hosam Ali Baeshen
- Department of Orthodontics, Faculty of Dentistry, King Abdulziz University, Jeddah, Saudi Arabia
| | - Heba Ashi
- Department of Dental Public Health, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Reem Nabil Hassan
- Biological Sciences Department (Genome), Faculty of Sciences, King Abdul-Aziz University, Jeddah, Saudi Arabia
| | - Mohammed Alshahrani
- Endodontic Department, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shankargouda Patil
- College of Dental Medicine, Roseman University of Health Sciences, South Jordan, UT, United States
| |
Collapse
|
63
|
Janes ME, Park KS, Gottlieb AP, Curreri A, Adebowale K, Kim J, Mitragotri S. Dendritic Cell Immune Modulation via Polyphenol Membrane Coatings. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28070-28079. [PMID: 38779939 DOI: 10.1021/acsami.4c01575] [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: 05/25/2024]
Abstract
Cellular hitchhiking is an emerging strategy for the in vivo control of adoptively transferred immune cells. Hitchhiking approaches are primarily mediated by adhesion of nano and microparticles to the cell membrane, which conveys an ability to modulate transferred cells via local drug delivery. Although T cell therapies employing this strategy have progressed into the clinic, phagocytic cells including dendritic cells (DCs) are much more challenging to engineer. DC vaccines hold great potential for a spectrum of diseases, and the combination drug delivery is an attractive strategy to manipulate their function and overcome in vivo plasticity. However, DCs are not compatible with current hitchhiking approaches due to their broad phagocytic capacity. In this work, we developed and validated META (membrane engineering using tannic acid) to enable DC cellular hitchhiking for the first time. META employs the polyphenol tannic acid (TA) to facilitate supramolecular assembly of protein drug cargoes on the cell membrane, enabling the creation of cell surface-bound formulations for local drug delivery to carrier DCs. We optimized META formulations to incorporate and release protein cargoes with varying physical properties alone and in combination and to preserve DC viability and critical functions such as migration. We further show that META loaded with either a pro- or anti-inflammatory cargo can influence the carrier cell phenotype, thus demonstrating the flexibility of the approach for applications from cancer to autoimmune disease. Overall, this approach illustrates a new platform for the local control of phagocytic immune cells as a next step to advance DC therapies in the clinic.
Collapse
Affiliation(s)
- Morgan E Janes
- Harvard-MIT Division of Health Sciences & Technology, Cambridge, Massachusetts 02139, United States
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| | - Kyung Soo Park
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| | - Alexander P Gottlieb
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| | - Alexander Curreri
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| | - Kolade Adebowale
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| | - Jayoung Kim
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied Sciences, Allston, Massachusetts 02134, United States
- Wyss Institute of Biologically Inspired Engineering, Boston, Massachusetts 02215, United States
| |
Collapse
|
64
|
Naser IH, Zaid M, Ali E, Jabar HI, Mustafa AN, Alubiady MHS, Ramadan MF, Muzammil K, Khalaf RM, Jalal SS, Alawadi AH, Alsalamy A. Unveiling innovative therapeutic strategies and future trajectories on stimuli-responsive drug delivery systems for targeted treatment of breast carcinoma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3747-3770. [PMID: 38095649 DOI: 10.1007/s00210-023-02885-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/02/2023] [Indexed: 05/23/2024]
Abstract
This comprehensive review delineates the latest advancements in stimuli-responsive drug delivery systems engineered for the targeted treatment of breast carcinoma. The manuscript commences by introducing mammary carcinoma and the current therapeutic methodologies, underscoring the urgency for innovative therapeutic strategies. Subsequently, it elucidates the logic behind the employment of stimuli-responsive drug delivery systems, which promise targeted drug administration and the minimization of adverse reactions. The review proffers an in-depth analysis of diverse types of stimuli-responsive systems, including thermoresponsive, pH-responsive, and enzyme-responsive nanocarriers. The paramount importance of material choice, biocompatibility, and drug loading strategies in the design of these systems is accentuated. The review explores characterization methodologies for stimuli-responsive nanocarriers and probes preclinical evaluations of their efficacy, toxicity, pharmacokinetics, and biodistribution in mammary carcinoma models. Clinical applications of stimuli-responsive systems, ongoing clinical trials, the potential of combination therapies, and the utility of multifunctional nanocarriers for the co-delivery of assorted drugs and therapies are also discussed. The manuscript addresses the persistent challenge of drug resistance in mammary carcinoma and the potential of stimuli-responsive systems in surmounting it. Regulatory and safety considerations, including FDA guidelines and biocompatibility assessments, are outlined. The review concludes by spotlighting future trajectories and emergent technologies in stimuli-responsive drug delivery, focusing on pioneering approaches, advancements in nanotechnology, and personalized medicine considerations. This review aims to serve as a valuable compendium for researchers and clinicians interested in the development of efficacious and safe stimuli-responsive drug delivery systems for the treatment of breast carcinoma.
Collapse
Affiliation(s)
- Israa Habeeb Naser
- Medical Laboratories Techniques Department, AL-Mustaqbal University, Hillah, Babil, Iraq
| | - Muhaned Zaid
- Department of Pharmacy, Al-Manara College for Medical Sciences, Maysan, Amarah, Iraq
| | - Eyhab Ali
- Al-Zahraa University for Women, Karbala, Iraq
| | - Hayder Imad Jabar
- Department of Pharmaceutics, College of Pharmacy, University of Al-Ameed, Karbala, Iraq
| | | | | | | | - Khursheed Muzammil
- Department of Public Health, College of Applied Medical Sciences, Khamis Mushait Campus, King Khalid University, Abha, Saudi Arabia
| | | | - Sarah Salah Jalal
- College of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Ahmed Hussien Alawadi
- College of Technical Engineering, the Islamic University, Najaf, Iraq
- College of Technical Engineering, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, the Islamic University of Babylon, Babylon, Iraq
| | - Ali Alsalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, Iraq.
| |
Collapse
|
65
|
Wang Y, Zhang H, Qiang H, Li M, Cai Y, Zhou X, Xu Y, Yan Z, Dong J, Gao Y, Pan C, Yin X, Gao J, Zhang T, Yu Z. Innovative Biomaterials for Bone Tumor Treatment and Regeneration: Tackling Postoperative Challenges and Charting the Path Forward. Adv Healthc Mater 2024; 13:e2304060. [PMID: 38429938 DOI: 10.1002/adhm.202304060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 02/24/2024] [Indexed: 03/03/2024]
Abstract
Surgical resection of bone tumors is the primary approach employed in the treatment of bone cancer. Simultaneously, perioperative interventions, particularly postoperative adjuvant anticancer strategies, play a crucial role in achieving satisfactory therapeutic outcomes. However, the occurrence of postoperative bone tumor recurrence, metastasis, extensive bone defects, and infection are significant risks that can result in unfavorable prognoses or even treatment failure. In recent years, there has been significant progress in the development of biomaterials, leading to the emergence of new treatment options for bone tumor therapy and bone regeneration. This progress report aims to comprehensively analyze the strategic development of unique therapeutic biomaterials with inherent healing properties and bioactive capabilities for bone tissue regeneration. These composite biomaterials, classified into metallic, inorganic non-metallic, and organic types, are thoroughly investigated for their responses to external stimuli such as light or magnetic fields, internal interventions including chemotherapy or catalytic therapy, and combination therapy, as well as their role in bone regeneration. Additionally, an overview of self-healing materials for osteogenesis is provided and their potential applications in combating osteosarcoma and promoting bone formation are explored. Furthermore, the safety concerns of integrated materials and current limitations are addressed, while also discussing the challenges and future prospects.
Collapse
Affiliation(s)
- Yu Wang
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, P. R. China
| | - Huaiyuan Zhang
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, P. R. China
| | - Huifen Qiang
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, P. R. China
| | - Meigui Li
- School of Pharmacy, Henan University, Kaifeng City, Henan, 475004, P. R. China
| | - Yili Cai
- Department of Gastroenterology, Naval Medical Center, Naval Medical University, Shanghai, 200052, P. R. China
| | - Xuan Zhou
- School of Pharmacy, Henan University, Kaifeng City, Henan, 475004, P. R. China
| | - Yanlong Xu
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, P. R. China
| | - Zhenzhen Yan
- Department of Burn Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
| | - Jinhua Dong
- The Women and Children Hospital Affiliated to Jiaxing University, Jiaxing, Zhejiang, 314000, P. R. China
| | - Yuan Gao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200433, P. R. China
| | - Chengye Pan
- Department of Gastroenterology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
| | - Xiaojing Yin
- Department of Gastroenterology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
| | - Jie Gao
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, P. R. China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, P. R. China
| | - Zuochong Yu
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, P. R. China
| |
Collapse
|
66
|
Wang T, Tan P, Tang Q, Zhou C, Ding Y, Xu S, Song M, Fu H, Zhang Y, Zhang X, Bai Y, Sun Z, Ma X. Phage-displayed heptapeptide sequence conjugation significantly improves the specific targeting ability of antimicrobial peptides against Staphylococcus aureus. MLIFE 2024; 3:251-268. [PMID: 38948143 PMCID: PMC11211671 DOI: 10.1002/mlf2.12123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 03/04/2024] [Accepted: 03/05/2024] [Indexed: 07/02/2024]
Abstract
Broad-spectrum antibacterial drugs often lack specificity, leading to indiscriminate bactericidal activity, which can disrupt the normal microbial balance of the host flora and cause unnecessary cytotoxicity during systemic administration. In this study, we constructed a specifically targeted antimicrobial peptide against Staphylococcus aureus by introducing a phage-displayed peptide onto a broad-spectrum antimicrobial peptide and explored its structure-function relationship through one-factor modification. SFK2 obtained by screening based on the selectivity index and the targeting index showed specific killing ability against S. aureus. Moreover, SFK2 showed excellent biocompatibility in mice and piglet, and demonstrated significant therapeutic efficacy against S. aureus infection. In conclusion, our screening of phage-derived heptapeptides effectively enhances the specific bactericidal ability of the antimicrobial peptides against S. aureus, providing a theoretical basis for developing targeted antimicrobial peptides.
