1
|
Sharma V, Kapil D, Singh B. Developing multifunctional zwitterionic-xanthan gum-anchored network copolymers for biomedical applications. SOFT MATTER 2025; 21:4177-4191. [PMID: 40326335 DOI: 10.1039/d5sm00250h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Herein, mucoadhesive and biocompatible network hydrogels were developed by grafting zwitterionic polymer poly(2-[(methacryoyloxy)ethyl]dimethyl (3-sulfopropyl) ammonium hydroxide (poly(MEDSAH)) onto bacterial-derived polysaccharide xanthan gum (XG) via free radical graft polymerization, followed by crosslinking with N,N'-methylene bisacrylamide (NNMBA). The developed polymeric hydrogels were characterized using XRD, FTIR, 13C-NMR, TGA-DSC, FESEM, EDX and AFM analyses. FESEM and AFM analyses of these hydrogels demonstrated morphological heterogeneous features with surface roughness. Augmentation in the atomic percentages of sulfur and nitrogen atoms in the EDX spectrum of the polymers and the appearance of the characteristic spectral peaks (FTIR and 13C-NMR) of the sulfonic and quaternary ammonium groups of poly(MEDSAH) confirmed the grafting of zwitterionic polymer on the XG backbone. The crosslinked product exhibited modified XRD and thermal degradation (TGA-DSC) patterns, as compared to native XG polysaccharide. The slow and gradual release of ertapenem was observed through supra-molecular interactions and occurred more frequently under neutral to slightly basic conditions. The drug release from the network hydrogels followed non-Fickian type diffusion and was best defined by the Higuchi model. The developed hydrogel-based drug delivery (DD) carriers were found to be bioadhesive with 100.0 ± 5.0 mN force of adhesion, biocompatible with 1.078 ± 0.073% haemolytic index and antioxidant in nature by displaying 28.384 ± 0.963% inhibition during DPPH assay. The physicochemical and biomedical properties of XG-based hydrogels are indicative features for their utilization in gastrointestinal DD applications.
Collapse
Affiliation(s)
- Vikrant Sharma
- Department of Chemistry, Himachal Pradesh University, Shimla 171005, India.
| | - Disha Kapil
- Department of Chemistry, Himachal Pradesh University, Shimla 171005, India.
| | - Baljit Singh
- Department of Chemistry, Himachal Pradesh University, Shimla 171005, India.
| |
Collapse
|
2
|
Zhu S, Yang Z, Liu Y, Cheng L, Long D, Dai F. Oral Lipid Nanoparticles for Improving the Efficiency of Drug Delivery Systems in Ulcerative Colitis: Recent Advances and Future Prospects. Pharmaceutics 2025; 17:547. [PMID: 40430840 PMCID: PMC12114620 DOI: 10.3390/pharmaceutics17050547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 04/12/2025] [Accepted: 04/19/2025] [Indexed: 05/29/2025] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease characterized by persistent, recurrent, and relapsing inflammation of the mucosal layer. Its pathogenesis is complex and not yet fully understood, with current treatments mainly focused on alleviating symptoms through pharmacological methods. Direct drug administration for UC often leads to poor intestinal bioavailability, suboptimal targeting, and an increased risk of resistance. Therefore, there is an urgent need for effective drug delivery systems. Lipid nanoparticles (LNPs) are promising candidates for UC drug delivery due to their high biocompatibility, stability, and customizable properties. Oral administration, as a preferred treatment approach for UC, offers benefits such as convenience, cost-effectiveness, and better patient compliance. However, oral drug delivery systems must navigate the complex gastrointestinal tract to effectively target colonic lesions, posing significant challenges for LNP-based systems. Researchers are exploring ways to enhance oral delivery efficiency by adjusting LNP composition, surface functionalization, and coating. This article reviews recent advancements in oral LNP research aimed at improving drug delivery efficiency for UC treatment and discusses future prospects.
Collapse
Affiliation(s)
- Siyu Zhu
- State Key Laboratory of Resource Insects, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400715, China; (S.Z.); (Z.Y.); (Y.L.); (L.C.)
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Zhenlin Yang
- State Key Laboratory of Resource Insects, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400715, China; (S.Z.); (Z.Y.); (Y.L.); (L.C.)
| | - Yulong Liu
- State Key Laboratory of Resource Insects, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400715, China; (S.Z.); (Z.Y.); (Y.L.); (L.C.)
| | - Lan Cheng
- State Key Laboratory of Resource Insects, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400715, China; (S.Z.); (Z.Y.); (Y.L.); (L.C.)
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
- Yibin Academy of Southwest University, Yibin 644000, China
| | - Dingpei Long
- State Key Laboratory of Resource Insects, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400715, China; (S.Z.); (Z.Y.); (Y.L.); (L.C.)
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
- Yibin Academy of Southwest University, Yibin 644000, China
| | - Fangyin Dai
- State Key Laboratory of Resource Insects, Key Laboratory of Sericultural Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400715, China; (S.Z.); (Z.Y.); (Y.L.); (L.C.)
- College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
- Yibin Academy of Southwest University, Yibin 644000, China
| |
Collapse
|
3
|
Subramanian D, Chin A, Shi Y, Liu GW, Langer R, Traverso G. Identification and Validation of Cyclic Peptides with Mucin-Selective, Location-Specific Binding in the Gastrointestinal Tract. ACS NANO 2025; 19:14693-14706. [PMID: 40216380 PMCID: PMC12020424 DOI: 10.1021/acsnano.4c13520] [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: 09/25/2024] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 04/23/2025]
Abstract
Oral drug delivery is a widely preferred method of drug administration due to its ease of use and convenience for patients. Localization of drug release in the gastrointestinal (GI) tract is important to treat localized diseases and maximize drug absorption. However, achieving drug localization in the dynamic GI tract is challenging. To address this challenge, we leveraged the geographic diversity of the GI tract by targeting its mucus layers, which coat the epithelial surfaces. These layers, composed of mucin glycoproteins, are synthesized with unique chemical compositions and expressed in different regions, making them ideal targets for drug localization. In this article, we identify cyclic peptides that bind selectively to MUC2 (in the intestines) and MUC5AC (in the stomach), serving as targeting ligands to these regions of the GI tract. We demonstrate the effectiveness of these peptides through in vitro, ex vivo, and in vivo experiments, showing that incorporating these targeting ligands can increase binding and selectivity 2-fold to the desired regions, thus potentially overcoming challenges with localizing drug distribution in oral delivery. These results indicate that cyclic peptides can be used to localize drug cargoes at certain sites in the body compared to free drugs.
Collapse
Affiliation(s)
- Deepak
A. Subramanian
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Austin Chin
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
| | - Yunhua Shi
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Gary W. Liu
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Robert Langer
- Department
of Chemical Engineering, Massachusetts Institute
of Technology, Cambridge, Massachusetts 02139, United States
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Giovanni Traverso
- David
H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Mechanical Engineering, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Division
of Gastroenterology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
4
|
Ahmad A, Akhtar J, Ahmad M, Wasim R, Khan MI. Drug Delivery Approaches for Buccal and Sublingual Administration. Drug Res (Stuttg) 2025. [PMID: 40262781 DOI: 10.1055/a-2560-9884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Both local and systemic medication delivery benefit greatly from the sublingual and buccal modes of administration. They have shown to be a successful substitute for the conventional oral route, particularly in situations requiring a quick commencement of action. Via venous drainage to the superior vena cava, drugs can enter the systemic circulation quickly and directly. They are therefore helpful for individuals who have trouble swallowing as well as for medications that are highly cleared by the liver or degraded in the gastrointestinal system. Traditionally, medications that are delivered through the buccal and sublingual channels are made in three different dose forms: liquid (such as sprays and drops), semi-solid (such as gels), and solid (such as pills, wafers, films, and patches). Physiological variables frequently influence conventional dose forms, which might decrease the formulation's interaction with the mucosa and result in unexpected medication absorption. Many formulation development advancements have been made to enhance medication absorption and retention in the buccal and sublingual areas. The physiological factors influencing buccal and sublingual drug delivery as well as developments in nanoparticulate drug delivery techniques for sublingual and buccal administration will be the main topics of this review. It also discusses about the clinical development pipeline, which includes formulations that have been authorized and are undergoing clinical studies.
Collapse
Affiliation(s)
- Asad Ahmad
- Faculty of Pharmacy, Integral University, Lucknow, India
| | - Juber Akhtar
- Faculty of Pharmacy, Integral University, Lucknow, India
| | - Mohammad Ahmad
- Faculty of Pharmacy, Integral University, Lucknow, India
| | - Rufaida Wasim
- Faculty of Pharmacy, Integral University, Lucknow, India
| | | |
Collapse
|
5
|
Obeidat WM, Lahlouh IK. Chitosan Nanoparticles: Approaches to Preparation, Key Properties, Drug Delivery Systems, and Developments in Therapeutic Efficacy. AAPS PharmSciTech 2025; 26:108. [PMID: 40244367 DOI: 10.1208/s12249-025-03100-z] [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: 12/31/2024] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
The integration of nanotechnology into drug delivery systems holds great promise for enhancing pharmaceutical effectiveness. This approach enables precise targeting, controlled release, improved patient compliance, reduced side effects, and increased bioavailability. Nanoparticles are vital for transporting biomolecules-such as proteins, enzymes, genes, and vaccines-through various administration routes, including oral, intranasal, vaginal, buccal, and pulmonary. Among biodegradable polymers, chitosan, a linear polysaccharide derived from chitin, stands out due to its biocompatibility, safety, biodegradability, mucoadhesive properties, and ability to enhance permeation. Its cationic nature supports strong molecular interactions and provides antimicrobial, anti-inflammatory, and hemostatic benefits. However, its solubility, influenced by pH and ionic sensitivity, poses challenges requiring effective solutions. This review explores chitosan, its modified derivatives and chitosan nanoparticles mainly, focusing on nanoparticles physicochemical properties, drug release mechanisms, preparation methods, and factors affecting their mean hydrodynamic diameter (particle size). It highlights their application in drug delivery systems and disease treatments across various routes. Key considerations include drug loading capacity, zeta potential, and stability, alongside the impact of molecular weight, degree of deacetylation, and drug solubility on nanoparticle properties. Recent advancements and studies underscore chitosan's potential, emphasizing its modified derivatives'versatility in improving therapeutic outcomes.