Collapse
Affiliation(s)
- Tao Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
- Luoyang Key Laboratory of Animal Genetic and Breeding, College of Animal ScienceHenan University of Science and TechnologyLuoyangChina
| | - Peng Tan
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Qi Tang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Chenlong Zhou
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Yakun Ding
- Key Laboratory of Innovative Utilization of Indigenous Cattle and Sheep Germplasm Resources (Co‐construction by Ministry and Province), Ministry of Agriculture and Rural AffairsZhengzhou UniversityZhengzhouChina
| | - Shenrui Xu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Mengda Song
- Key Laboratory of Innovative Utilization of Indigenous Cattle and Sheep Germplasm Resources (Co‐construction by Ministry and Province), Ministry of Agriculture and Rural AffairsZhengzhou UniversityZhengzhouChina
| | - Huiyang Fu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Yucheng Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Xiaohui Zhang
- Luoyang Key Laboratory of Animal Genetic and Breeding, College of Animal ScienceHenan University of Science and TechnologyLuoyangChina
| | - Yueyu Bai
- Key Laboratory of Innovative Utilization of Indigenous Cattle and Sheep Germplasm Resources (Co‐construction by Ministry and Province), Ministry of Agriculture and Rural AffairsZhengzhou UniversityZhengzhouChina
- Animal Health Supervision in Henan ProvinceZhengzhouChina
| | - Zhihong Sun
- Laboratory for Bio‐Feed and Molecular Nutrition, Department of Animal Science and TechnologySouthwest UniversityChongqingChina
| | - Xi Ma
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| |
Collapse
|
67
|
Sun T, Zhao H, Hu L, Shao X, Lu Z, Wang Y, Ling P, Li Y, Zeng K, Chen Q. Enhanced optical imaging and fluorescent labeling for visualizing drug molecules within living organisms. Acta Pharm Sin B 2024; 14:2428-2446. [PMID: 38828150 PMCID: PMC11143489 DOI: 10.1016/j.apsb.2024.01.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/07/2024] [Accepted: 01/25/2024] [Indexed: 06/05/2024] Open
Abstract
The visualization of drugs in living systems has become key techniques in modern therapeutics. Recent advancements in optical imaging technologies and molecular design strategies have revolutionized drug visualization. At the subcellular level, super-resolution microscopy has allowed exploration of the molecular landscape within individual cells and the cellular response to drugs. Moving beyond subcellular imaging, researchers have integrated multiple modes, like optical near-infrared II imaging, to study the complex spatiotemporal interactions between drugs and their surroundings. By combining these visualization approaches, researchers gain supplementary information on physiological parameters, metabolic activity, and tissue composition, leading to a comprehensive understanding of drug behavior. This review focuses on cutting-edge technologies in drug visualization, particularly fluorescence imaging, and the main types of fluorescent molecules used. Additionally, we discuss current challenges and prospects in targeted drug research, emphasizing the importance of multidisciplinary cooperation in advancing drug visualization. With the integration of advanced imaging technology and molecular design, drug visualization has the potential to redefine our understanding of pharmacology, enabling the analysis of drug micro-dynamics in subcellular environments from new perspectives and deepening pharmacological research to the levels of the cell and organelles.
Collapse
Affiliation(s)
- Ting Sun
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Huanxin Zhao
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Luyao Hu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xintian Shao
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- School of Life Sciences, Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Zhiyuan Lu
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Yuli Wang
- Tianjin Pharmaceutical DA REN TANG Group Corporation Limited Traditional Chinese Pharmacy Research Institute, Tianjin 300457, China
- Key Laboratory of Systems Bioengineering (Ministry of Education), School of Chemistry Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Peixue Ling
- Institute of Biochemical and Biotechnological Drugs, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
- Key Laboratory of Biopharmaceuticals, Postdoctoral Scientific Research Workstation, Shandong Academy of Pharmaceutical Science, Jinan 250098, China
| | - Yubo Li
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Kewu Zeng
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Qixin Chen
- School of Pharmaceutical Sciences, National Key Laboratory of Advanced Drug Delivery System, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore 119074, Singapore
| |
Collapse
|
68
|
Bai H, Huang W, Li J, Ji Y, He S, Hu H. Enhancing anticancer treatment: Development of cRGD-Conjugated F-OH-Evo prodrugs for targeted delivery. Bioorg Med Chem 2024; 107:117759. [PMID: 38795572 DOI: 10.1016/j.bmc.2024.117759] [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: 02/29/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/28/2024]
Abstract
Small molecule drugs sourced from natural products are pivotal for novel therapeutic discoveries. However, their clinical deployment is often impeded by non-specific activity and severe adverse effects. This study focused on 3-fluoro-10-hydroxy-Evodiamine (F-OH-Evo), a potent derivative of Evodiamine, whose development is curtailed due to suboptimal tumor selectivity and heightened cytotoxicity. By harnessing the remarkable stability, specificity, and αvβ3 integrin affinity of c(RGDFK), a novel prodrug by conjugating F-OH-Evo with cRGD was synthesized. This innovative prodrug substantially enhanced the tumor-specific targeting of F-OH-Evo and improved the anti-tumor activities. Among them, compound 3c demonstrated the best selective inhibitory activity toward U87 cancer cells in vitro. It selectively enterd U87 cells by binding to αvβ3 integrin, releasing the parent molecule under the dual response of ROS and GSH to exert inhibitory activity on topo I. The results highlight the potential of cRGD-conjugated prodrugs in targeted cancer therapy. This approach signifies a significant advancement in developing safer and more effective chemotherapy drugs, emphasizing the role of prodrug strategies in overcoming the limitations of traditional cancer treatments.
Collapse
Affiliation(s)
- Haohao Bai
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China
| | - Wenjing Huang
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China
| | - Jinqiu Li
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China
| | - Yajing Ji
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China
| | - Shipeng He
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China.
| | - Honggang Hu
- Institute of Translational Medicine, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai 200444, PR China.
| |
Collapse
|
69
|
Liu R, Hou W, Li J, Gou X, Gao M, Wang H, Zhang Y, Deng H, Yang X, Zhang W. Co-assembly of cisplatin and dasatinib in hyaluronan nanogel to combat triple negative breast cancer with reduced side effects. Int J Biol Macromol 2024; 269:132074. [PMID: 38705320 DOI: 10.1016/j.ijbiomac.2024.132074] [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: 01/29/2024] [Revised: 04/14/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Treatment for triple negative breast cancer (TNBC) remains a huge challenge due to the lack of targeted therapeutics and tumor heterogenicity. Cisplatin (Cis) have demonstrated favorable therapeutic response in TNBC and thus is used together with various kinase inhibitors to fight the heterogenicity of TNBC. The combination of Cis with SRC inhibitor dasatinib (DAS) has shown encouraging anti-TNBC efficacy although the additive toxicity was commonly observed. To overcome the severe side effects of this Cis involved therapy, here we co-encapsulated Cis and DAS into a self-assembled hyaluronan (HA) nanogel (designated as HA/Cis/DAS (HCD) nanogel) to afford the TNBC targeted delivery by using the 4T1 mouse model. The acquired HCD nanogel was around 181 nm in aqueous solution, demonstrating the pharmacological activities of both Cis and DAS. Taking advantages of HA's targeting capability towards CD44 that is overexpressed on many TNBC cells, the HCD could well maintain the anticancer efficacy of the Cis and DAS combination, significantly increase the maximum tolerated dose and relieve the renal toxicity in vivo. The current HCD nanogel provides a potent strategy to improve the therapeutic outcome of Cis and DAS combination and thus representing a new targeted treatment option for TNBC.
Collapse
Affiliation(s)
- Runmeng Liu
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Wei Hou
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Jiayi Li
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Xiaorong Gou
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Menghan Gao
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Huimin Wang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Yiyi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Hong Deng
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China
| | - Xue Yang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, PR China.
| | - Weiqi Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, PR China.
| |
Collapse
|
70
|
Huster D, Maiti S, Herrmann A. Phospholipid Membranes as Chemically and Functionally Tunable Materials. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312898. [PMID: 38456771 DOI: 10.1002/adma.202312898] [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: 11/29/2023] [Revised: 02/12/2024] [Indexed: 03/09/2024]
Abstract
The sheet-like lipid bilayer is the fundamental structural component of all cell membranes. Its building blocks are phospholipids and cholesterol. Their amphiphilic structure spontaneously leads to the formation of a bilayer in aqueous environment. Lipids are not just structural elements. Individual lipid species, the lipid membrane structure, and lipid dynamics influence and regulate membrane protein function. An exciting field is emerging where the membrane-associated material properties of different bilayer systems are used in designing innovative solutions for widespread applications across various fields, such as the food industry, cosmetics, nano- and biomedicine, drug storage and delivery, biotechnology, nano- and biosensors, and computing. Here, the authors summarize what is known about how lipids determine the properties and functions of biological membranes and how this has been or can be translated into innovative applications. Based on recent progress in the understanding of membrane structure, dynamics, and physical properties, a perspective is provided on how membrane-controlled regulation of protein functions can extend current applications and even offer new applications.
Collapse
Affiliation(s)
- Daniel Huster
- Institute of Medical Physics and Biophysics, University of Leipzig, Härtelstr. 16/18, D-04107, Leipzig, Germany
| | - Sudipta Maiti
- Department of Chemical Sciences, Tata Institute of Fundamental Research, Homi Bhabha Road, Colaba, Mumbai, 400 005, India
| | - Andreas Herrmann
- Freie Universität Berlin, Department Chemistry and Biochemistry, SupraFAB, Altensteinstr. 23a, D-14195, Berlin, Germany
| |
Collapse
|
71
|
Lin J, Lin Z, Liu L, Lin W, Xie X, Zhang X. Enhancing glioma-specific drug delivery through self-assembly of macrophage membrane and targeted polymer assisted by low-frequency ultrasound irradiation. Mater Today Bio 2024; 26:101067. [PMID: 38706730 PMCID: PMC11068854 DOI: 10.1016/j.mtbio.2024.101067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 05/07/2024] Open
Abstract
The blood-brain Barrier (BBB), combined with immune clearance, contributes to the low efficacy of drug delivery and suboptimal treatment outcomes in glioma. Here, we propose a novel approach that combines the self-assembly of mouse bone marrow-derived macrophage membrane with a targeted positive charge polymer (An-PEI), along with low-frequency ultrasound (LFU) irradiation, to achieve efficient and safe therapy for glioma. Our findings demonstrate the efficacy of a charge-induced self-assembly strategy, resulting in a stable co-delivery nanosystem with a high drug loading efficiency of 44.2 %. Moreover, this structure triggers a significant release of temozolomide in the acidic environment of the tumor microenvironment. Additionally, the macrophage membrane coating expresses Spyproteins, which increase the amount of An-BMP-TMZ that can evade the immune system by 40 %, while LFU irradiation treatment facilitates the opening of the BBB, allowing for enormously increased entry of An-BMP-TMZ (approximately 400 %) into the brain. Furthermore, after crossing the BBB, the Angiopep-2 peptide-modified An-BMP-TMZ exhibits the ability to selectively target glioma cells. These advantages result in an obvious tumor inhibition effect in animal experiments and significantly improve the survival of glioma-bearing mice. These results suggest that combining the macrophage membrane-coated drug delivery system with LFU irradiation offers a feasible approach for the accurate, efficient and safe treatment of brain disease.
Collapse
Affiliation(s)
- Junqing Lin
- Department of Interventional Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Zhenhu Lin
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Leilei Liu
- Department of Ultrasound, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, 350001, Fujian, China
| | - Wenjin Lin
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| | - Xiaodong Xie
- Fujian-Taiwan-Hongkong-Macao Science and Technology Cooperation Base of Intelligent Pharmaceutics, College of Materials and Chemical Engineering, Minjiang University, Fuzhou, 350001, Fujian, China
| | - Xiujuan Zhang
- Department of Ultrasound, Fujian Medical University Union Hospital, Fuzhou, 350001, Fujian, China
| |
Collapse
|
72
|
Zhou M, Wang Y, Xia Y, Li Y, Bao J, Zhang Y, Cheng J, Shi Y. MRI-guided cell membrane-camouflaged bimetallic coordination nanoplatform for combined tumor phototherapy. Mater Today Bio 2024; 26:101019. [PMID: 38516170 PMCID: PMC10950690 DOI: 10.1016/j.mtbio.2024.101019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/18/2024] [Accepted: 03/05/2024] [Indexed: 03/23/2024] Open
Abstract
Nanotechnology for tumor diagnosis and optical therapy has attracted widespread interest due to its low toxicity and convenience but is severely limited due to uncontrollable tumor targeting. In this work, homologous cancer cell membrane-camouflaged multifunctional hybrid metal coordination nanoparticles (DRu/Gd@CM) were prepared for MRI-guided photodynamic therapy (PDT) and photothermal therapy (PTT) of tumors. Bimetallic coordination nanoparticles are composed of three functional modules: dopamine, Ru(dcbpy)3Cl2 and GdCl3, which are connected through 1,4-Bis[(1H-imidazole-1-yl)methyl]benzene (BIX). Their morphology can be easily controlled by adjusting the ratio of precursors. Optimistically, the intrinsic properties of the precursors, including the photothermal properties of polydopamine (PDA), the magnetic resonance (MR) response of Gd3+, and the singlet oxygen generation of Ru(dcbpy)3Cl2, are well preserved in the hybrid metal nanoparticles. Furthermore, the targeting of homologous cancer cell membranes enables these coordinated nanoparticles to precisely target tumor cells. The MR imaging capabilities and the combination of PDT and PTT were demonstrated in in vitro experiments. In addition, in vivo experiments indicated that the nanoplatform showed excellent tumor accumulation and therapeutic effects on mice with subcutaneous tumors, and could effectively eliminate tumors within 14 days. Therefore, it expanded the new horizon for the preparation of modular nanoplatform and imaging-guided optical therapy of tumors.
Collapse
Affiliation(s)
| | | | - Yaning Xia
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou, 450052, China
| | - Yinhua Li
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou, 450052, China
| | - Jianfeng Bao
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou, 450052, China
| | - Yong Zhang
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou, 450052, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou, 450052, China
| | - Yupeng Shi
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Henan Key Laboratory of Functional Magnetic Resonance Imaging and Molecular Imaging, Zhengzhou, 450052, China
| |
Collapse
|
73
|
Jones Lipinski RA, Stancill JS, Nuñez R, Wynia-Smith SL, Sprague DJ, Nord JA, Bird A, Corbett JA, Smith BC. Zinc-chelating BET bromodomain inhibitors equally target islet endocrine cell types. Am J Physiol Regul Integr Comp Physiol 2024; 326:R515-R527. [PMID: 38618911 PMCID: PMC11381023 DOI: 10.1152/ajpregu.00259.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 03/19/2024] [Accepted: 04/07/2024] [Indexed: 04/16/2024]
Abstract
Inhibition of the bromodomain and extraterminal domain (BET) protein family is a potential strategy to prevent and treat diabetes; however, the clinical use of BET bromodomain inhibitors (BETis) is associated with adverse effects. Here, we explore a strategy for targeting BETis to β cells by exploiting the high-zinc (Zn2+) concentration in β cells relative to other cell types. We report the synthesis of a novel, Zn2+-chelating derivative of the pan-BETi (+)-JQ1, (+)-JQ1-DPA, in which (+)-JQ1 was conjugated to dipicolyl amine (DPA). As controls, we synthesized (+)-JQ1-DBA, a non-Zn2+-chelating derivative, and (-)-JQ1-DPA, an inactive enantiomer that chelates Zn2+. Molecular modeling and biophysical assays showed that (+)-JQ1-DPA and (+)-JQ1-DBA retain potent binding to BET bromodomains in vitro. Cellular assays demonstrated (+)-JQ1-DPA attenuated NF-ĸB target gene expression in β cells stimulated with the proinflammatory cytokine interleukin 1β. To assess β-cell selectivity, we isolated islets from a mouse model that expresses green fluorescent protein in insulin-positive β cells and mTomato in insulin-negative cells (non-β cells). Surprisingly, Zn2+ chelation did not confer β-cell selectivity as (+)-JQ1-DPA was equally effective in both β and α cells; however, (+)-JQ1-DPA was less effective in macrophages, a nonendocrine islet cell type. Intriguingly, the non-Zn2+-chelating derivative (+)-JQ1-DBA displayed the opposite selectivity, with greater effect in macrophages compared with (+)-JQ1-DPA, suggesting potential as a macrophage-targeting molecule. These findings suggest that Zn2+-chelating small molecules confer endocrine cell selectivity rather than β-cell selectivity in pancreatic islets and provide valuable insights and techniques to assess Zn2+ chelation as an approach to selectively target small molecules to pancreatic β cells.NEW & NOTEWORTHY Inhibition of BET bromodomains is a novel potential strategy to prevent and treat diabetes mellitus. However, BET inhibitors have negative side effects. We synthesized a BET inhibitor expected to exploit the high zinc concentration in β cells to accumulate in β cells. We show our inhibitor targeted pancreatic endocrine cells; however, it was less effective in immune cells. A control inhibitor showed the opposite effect. These findings help us understand how to target specific cells in diabetes treatment.
Collapse
Affiliation(s)
- Rachel A Jones Lipinski
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Jennifer S Stancill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Raymundo Nuñez
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Sarah L Wynia-Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Daniel J Sprague
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Joshua A Nord
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Amir Bird
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - John A Corbett
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| | - Brian C Smith
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Program in Chemical Biology, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, United States
| |
Collapse
|
74
|
Wang KN, Zhou K, Zhong NN, Cao LM, Li ZZ, Xiao Y, Wang GR, Huo FY, Zhou JJ, Liu B, Bu LL. Enhancing cancer therapy: The role of drug delivery systems in STAT3 inhibitor efficacy and safety. Life Sci 2024; 346:122635. [PMID: 38615745 DOI: 10.1016/j.lfs.2024.122635] [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: 01/16/2024] [Revised: 03/14/2024] [Accepted: 04/10/2024] [Indexed: 04/16/2024]
Abstract
The signal transducer and activator of transcription 3 (STAT3), a member of the STAT family, resides in the nucleus to regulate genes essential for vital cellular functions, including survival, proliferation, self-renewal, angiogenesis, and immune response. However, continuous STAT3 activation in tumor cells promotes their initiation, progression, and metastasis, rendering STAT3 pathway inhibitors a promising avenue for cancer therapy. Nonetheless, these inhibitors frequently encounter challenges such as cytotoxicity and suboptimal biocompatibility in clinical trials. A viable strategy to mitigate these issues involves delivering STAT3 inhibitors via drug delivery systems (DDSs). This review delineates the regulatory mechanisms of the STAT3 signaling pathway and its association with cancer. It offers a comprehensive overview of the current application of DDSs for anti-STAT3 inhibitors and investigates the role of DDSs in cancer treatment. The conclusion posits that DDSs for anti-STAT3 inhibitors exhibit enhanced efficacy and reduced adverse effects in tumor therapy compared to anti-STAT3 inhibitors alone. This paper aims to provide an outline of the ongoing research and future prospects of DDSs for STAT3 inhibitors. Additionally, it presents our insights on the merits and future outlook of DDSs in cancer treatment.
Collapse
Affiliation(s)
- Kang-Ning Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Kan Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Fang-Yi Huo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jun-Jie Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial, Anyang Sixth People's Hospital, Anyang 45500, China.
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
75
|
Dourson AJ, Fadaka AO, Warshak AM, Paranjpe A, Weinhaus B, Queme LF, Hofmann MC, Evans HM, Donmez OA, Forney C, Weirauch MT, Kottyan LC, Lucas D, Deepe GS, Jankowski MP. Macrophage memories of early-life injury drive neonatal nociceptive priming. Cell Rep 2024; 43:114129. [PMID: 38640063 PMCID: PMC11197107 DOI: 10.1016/j.celrep.2024.114129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 01/05/2024] [Accepted: 04/04/2024] [Indexed: 04/21/2024] Open
Abstract
The developing peripheral nervous and immune systems are functionally distinct from those of adults. These systems are vulnerable to early-life injury, which influences outcomes related to nociception following subsequent injury later in life (i.e., "neonatal nociceptive priming"). The underpinnings of this phenomenon are unclear, although previous work indicates that macrophages are trained by inflammation and injury. Our findings show that macrophages are both necessary and partially sufficient to drive neonatal nociceptive priming, possibly due to a long-lasting remodeling in chromatin structure. The p75 neurotrophic factor receptor is an important effector in regulating neonatal nociceptive priming through modulation of the inflammatory profile of rodent and human macrophages. This "pain memory" is long lasting in females and can be transferred to a naive host to alter sex-specific pain-related behaviors. This study reveals a mechanism by which acute, neonatal post-surgical pain drives a peripheral immune-related predisposition to persistent pain following a subsequent injury.
Collapse
Affiliation(s)
- Adam J Dourson
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Adewale O Fadaka
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anna M Warshak
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Aditi Paranjpe
- Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Benjamin Weinhaus
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, USA
| | - Luis F Queme
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Megan C Hofmann
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Heather M Evans
- Division of Infectious Diseases, University of Cincinnati, Cincinnati, OH, USA
| | - Omer A Donmez
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Carmy Forney
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Matthew T Weirauch
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Divisions of Biomedical Informatics and Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Leah C Kottyan
- Center for Autoimmune Genomics and Etiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Daniel Lucas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - George S Deepe
- Division of Infectious Diseases, Department of Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael P Jankowski
- Department of Anesthesia, Division of Pain Management, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA; Pediatric Pain Research Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
76
|
Huang G, Zhu G, Lin R, Chen W, Chen R, Sun Y, Chen L, Hong D, Chen L. Magnetotactic Bacteria AMB-1 with Active Deep Tumor Penetrability for NIR-II Photothermal Tumor Therapy. ACS OMEGA 2024; 9:23060-23068. [PMID: 38826521 PMCID: PMC11137709 DOI: 10.1021/acsomega.4c02914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 06/04/2024]
Abstract
The complex tumor structure and microenvironment such as abnormal tumor vasculature, dense tumor matrix, and elevated interstitial fluid pressure greatly hinder the penetration and retention of therapeutic agents in solid tumors. The development of an advanced method for robust penetration and retention of therapeutic agents in tumors is of great significance for efficient tumor treatments. In this work, we demonstrated that magnetotactic bacteria AMB-1 with hypoxic metabolism characteristics can actively penetrate the tumor to selectively colonize deep hypoxic regions, which emerge as a promising intelligent drug carrier. Furthermore, AMB-1 presents intrinsic second near-infrared (NIR-II) photothermal performance that can efficiently convert a 1064 nm laser into heat for tumor thermal ablation. We believe that our investigations not only develop a novel bacteria-based photothermal agent but also provide useful insights for the development of advanced tumor microbial therapies.