Collapse
Affiliation(s)
- Wasfy M Obeidat
- Jordan University of Science and Technology, 3030, Irbid, 22110, Jordan.
| | - Ishraq K Lahlouh
- Jordan University of Science and Technology, 3030, Irbid, 22110, Jordan
| |
Collapse
|
6
|
Jeong HS, Choi Y, Kim DW, Je JY, Lee SJ, Choi CH. Multicompartmental Hydrogel Microspheres with a Concentric Thin Oil Layer: Protecting and Targeting Therapeutic Agents for Inflammatory Bowel Disease. ACS APPLIED BIO MATERIALS 2025; 8:2251-2262. [PMID: 40030207 DOI: 10.1021/acsabm.4c01763] [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: 03/18/2025]
Abstract
Although oral delivery of therapeutic agents offers numerous benefits, its application is limited due to the digestive tract's harsh conditions (e.g., strong acidity and high osmolarity), which impair activity and create challenges in achieving targeted release into the intestine. Here, we present multicompartmental hydrogel microspheres equipped with a concentric oil layer to significantly enhance the oral drug delivery efficiency for treating inflammatory bowel disease (IBD). These microspheres are created through the utilization of triple-emulsion droplets, featuring intermediate oil layers that distinctively separate two prepolymer phases, allowing us to fine-tune the composition of each compartment through a tailored polymerization strategy. We demonstrate that the oil layer can protect the encapsulated material by preventing exposure to the acidic environment of the stomach during the digestive process. Unlike aqueous core capsules, the core is composed of hydrogel, which provides high stability even under high osmolarity conditions in the stomach. By fine-tuning the shell's composition, we can develop capsules that release selectively in response to the gut's pH conditions. We demonstrate the system's efficacy by preserving the anti-inflammatory activities of 5-aminosalicylic acid (5-ASA) and Lys-Pro-Val (KPV) under stomach conditions and maintaining their therapeutic effects on colonic epithelial cell migration and proliferation.
Collapse
Affiliation(s)
- Hye-Seon Jeong
- School of Chemical Engineering, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Yoon Choi
- Division of Cosmetic Science and Technology, Daegu Haany University, 1 Haanydaero, Gyeongsan, Gyeongbuk 38610, Republic of Korea
| | - Do-Wan Kim
- Department of Pharmaceutical Engineering, Daegu Haany University, Gyeongsan 38610, Republic of Korea
| | - Jae-Young Je
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Sei-Jung Lee
- Major of Human Bio-convergence, Division of Smart Healthcare, Pukyong National University, Busan 48513, Republic of Korea
| | - Chang-Hyung Choi
- School of Chemical Engineering, Yeungnam University, 280, Daehak-ro, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| |
Collapse
|
7
|
Cardoso VDO, Bistaffa MJ, Sterman RG, Lima LLD, Toldo GS, Cancino-Bernardi J, Zucolotto V. Nanomedicine Innovations for Lung Cancer Diagnosis and Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:13197-13220. [PMID: 40045524 PMCID: PMC11891907 DOI: 10.1021/acsami.4c16840] [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] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 03/12/2025]
Abstract
Lung cancer remains a challenge within the realm of oncology. Characterized by late-stage diagnosis and resistance to conventional treatments, the currently available therapeutic strategies encompass surgery, radiotherapy, chemotherapy, immunotherapy, and biological therapy; however, overall patient survival remains suboptimal. Nanotechnology has ushered in a new era by offering innovative nanomaterials with the potential to precisely target cancer cells while sparing healthy tissues. It holds the potential to reshape the landscape of cancer management, offering hope for patients and clinicians. The assessment of these nanotechnologies follows a rigorous evaluation process similar to that applied to chemical drugs, which includes considerations of their pharmacokinetics, pharmacodynamics, toxicology, and clinical effectiveness. However, because of the characteristics of nanoparticles, standard toxicological tests require modifications to accommodate their unique characteristics. Effective therapeutic strategies demand a profound understanding of the disease and consideration of clinical outcomes, physicochemical attributes of nanomaterials, nanobiointeractions, nanotoxicity, and regulatory compliance to ensure patient safety. This review explores the promise of nanomedicine in lung cancer treatment by capitalizing on its unique physicochemical properties. We address the multifaceted challenges of lung cancer and its tumor microenvironment and provide an overview of recent developments in nanoplatforms for early diagnosis and treatment that can enhance patient outcomes and overall quality of life.
Collapse
Affiliation(s)
- Valéria
Maria de Oliveira Cardoso
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, 13560-970 São Carlos, São Paulo, Brazil
| | - Maria Julia Bistaffa
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, 13560-970 São Carlos, São Paulo, Brazil
| | - Raquel González Sterman
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, 13560-970 São Carlos, São Paulo, Brazil
| | - Lorena Leticia
Peixoto de Lima
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, 13560-970 São Carlos, São Paulo, Brazil
| | - Gustavo Silveira Toldo
- Chemistry
Department, Laboratory in Bioanalytical of Nanosystems, Faculty of
Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, 14040-901 Ribeirão Preto, São Paulo, Brazil
| | - Juliana Cancino-Bernardi
- Chemistry
Department, Laboratory in Bioanalytical of Nanosystems, Faculty of
Philosophy, Sciences and Letters of Ribeirão Preto, University of São Paulo, 14040-901 Ribeirão Preto, São Paulo, Brazil
| | - Valtencir Zucolotto
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, 13560-970 São Carlos, São Paulo, Brazil
- Comprehensive
Center for Precision Oncology, C2PO, University of São Paulo, São Paulo 01246-000, Brazil
| |
Collapse
|
8
|
Gvozdeva Y, Staynova R. pH-Dependent Drug Delivery Systems for Ulcerative Colitis Treatment. Pharmaceutics 2025; 17:226. [PMID: 40006593 PMCID: PMC11858926 DOI: 10.3390/pharmaceutics17020226] [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: 12/30/2024] [Revised: 01/23/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Inflammatory bowel diseases (IBDs), such as ulcerative colitis (UC) or Crohn's disease, are becoming a growing global problem due to the limitations of current treatments, which fail to address the needs of patients effectively. UC is characterized by the widespread inflammation of the mucosal lining, affecting both the rectum and the entire length of the colon. Over the past forty years, traditional treatments for IBDs have primarily relied on anti-inflammatory drugs and immunosuppressive medications. Treatment could be more effective if drugs could be specifically targeted to act directly on the colon. Conventional drug delivery systems for IBDs encounter numerous challenges on their way to the colon, such as physiological barriers and disease severity. To address these issues, pH-dependent carriers have emerged as a promising advancement, offering a more effective and tolerable treatment for UC. These carriers enable localized, targeted action, reducing side effects and preventing the premature clearance of drugs from inflamed colon tissues. pH-responsive systems are a leading approach for targeted drug release in colitis treatment as they take advantage of the varying pH levels throughout the gastrointestinal tract (GIT). By incorporating pH-sensitive polymers, they ensure drug protection and controlled release in the lower GIT. This review will discuss the advantages and limitations of pH-dependent drug delivery systems for colon-targeted drug delivery.
Collapse
Affiliation(s)
- Yana Gvozdeva
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Radiana Staynova
- Department of Organisation and Economics of Pharmacy, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria;
| |
Collapse
|
9
|
Sriram A, Ithape H, Singh PK. Deep-insights: Nanoengineered gel-based localized drug delivery for arthritis management. Asian J Pharm Sci 2025; 20:101012. [PMID: 39995751 PMCID: PMC11848107 DOI: 10.1016/j.ajps.2024.101012] [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: 07/17/2023] [Revised: 05/15/2024] [Accepted: 07/03/2024] [Indexed: 02/26/2025] Open
Abstract
Arthritis is an inflammatory joint disorder that progressively impairs function and diminishes quality of life. Conventional therapies often prove ineffective, as oral administration lacks specificity, resulting in off-target side effects like hepatotoxicity and GIT-related issues. Intravenous administration causes systemic side effects. The characteristic joint-localized symptoms such as pain, stiffness, and inflammation make the localized drug delivery suitable for managing arthritis. Topical/transdermal/intra-articular routes have become viable options for drug delivery in treating arthritis. However, challenges with those localized drug delivery routes include skin barrier and cartilage impermeability. Additionally, conventional intra-articular drug delivery also leads to rapid clearance of drugs from the synovial joint tissue. To circumvent these limitations, researchers have developed nanocarriers that enhance drug permeability through skin and cartilage, influencing localized action. Gel-based nanoengineered therapy employs a gel matrix to incorporate the drug-encapsulated nanocarriers. This approach combines the benefits of gels and nanocarriers to enhance therapeutic effects and improve patient compliance. This review emphasizes deep insights into drug delivery using diverse gel-based novel nanocarriers, exploring their various applications embedded in hyaluronic acid (biopolymer)-based gels, carbopol-based gels, and others. Furthermore, this review discusses the influence of nanocarrier pharmacokinetics on the localization and therapeutic manipulation of macrophages mediated by nanocarriers. The ELVIS (extravasation through leaky vasculature and inflammatory cell-mediated sequestration) effect associated with arthritis is advantageous in drug delivery. Simply put, the ELVIS effect refers to the extravasation of nanocarriers through leaky vasculatures, which finally results in the accumulation of nanocarriers in the joint cavity.
Collapse
Affiliation(s)
| | | | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Telangana 500037, India
| |
Collapse
|
10
|
Abomosallam M, Hendam BM, Shouman Z, Refaat R, Hashem NMA, Sakr SA, Wahed NM. Rutin Nanoparticles Alleviate Cadmium-Induced Oxidative and Immune Damage in Broilers' Bursa of Fabricius via Modulating Hsp70/TLR4/NF-κB Signaling Pathway. Biol Trace Elem Res 2025; 203:1016-1034. [PMID: 38703309 PMCID: PMC11750906 DOI: 10.1007/s12011-024-04199-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/22/2024] [Indexed: 05/06/2024]
Abstract
Cadmium (Cd) is a serious environmental pollutant affecting various tissues/organs in broilers and compromising their immunological function and productivity. Therefore, the current study aimed to investigate Cd-induced immunotoxicity and potential immunoprotective effect of rutin nanoparticles (RNPs) in the bursal tissue of broilers. A total number of 150 chicks from the Hubbard breed were randomly divided into 5 groups. Group I was fed on standard basal diet (SD) with normal drinking water (DW), Group II received SD containing RNPs (50 mg/kg feed) with DW, Group III fed on SD and DW containing Cd (150 mg/L), Group IV co-treated with rutin-enforced SD (50 mg/kg diet) and DW containing Cd (150 mg/L), and finally, Group V co-supplemented with RNP-enhanced SD (50 mg/kg diet) DW containing Cd (150 mg/L). Productive performance, economic efficiency, oxidative biomarkers, histopathological changes, and the expression level of TLR-4, HSP-70, caspase 3, NF-κB, Bcl-2, and Bax were assessed in the BF tissue. Cd led to severe production and economic losses in exposed birds with a marked surge of oxidative biomarkers, pro-inflammatory cytokines, and histopathological changes in the bursal tissue which could be explained through upregulation of the Hsp70/TLR4/NF-κB molecular pathway in the BF tissue. Meanwhile, RNPs could alleviate most of these changes and prevail optimistic immunomodulatory properties which subsequently could enhance broilers' productivity when incorporated in their diets.
Collapse
Affiliation(s)
- Mohamed Abomosallam
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt.
| | - Basma M Hendam
- Department of Animal Wealth Development, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Zeinab Shouman
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Rasha Refaat
- Phytochemistry and Plant Systematics Department, National Research Center, Dokki, Giza, 12622, Egypt
| | - Nada M A Hashem
- Department of Physiology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Shimaa A Sakr
- Department of Animal Wealth Development, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| | - Noha M Wahed
- Department of Animal Wealth Development, Faculty of Veterinary Medicine, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
11
|
Minaiyan P, Varshosaz J, Minaiyan M. Colon delivery of agomelatine nanoparticles in the treatment of TNBS induced ulcerative colitis. Drug Deliv Transl Res 2025:10.1007/s13346-025-01794-z. [PMID: 39856440 DOI: 10.1007/s13346-025-01794-z] [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] [Accepted: 01/08/2025] [Indexed: 01/27/2025]
Abstract
Agomelatine is an atypical antidepressant with a long half-life and the mechanism of action similar to melatonin. Agomelatine is a strong antioxidant and its anti-inflammatory effect has been reported in many studies. The current study aimed to evaluate the anti-inflammatory effect of agomelatine loaded in targeted nanoparticles (NPs) in an experimental colitis model induced by trinitrobenzene sulfonic acid (TNBS). Poly(1-vinylpyrrolidone)-graft-(1-triacontene) (PVP-TA) and Eudragit®-FS30D polymers were used alone and in combination as time, pH and time/pH dependent formulations respectively. The optimal formula was selected according to their physicochemical properties such as particle size, morphology, and drug release pattern. Six separate groups of rats were induced with 0.5 ml of TNBS. The designed groups were: normal, untreated, agomelatine (25 mg/kg/d), agomelatine/ Eudragit®-FS30D NPs, agomelatine/ Eudragit-FS30D/PVP-TA NPs, and dexamethasone (Dex., 1 mg/kg/d). Twenty-four hours after the last administration, colonic tissue was analyzed for macroscopic and histopathological evaluations, along with quantification of malondialdehyde (MDA) and myeloperoxidase (MPO) levels. The results showed that the PVP-TA NPs alone was not suitable regarding to release profile and particle size distribution. However, Eudragit-FS30D NPs alone and Eudragit-FS30D + PVP-TA NPs passed physicochemical evaluations and were both effective in reducing the symptoms and indices of experimental colitis. Taken together, targeted NPs of agomelatine are potentially effective in treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Parinaz Minaiyan
- Department of Pharmaceutics, Faculty of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jaleh Varshosaz
- Department of Pharmaceutics, Faculty of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran.
- Novel Drug Delivery Systems Research Center, Department of Pharmaceutics, Faculty of Pharmacy, Isfahan University of Medical Sciences, PO Box 81745-359, Isfahan, Iran.
| | - Mohsen Minaiyan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
12
|
Kiilerich KF, Andresen T, Darbani B, Gregersen LHK, Liljensøe A, Bennike TB, Holm R, Moeller JB, Andersen V. Advancing Inflammatory Bowel Disease Treatment by Targeting the Innate Immune System and Precision Drug Delivery. Int J Mol Sci 2025; 26:575. [PMID: 39859291 PMCID: PMC11765494 DOI: 10.3390/ijms26020575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/04/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Inflammatory bowel disease (IBD), encompassing Crohn's disease and ulcerative colitis, involves chronic inflammation of the gastrointestinal tract. Current immune-modulating therapies are insufficient for 30-50% of patients or cause significant side effects, emphasizing the need for new treatments. Targeting the innate immune system and enhancing drug delivery to inflamed gut regions are promising strategies. Neutrophils play a central role in IBD by releasing reactive oxygen species (ROS) and neutrophil extracellular traps (NETs) -DNA-based structures with cytotoxic proteins-that contribute to mucosal damage and inflammation. Recent studies linking ROS production, DNA repair, and NET formation have identified NETs as potential therapeutic targets, with preclinical models showing positive outcomes from NET inhibition. Innovative oral drug delivery systems designed to target gut inflammation directly-without systemic absorption-could improve treatment precision and reduce side effects. Advanced formulations utilize properties such as particle size, surface modifications, and ROS-triggered release to selectively target the distal ileum and colon. A dual strategy that combines a deeper understanding of IBD pathophysiology to identify inflammation-related therapeutic targets with advanced drug delivery systems may offer significant promise. For instance, pairing NET inhibition with ROS-responsive nanocarriers could enhance treatment efficacy, though further research is needed. This synergistic approach has the potential to greatly improve outcomes for IBD patients.
Collapse
Affiliation(s)
- Kat F. Kiilerich
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (K.F.K.); (J.B.M.)
| | - Trine Andresen
- Department of Health Science and Technology, The Faculty of Medicine, Aalborg University, 9220 Aalborg Ø, Denmark; (T.A.); (T.B.B.)
| | - Behrooz Darbani
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
| | - Laura H. K. Gregersen
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark
| | - Anette Liljensøe
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
| | - Tue B. Bennike
- Department of Health Science and Technology, The Faculty of Medicine, Aalborg University, 9220 Aalborg Ø, Denmark; (T.A.); (T.B.B.)
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
| | - René Holm
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, 5000 Odense, Denmark;
| | - Jesper B. Moeller
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (K.F.K.); (J.B.M.)
- Danish Institute for Advanced Study, University of Southern Denmark, 5000 Odense, Denmark
| | - Vibeke Andersen
- Molecular Diagnostic and Clinical Research Unit, University Hospital of Southern Denmark, 6200 Aabenraa, Denmark; (B.D.); (L.H.K.G.); (A.L.)
- Department of Regional Health Research, University of Southern Denmark, 5000 Odense, Denmark
| |
Collapse
|
13
|
Kendre PN, Kayande DR, Pote AK, Kanthale SB, Prajapati BG, Kendre Y, Jain S. Emerging Lipid-based Carriers for Systematic Utilization in the Pharmaceutical and Biomedical Sciences: A Review. Pharm Nanotechnol 2025; 13:2-21. [PMID: 38284709 DOI: 10.2174/0122117385268268231204061938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/07/2023] [Accepted: 10/16/2023] [Indexed: 01/30/2024]
Abstract
Emerging lipid-based carriers are revolutionizing drug delivery in the pharmaceutical and biomedical sciences. These innovative carriers harness the unique properties of lipids to improve the solubility, stability, and targeted delivery of therapeutic agents, ushering in a new era of precision medicine. Lipid- based carriers, such as liposomes, lipid nanoparticles, and solid lipid nanoparticles, offer several advantages. They can encapsulate both hydrophilic and hydrophobic drugs, enabling the delivery of a wide range of compounds. Additionally, lipids are biocompatible and biodegradable, minimizing the risk of toxicity. Their ability to mimic cell membranes allows for enhanced cellular uptake and controlled release, optimizing drug efficacy while minimizing side effects. Furthermore, lipid-based carriers are ideal for delivering drugs to specific sites within the body. By modifying the lipid composition, surface charge, and size, researchers can tailor these carriers to target tumours, inflamed tissues, or specific cells, improving therapeutic outcomes and reducing systemic toxicity. In summary, emerging lipid-based carriers are poised to transform pharmaceutical and biomedical sciences by addressing critical challenges in drug delivery. These carriers enhance drug stability, bioavailability, and targeted delivery, offering the potential to revolutionize the treatment of various diseases and improve patient outcomes. As research in this field continues to advance, we can expect even more sophisticated lipid-based carrier systems to emerge, further expanding the possibilities for precision medicine. This review focuses on the contribution of lipid carriers in the pharmaceutical and biomedical sciences.
Collapse
Affiliation(s)
- Prakash N Kendre
- Rajarshi Shahu College of Pharmacy, Buldhana, 443001, Maharashtra, India
| | - Dhiraj R Kayande
- Rajarshi Shahu College of Pharmacy, Buldhana, 443001, Maharashtra, India
| | - Ajinkya K Pote
- Department of Pharmaceutics, Rajarshi Shahu College of Pharmacy, Khamgaon-Botha Road, Malvihir, Buldhana, 443001, India
| | | | - Bhupendra G Prajapati
- S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Ganpat Vidya Nagar, Mehsana, 384012, Gujrat, India
| | - Yuvraj Kendre
- Podar International School, Buldhana, 443001, Maharashtra, India
| | - Shirish Jain
- Rajarshi Shahu College of Pharmacy, Buldhana, 443001, Maharashtra, India
| |
Collapse
|
14
|
Yu T, Bai R, Wang Z, Qin Y, Wang J, Wei Y, Zhao R, Nie G, Han B. Colon-targeted engineered postbiotics nanoparticles alleviate osteoporosis through the gut-bone axis. Nat Commun 2024; 15:10893. [PMID: 39738035 PMCID: PMC11686147 DOI: 10.1038/s41467-024-55263-1] [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: 04/23/2024] [Accepted: 12/06/2024] [Indexed: 01/01/2025] Open
Abstract
The potential for mitigating intestinal inflammation through the gut-bone axis in the treatment of osteoporosis is significant. While various gut-derived postbiotics or bacterial metabolites have been created as dietary supplements to prevent or reverse bone loss, their efficacy and safety still need improvement. Herein, a colon-targeted drug delivery system is developed using surface engineering of polyvinyl butyrate nanoparticles by shellac resin to achieve sustained release of postbiotics butyric acid at the colorectal site. These engineered postbiotics nanoparticles can effectively suppress macrophage inflammatory activation, modulate the redox balance, and regulate the composition of the gut microbiota, thereby restoring epithelial barriers, inhibiting bacterial invasion, and down-regulating pro-inflammatory responses. As a result, the remission of systemic inflammation is accompanied by a rebalancing of osteoblast and osteoclast activity, alleviating inflammatory bowel disease-related and post-menopausal bone loss. Specifically, the treatment of engineered postbiotics nanoparticles can also improve the quality and quantity of bone with restoration of deteriorative mechanical properties, which indicating a therapeutic potential on fracture prevention. This study provides valuable insights into the gut-bone axis and establishes a promising and safe therapeutic strategy for osteoporosis.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
| | - Rushui Bai
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Zeming Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yuting Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Jingwei Wang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China
- National Center for Stomatology, Beijing, China
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Yaohua Wei
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China
| | - Ruifang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China.
| | - Bing Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, China.
- National Center for Stomatology, Beijing, China.