Collapse
Affiliation(s)
- Guoming Huang
- College
of Biological Science and Engineering, Fuzhou
University, Fuzhou 350116, P. R. China
| | - Guifen Zhu
- College
of Biological Science and Engineering, Fuzhou
University, Fuzhou 350116, P. R. China
| | - Ruipeng Lin
- College
of Biological Science and Engineering, Fuzhou
University, Fuzhou 350116, P. R. China
| | - Wenwen Chen
- College
of Biological Science and Engineering, Fuzhou
University, Fuzhou 350116, P. R. China
| | - Rong Chen
- College
of Biological Science and Engineering, Fuzhou
University, Fuzhou 350116, P. R. China
| | - Yutong Sun
- College
of Biological Science and Engineering, Fuzhou
University, Fuzhou 350116, P. R. China
| | - Liqun Chen
- College
of Biological Science and Engineering, Fuzhou
University, Fuzhou 350116, P. R. China
| | - Dengke Hong
- Department
of Vascular Surgery, Fujian Medical University
Union Hospital, Fuzhou 350001, P. R. China
| | - Lihong Chen
- Department
of Radiology, Fujian Medical University
Union Hospital, Fuzhou 350001, P. R. China
| |
Collapse
|
77
|
Kakinen A, Jiang Y, Davis TP, Teesalu T, Saarma M. Brain Targeting Nanomedicines: Pitfalls and Promise. Int J Nanomedicine 2024; 19:4857-4875. [PMID: 38828195 PMCID: PMC11143448 DOI: 10.2147/ijn.s454553] [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: 12/12/2023] [Accepted: 04/15/2024] [Indexed: 06/05/2024] Open
Abstract
Brain diseases are the most devastating problem among the world's increasingly aging population, and the number of patients with neurological diseases is expected to increase in the future. Although methods for delivering drugs to the brain have advanced significantly, none of these approaches provide satisfactory results for the treatment of brain diseases. This remains a challenge due to the unique anatomy and physiology of the brain, including tight regulation and limited access of substances across the blood-brain barrier. Nanoparticles are considered an ideal drug delivery system to hard-to-reach organs such as the brain. The development of new drugs and new nanomaterial-based brain treatments has opened various opportunities for scientists to develop brain-specific delivery systems that could improve treatment outcomes for patients with brain disorders such as Alzheimer's disease, Parkinson's disease, stroke and brain tumors. In this review, we discuss noteworthy literature that examines recent developments in brain-targeted nanomedicines used in the treatment of neurological diseases.
Collapse
Affiliation(s)
- Aleksandr Kakinen
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Yuhao Jiang
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Thomas Paul Davis
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD, Australia
| | - Tambet Teesalu
- Institute of Biomedicine and Translational Medicine, Faculty of Medicine, University of Tartu, Tartu, Estonia
- Materials Research Laboratory, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Mart Saarma
- Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| |
Collapse
|
78
|
Moaveni AK, Amiri M, Shademan B, Farhadi A, Behroozi J, Nourazarian A. Advances and challenges in gene therapy strategies for pediatric cancer: a comprehensive update. Front Mol Biosci 2024; 11:1382190. [PMID: 38836106 PMCID: PMC11149429 DOI: 10.3389/fmolb.2024.1382190] [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: 02/05/2024] [Accepted: 03/27/2024] [Indexed: 06/06/2024] Open
Abstract
Pediatric cancers represent a tragic but also promising area for gene therapy. Although conventional treatments have improved survival rates, there is still a need for targeted and less toxic interventions. This article critically analyzes recent advances in gene therapy for pediatric malignancies and discusses the challenges that remain. We explore the innovative vectors and delivery systems that have emerged, such as adeno-associated viruses and non-viral platforms, which show promise in addressing the unique pathophysiology of pediatric tumors. Specifically, we examine the field of chimeric antigen receptor (CAR) T-cell therapies and their adaptation for solid tumors, which historically have been more challenging to treat than hematologic malignancies. We also discuss the genetic and epigenetic complexities inherent to pediatric cancers, such as tumor heterogeneity and the dynamic tumor microenvironment, which pose significant hurdles for gene therapy. Ethical considerations specific to pediatric populations, including consent and long-term follow-up, are also analyzed. Additionally, we scrutinize the translation of research from preclinical models that often fail to mimic pediatric cancer biology to the regulatory landscapes that can either support or hinder innovation. In summary, this article provides an up-to-date overview of gene therapy in pediatric oncology, highlighting both the rapid scientific progress and the substantial obstacles that need to be addressed. Through this lens, we propose a roadmap for future research that prioritizes the safety, efficacy, and complex ethical considerations involved in treating pediatric patients. Our ultimate goal is to move from incremental advancements to transformative therapies.
Collapse
Affiliation(s)
- Amir Kian Moaveni
- Pediatric Urology and Regenerative Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Amiri
- Pediatric Urology and Regenerative Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Behrouz Shademan
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arezoo Farhadi
- Department of Genetics and Molecular Medicine, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Javad Behroozi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Alireza Nourazarian
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| |
Collapse
|
79
|
Moreira R, Nóbrega C, de Almeida LP, Mendonça L. Brain-targeted drug delivery - nanovesicles directed to specific brain cells by brain-targeting ligands. J Nanobiotechnology 2024; 22:260. [PMID: 38760847 PMCID: PMC11100082 DOI: 10.1186/s12951-024-02511-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/29/2024] [Indexed: 05/19/2024] Open
Abstract
Neurodegenerative diseases are characterized by extensive loss of function or death of brain cells, hampering the life quality of patients. Brain-targeted drug delivery is challenging, with a low success rate this far. Therefore, the application of targeting ligands in drug vehicles, such as lipid-based and polymeric nanoparticles, holds the promise to overcome the blood-brain barrier (BBB) and direct therapies to the brain, in addition to protect their cargo from degradation and metabolization. In this review, we discuss the barriers to brain delivery and the different types of brain-targeting ligands currently in use in brain-targeted nanoparticles, such as peptides, proteins, aptamers, small molecules, and antibodies. Moreover, we present a detailed review of the different targeting ligands used to direct nanoparticles to specific brain cells, like neurons (C4-3 aptamer, neurotensin, Tet-1, RVG, and IKRG peptides), astrocytes (Aquaporin-4, D4, and Bradykinin B2 antibodies), oligodendrocytes (NG-2 antibody and the biotinylated DNA aptamer conjugated to a streptavidin core Myaptavin-3064), microglia (CD11b antibody), neural stem cells (QTRFLLH, VPTQSSG, and NFL-TBS.40-63 peptides), and to endothelial cells of the BBB (transferrin and insulin proteins, and choline). Reports demonstrated enhanced brain-targeted delivery with improved transport to the specific cell type targeted with the conjugation of these ligands to nanoparticles. Hence, this strategy allows the implementation of high-precision medicine, with reduced side effects or unwanted therapy clearance from the body. Nevertheless, the accumulation of some of these nanoparticles in peripheral organs has been reported indicating that there are still factors to be improved to achieve higher levels of brain targeting. This review is a collection of studies exploring targeting ligands for the delivery of nanoparticles to the brain and we highlight the advantages and limitations of this type of approach in precision therapies.
Collapse
Grants
- under BrainHealth2020 projects (CENTRO-01-0145-FEDER-000008), through the COMPETE 2020 - Operational Programme for Competitiveness and Internationalization and Portuguese national funds via FCT - Fundação para a Ciência e a Tecnologia, under projects - UIDB/04539/2020 and UIDP/04539/2020, POCI-01-0145-FEDER-030737 (NeuroStemForMJD, PTDC/BTM-ORG/30737/2017), CEECIND/04242/2017, and PhD Scholarship European Regional Development Fund (ERDF) through the Centro 2020 Regional Operational Programme
- under BrainHealth2020 projects (CENTRO-01-0145-FEDER-000008), through the COMPETE 2020 - Operational Programme for Competitiveness and Internationalization and Portuguese national funds via FCT - Fundação para a Ciência e a Tecnologia, under projects - UIDB/04539/2020 and UIDP/04539/2020, POCI-01-0145-FEDER-030737 (NeuroStemForMJD, PTDC/BTM-ORG/30737/2017), CEECIND/04242/2017, and PhD Scholarship European Regional Development Fund (ERDF) through the Centro 2020 Regional Operational Programme
Collapse
Affiliation(s)
- Ricardo Moreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, polo 1, Coimbra, FMUC, 3004-504, Portugal
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-504, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, 3000-548, Portugal
| | - Clévio Nóbrega
- Algarve Biomedical Center Research Institute (ABC-RI), University of Algarve, Faro, 8005-139, Portugal
- Faculty of Medicine and Biomedical Sciences, University of Algarve, Faro, 8005-139, Portugal
| | - Luís Pereira de Almeida
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, polo 1, Coimbra, FMUC, 3004-504, Portugal
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-504, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, 3000-548, Portugal
- Institute of Interdisciplinary Research, University of Coimbra, Coimbra, 3030-789, Portugal
| | - Liliana Mendonça
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Rua Larga, polo 1, Coimbra, FMUC, 3004-504, Portugal.
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, 3004-504, Portugal.
- Institute of Interdisciplinary Research, University of Coimbra, Coimbra, 3030-789, Portugal.
| |
Collapse
|
80
|
Li X, Gao J, Wu C, Wang C, Zhang R, He J, Xia ZJ, Joshi N, Karp JM, Kuai R. Precise modulation and use of reactive oxygen species for immunotherapy. SCIENCE ADVANCES 2024; 10:eadl0479. [PMID: 38748805 PMCID: PMC11095489 DOI: 10.1126/sciadv.adl0479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 04/10/2024] [Indexed: 05/19/2024]
Abstract
Reactive oxygen species (ROS) play an important role in regulating the immune system by affecting pathogens, cancer cells, and immune cells. Recent advances in biomaterials have leveraged this mechanism to precisely modulate ROS levels in target tissues for improving the effectiveness of immunotherapies in infectious diseases, cancer, and autoimmune diseases. Moreover, ROS-responsive biomaterials can trigger the release of immunotherapeutics and provide tunable release kinetics, which can further boost their efficacy. This review will discuss the latest biomaterial-based approaches for both precise modulation of ROS levels and using ROS as a stimulus to control the release kinetics of immunotherapeutics. Finally, we will discuss the existing challenges and potential solutions for clinical translation of ROS-modulating and ROS-responsive approaches for immunotherapy, and provide an outlook for future research.
Collapse
Affiliation(s)
- Xinyan Li
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jingjing Gao
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Biomedical Engineering, Material Science and Engineering Graduate Program and The Center for Bioactive Delivery-Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
| | - Chengcheng Wu
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Chaoyu Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ruoshi Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Jia He
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Ziting Judy Xia
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Nitin Joshi
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jeffrey M. Karp
- Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Rui Kuai
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China
- Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| |
Collapse
|
81
|
Li Z, Xie HY, Nie W. Nano-Engineering Strategies for Tumor-Specific Therapy. ChemMedChem 2024; 19:e202300647. [PMID: 38356248 DOI: 10.1002/cmdc.202300647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/09/2024] [Accepted: 02/14/2024] [Indexed: 02/16/2024]
Abstract
Nanodelivery systems (NDSs) provide promising prospects for decreasing drug doses, reducing side effects, and improving therapeutic effects. However, the bioapplications of NDSs are still compromised by their fast clearance, indiscriminate biodistribution, and limited tumor accumulation. Hence, engineering modification of NDSs aiming at promoting tumor-specific therapy and avoiding systemic toxicity is usually needed. An NDS integrating various functionalities, including flexible camouflage, specific biorecognition, and sensitive stimuli-responsiveness, into one sequence would be "smart" and highly effective. Herein, we systematically summarize the related principles, methods, and progress. At the end of the review, we predict the obstacles to precise nanoengineering and prospects for the future application of NDSs.
Collapse
Affiliation(s)
- Zijin Li
- School of Life Science, Beijing Institute of Technology, No. 5, Zhongguancun South Street, Beijing, 100081, China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Chemical Biology Center, Peking University, Beijing, 100191, China
| | - Weidong Nie
- School of Life Science, Beijing Institute of Technology, No. 5, Zhongguancun South Street, Beijing, 100081, China
| |
Collapse
|
82
|
Kumar M, Kumar D, Kumar D, Garg Y, Chopra S, Bhatia A. Therapeutic Potential of Nanocarrier Mediated Delivery of Peptides for Wound Healing: Current Status, Challenges and Future Prospective. AAPS PharmSciTech 2024; 25:108. [PMID: 38730090 DOI: 10.1208/s12249-024-02827-5] [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: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
Wound healing presents a complex physiological process that involves a sequence of events orchestrated by various cellular and molecular mechanisms. In recent years, there has been growing interest in leveraging nanomaterials and peptides to enhance wound healing outcomes. Nanocarriers offer unique properties such as high surface area-to-volume ratio, tunable physicochemical characteristics, and the ability to deliver therapeutic agents in a controlled manner. Similarly, peptides, with their diverse biological activities and low immunogenicity, hold great promise as therapeutics in wound healing applications. In this review, authors explore the potential of peptides as bioactive components in wound healing formulations, focusing on their antimicrobial, anti-inflammatory, and pro-regenerative properties. Despite the significant progress made in this field, several challenges remain, including the need for standardized characterization methods, optimization of biocompatibility and safety profiles, and translation from bench to bedside. Furthermore, developing multifunctional nanomaterial-peptide hybrid systems represents promising avenues for future research. Overall, the integration of nanomaterials made up of natural or synthetic polymers with peptide-based formulations holds tremendous therapeutic potential in advancing the field of wound healing and improving clinical outcomes for patients with acute and chronic wounds.
Collapse
Affiliation(s)
- Mohit Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Dikshant Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Devesh Kumar
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Yogesh Garg
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Shruti Chopra
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India
| | - Amit Bhatia
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University (MRSPTU), Bathinda, 151001, Punjab, India.
| |
Collapse
|
83
|
Beach M, Nayanathara U, Gao Y, Zhang C, Xiong Y, Wang Y, Such GK. Polymeric Nanoparticles for Drug Delivery. Chem Rev 2024; 124:5505-5616. [PMID: 38626459 PMCID: PMC11086401 DOI: 10.1021/acs.chemrev.3c00705] [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: 04/18/2024]
Abstract
The recent emergence of nanomedicine has revolutionized the therapeutic landscape and necessitated the creation of more sophisticated drug delivery systems. Polymeric nanoparticles sit at the forefront of numerous promising drug delivery designs, due to their unmatched control over physiochemical properties such as size, shape, architecture, charge, and surface functionality. Furthermore, polymeric nanoparticles have the ability to navigate various biological barriers to precisely target specific sites within the body, encapsulate a diverse range of therapeutic cargo and efficiently release this cargo in response to internal and external stimuli. However, despite these remarkable advantages, the presence of polymeric nanoparticles in wider clinical application is minimal. This review will provide a comprehensive understanding of polymeric nanoparticles as drug delivery vehicles. The biological barriers affecting drug delivery will be outlined first, followed by a comprehensive description of the various nanoparticle designs and preparation methods, beginning with the polymers on which they are based. The review will meticulously explore the current performance of polymeric nanoparticles against a myriad of diseases including cancer, viral and bacterial infections, before finally evaluating the advantages and crucial challenges that will determine their wider clinical potential in the decades to come.
Collapse
Affiliation(s)
- Maximilian
A. Beach
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Umeka Nayanathara
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yanting Gao
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Changhe Zhang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yijun Xiong
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yufu Wang
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Georgina K. Such
- School
of Chemistry, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
84
|
Heyns IM, Arora M, Ganugula R, Allamreddy SR, Tiwari S, Shah DK, Basu R, Ravi Kumar MNV. Polyester Nanoparticles with Controlled Topography for Peroral Drug Delivery Using Insulin as a Model Protein. ACS NANO 2024; 18:11863-11875. [PMID: 38622996 PMCID: PMC11145941 DOI: 10.1021/acsnano.4c01027] [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: 04/17/2024]
Abstract
Receptor-mediated polyester drug delivery systems have tremendous potential for improving the clinical performance of existing pharmaceutical drugs. Despite significant progress made in this area, it remains unclear how and to what extent the polyester nanoparticle surface topography would affect the in vitro, ex vivo and in vivo performance of a drug, and if there exists a correlation between in vitro and in vivo, as well as healthy versus pathophysiological states. Herein, we report a systematic investigation of the interactions between ligands and receptors as a function of the linker length, two-carbon (2C) versus four-carbon (4C). The in vitro, ex vivo and in vivo in healthy models validate the hypothesis that 4C has better reach and binding to the receptors. The results indicate that 4C offered better performance over 2C in vivo in improving the oral bioavailability of insulin (INS) by 1.1-fold (3.5-fold compared to unfunctionalized nanoparticles) in a healthy rat model. Similar observations were made in pathophysiological models; however, the effects were less prominent compared to those in healthy models. Throughout, ligand decorated nanoparticles outperformed unfunctionalized nanoparticles. Finally, a semimechanistic pharmacokinetic and pharmacodynamic (PKPD) model was developed using the experimental data sets to quantitatively evaluate the effect of P2Ns-GA on oral bioavailability and efficacy of insulin. The study presents a sophisticated oral delivery system for INS or hydrophilic therapeutic cargo, highlighting the significant impact on bioavailability that minor adjustments to the surface chemistry can have.
Collapse
Affiliation(s)
- Ingrid Marie Heyns
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States
- Division of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL 35401, United States
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35401, United States
| | - Meenakshi Arora
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States
- Division of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL 35401, United States
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35401, United States
- Department of Biological Sciences, The University of Alabama, SEC 1325, Tuscaloosa, AL 35487, United States
| | - Raghu Ganugula
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States
- Division of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL 35401, United States
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35401, United States
- Department of Biological Sciences, The University of Alabama, SEC 1325, Tuscaloosa, AL 35487, United States
| | - Swetha Reddy Allamreddy
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States
- Division of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL 35401, United States
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35401, United States
| | - Shrusti Tiwari
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY 14214, United States
| | - Dhaval K. Shah
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, The State University of New York at Buffalo, 455 Pharmacy Building, Buffalo, NY 14214, United States
| | - Rita Basu
- Division of Endocrinology, Diabetes, and Metabolism, School of Medicine, Marnix E. Heersink School of Medicine, The University of Alabama, Birmingham, AL 35294, United States
| | - M. N. V. Ravi Kumar
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, AL 35401, United States
- Division of Translational Science and Medicine, College of Community Health Sciences, The University of Alabama, Tuscaloosa, AL 35401, United States
- Alabama Life Research Institute, The University of Alabama, Tuscaloosa, AL 35401, United States
- Department of Biological Sciences, The University of Alabama, SEC 1325, Tuscaloosa, AL 35487, United States
- Chemical and Biological Engineering, University of Alabama, SEC 3448, Tuscaloosa, AL 35487, United States
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL 35294, United States
- Nephrology Research and Training Center, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| |
Collapse
|
85
|
Switala L, Di L, Gao H, Asase C, Klos M, Rengasamy P, Fedyukina D, Maiseyeu A. Engineered nanoparticles promote cardiac tropism of AAV vectors. J Nanobiotechnology 2024; 22:223. [PMID: 38702815 PMCID: PMC11067271 DOI: 10.1186/s12951-024-02485-6] [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: 01/23/2024] [Accepted: 04/17/2024] [Indexed: 05/06/2024] Open
Abstract
Cardiac muscle targeting is a notoriously difficult task. Although various nanoparticle (NP) and adeno-associated viral (AAV) strategies with heart tissue tropism have been developed, their performance remains suboptimal. Significant off-target accumulation of i.v.-delivered pharmacotherapies has thwarted development of disease-modifying cardiac treatments, such as gene transfer and gene editing, that may address both rare and highly prevalent cardiomyopathies and their complications. Here, we present an intriguing discovery: cargo-less, safe poly (lactic-co-glycolic acid) particles that drastically improve heart delivery of AAVs and NPs. Our lead formulation is referred to as ePL (enhancer polymer). We show that ePL increases selectivity of AAVs and virus-like NPs (VLNPs) to the heart and de-targets them from the liver. Serotypes known to have high (AAVrh.74) and low (AAV1) heart tissue tropisms were tested with and without ePL. We demonstrate up to an order of magnitude increase in heart-to-liver accumulation ratios in ePL-injected mice. We also show that ePL exhibits AAV/NP-independent mechanisms of action, increasing glucose uptake in the heart, increasing cardiac protein glycosylation, reducing AAV neutralizing antibodies, and delaying blood clearance of AAV/NPs. Current approaches utilizing AAVs or NPs are fraught with challenges related to the low transduction of cardiomyocytes and life-threatening immune responses; our study introduces an exciting possibility to direct these modalities to the heart at reduced i.v. doses and, thus, has an unprecedented impact on drug delivery and gene therapy. Based on our current data, the ePL system is potentially compatible with any therapeutic modality, opening a possibility of cardiac targeting with numerous pharmacological approaches.
Collapse
Affiliation(s)
- Lauren Switala
- Department of Medicine, School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA
| | - Lin Di
- Department of Medicine, School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, USA
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA
| | - Huiyun Gao
- Department of Medicine, School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, USA
| | - Courteney Asase
- Department of Medicine, School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, USA
| | - Matthew Klos
- Department of Pediatrics, Case Western Reserve University, Cleveland, USA
| | - Palanivel Rengasamy
- Department of Medicine, School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, USA
| | - Daria Fedyukina
- Bioheights LLC, Cleveland, USA
- Advanced Research Projects Agency for Health, ARPA-H, Washington, USA
| | - Andrei Maiseyeu
- Department of Medicine, School of Medicine, Cardiovascular Research Institute, Case Western Reserve University, Cleveland, USA.
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, USA.
| |
Collapse
|
86
|
Zhu P, Simon I, Kokalari I, Kohane DS, Rwei AY. Miniaturized therapeutic systems for ultrasound-modulated drug delivery to the central and peripheral nervous system. Adv Drug Deliv Rev 2024; 208:115275. [PMID: 38442747 PMCID: PMC11031353 DOI: 10.1016/j.addr.2024.115275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/19/2024] [Accepted: 03/01/2024] [Indexed: 03/07/2024]
Abstract
Ultrasound is a promising technology to address challenges in drug delivery, including limited drug penetration across physiological barriers and ineffective targeting. Here we provide an overview of the significant advances made in recent years in overcoming technical and pharmacological barriers using ultrasound-assisted drug delivery to the central and peripheral nervous system. We commence by exploring the fundamental principles of ultrasound physics and its interaction with tissue. The mechanisms of ultrasonic-enhanced drug delivery are examined, as well as the relevant tissue barriers. We highlight drug transport through such tissue barriers utilizing insonation alone, in combination with ultrasound contrast agents (e.g., microbubbles), and through innovative particulate drug delivery systems. Furthermore, we review advances in systems and devices for providing therapeutic ultrasound, as their practicality and accessibility are crucial for clinical application.