- National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, China.
| |
Collapse
|
15
|
Li JH, Xu J, Huang C, Hu JX, Xu HM, Guo X, Zhang Y, Xu JK, Peng Y, Zhang Y, Zhu MZ, Zhou YL, Nie YQ. Houttuynia cordata-Derived Exosome-Like Nanoparticles Mitigate Colitis in Mice via Inhibition of the NLRP3 Signaling Pathway and Modulation of the Gut Microbiota. Int J Nanomedicine 2024; 19:13991-14018. [PMID: 39742094 PMCID: PMC11687308 DOI: 10.2147/ijn.s493434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/19/2024] [Indexed: 01/03/2025] Open
Abstract
BACKGROUND Plant-derived exosome-like nanoparticles (PELNs) have received widespread attention in treating ulcerative colitis (UC). However, the role of Houttuynia cordata-derived exosome-like nanoparticles (HELNs) in UC remains unclear. This study aims to evaluate the efficacy of HELNs in treating colitis in mice and investigate its potential mechanisms. METHODS HELNs were isolated from H. cordata for characterization, and their safety and stability were evaluated. A dextran sulfate sodium (DSS)-induced colitis mouse model was utilized to assess the therapeutic potential of HELNs in UC. In vivo, imaging and flow cytometry were utilized to investigate the targeting effect of HELNs on inflamed colonic sites and their modulation of the immune environment. RNA-seq analysis and molecular docking were performed to identify potential pathways recruited by HELNs. Guided by transcriptomic findings, NLRP3-/- mice were used in conjunction with Western blotting, qPCR, immunofluorescence, and other techniques to verify that HELNs alleviated DSS-induced colitis by inhibiting NLRP3/NOD-like receptor signaling pathways. Lastly, the impact of HELNs on the gut microbiota was investigated through 16S rRNA sequencing. RESULTS HELNs significantly reduced the severity of DSS-induced colitis in mice, alleviating colitis symptoms and histopathological damage. Furthermore, HELNs can specifically target inflamed colon tissue, regulate the immune environment, and decrease inflammation. RNA-seq analysis, coupled with the use of NLRP3-/- mice, demonstrated that HELNs inhibited the NLRP3/NOD-like receptor signaling pathways. Lastly, HELNs balanced the gut microbiota composition in mice with colitis, decreasing the abundance of harmful bacteria and increasing the abundance of beneficial bacteria in the intestinal tract of these mice. CONCLUSION In summary, HELNs exhibit the potential to protect the colon from DSS-induced damage by inhibiting the NLRP3/NOD-like receptor signaling pathway and modulating the gut microbiota, presenting a promising therapeutic option for the management of UC.
Collapse
Affiliation(s)
- Jian-Hong Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Chen Huang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Jin-Xia Hu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Xue Guo
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yan Zhang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Jing-Kui Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yao Peng
- Department of Gastroenterology and Hepatology, Shenzhen General Hospital, Shenzhen, People’s Republic of China
| | - Yong Zhang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - You-Lian Zhou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yu-Qiang Nie
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
16
|
Ranjan AP, Czyzyk DJ, Martinez-Traverso G, Sadiqova A, Valhondo M, Schaefer DA, Spasov KA, Jorgensen WL, Vishwanatha JK, Riggs MW, Castellanos-Gonzalez A, Anderson KS. Prodrug nanotherapy demonstrates in vivo anticryptosporidial efficacy in a mouse model of chronic Cryptosporidium infection. RSC PHARMACEUTICS 2024; 1:963-975. [PMID: 39372445 PMCID: PMC11447440 DOI: 10.1039/d4pm00093e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/16/2024] [Indexed: 10/08/2024]
Abstract
The gastrointestinal disease cryptosporidiosis, caused by the genus Cryptosporidium, is a common cause of diarrheal diseases in children, particularly in developing countries and frequently fatal in immunocompromised individuals. Cryptosporidium hominis (Ch)-specific bifunctional dihydrofolate reductase-thymidylate synthase (DHFR-TS) has been a molecular target for inhibitor design. (Note that this bifunctional enzyme has also been referred to as TS-DHFR in previous literature since the functional biochemical reaction first involves the conversion of methylene tetrahydrofolate to dihydrofolate at the TS site.) While nanomolar inhibitors of Ch DHFR-TS have been identified at the biochemical level, effective delivery of these compounds to achieve anticryptosporidial activity in cell culture and in vivo models of parasite infection remains a major challenge in developing new therapies. Previous studies, using a nanotherapy approach, have shown a promising Ch DHFR-TS inhibitor, 906, that can successfully target Cryptosporidium parasites in cell culture with nanomolar anticryptosporidial activity. This formulation utilized poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) loaded with 906 (NP-906) and conjugated with a Cryptosporidium monoclonal antibody (MAb) on the nanoparticle surface to specifically target the glycoprotein GP25-200 in excysting oocysts. However, a limitation for in vivo use is antibody susceptibility to gastric acidity. To address this gap, a prodrug diethyl ester form of 906 (MAb-NP-Prodrug) was synthesized that allowed higher compound loading in the MAb-coated PLGA nanoparticles. An oral formulation was prepared by loading lyophilized MAb-NP-Prodrug into gelatin capsules with an enteric coating for gastric stability. Proof-of-concept studies with this oral formulation demonstrated antiparasitic activity in a chronic mouse model of Cryptosporidium infection. Efficacy was observed after a low daily dose of 2 × 8 mg kg-1 for 5 days, when examined 6 and 20 days postinfection, offering a new avenue of drug delivery to be further explored.
Collapse
Affiliation(s)
- Amalendu P Ranjan
- Department of Microbiology, Immunology and Genetics, College of Biomedical and Translational Sciences, University of North Texas Health Science Center Fort Worth TX 76107 USA
| | - Daniel J Czyzyk
- Department of Pharmacology, Yale University School of Medicine 333 Cedar Street New Haven CT 06520 USA +1 (203) 785-4526
| | - Griselle Martinez-Traverso
- Infectious Disease Division, Department of Internal Medicine, University of Texas Medical Branch Galveston TX 77555 USA +1 (409) 772-3729
| | - Aygul Sadiqova
- Infectious Disease Division, Department of Internal Medicine, University of Texas Medical Branch Galveston TX 77555 USA +1 (409) 772-3729
| | - Margarita Valhondo
- Department of Chemistry, Yale University 225 Prospect Street PO Box 208107 New Haven CT 06520 USA
| | - Deborah A Schaefer
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona Tucson AZ 85721 USA
| | - Krasimir A Spasov
- Department of Pharmacology, Yale University School of Medicine 333 Cedar Street New Haven CT 06520 USA +1 (203) 785-4526
| | - William L Jorgensen
- Department of Chemistry, Yale University 225 Prospect Street PO Box 208107 New Haven CT 06520 USA
| | - Jamboor K Vishwanatha
- Department of Microbiology, Immunology and Genetics, College of Biomedical and Translational Sciences, University of North Texas Health Science Center Fort Worth TX 76107 USA
| | - Michael W Riggs
- School of Animal and Comparative Biomedical Sciences, College of Agriculture and Life Sciences, University of Arizona Tucson AZ 85721 USA
| | - Alejandro Castellanos-Gonzalez
- Infectious Disease Division, Department of Internal Medicine, University of Texas Medical Branch Galveston TX 77555 USA +1 (409) 772-3729
| | - Karen S Anderson
- Department of Pharmacology, Yale University School of Medicine 333 Cedar Street New Haven CT 06520 USA +1 (203) 785-4526
- Department of Molecular Biophysics and Biochemistry, Yale University School of Medicine 333 Cedar Street New Haven CT 06520 USA
| |
Collapse
|
17
|
Zhang H, Wu A, Nan X, Yang L, Zhang D, Zhang Z, Liu H. The Application and Pharmaceutical Development of Etomidate: Challenges and Strategies. Mol Pharm 2024; 21:5989-6006. [PMID: 39495089 DOI: 10.1021/acs.molpharmaceut.4c00325] [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: 11/05/2024]
Abstract
Etomidate is a synthetic imidazole anesthetic that exerts hypnotic effects by potentiating the action of the inhibitory neurotransmitter γ-aminobutyric acid (GABA) or directly activating the anionic GABA (GABAA) receptor. It stands out among many anesthetics because of its multiple advantages, such as good hemodynamic stability and minimal inhibition of spontaneous respiration. However, its low water solubility and side effects, such as adrenal cortex inhibition and myoclonus, have limited the clinical application of this drug. To address these issues, extensive research has been conducted on the drug delivery of etomidate in recent decades, which has led to the emergence of different etomidate preparations. Despite so many etomidate preparations, so far some of the toxic side effects have not yet been effectively addressed. Herein we discuss the pharmaceutical design of etomidate that may resolve the above problem. We also propose targeted strategies for future research on etomidate preparations and discuss the feasibility of different administration routes and dosage forms to expand the application of this drug. Through this review, we hope to draw more attention to the potential of etomidate and its application challenges and provide valuable insights into the development of new etomidate preparations.
Collapse
Affiliation(s)
- Hao Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
- Department of Pharmacy, Zigong First People's Hospital, Zigong, Sichuan 643000, People's Republic of China
| | - Ailing Wu
- Department of Anesthesiology, Second People's Hospital of Neijiang, Southwest Medical University, Neijiang, Sichuan 641000, People's Republic of China
- Department of Anesthesiology, First People's Hospital of Neijiang, Neijiang, Sichuan 641099, People's Republic of China
| | - Xichen Nan
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Luhan Yang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Dan Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Zhuo Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| | - Hao Liu
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, People's Republic of China
| |
Collapse
|
18
|
Li S, Wu T, Wu J, Chen W, Zhang D. Recognizing the biological barriers and pathophysiological characteristics of the gastrointestinal tract for the design and application of nanotherapeutics. Drug Deliv 2024; 31:2415580. [PMID: 39404464 PMCID: PMC11485891 DOI: 10.1080/10717544.2024.2415580] [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: 05/07/2024] [Revised: 10/04/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
The gastrointestinal tract (GIT) is an important and complex system by which humans to digest food and absorb nutrients. The GIT is vulnerable to diseases, which may led to discomfort or even death in humans. Therapeutics for GIT disease treatment face multiple biological barriers, which significantly decrease the efficacy of therapeutics. Recognizing the biological barriers and pathophysiological characteristics of GIT may be helpful to design innovative therapeutics. Nanotherapeutics, which have special targeting and controlled therapeutic release profiles, have been widely used for the treatment of GIT diseases. Herein, we provide a comprehensive review of the biological barrier and pathophysiological characteristics of GIT, which may aid in the design of promising nanotherapeutics for GIT disease treatment. Furthermore, several typical diseases of the upper and lower digestive tracts, such as Helicobacter pylori infection and inflammatory bowel disease, were selected to investigate the application of nanotherapeutics for GIT disease treatment.