Collapse
Affiliation(s)
- Pancheng Zhu
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands; State Key Laboratory of Mechanics and Control of Aerospace Structures, Nanjing University of Aeronautics & Astronautics, 210016, Nanjing, China; Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China
| | - Ignasi Simon
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands
| | - Ida Kokalari
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands
| | - Daniel S Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Alina Y Rwei
- Department of Chemical Engineering, Delft University of Technology, 2629 HZ, Delft, the Netherlands.
| |
Collapse
|
87
|
Singh D. Beyond the Maze: Recent Advancements in Molecular and Cellular Tethered Drug Delivery Systems. Assay Drug Dev Technol 2024; 22:203-215. [PMID: 38717194 DOI: 10.1089/adt.2024.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/19/2024] Open
Abstract
The relentless pursuit of precision medicine has catalyzed the development of molecular and cellular tethered drug delivery systems, a burgeoning field that stands to redefine the paradigms of therapeutic delivery. This review encapsulates the cutting-edge advancements within this domain, emphasizing the engineering of molecular tethers and cellular vectors designed to ferry therapeutics directly to their target sites with unparalleled specificity and efficiency. By exploiting the unique biochemical signatures of disease states, these systems promise a substantial reduction in off-target effects and an enhancement in drug bioavailability, thereby mitigating the systemic side effects that are often associated with conventional drug therapies. Through a synthesis of recent research findings, this review highlights the innovative approaches being explored in the design and application of these tethered systems, ranging from nanotechnology-based solutions to genetically engineered cellular carriers. The potential of these systems to provide targeted therapy for a wide array of diseases, including cancer, autoimmune disorders, and neurological conditions, is thoroughly examined. This abstract aims to provide a succinct overview of the current state and future prospects of molecular and cellular tethered drug delivery systems in advancing the frontiers of precision medicine.
Collapse
Affiliation(s)
- Dilpreet Singh
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, India
- University Centre for Research and Development, Chandigarh University, Gharuan, India
| |
Collapse
|
88
|
Demartis S, Rassu G, Anjani QK, Volpe-Zanutto F, Hutton ARJ, Sabri AB, McCarthy HO, Giunchedi P, Donnelly RF, Gavini E. Improved pharmacokinetic and lymphatic uptake of Rose Bengal after transfersome intradermal deposition using hollow microneedles. J Control Release 2024; 369:363-375. [PMID: 38554770 DOI: 10.1016/j.jconrel.2024.03.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
The lymphatic system is active in several processes that regulate human diseases, among which cancer progression stands out. Thus, various drug delivery systems have been investigated to promote lymphatic drug targeting for cancer therapy; mainly, nanosized particles in the 10-150 nm range quickly achieve lymphatic vessels after an interstitial administration. Herein, a strategy to boost the lymphotropic delivery of Rose Bengal (RB), a hydrosoluble chemotherapeutic, is proposed, and it is based on the loading into Transfersomes (RBTF) and their intradermal deposition in vivo by microneedles. RBTF of 96.27 ± 13.96 nm (PDI = 0.29 ± 0.02) were prepared by a green reverse-phase evaporation technique, and they showed an RB encapsulation efficiency of 98.54 ± 0.09%. In vitro, RBTF remained physically stable under physiological conditions and avoided the release of RB. In vivo, intravenous injection of RBTF prolonged RB half-life of 50 min in healthy rats compared to RB intravenous injection; the RB half-life in rat body was further increased after intradermal injection reaching 24 h, regardless of the formulation used. Regarding lymphatic targeting, RBTF administered intravenously provided an RB accumulation in the lymph nodes of 12.3 ± 0.14 ng/mL after 2 h, whereas no RB accumulation was observed after RB intravenous injection. Intradermally administered RBTF resulted in the highest RB amount detected in lymph nodes after 2 h from the injection (84.2 ± 25.10 ng/mL), which was even visible to the naked eye based on the pink colouration of the drug. In the case of intradermally administered RB, RB in lymph node was detected only at 24 h (13.3 ± 1.41 ng/mL). In conclusion, RBTF proved an efficient carrier for RB delivery, enhancing its pharmacokinetics and promoting lymph-targeted delivery. Thus, RBTF represents a promising nanomedicine product for potentially facing the medical need for novel strategies for cancer therapy.
Collapse
Affiliation(s)
- Sara Demartis
- Department of Chemical, Mathematical, Natural and Physical Sciences, University of Sassari, Sassari 07100, Italy
| | - Giovanna Rassu
- Department of Medicine and Surgery, University of Sassari, Sassari 07100, Italy.
| | - Qonita Kurnia Anjani
- School of Pharmacy, Queen's University, Belfast 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Fabiana Volpe-Zanutto
- School of Pharmacy, Queen's University, Belfast 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Aaron R J Hutton
- School of Pharmacy, Queen's University, Belfast 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Akmal B Sabri
- School of Pharmacy, Queen's University, Belfast 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Helen O McCarthy
- School of Pharmacy, Queen's University, Belfast 97 Lisburn Road, Belfast BT9 7BL, United Kingdom
| | - Paolo Giunchedi
- Department of Medicine and Surgery, University of Sassari, Sassari 07100, Italy
| | - Ryan F Donnelly
- School of Pharmacy, Queen's University, Belfast 97 Lisburn Road, Belfast BT9 7BL, United Kingdom.
| | - Elisabetta Gavini
- Department of Medicine and Surgery, University of Sassari, Sassari 07100, Italy
| |
Collapse
|
89
|
Fralish Z, Chen A, Khan S, Zhou P, Reker D. The landscape of small-molecule prodrugs. Nat Rev Drug Discov 2024; 23:365-380. [PMID: 38565913 DOI: 10.1038/s41573-024-00914-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 04/04/2024]
Abstract
Prodrugs are derivatives with superior properties compared with the parent active pharmaceutical ingredient (API), which undergo biotransformation after administration to generate the API in situ. Although sharing this general characteristic, prodrugs encompass a wide range of different chemical structures, therapeutic indications and properties. Here we provide the first holistic analysis of the current landscape of approved prodrugs using cheminformatics and data science approaches to reveal trends in prodrug development. We highlight rationales that underlie prodrug design, their indications, mechanisms of API release, the chemistry of promoieties added to APIs to form prodrugs and the market impact of prodrugs. On the basis of this analysis, we discuss strengths and limitations of current prodrug approaches and suggest areas for future development.
Collapse
Affiliation(s)
- Zachary Fralish
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Ashley Chen
- Department of Computer Science, Duke University, Durham, NC, USA
| | | | - Pei Zhou
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, USA
| | - Daniel Reker
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
90
|
Song YH, Cho HM, Ryu YC, Hwang BH, Seo JH. Electrosprayable Levan-Coated Nanoclusters and Ultrasound-Responsive Drug Delivery for Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:21509-21521. [PMID: 38642038 DOI: 10.1021/acsami.3c18774] [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: 04/22/2024]
Abstract
In this study, we synthesized levan shell hydrophobic silica nanoclusters encapsulating doxorubicin (L-HSi-Dox) and evaluated their potential as ultrasound-responsive drug delivery systems for cancer treatment. L-HSi-Dox nanoclusters were successfully fabricated by integrating a hydrophobic silica nanoparticle-doxorubicin complex as the core and an amphiphilic levan carbohydrate polymer as the shell by using an electrospray technique. Characterization analyses confirmed the stability, size, and composition of the nanoclusters. In particular, the nanoclusters exhibited a controlled release of Dox under aqueous conditions, demonstrating their potential as efficient drug carriers. The levanic groups of the nanoclusters enhanced the targeted delivery of Dox to specific cancer cells. Furthermore, the synergism between the nanoclusters and ultrasound effectively reduced cell viability and induced cell death, particularly in the GLUT5-overexpressing MDA-MB-231 cells. In a tumor xenograft mouse model, treatment with the nanoclusters and ultrasound significantly reduced the tumor volume and weight without affecting the body weight. Collectively, these results highlight the potential of the L-HSi-Dox nanoclusters and ultrasound as promising drug delivery systems with an enhanced therapeutic efficacy for biomedical applications.
Collapse
Affiliation(s)
- Young Hoon Song
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, South Korea
| | - Hye Min Cho
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Incheon 22012, South Korea
| | - Yeong Chae Ryu
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Incheon 22012, South Korea
| | - Byeong Hee Hwang
- Department of Bioengineering and Nano-bioengineering, Incheon National University, Incheon 22012, South Korea
- Division of Bioengineering, Incheon National University, Incheon 22012, South Korea
| | - Jeong Hyun Seo
- School of Chemical Engineering, Yeungnam University, Gyeongsan 38541, South Korea
| |
Collapse
|
91
|
Zahiri M, Kamali H, Abnous K, Mohammad Taghdisi S, Nekooei S, Nekooei N, Ramezani M, Alibolandi M. Synthesis of folate targeted theranostic cubosomal platform for co-delivery of bismuth oxide and doxorubicin to melanoma in vitro and in vivo. Eur J Pharm Biopharm 2024; 198:114259. [PMID: 38479563 DOI: 10.1016/j.ejpb.2024.114259] [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: 01/04/2024] [Revised: 02/21/2024] [Accepted: 03/11/2024] [Indexed: 04/19/2024]
Abstract
Liquid crystalline nanoparticles (LCNPs) have gained much attention in cancer nanomedicines due to their unique features such as high surface area, storage stability, and sustained-release profile. In the current study, a novel LCNP for co-encapsulation of Bi2O3 and hydrophilic doxorubicin (DOX) was fabricated and functionalized with folic acid (FA) to achieve efficient tumor targeting toward CT-scan imaging and chemotherapy of melanoma in vitro and in vivo. LCNPs Bi2O3 NPs were prepared using glycerol monooleate-pluronic F-127 (GMO/PF127/water). Firstly, GMO/water were homogenized to prepare LC gel. Then, the stabilizer aqueous solution (PF127/Bi2O3/DOX) was added to the prepared LC gel and homogenized using homogenization and ultrasonication. The formulated NPs exhibited superior stability with encapsulation efficiency. High cytotoxicity and cellular internalization of the FA-Bi2O3-DOX-NPs were observed in comparison with Bi2O3-DOX-NPs and the free DOX in folate-receptor (FR) overexpressing cells (B16F10) in vitro. Moreover, ideal tumor suppression with increased survival rate were observed in tumorized mice treated with FA-Bi2O3-DOX-NPs compared to those treated with non-targeted one. On the other hand, the CT-imaging ability of the Bi2O3-DOX-NPs was tested inB16F10 tumor-bearing mice. The obtained data indicated a high potential of the developed targeted theranostic FA-Bi2O3-DOX-NPs for diagnostics and treatment of melanoma.
Collapse
Affiliation(s)
- Mahsa Zahiri
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hosein Kamali
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Khalil Abnous
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mohammad Taghdisi
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sirous Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Nekooei
- Department of Radiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Ramezani
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
92
|
Xiong W, Han Z, Ding S, Wang H, Du Y, Cui W, Zhang M. In Situ Remodeling of Efferocytosis via Lesion-Localized Microspheres to Reverse Cartilage Senescence. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400345. [PMID: 38477444 PMCID: PMC11109622 DOI: 10.1002/advs.202400345] [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: 01/30/2024] [Revised: 02/19/2024] [Indexed: 03/14/2024]
Abstract
Efferocytosis, an intrinsic regulatory mechanism to eliminate apoptotic cells, will be suppressed due to the delayed apoptosis process in aging-related diseases, such as osteoarthritis (OA). In this study, cartilage lesion-localized hydrogel microspheres are developed to remodel the in situ efferocytosis to reverse cartilage senescence and recruit endogenous stem cells to accelerate cartilage repair. Specifically, aldehyde- and methacrylic anhydride (MA)-modified hyaluronic acid hydrogel microspheres (AHM), loaded with pro-apoptotic liposomes (liposomes encapsulating ABT263, A-Lipo) and PDGF-BB, namely A-Lipo/PAHM, are prepared by microfluidic and photo-cross-linking techniques. By a degraded porcine cartilage explant OA model, the in situ cartilage lesion location experiment illustrated that aldehyde-functionalized microspheres promote affinity for degraded cartilage. In vitro data showed that A-Lipo induced apoptosis of senescent chondrocytes (Sn-chondrocytes), which can then be phagocytosed by the efferocytosis of macrophages, and remodeling efferocytosis facilitated the protection of normal chondrocytes and maintained the chondrogenic differentiation capacity of MSCs. In vivo experiments confirmed that hydrogel microspheres localized to cartilage lesion reversed cartilage senescence and promoted cartilage repair in OA. It is believed this in situ efferocytosis remodeling strategy can be of great significance for tissue regeneration in aging-related diseases.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730P. R. China
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Zeyu Han
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730P. R. China
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Sheng‐Long Ding
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730P. R. China
| | - Haoran Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Yawei Du
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of Medicine197 Ruijin 2nd RoadShanghai200025P. R. China
| | - Ming‐Zhu Zhang
- Department of Foot and Ankle SurgeryBeijing Tongren HospitalCapital Medical UniversityBeijing100730P. R. China
| |
Collapse
|
93
|
Eling CJ, Bruce N, Gunasekar NK, Alves PU, Edwards PR, Martin RW, Laurand N. Biotinylated Photocleavable Semiconductor Colloidal Quantum Dot Supraparticle Microlaser. ACS APPLIED NANO MATERIALS 2024; 7:9159-9166. [PMID: 38694721 PMCID: PMC11059076 DOI: 10.1021/acsanm.4c00668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/04/2024] [Accepted: 04/08/2024] [Indexed: 05/04/2024]
Abstract
Luminescent supraparticles of colloidal semiconductor nanocrystals can act as microscopic lasers and are hugely attractive for biosensing, imaging, and drug delivery. However, biointerfacing these to increase functionality while retaining their main optical properties remains an unresolved challenge. Here, we propose and demonstrate red-emitting, silica-coated CdSxSe1-x/ZnS colloidal quantum dot supraparticles functionalized with a biotinylated photocleavable ligand. The success of each step of the synthesis is confirmed by scanning electron microscopy, energy dispersive X-ray and Fourier transform infrared spectroscopy, ζ-potential, and optical pumping measurements. The capture and release functionality of the supraparticle system is proven by binding to a neutravidin functionalized glass slide and subsequently cleaving off after UV-A irradiation. The biotinylated supraparticles still function as microlasers; e.g., a 9 μm diameter supraparticle has oscillating modes around 625 nm at a threshold of 58 mJ/cm2. This work is a first step toward using supraparticle lasers as enhanced labels for bionano applications.
Collapse
Affiliation(s)
- Charlotte J. Eling
- Institute
of Photonics, Department of Physics, SUPA,
University of Strathclyde, Glasgow G1 1RD, U.K.
| | - Natalie Bruce
- Institute
of Photonics, Department of Physics, SUPA,
University of Strathclyde, Glasgow G1 1RD, U.K.
- Fraunhofer
Centre for Applied Photonics, 99 George Street, Glasgow G1 1RD, U.K.
| | - Naresh-Kumar Gunasekar
- Department
of Physics, SUPA, University of Strathclyde, Glasgow G4 0NG, U.K.
- Institute
for Compound Semiconductors, School of Physics and Astronomy, Cardiff University, Cardiff CF24 3AA, U.K.
| | - Pedro Urbano Alves
- Institute
of Photonics, Department of Physics, SUPA,
University of Strathclyde, Glasgow G1 1RD, U.K.
| | - Paul R. Edwards
- Department
of Physics, SUPA, University of Strathclyde, Glasgow G4 0NG, U.K.
| | - Robert W. Martin
- Department
of Physics, SUPA, University of Strathclyde, Glasgow G4 0NG, U.K.
| | - Nicolas Laurand
- Institute
of Photonics, Department of Physics, SUPA,
University of Strathclyde, Glasgow G1 1RD, U.K.
| |
Collapse
|
94
|
Schauenburg D, Gao B, Rochet LNC, Schüler D, Coelho JAS, Ng DYW, Chudasama V, Kuan SL, Weil T. Macrocyclic Dual-Locked "Turn-On" Drug for Selective and Traceless Release in Cancer Cells. Angew Chem Int Ed Engl 2024; 63:e202314143. [PMID: 38179812 DOI: 10.1002/anie.202314143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/21/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
Drug safety and efficacy due to premature release into the bloodstream and poor biodistribution remains a problem despite seminal advances in this area. To circumvent these limitations, we report drug cyclization based on dynamic covalent linkages to devise a dual lock for the small-molecule anticancer drug, camptothecin (CPT). Drug activity is "locked" within the cyclic structure by the redox responsive disulfide and pH-responsive boronic acid-salicylhydroxamate and turns on only in the presence of acidic pH, reactive oxygen species and glutathione through traceless release. Notably, the dual-responsive CPT is more active (100-fold) than the non-cleavable (permanently closed) analogue. We further include a bioorthogonal handle in the backbone for functionalization to generate cyclic-locked, cell-targeting peptide- and protein-CPTs, for targeted delivery of the drug and traceless release in triple negative metastatic breast cancer cells to inhibit cell growth at low nanomolar concentrations.
Collapse
Affiliation(s)
- Dominik Schauenburg
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Bingjie Gao
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Léa N C Rochet
- Department of Chemistry, University College London, London, WC1H 0AJ, UK
| | - Darijan Schüler
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Jaime A S Coelho
- Centro de Química Estrutural, Institute of Molecular Sciences, Faculty of Sciences, University of Lisbon, 1749-016, Lisbon, Portugal
| | - David Y W Ng
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, 55128, Mainz, Germany
| | - Vijay Chudasama
- Department of Chemistry, University College London, London, WC1H 0AJ, UK
| | - Seah Ling Kuan
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, 55128, Mainz, Germany
- Institute of Inorganic Chemistry I, Ulm University, 89081, Ulm, Germany
| | - Tanja Weil
- Synthesis of Macromolecules, Max Planck Institute for Polymer Research, 55128, Mainz, Germany
- Institute of Inorganic Chemistry I, Ulm University, 89081, Ulm, Germany
| |
Collapse
|
95
|
Jaye S, Sandau US, Saugstad JA. Clathrin mediated endocytosis in Alzheimer's disease: cell type specific involvement in amyloid beta pathology. Front Aging Neurosci 2024; 16:1378576. [PMID: 38694257 PMCID: PMC11061891 DOI: 10.3389/fnagi.2024.1378576] [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: 01/29/2024] [Accepted: 04/03/2024] [Indexed: 05/04/2024] Open
Abstract
This review provides a comprehensive examination of the role of clathrin-mediated endocytosis (CME) in Alzheimer's disease (AD) pathogenesis, emphasizing its impact across various cellular contexts beyond neuronal dysfunction. In neurons, dysregulated CME contributes to synaptic dysfunction, amyloid beta (Aβ) processing, and Tau pathology, highlighting its involvement in early AD pathogenesis. Furthermore, CME alterations extend to non-neuronal cell types, including astrocytes and microglia, which play crucial roles in Aβ clearance and neuroinflammation. Dysregulated CME in these cells underscores its broader implications in AD pathophysiology. Despite significant progress, further research is needed to elucidate the precise mechanisms underlying CME dysregulation in AD and its therapeutic implications. Overall, understanding the complex interplay between CME and AD across diverse cell types holds promise for identifying novel therapeutic targets and interventions.
Collapse
Affiliation(s)
| | | | - Julie A. Saugstad
- Department of Anesthesiology & Perioperative Medicine, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
96
|
Aschmann D, Knol RA, Kros A. Lipid-Based Nanoparticle Functionalization with Coiled-Coil Peptides for In Vitro and In Vivo Drug Delivery. Acc Chem Res 2024; 57:1098-1110. [PMID: 38530194 PMCID: PMC11025025 DOI: 10.1021/acs.accounts.3c00769] [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: 12/06/2023] [Revised: 03/12/2024] [Accepted: 03/12/2024] [Indexed: 03/27/2024]
Abstract
For the delivery of drugs, different nanosized drug carriers (e.g., liposomes, lipid nanoparticles, and micelles) have been developed in order to treat diseases that afflict society. Frequently, these vehicles are formed by the self-assembly of small molecules to encapsulate the therapeutic cargo of interest. Over decades, nanoparticles have been optimized to make them more efficient and specific to fulfill tailor-made tasks, such as specific cell targeting or enhanced cellular uptake. In recent years, lipid-based nanoparticles in particular have taken center stage; however, off-targeting side effects and poor endosomal escape remain major challenges since therapies require high efficacy and acceptable toxicity.To overcome these issues, many different approaches have been explored to make drug delivery more specific, resulting in reduced side effects, to achieve an optimal therapeutic effect and a lower required dose. The fate of nanoparticles is largely dependent on size, shape, and surface charge. A common approach to designing drug carriers with targeting capability is surface modification. Different approaches to functionalize nanoparticles have been investigated since the attachment of targeting moieties plays a significant role in whether they can later interact with surface-exposed receptors of cells. To this end, various strategies have been used involving different classes of biomolecules, such as small molecules, nucleic acids, antibodies, aptamers, and peptides.Peptides in particular are often used since there are many receptors overexpressed in different specific cell types. Furthermore, peptides can be produced and modified at a low cost, enabling high therapeutic screening. Cell-penetrating peptides (CPPs) and cell-targeting peptides (CTPs) are frequently used for this purpose. Less studied in this context are fusogenic coiled-coil peptides. Lipid-based nanoparticles functionalized with these peptides are able to avoid the endolysosomal pathway; instead such particles can be taken up by membrane fusion, resulting in increased delivery of payload. Furthermore, they can be used for targeting cells/organs but are not directed at surface-exposed receptors. Instead, they recognize complementary peptide sequences, facilitating their uptake into cells.In this Account, we will discuss peptides as moieties for enhanced cytosolic delivery, targeted uptake, and how they can be attached to lipid-based nanoparticles to alter their properties. We will discuss the properties imparted to the particles by peptides, surface modification approaches, and recent examples showing the power of peptides for in vitro and in vivo drug delivery. The main focus will be on the functionalization of lipid-based nanoparticles by fusogenic coiled-coil peptides, highlighting the relevance of this concept for the development of future therapeutics.