Collapse
Affiliation(s)
- Shan Li
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Army 953 Hospital, Shigatse Branch of Xinqiao Hospital, Army Medical University (Third Military Medical University), Shigatse, Tibet Autonomous Region, China
| | - Tianyu Wu
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jingfeng Wu
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wensheng Chen
- Department of Gastroenterology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Dinglin Zhang
- Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
19
|
Pereira AAR, Aparecida JVM, Ramalho ME, Ferreira LMB, Gremião MPD. Tailoring Mesalazine Nanosuspension Using Chitosan Polyelectrolyte Complexes with Alginate and Alginate/Hydroxypropyl-Methylcellulose Phthalate. Pharmaceutics 2024; 16:1489. [PMID: 39771469 PMCID: PMC11676705 DOI: 10.3390/pharmaceutics16121489] [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: 10/18/2024] [Revised: 11/16/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025] Open
Abstract
Background/Objectives: This study evaluated how the relative proportion of chitosan (CS) to the polyanions alginate (ALG) and hydroxypropyl-methylcellulose phthalate (HP) affects the colloidal properties of mesalazine (MSZ) nanosuspensions as a strategy to produce particles with specific characteristics. Methods: Nanosuspensions were prepared using a bottom-up approach based on acid-base reactions and were modified with CS in a binary mixture with ALG or a ternary mixture with ALG and HP. The particle size, polydispersity index (PDI), zeta potential, morphology, and drug association efficiency were analyzed. Results: Higher proportions of CS relative to the polyanions resulted in smaller, less polydisperse particles. The zeta potential inversion was influenced by the relative proportion of CS in the system. These results were consistent over 30 days and pH exerted an influence on the magnitude of the observed effect. The optimized NS modified with binary CS/ALG blends had the following properties at pH 6.0: an average particle size of 324.9 nm, PDI of 0.5, and zeta potential of +40.8 mV; at pH 4.0, it had an average particle size of 310.4 nm, PDI of 0.4, and zeta potential of +43.6 mV. The optimized NS modified with ternary CS/ALG/HP had the following properties at pH 6.0: an average particle size of 316.7 nm, PDI of 0.5, and zeta potential of +33.9 mV; at pH 4.0, it had an average particle size of 363.5 nm, PDI of 0.6, and zeta potential of +33.9 mV. Conclusions: CS-based polyelectrolyte complexes with ALG and ALG/HP offer an approach to modulating the properties of MSZ nanosuspensions, enabling the production of particles with tailored characteristics.
Collapse
Affiliation(s)
| | | | | | - Leonardo Miziara Barboza Ferreira
- Faculty of Pharmaceutical Science, UNESP—São Paulo State University, Rodovia Araraquara-Jaú, Km 01, Araraquara 14801-902, Brazil; (A.A.R.P.); (J.V.M.A.)
| | - Maria Palmira Daflon Gremião
- Faculty of Pharmaceutical Science, UNESP—São Paulo State University, Rodovia Araraquara-Jaú, Km 01, Araraquara 14801-902, Brazil; (A.A.R.P.); (J.V.M.A.)
| |
Collapse
|
20
|
Yao H, Ma S, Huang J, Si X, Yang M, Song W, Lv G, Wang G. Trojan-Horse Strategy Targeting the Gut-Liver Axis Modulates Gut Microbiome and Reshapes Microenvironment for Orthotopic Hepatocellular Carcinoma Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2310002. [PMID: 39373804 PMCID: PMC11600211 DOI: 10.1002/advs.202310002] [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: 12/19/2023] [Revised: 08/06/2024] [Indexed: 10/08/2024]
Abstract
Reversing the hepatic inflammatory and immunosuppressive microenvironment caused by gut microbiota-derived lipopolysaccharides (LPS), accumulating to the liver through the gut-liver axis, is crucial for suppressing hepatocellular carcinoma (HCC) and metastasis. However, synergistically manipulating LPS-induced inflammation and gut microbiota remains a daunting task. Herein, a Trojan-horse strategy is proposed using an oral dextran-carbenoxolone (DEX-CBX) conjugate, which combines prebiotic and glycyrrhetinic acid (GA) homologs, to targeted delivery GA to HCC through the gut-liver axis for simultaneous modulation of hepatic inflammation and gut microbiota. In the orthotopic HCC model, a 95-45% reduction in the relative abundances of LPS-associated microbiota is observed, especially Helicobacter, caused by DEX-CBX treatment over phosphate-buffered saline (PBS) treatment. Notably, a dramatic increase (37-fold over PBS) in the abundance of Akkermansia, which is known to strengthen systemic immune response, is detected. Furthermore, DEX-CBX significantly increased natural killer T cells (5.7-fold) and CD8+ T cells (3.9-fold) as well as decreased M2 macrophages (59% reduction) over PBS treatment, resulting in a tumor suppression rate of 85.4%. DEX-CBX is anticipated to offer a novel strategy to precisely modulate hepatic inflammation and the gut microbiota to address both the symptoms and root causes of LPS-induced immunosuppression in HCC.
Collapse
Affiliation(s)
- Haochen Yao
- Hepatobiliary and Pancreatic Surgery DepartmentGeneral Surgery CenterFirst Hospital of Jilin UniversityNo.1 Xinmin StreetChangchunJilin130021China
- Key Laboratory of ZoonosisChinese Ministry of EducationCollege of Basic Medical SciencesJilin UniversityChangchunJilin130021China
| | - Sheng Ma
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin RoadChangchun130022China
- Jilin Biomedical Polymers Engineering LaboratoryChangchun Institute of Applied Chemistry5625 Renmin RoadChangchun130022China
| | - Juanjuan Huang
- Key Laboratory of ZoonosisChinese Ministry of EducationCollege of Basic Medical SciencesJilin UniversityChangchunJilin130021China
- Department of Computational MathematicsSchool of MathematicsJilin UniversityChangchun130012China
| | - Xinghui Si
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin RoadChangchun130022China
- Jilin Biomedical Polymers Engineering LaboratoryChangchun Institute of Applied Chemistry5625 Renmin RoadChangchun130022China
| | - Ming Yang
- Department of Molecular BiologyCollege of Basic Medical SciencesJilin UniversityChangchun130021China
| | - Wantong Song
- Key Laboratory of Polymer EcomaterialsChangchun Institute of Applied ChemistryChinese Academy of Sciences5625 Renmin RoadChangchun130022China
- Jilin Biomedical Polymers Engineering LaboratoryChangchun Institute of Applied Chemistry5625 Renmin RoadChangchun130022China
| | - Guoyue Lv
- Hepatobiliary and Pancreatic Surgery DepartmentGeneral Surgery CenterFirst Hospital of Jilin UniversityNo.1 Xinmin StreetChangchunJilin130021China
| | - Guoqing Wang
- Key Laboratory of ZoonosisChinese Ministry of EducationCollege of Basic Medical SciencesJilin UniversityChangchunJilin130021China
| |
Collapse
|
21
|
Long J, Liang X, Ao Z, Tang X, Li C, Yan K, Yu X, Wan Y, Li Y, Li C, Zhou M. Stimulus-responsive drug delivery nanoplatforms for inflammatory bowel disease therapy. Acta Biomater 2024; 188:27-47. [PMID: 39265673 DOI: 10.1016/j.actbio.2024.09.007] [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: 05/22/2024] [Revised: 08/26/2024] [Accepted: 09/06/2024] [Indexed: 09/14/2024]
Abstract
Inflammatory bowel disease (IBD) manifests as inflammation in the colon, rectum, and ileum, presenting a global health concern with increasing prevalence. Therefore, effective anti-inflammatory therapy stands as a promising strategy for the prevention and management of IBD. However, conventional nano drug delivery systems (NDDSs) for IBD face many challenges in targeting the intestine, such as physiological and pathological barriers, genetic variants, disease severity, and nutritional status, which often result in nonspecific tissue distribution and uncontrolled drug release. To address these limitations, stimulus-responsive NDDSs have received considerable attention in recent years due to their advantages in providing controlled release and enhanced targeting. This review provides an overview of the pathophysiological mechanisms underlying IBD and summarizes recent advancements in microenvironmental stimulus-responsive nanocarriers for IBD therapy. These carriers utilize physicochemical stimuli such as pH, reactive oxygen species, enzymes, and redox substances to deliver drugs for IBD treatment. Additionally, pivotal challenges in the future development and clinical translation of stimulus-responsive NDDSs are emphasized. By offering insights into the development and optimization of stimulus-responsive drug delivery nanoplatforms, this review aims to facilitate their application in treating IBD. STATEMENT OF SIGNIFICANCE: This review highlights recent advancements in stimulus-responsive nano drug delivery systems (NDDSs) for the treatment of inflammatory bowel disease (IBD). These innovative nanoplatforms respond to specific environmental triggers, such as pH reactive oxygen species, enzymes, and redox substances, to release drugs directly at the inflammation site. By summarizing the latest research, our work underscores the potential of these technologies to improve drug targeting and efficacy, offering new directions for IBD therapy. This review is significant as it provides a comprehensive overview for researchers and clinicians, facilitating the development of more effective treatments for IBD and other chronic inflammatory diseases.
Collapse
Affiliation(s)
- Jiang Long
- Department of Cardiology, Xuyong County People's Hospital, Luzhou, Sichuan 646000, China
| | - Xiaoya Liang
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zuojin Ao
- Analysis and Testing Center, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xiao Tang
- College of Integrated Chinese and Western Medicine, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chuang Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Kexin Yan
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xin Yu
- Chinese Pharmacy Laboratory, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Ying Wan
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yao Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Science and Technology Department, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Chunhong Li
- Department of Pharmaceutical Sciences, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Meiling Zhou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| |
Collapse
|
22
|
Liu X, Dong Y, Wang C, Guo Z. Application of chitosan as nano carrier in the treatment of inflammatory bowel disease. Int J Biol Macromol 2024; 278:134899. [PMID: 39187100 DOI: 10.1016/j.ijbiomac.2024.134899] [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: 12/11/2023] [Revised: 08/13/2024] [Accepted: 08/18/2024] [Indexed: 08/28/2024]
Abstract
Inflammatory bowel disease (IBD), encompassing ulcerative colitis (UC) and Crohn's disease (CD), is characterized by persistent and recurrent gastrointestinal inflammation. Conventional IBD therapies often involve the use of antibiotics, NSAIDs, biological agents, and immunomodulators. While these medications can mitigate acute inflammatory symptoms, their long-term efficacy is frequently compromised due to cumulative toxic effects. In recent years, significant attention has shifted toward nanoparticle (NP)-based therapies as potential alternatives for IBD management. Various drug delivery strategies, including those targeting microbiota interactions, ligand-receptor binding, pH sensitivity, biodegradability, pressure response, and specific charge and size parameters, have been explored and optimized in animal studies. This review provides a comprehensive overview of the current landscape of chitosan NP-mediated drug delivery systems for IBD treatment. Additionally, it will discuss the prevailing challenges and propose future research directions to advance chitosan NP-based therapeutic strategies for IBD.