Collapse
Affiliation(s)
- Dennis Aschmann
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| | - Renzo A. Knol
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| | - Alexander Kros
- Leiden Institute of Chemistry, Leiden University, Einsteinweg 55, 2333CC Leiden, The Netherlands
| |
Collapse
|
97
|
Yu L, Zhang M, He J, Sun X, Ni P. A nanomedicine composed of polymer-ss-DOX and polymer-Ce6 prodrugs with monoclonal antibody targeting effect for anti-tumor chemo-photodynamic synergetic therapy. Acta Biomater 2024; 179:272-283. [PMID: 38460931 DOI: 10.1016/j.actbio.2024.02.048] [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: 12/09/2023] [Revised: 02/07/2024] [Accepted: 02/29/2024] [Indexed: 03/11/2024]
Abstract
Anticancer drugs used for systemic chemotherapy often exhibit off-target toxicity and uncontrolled drug release due to their lack of targeting. To improve the bioavailability of drugs and reduce side effects, we have developed a mixed micelle of nanomedicine composed of two prodrugs with surface modified monoclonal antibody for cancer therapy. In this system, Nimotuzumab was used as targeting ligands of the mixed micelles (named as DCMMs) that is composed of polymer-doxorubicin prodrug (abbreviated as PEG-b-P(GMA-ss-DOX)) and maleimide polyethylene glycol-chlorin e6 (abbreviated as Mal-PEG-Ce6). The mixed micelles modified with Nimotuzumab (named as NTZ-DCMMs) bind to overexpressed EGFR receptors on Hepatoma-22 (H22) cells. Disulfide bonds in PEG-b-P(GMA-ss-DOX) are disrupted in tumor microenvironment, inducing the reduction-responsive release of DOX and leading to tumor cell apoptosis. Simultaneously, Chlorin e6 (Ce6) produced plenty of singlet oxygen (1O2) under laser irradiation to kill tumor cells. In vivo biological distribution and antineoplastic effect experiments demonstrate that NTZ-DCMMs enhanced drug enrichment at tumor sites through targeting function of antibody, dramatically suppressing tumor growth and mitigating cardiotoxicity of drugs. All results prove that NTZ-DCMMs have the ability to actively target H22 cells and quickly respond to tumor microenvironment, which is expected to become an intelligent and multifunctional drug delivery carrier for efficient chemotherapy and photodynamic therapy of hepatoma. STATEMENT OF SIGNIFICANCE: Anticancer drugs used for systemic chemotherapy often exhibit off-target toxicity due to their lack of targeting. Therefore, it's necessary to develop effective, targeted, and collaborative treatment strategies. We construct a mixed micelle of nanomedicine based on two polymer prodrugs and modified with monoclonal antibody on surface for cancer therapy. Under the tumor cell microenvironment, the disulfide bonds of polymer-ss-DOX were broken, effectively triggering DOX release. The photosensitizer Ce6 could generate a large amount of ROS under light, which synergistically promotes tumor cell apoptosis. By coupling antibodies to the hydrophilic segments of polymer micelles, drugs can be specifically delivered. Compared with monotherapy, the combination of chemotherapy and photodynamic therapy can significantly enhance the therapeutic effect of liver cancer.
Collapse
Affiliation(s)
- Liang Yu
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou, 215123, PR China
| | - Mingzu Zhang
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou, 215123, PR China
| | - Jinlin He
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou, 215123, PR China
| | - Xingwei Sun
- Intervention Department, The Second Affiliated Hospital of Soochow University, Suzhou, 215004, PR China.
| | - Peihong Ni
- College of Chemistry, Chemical Engineering and Materials Science, State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, Suzhou Key Laboratory of Macromolecular Design and Precision Synthesis, Soochow University, Suzhou, 215123, PR China.
| |
Collapse
|
98
|
Zhang FZ, Tan M, Zeng J, Qi XW, Zhang YT, Che YT, Zhang S, Li BJ. A Supramolecular Assembly of EGCG for Long-Term Treatment of Allergic Rhinitis. ACS Biomater Sci Eng 2024; 10:2282-2298. [PMID: 38526450 DOI: 10.1021/acsbiomaterials.4c00091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
Abstract
Allergic rhinitis (AR) is a type-I hypersensitivity disease mediated by immunoglobulin E (IgE). Although antihistamines, glucocorticoids, leukotriene receptor antagonists, and other drugs are widely used to treat AR, the various adverse side effects of long-term use of these drugs should not be ignored. Therefore, more effective and safe natural alternative strategies are urgently needed. To this end, this study designed a nanosupramolecular delivery system composed of β-cyclodextrin supramolecular polymer (PCD), thiolated chitosan (TCS), and natural polyphenol epigallocatechin gallate (EGCG) for intranasal topical continuous treatment of AR. The TCS/PCD@EGCG nanocarriers exhibited an excellent performance in terms of retention and permeability in the nasal mucosa and released the vast majority of EGCG responsively in the nasal microenvironment, thus resulting in the significantly high antibacterial and antioxidant capacities. According to the in vitro model, compared with free EGCG, TCS/PCD@EGCG inhibited mast cell activity and abnormal histamine secretion in a more long-term and sustained manner. According to the in vivo model, whether in the presence of continuous or intermittent administration, TCS/PCD@EGCG substantially inhibited the secretion of allergenic factors and inflammatory factors, mitigated the pathological changes of nasal mucosa, alleviated the symptoms of rhinitis in mice, and produced a satisfactory therapeutic effect on AR. In particular, the therapeutic effect of TCS/PCD@EGCG systems were even superior to that of budesonide during intermittent treatment. Therefore, the TCS/PCD@EGCG nanocarrier is a potential long-lasting antiallergic medicine for the treatment of AR.
Collapse
Affiliation(s)
- Fu Zhong Zhang
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Min Tan
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Zeng
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xu-Wei Qi
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ye-Tao Zhang
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yu-Ting Che
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sheng Zhang
- State Key Laboratory of Polymer Materials Engineering, Polymer Research Institute of Sichuan University, Sichuan University, Chengdu 610065, China
| | - Bang-Jing Li
- Key Laboratory of Mountain Ecological Restoration and Bioresource Utilization, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610041, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
99
|
Bai S, Chen H, Fu S, Liu C, Gao X, Li S, Chen Y, Lan Y, Xia Y, Dai Q, He P, Zhang Y, Zhao Q, Mao J, Lu Z, Liu G. Bioinspired Tumor Calcification-Guided Early Diagnosis and Eradication of Hepatocellular Carcinoma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310818. [PMID: 38190432 DOI: 10.1002/adma.202310818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/03/2024] [Indexed: 01/10/2024]
Abstract
Tumor calcification is found to be associated with the benign prognostic, and which shows considerable promise as a somewhat predictive index of the tumor response clinically. However, calcification is still a missing area in clinical cancer treatment. A specific strategy is proposed for inducing tumor calcification through the synergy of calcium peroxide (CaO2)-based microspheres and transcatheter arterial embolization for the treatment of hepatocellular carcinoma (HCC). The persistent calcium stress in situ specifically leads to powerful tumor calcioptosis, resulting in diffuse calcification and a high-density shadow on computed tomography that enables clear localization of the in vivo tumor site and partial delineation of tumor margins in an orthotopic HCC rabbit model. This osmotic calcification can facilitate tumor clinical diagnosis, which is of great significance in differentiating tumor response during early follow-up periods. Proteome and phosphoproteome analysis identify that calreticulin (CALR) is a crucial target protein involved in tumor calcioptosis. Further fluorescence molecular imaging analysis also indicates that CALR can be used as a prodromal marker of calcification to predict tumor response at an earlier stage in different preclinical rodent models. These findings suggest that upregulated CALR in association with tumor calcification, which may be broadly useful for quick visualization of tumor response.
Collapse
Affiliation(s)
- Shuang Bai
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- Shaanxi Province Center for Regenerative Medicine and Surgery Engineering Research, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hu Chen
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Shiying Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Chao Liu
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Xing Gao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Shuo Li
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Yulun Chen
- Department of Radiology, Xiang'an Hospital of Xiamen University, Xiamen, 361102, China
| | - Yulu Lan
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Yutian Xia
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Qixuan Dai
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Pan He
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Yang Zhang
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Qingliang Zhao
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Jingsong Mao
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- Department of Radiology, Xiang'an Hospital of Xiamen University, Xiamen, 361102, China
| | - Zhixiang Lu
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361102, China
| | - Gang Liu
- State Key Laboratory of Infectious Disease Vaccine Development, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Biology, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| |
Collapse
|
100
|
Huang HC, Wang TY, Rousseau J, Orlando M, Mungaray M, Michaud C, Plaisier C, Chen ZB, Wang KC. Biomimetic nanodrug targets inflammation and suppresses YAP/TAZ to ameliorate atherosclerosis. Biomaterials 2024; 306:122505. [PMID: 38359507 PMCID: PMC11479593 DOI: 10.1016/j.biomaterials.2024.122505] [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: 05/14/2023] [Revised: 01/21/2024] [Accepted: 02/07/2024] [Indexed: 02/17/2024]
Abstract
Atherosclerosis, a chronic inflammatory disease, is the primary cause of myocardial infarction and ischemic stroke. Recent studies have demonstrated that dysregulation of yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding domain (TAZ) contributes to plaque development, making YAP/TAZ potential therapeutic targets. However, systemic modulation of YAP/TAZ expression or activities risks serious off-target effects, limiting clinical applicability. To address the challenge, this study develops monocyte membrane-coated nanoparticles (MoNP) as a targeted delivery system for activated and inflamed endothelium lining the plaque surface. The MoNP system is used to deliver verteporfin (VP), aimed at inhibiting YAP/TAZ specifically within arterial regions prone to atherosclerosis. The results reveal that MoNP significantly enhance payload delivery to inflamed endothelial cells (EC) while avoiding phagocytic cells. When administered in mice, MoNP predominantly accumulate in intima of the atheroprone artery. MoNP-mediated delivery of VP substantially reduces YAP/TAZ expression, thereby suppressing inflammatory gene expression and macrophage infiltration in cultured EC and mouse arteries exposed to atherogenic stimuli. Importantly, this targeted VP nanodrug effectively decreases plaque development in mice without causing noticeable histopathological changes in major organs. Collectively, these findings demonstrate a lesion-targeted and pathway-specific biomimetic nanodrug, potentially leading to safer and more effective treatments for atherosclerosis.
Collapse
Affiliation(s)
- Hui-Chun Huang
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Ting-Yun Wang
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA; School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, 85287, USA
| | - Joshua Rousseau
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Mark Orlando
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Michelle Mungaray
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Chamonix Michaud
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Christopher Plaisier
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Zhen Bouman Chen
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Kuei-Chun Wang
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|