Collapse
Affiliation(s)
- Xiaoming Liu
- Department of Gastroenterology, Huaihe Hospital of Henan University, 115 Ximen Street, Kaifeng 475000, Henan, China
| | - Yunrui Dong
- Hubei University of Science and Technology, 88 Xianning Road, Xianning 437100, Hubei, China
| | - Chenyu Wang
- Department of General Surgery, Huaihe Hospital of Henan University, 115 Ximen Street, Kaifeng 475000, Henan, China
| | - Zhiguo Guo
- Department of Gastroenterology, Suzhou Hospital of Anhui Medical University (Suzhou Municipal Hospital of Anhui Province), No.616 Bianyangsan Road, Suzhou 234000, Anhui, China.
| |
Collapse
|
23
|
González-Fuentes J, Plaza-Oliver M, Santander-Ortega MJ, Lozano MV. Understanding the role of the structure of single-stimuli hybrid systems on their behaviour as platforms for colonic delivery. Drug Deliv Transl Res 2024; 14:2598-2614. [PMID: 38856952 DOI: 10.1007/s13346-024-01641-7] [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] [Accepted: 05/20/2024] [Indexed: 06/11/2024]
Abstract
The success of colon-targeted oral hybrid systems relies in the proper control over the release of the entrapped nanostructures at the colon. This work describes the design of hybrid systems for their colonic enzyme-triggered release. The hybrid systems were constituted by nanoemulsions, with adequate characteristics for the treatment of ulcerative colitis, included in a pectin hydrogel-like matrix. For that purpose, pectins with similar degrees of methylation (< 50%) and increasing degree of amidation, i.e. 0, 13 and 20%, were selected. Hybrid systems were formulated by a novel aggregation induced gelation method, using Ca2+, Ba2+ or Zn2+ as aggregating agents, as well as by a polyelectrolyte condensation approach, obtaining structures in the micrometric range (< 10 μm). Despite the resistance of pectins to the upper gastrointestinal tract stimuli, the analysis of the behaviour of the different prototypes showed that the non-covalent crosslinks that allow the formation of the hybrid structure may play a relevant role on the performance of the formulation.Our results indicated that the partial disassembling of the hybrid system's microstructure due to the intestinal conditions may facilitate the stimuli-triggered release of the nanoemulsions at the colon. More interestingly, the particle tracking experiments showed that the condensation process that occurs during the formation of the system may affect to the enzymatic degradation of pectin. In this sense, the effect of the high degree of amidation of pectin may be more prevalent as structural feature rather than as a promoter of the enzyme-triggered release.
Collapse
Affiliation(s)
- Joaquín González-Fuentes
- Development and Evaluation of Nanomedicines (DEVANA), Faculty of Pharmacy, Albacete, Spain
- Instituto de Biomedicina (IB), Universidad de Castilla-La Mancha (UCLM), Albacete, 02008, Spain
| | - María Plaza-Oliver
- Development and Evaluation of Nanomedicines (DEVANA), Faculty of Pharmacy, Albacete, Spain
- Instituto de Biomedicina (IB), Universidad de Castilla-La Mancha (UCLM), Albacete, 02008, Spain
| | - Manuel Jesús Santander-Ortega
- Development and Evaluation of Nanomedicines (DEVANA), Faculty of Pharmacy, Albacete, Spain.
- Instituto de Biomedicina (IB), Universidad de Castilla-La Mancha (UCLM), Albacete, 02008, Spain.
| | - María Victoria Lozano
- Development and Evaluation of Nanomedicines (DEVANA), Faculty of Pharmacy, Albacete, Spain.
- Instituto de Biomedicina (IB), Universidad de Castilla-La Mancha (UCLM), Albacete, 02008, Spain.
| |
Collapse
|
24
|
Maurya R, Vikal A, Patel P, Narang RK, Kurmi BD. "Enhancing Oral Drug Absorption: Overcoming Physiological and Pharmaceutical Barriers for Improved Bioavailability". AAPS PharmSciTech 2024; 25:228. [PMID: 39354282 DOI: 10.1208/s12249-024-02940-5] [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: 05/18/2024] [Accepted: 09/11/2024] [Indexed: 10/03/2024] Open
Abstract
The oral route stands out as the most commonly used method for drug administration, prized for its non-invasive nature, patient compliance, and easy administration. Several elements influence the absorption of oral medications, including their solubility, permeability across mucosal membranes, and stability within the gastrointestinal (GI) environment. Research has delved into comprehending physicochemical, biochemical, metabolic, and biological obstacles that impact the bioavailability of a drug. To improve oral drug absorption, several pharmaceutical technologies and delivery methods have been studied, including cyclodextrins, micelles, nanocarriers, and lipid-based carriers. This review examines both traditional and innovative drug delivery methods, as well as the physiological and pharmacological barriers influencing medication bioavailability when taken orally. Additionally, it describes the challenges and advancements in developing formulations suitable for oral use.
Collapse
Affiliation(s)
- Rashmi Maurya
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Akash Vikal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Raj Kumar Narang
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
- ISF College of Pharmacy and Research, Rattian Road, Moga, 142048, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
25
|
Di Rienzo A, Marinelli L, Dimmito MP, Toto EC, Di Stefano A, Cacciatore I. Advancements in Inflammatory Bowel Disease Management: From Traditional Treatments to Monoclonal Antibodies and Future Drug Delivery Systems. Pharmaceutics 2024; 16:1185. [PMID: 39339221 PMCID: PMC11435298 DOI: 10.3390/pharmaceutics16091185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/29/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal inflammatory disorder with two main subtypes: ulcerative colitis (UC) and Crohn's disease (CD). The pathogenesis involves genetic predisposition, dysbiosis, and immune dysregulation. Complications include perianal lesions, strictures, fistulas, perforations, and an increased risk of colon cancer. Clinical classification ranges from mild to fulminant and recurrent disease, with common symptoms such as abdominal discomfort, rectal bleeding, diarrhea, and weight loss. Extraintestinal manifestations include arthritis, erythema nodosum, pyoderma gangrenosum, and uveitis. Conventional treatments using aminosalicylates, corticosteroids, and immunomodulators have limitations. Biologics, introduced in the 1990s, offer improved efficacy and specificity, targeting factors like TNF-α, integrins, and cytokines. Monoclonal antibodies play a crucial role in IBD management, aiming to reduce relapses, hospitalizations, and surgeries. In conclusion, this review is aimed at summarizing the latest knowledge, advantages, and drawbacks of IBD therapies, such as small molecules, biologics, and monoclonal antibodies, to provide a basis for further research in the IBD field.
Collapse
Affiliation(s)
| | - Lisa Marinelli
- Department of Pharmacy, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy; (A.D.R.); (M.P.D.); (E.C.T.); (A.D.S.); (I.C.)
| | | | | | | | | |
Collapse
|
26
|
Wu T, Bai X, Zhang Y, Dai E, Ma J, Yu C, He C, Li Q, Yang Y, Kong H, Qu H, Zhao Y. Natural medicines-derived carbon dots as novel oral antioxidant administration strategy for ulcerative colitis therapy. J Nanobiotechnology 2024; 22:511. [PMID: 39187876 PMCID: PMC11348712 DOI: 10.1186/s12951-024-02702-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/05/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic intestinal inflammation, resulting in a global healthcare challenge with no real specific medicine. Natural medicines are recognized as a potential clinical alternative therapy, but their applications are limited by poor solubility and low bioavailability. RESULTS In this work, inspired by the natural medicines of ancient China, novel functional carbon dots derived from Magnetite and Medicated Leaven (MML-CDs) were synthesized by hydrothermal method, and confirmed their ultrasmall nano-size (3.2 ± 0.6 nm) and Fe doped surface structure, thereby with excellent gastrointestinal stability, remarkable capabilities in eliminating ROS, and highly biocompatibility. With no external stimuli, the oral administration of MML-CDs demonstrated obvious alleviation to UC. Further experiments pointed that MML-CDs could improve hemostasis capability, suppress inflammation reactions and oxidative stress, and up-regulate the expression of tight junction proteins. Furthermore, MML-CDs also showed well regulation in the dysbiosis of intestinal flora. CONCLUSION Overall, above evidence reveals that green-synthesized MML-CDs can significantly alleviate intestinal bleeding, inhibit colon inflammation, and repair colonic barrier damage, further regulating intestinal flora and intestinal inflammation microenvironment. Our findings provide an efficient oral administration of MML-CDs as a novel therapy strategy for ulcerative colitis.
Collapse
Affiliation(s)
- Tong Wu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xue Bai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yue Zhang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ertong Dai
- Qingdao Eighth People's Hospital, Qingdao, 266100, China
| | - Jinyu Ma
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cai Yu
- Department of Endocrine, Beijing Daxing District Hospital of Integrated Chinese and Western Medicine, Beijing, 100163, China
| | - Chenxin He
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qiannan Li
- Department of Traditional Chinese Medicine, Beijing Daxing District Hospital of Integrated Chinese and Western Medicine, Beijing, 100163, China
| | - Yingxin Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hui Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Huihua Qu
- Centre of Scientific Experiment, Beijing University of Chinese Medicine, Beijing, 100029, China.
| | - Yan Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
27
|
Vaezi Z, Baradaran Ghavami S, Farmani M, Mahdavian R, Asadzadeh Aghdaei H, Naderi-Manesh H. Oral Formulation of 5-Aminosalicylic Acid-Hemoglobin Bio-Adhesive Nanoparticles Enhance Therapeutic Efficiency in Ulcerative Colitis Mice: A Preclinical Evaluation. J Pharm Sci 2024; 113:2331-2341. [PMID: 38582281 DOI: 10.1016/j.xphs.2024.03.028] [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: 01/05/2024] [Revised: 03/31/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
The oral formulation design for colon-specific drug delivery brings some therapeutic benefits in the ulcerative colitis treatment. We recently reported the specific delivery of hemoglobin nanoparticles-conjugating 5-aminosalicylic acid (5-ASA-HbNPs) to the inflamed site. In the current study, the therapeutic effect of the 5-ASA-HbNPs formulation was confirmed in vivo. This evaluation of 5-ASA-HbNPs not only shows longer colonic retention time due to adhesive properties, also provides full support for it as compared with free 5-ASA. It was considered as a suitable bio-adhesive nanoparticle with mucoadhesive property to pass through the mucus layer and accumulate into the mucosa. In UC model mice, a two-fold decrease in the disease activity indexes and colon weight/length ratios was significantly observed in the group treated with 5-ASA-HbNPs. This group received one percent of the standard dosage of 5-ASA (50 μg/kg), while, a similar result was observed for a significant amount of free 5-ASA (5 mg/kg). Furthermore, microscopic images of histological sections of the extracted colons demonstrated that the 5-ASA-HbNPs and 5-ASA groups displayed instances of inflammatory damage within the colon. However, in comparison to the colitis group, the extent of this damage was relatively moderate, suggesting 5-ASA-HbNPs improved therapeutic efficacy with the lower dosage form.
Collapse
Affiliation(s)
- Zahra Vaezi
- Department of Bioactive compounds, Faculty of Interdisciplinary Sciences and Technologies, Tarbiat Modares University, Postal codes: 14115-154, Tehran, Iran.
| | - Shaghayegh Baradaran Ghavami
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, P.O.BOX: 1985717411, Tehran, Iran
| | - Maryam Farmani
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, P.O.BOX: 1985717411, Tehran, Iran
| | - Reza Mahdavian
- Department of Biophysics, Faculty of Biological Science, Tarbiat Modares University, Postal codes: 14115-154, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, P.O.BOX: 1985717411, Tehran, Iran.
| | - Hossein Naderi-Manesh
- Department of Bioactive compounds, Faculty of Interdisciplinary Sciences and Technologies, Tarbiat Modares University, Postal codes: 14115-154, Tehran, Iran; Department of Biophysics, Faculty of Biological Science, Tarbiat Modares University, Postal codes: 14115-154, Tehran, Iran.
| |
Collapse
|
28
|
Li Y, Wang Y, Li Y, Yan S, Gao X, Li P, Zheng X, Gu Q. Dress me an outfit: advanced probiotics hybrid systems for intelligent IBD therapy. Crit Rev Food Sci Nutr 2024:1-24. [PMID: 39007752 DOI: 10.1080/10408398.2024.2359135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Inflammation bowel disease (IBD) has emerged as a public health challenge worldwide; with high incidence and rapid prevalence, it has troubled billions of people and further induced multitudinous systemic complications. Recent decade has witnessed the vigorous application of food-borne probiotics for IBD therapy; however, the complicated and changeable environments of digestive tract have forced probiotics to face multiple in vivo pressures, consequently causing unsatisfied prophylactic or therapeutic efficacy attributed to off-targeted arrival, damaged viability, insufficient colonization efficiency, etc. Fortunately, arisen hybrid technology has provided versatile breakthroughs for the targeted transplantation of probiotics. By ingeniously modifying probiotics to form probiotics hybrid systems (PHS), the biological behaviors of probiotics in vivo could be mediated, the interactions between probiotics with intestinal components can be facilitated, and diverse advanced probiotic-based therapies for IBD challenge can be developed, which attribute to the intelligent response to microenvironment of PHS, and intelligent design of PHS for multiple functions combination. In this review, various PHS were categorized and their intestinal behaviors were elucidated systematically, their therapeutic effects and intrinsic mechanism were further analyzed. Besides, shortages of present PHS and the corresponding solutions have been discussed, based on which the future perspectives of this field have also been proposed. The undeniable fact is that PHS show an incomparable future to bring the next generation of advanced food science.
Collapse
Affiliation(s)
- Yonglu Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Yadi Wang
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Yapeng Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Shihai Yan
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Xin Gao
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Ping Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| | - Xiaodong Zheng
- Department of Food Science and Nutrition; Zhejiang Key Laboratory for Agro-food Processing; Fuli Institute of Food Science; National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou, People's Republic of China
| | - Qing Gu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, People's Republic of China
- Key Laboratory for Food Microbial Technology of Zhejiang Province, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
29
|
Zhang D, Wan H, Zhao R, Zhang Y, Chen H. Eudragit S100 coated iron oxide-chitosan nanocomposites for colon targeting of 5-aminosalicylic acid ameliorate ulcerative colitis by improving intestinal barrier function and inhibiting NLRP3 inflammasome. Int Immunopharmacol 2024; 139:112661. [PMID: 39008936 DOI: 10.1016/j.intimp.2024.112661] [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: 04/15/2024] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
The therapeutic effect of 5-amino salicylic acid (5-ASA), a first-line therapeutic agent for the treatment of ulcerative colitis (UC), is limited by the modest bioavailability afforded by its oral administration. In this study, a 5-ASA oral delivery system was developed using Eudragit S100-coated iron oxide-chitosan nanocomposites (ES-IOCS/5-ASA) to address this issue. According to drug release studies in vitro, ES-IOCS/5-ASA only released a small amount of drug in simulated gastric fluid with a pH of 1.2. However, in a medium with a pH of 7.5, a relatively rapid and complete release was noted. 5-ASA-loaded iron oxide-chitosan nanocomposites (IOCS/5-ASA) could be effectively taken up by NCM460 cells and performed better anti-inflammatory effects than free 5-ASA. At the same time, IOCS/5-ASA improved barrier damage in DSS-induced NCM460 cells. In vivo models of dextran sulphate sodium (DSS)-induced colitis were used to assess the therapeutic efficacy of oral administration of ES-IOCS/5-ASA. ES-IOCS/5-ASA significantly relieved DSS-induced colitis and enhanced the integrity of the intestinal epithelial barrier. ES-IOCS/5-ASA also reduced the expression of NLRP3, ASC and IL-1β. Additionally, iron oxide nanoparticles used as nanozymes could alleviate inflammation. In summary, this study indicates that ES-IOCS/5-ASA exert anti-inflammatory effects on DSS-induced colitis by improving intestinal barrier function and inhibiting NLRP3 inflammasome expression, presenting a viable therapeutic choice for the treatment of UC.
Collapse
Affiliation(s)
- Dandan Zhang
- Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Hao Wan
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, Jiangsu, People's Republic of China
| | - Ran Zhao
- Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Yu Zhang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, Jiangsu, People's Republic of China.
| | - Hong Chen
- Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China.
| |
Collapse
|
30
|
Ma S, Yao H, Si X, Huang Z, Wang R, Wan R, Tang Z, Wang G, Song W. Orally available dextran-aspirin nanomedicine modulates gut inflammation and microbiota homeostasis for primary colorectal cancer therapy. J Control Release 2024; 370:528-542. [PMID: 38705520 DOI: 10.1016/j.jconrel.2024.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/23/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Reversing the aggravated immunosuppression hence overgrowth of colorectal cancer (CRC) caused by the gut inflammation and microbiota dysbiosis is pivotal for effective CRC therapy and metastasis inhibition. However, the low delivery efficiency and severe dose-limiting off-target toxicities caused by unsatisfied drug delivery systems remain the major obstacles in precisely modulating gut inflammation and microbiota in CRC therapy. Herein, a multifunctional oral dextran-aspirin nanomedicine (P3C-Asp) was utilized for oral treatment of primary CRC, as it could release salicylic acid (SA) while scavenging reactive oxygen species (ROS) and held great potential in modulating gut microbiota with prebiotic (dextran). Oral P3C-Asp retained in CRC tissues for over 12 h and significantly increased SA accumulation in CRC tissues over free aspirin (10.8-fold at 24 h). The enhanced SA accumulation and ROS scavenging of P3C-Asp cooperatively induced more potent inflammation relief over free aspirin, characterized as lower level of cyclooxygenase-2 and immunosuppressive cytokines. Remarkably, P3C-Asp promoted the microbiota homeostasis and notably increased the relative abundance of strengthening systemic anti-cancer immune response associated microbiota, especially lactobacillus and Akkermansia to 6.66- and 103- fold over the control group. Additionally, a demonstrable reduction in pathogens associated microbiota (among 96% to 79%) including Bacteroides could be detected. In line with our findings, inflammation relief along with enhanced abundance of lactobacillus was positively correlated with CRC inhibition. In primary CRC model, P3C-Asp achieved 2.1-fold tumor suppression rate over free aspirin, with an overall tumor suppression rate of 85%. Moreover, P3C-Asp cooperated with αPD-L1 further reduced the tumor weight of each mouse and extended the median survival of mice by 29 days over αPD-L1 alone. This study unravels the synergistic effect of gut inflammation and microbiota modulation in primary CRC treatment, and unlocks an unconventional route for immune regulation in TME with oral nanomedicine.
Collapse
Affiliation(s)
- Sheng Ma
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Haochen Yao
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Xinghui Si
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China
| | - Zichao Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Ruoyi Wang
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun 130041, China
| | - Renming Wan
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Zhaohui Tang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China
| | - Guoqing Wang
- Department of Pathogenobiology, The Key Laboratory of Zoonosis, Chinese Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
31
|
Sahle Z, Engidaye G, Shenkute Gebreyes D, Adenew B, Abebe TA. Fecal microbiota transplantation and next-generation therapies: A review on targeting dysbiosis in metabolic disorders and beyond. SAGE Open Med 2024; 12:20503121241257486. [PMID: 38826830 PMCID: PMC11143861 DOI: 10.1177/20503121241257486] [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: 09/15/2023] [Accepted: 05/09/2024] [Indexed: 06/04/2024] Open
Abstract
The human microbiome, particularly the gut microbiome, has emerged as a central determinant of health and disease. Dysbiosis, an imbalance in the microbial composition of the gut, is associated with a variety of metabolic and other diseases, highlighting the potential for microbiota-targeted treatments. Fecal microbiota transplantation has received considerable attention as a promising therapy to modulate the gut microbiome and restore microbial homeostasis. However, challenges remain, including standardization, safety, and long-term efficacy. This review summarizes current knowledge on fecal microbiota transplantation and describes the next generation therapies targeting microbiome. This review looked at the mechanistic understanding of fecal microbiota transplantation and alternative strategies, elucidating their potential role in improving dysbiosis-associated metabolic disorders, such as obesity, and type 2 diabetes and others. Additionally, this review discussed the growing application of therapies targeting the gut microbiome. Insights from clinical trials, preclinical studies, and emerging technologies provide a comprehensive overview of the evolving landscape of microbiome-based interventions. Through a critical assessment of current advances and prospects, this review aims to highlight the therapeutic potential of targeting gut microbiome and pave the way for innovative approaches in precision medicine and personalized treatments.
Collapse
Affiliation(s)
- Zenawork Sahle
- Department of Medical Laboratory Science, Asrat Weldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Getabalew Engidaye
- Department of Medical Laboratory Science, Asrat Weldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Demissew Shenkute Gebreyes
- Department of Medical Laboratory Science, Asrat Weldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Ethiopia
| | - Behailu Adenew
- Department of Medical Laboratory Science, Debre Berhan Compressive Specialized Hospital, Debre Berhan, Ethiopia
| | - Tsegahun Asfaw Abebe
- Department of Medical Laboratory Science, Asrat Weldeyes Health Science Campus, Debre Berhan University, Debre Berhan, Ethiopia
| |
Collapse
|
32
|
Li N, Niu L, Liu Y, Wang Y, Su X, Xu C, Sun Z, Guo H, Gong J, Shen S. Taking SCFAs produced by Lactobacillus reuteri orally reshapes gut microbiota and elicits antitumor responses. J Nanobiotechnology 2024; 22:241. [PMID: 38735933 PMCID: PMC11089779 DOI: 10.1186/s12951-024-02506-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 04/29/2024] [Indexed: 05/14/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) incidence is increasing in recent years due to intestinal flora imbalance, making oral probiotics a hotspot for research. However, numerous studies related to intestinal flora regulation ignore its internal mechanisms without in-depth research. RESULTS Here, we developed a probiotic microgel delivery system (L.r@(SA-CS)2) through the layer-by-layer encapsulation technology of alginate (SA) and chitosan (CS) to improve gut microbiota dysbiosis and enhance anti-tumor therapeutic effect. Short chain fatty acids (SCFAs) produced by L.r have direct anti-tumor effects. Additionally, it reduces harmful bacteria such as Proteobacteria and Fusobacteriota, and through bacteria mutualophy increases beneficial bacteria such as Bacteroidota and Firmicutes which produce butyric acid. By binding to the G protein-coupled receptor 109A (GPR109A) on the surface of colonic epithelial cells, butyric acid can induce apoptosis in abnormal cells. Due to the low expression of GPR109A in colon cancer cells, MK-6892 (MK) can be used to stimulate GPR109A. With increased production of butyrate, activated GPR109A is able to bind more butyrate, which further promotes apoptosis of cancer cells and triggers an antitumor response. CONCLUSION It appears that the oral administration of L.r@(SA-CS)2 microgels may provide a treatment option for CRC by modifying the gut microbiota.
Collapse
Affiliation(s)
- Nannan Li
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116021, China
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Lili Niu
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116021, China
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Yao Liu
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
- Clinical Oncology Center, Shanghai Municipal Hospital of TCM, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Yang Wang
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116021, China
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Xiaomin Su
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116021, China
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Ce Xu
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116021, China
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Zanya Sun
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116021, China
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Huishu Guo
- Central Laboratory, First Affiliated Hospital, Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, 116021, China.
| | - Jingru Gong
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| | - Shun Shen
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China.
| |
Collapse
|
33
|
Gómez-Labrador C, Ricart E, Iborra M, Iglesias E, Martín-Arranz MD, de Castro L, De Francisco R, García-Alonso FJ, Sanahuja A, Gargallo-Puyuelo CJ, Mesonero F, Casanova MJ, Mañosa M, Rivero M, Calvo M, Sierra-Ausin M, González-Muñoza C, Calvet X, García-López S, Guardiola J, Arias García L, Márquez-Mosquera L, Gutiérrez A, Zabana Y, Navarro-Llavat M, Lorente Poyatos R, Piqueras M, Torrealba L, Bermejo F, Ponferrada-Díaz Á, Pérez-Calle JL, Acosta MBD, Tejido C, Cabriada JL, Marín-Jiménez I, Roncero Ó, Ber Y, Fernández-Salazar L, Camps Aler B, Lucendo AJ, Llaó J, Bujanda L, Muñoz Villafranca C, Domènech E, Chaparro M, Gisbert JP. Trends in Targeted Therapy Usage in Inflammatory Bowel Disease: TRENDY Study of ENEIDA. Pharmaceutics 2024; 16:629. [PMID: 38794292 PMCID: PMC11124941 DOI: 10.3390/pharmaceutics16050629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/26/2024] Open
Abstract
Markers that allow for the selection of tailored treatments for individual patients with inflammatory bowel diseases (IBD) are yet to be identified. Our aim was to describe trends in real-life treatment usage. For this purpose, patients from the ENEIDA registry who received their first targeted IBD treatment (biologics or tofacitinib) between 2015 and 2021 were included. A subsequent analysis with Machine Learning models was performed. The study included 10,009 patients [71% with Crohn's disease (CD) and 29% with ulcerative colitis (UC)]. In CD, anti-TNF (predominantly adalimumab) were the main agents in the 1st line of treatment (LoT), although their use declined over time. In UC, anti-TNF (mainly infliximab) use was predominant in 1st LoT, remaining stable over time. Ustekinumab and vedolizumab were the most prescribed drugs in 2nd and 3rd LoT in CD and UC, respectively. Overall, the use of biosimilars increased over time. Machine Learning failed to identify a model capable of predicting treatment patterns. In conclusion, drug positioning is different in CD and UC. Anti-TNF were the most used drugs in IBD 1st LoT, being adalimumab predominant in CD and infliximab in UC. Ustekinumab and vedolizumab have gained importance in CD and UC, respectively. The approval of biosimilars had a significant impact on treatment.
Collapse
Affiliation(s)
- Celia Gómez-Labrador
- Gastroenterology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28006 Madrid, Spain; (M.J.C.); (M.C.); (J.P.G.)
| | - Elena Ricart
- Gastroenterology Unit, Hospital Clinic of Barcelona, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) and CIBEREHD, 08036 Barcelona, Spain
| | - Marisa Iborra
- Gastroenterology Unit, Hospital Universitario y Politécnico La Fe, 46026 Valencia, Spain;
| | - Eva Iglesias
- Gastroenterology Unit, Hospital Universitario Reina Sofía, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), 14004 Córdoba, Spain;
| | - María Dolores Martín-Arranz
- Gastroenterology Unit, Hospital Universitario La Paz, Universidad Autónoma de Madrid, Hospital La Paz Institute for Health Research, 28046 Madrid, Spain;
| | - Luisa de Castro
- Gastroenterology Unit, Hospital Álvaro Cunqueiro, Grupo de Investigación en Patología Digestiva, Instituto de Investigación Sanitaria Galicia Sur (IIS Galicia Sur) SERGAS, UVIGO, 36312 Vigo, Spain;
| | - Ruth De Francisco
- Gastroenterology Unit, Hospital Universitario Central de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain;
| | | | - Ana Sanahuja
- Gastroenterology Unit, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain;
| | - Carla J. Gargallo-Puyuelo
- Gastroenterology Unit, Hospital Clínico Universitario Lozano Blesa, IIS Aragón, 50009 Zaragoza, Spain;
| | - Francisco Mesonero
- Gastroenterology Unit, Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain;
| | - María José Casanova
- Gastroenterology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28006 Madrid, Spain; (M.J.C.); (M.C.); (J.P.G.)
| | - Míriam Mañosa
- Gastroenterology Unit, Hospital Universitari Germans Trias i Pujol, and CIBEREHD, 08916 Badalona, Spain; (M.M.); (E.D.)
| | - Montserrat Rivero
- Gastroenterology Unit, Hospital Universitario de Valdecilla and Instituto de Investigación Sanitaria Valdecilla (IDIVAL), 39008 Santander, Spain;
| | - Marta Calvo
- Gastroenterology Unit, Hospital Universitario Puerta de Hierro Majadahonda, 28222 Madrid, Spain;
| | - Mónica Sierra-Ausin
- Gastroenterology Unit, Complejo Asistencial Universitario de León, 24008 León, Spain;
| | | | - Xavier Calvet
- Gastroenterology Unit, Parc Taulí, Hospital Universitari, Institut d’Investigació i Innovació Parc Taulí, Departament de Medicina, Universitat Autònoma de Barcelona and CIBEREHD, 08208 Sabadell, Spain;
| | - Santiago García-López
- Gastroenterology Unit, Hospital Universitario Miguel Servet, e Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain;
| | - Jordi Guardiola
- Gastroenterology Unit, Hospital Universitario de Bellvitge, Instituto de Investigación Biomédica de Bellvitge (IDIBELL), Universitat de Barcelona, 08907 Barcelona, Spain;
| | - Lara Arias García
- Gastroenterology Unit, Hospital Universitario de Burgos, 09006 Burgos, Spain;
| | - Lucía Márquez-Mosquera
- Gastroenterology Unit, Hospital del Mar, Barcelona and IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain;
| | - Ana Gutiérrez
- Gastroenterology Unit, Hospital General Universitario de Alicante, CIBEREHD, Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain;
| | - Yamile Zabana
- Gastroenterology Unit, Hospital Universitario Mútua Terrassa, and CIBEREHD, 08221 Terrassa, Spain;
| | - Mercè Navarro-Llavat
- Gastroenterology Unit, Complex Hospitalari Universitari Moisès Broggi, 08970 Barcelona, Spain;
| | - Rufo Lorente Poyatos
- Gastroenterology Unit, Hospital General Universitario de Ciudad Real, 13005 Ciudad Real, Spain;
| | - Marta Piqueras
- Gastroenterology Unit, Consorci Sanitari Terrassa, 08227 Terrassa, Spain;
| | - Leyanira Torrealba
- Gastroenterology Unit, Hospital Universitario Dr. Josep Trueta, 17007 Girona, Spain;
| | - Fernando Bermejo
- Gastroenterology Unit, Hospital Universitario de Fuenlabrada, 28942 Madrid, Spain;
| | | | - José L. Pérez-Calle
- Gastroenterology Unit, Hospital Universitario Fundación Alcorcón, 28922 Madrid, Spain;
| | - Manuel Barreiro-de Acosta
- Gastroenterology Unit, Hospital Clínico Universitario de Santiago de Compostela, 15706 Santiago de Compostela, Spain;
| | - Coral Tejido
- Gastroenterology Unit, Complexo Hospitalario Universitario de Ourense, 32005 Ourense, Spain;
| | - José Luis Cabriada
- Gastroenterology Unit, Hospital de Galdakao-Usansolo, 48960 Galdakao, Spain;
| | - Ignacio Marín-Jiménez
- Gastroenterology Unit, Hospital Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense de Madrid, 28007 Madrid, Spain;
| | - Óscar Roncero
- Gastroenterology Unit, Complejo Hospitalario la Mancha Centro, 13600 Alcázar de San Juan, Spain;
| | - Yolanda Ber
- Gastroenterology Unit, Hospital Universitario San Jorge, 22004 Huesca, Spain;
| | - Luis Fernández-Salazar
- Gastroenterology Unit, Hospital Clínico Universitario de Valladolid (SACYL), Universidad de Valladolid, 47003 Valladolid, Spain;
| | - Blau Camps Aler
- Gastroenterology Unit, Hospital General de Granollers, 08402 Granollers, Spain;
| | - Alfredo J. Lucendo
- Gastroenterology Unit, Hospital General de Tomelloso, Instituto de Investigación Sanitaria de Castilla-La Mancha (IDISCAM), and CIBEREHD, 13700 Tomelloso, Spain;
| | - Jordina Llaó
- Gastroenterology Unit, Althaia Xarxa Assistencial Universitaria de Manresa, 08243 Barcelona, Spain;
| | - Luis Bujanda
- Gastroenterology Unit, Hospital Universitario Donostia, Instituto Biodonostia, CIBEREHD and Universidad del País Vasco (UPV/EHU), 20014 San Sebastián, Spain;
| | | | - Eugeni Domènech
- Gastroenterology Unit, Hospital Universitari Germans Trias i Pujol, and CIBEREHD, 08916 Badalona, Spain; (M.M.); (E.D.)
- Universitat Autònoma de Barcelona, 08193 Barcelona, Spain
| | - María Chaparro
- Gastroenterology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28006 Madrid, Spain; (M.J.C.); (M.C.); (J.P.G.)
| | - Javier P. Gisbert
- Gastroenterology Unit, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), 28006 Madrid, Spain; (M.J.C.); (M.C.); (J.P.G.)
| |
Collapse
|
34
|
Huang D, Wang Y, Xu C, Zou M, Ming Y, Luo F, Xu Z, Miao Y, Wang N, Lin Z, Weng Z. Colon-targeted hydroxyethyl starch-curcumin microspheres with high loading capacity ameliorate ulcerative colitis via alleviating oxidative stress, regulating inflammation, and modulating gut microbiota. Int J Biol Macromol 2024; 266:131107. [PMID: 38527677 DOI: 10.1016/j.ijbiomac.2024.131107] [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: 01/10/2024] [Revised: 03/08/2024] [Accepted: 03/21/2024] [Indexed: 03/27/2024]
Abstract
Curcumin (CUR) is a natural polyphenol that holds promise for treating ulcerative colitis (UC), yet oral administration of CUR exhibits limited bioavailability and existing formulations for oral delivery of CUR often suffer from unsatisfactory loading capacity. This study presents hydroxyethyl starch-curcumin microspheres (HC-MSs) with excellent CUR loading capacity (54.52 %), and the HC-MSs can further encapsulate anti-inflammatory drugs dexamethasone (DEX) to obtain a combination formulation (DHC-MSs) with high DEX loading capacity (19.91 %), for combination therapy of UC. The microspheres were successfully engineered, retaining the anti-oxidative and anti-inflammatory activities of parental CUR and demonstrating excellent biocompatibility and controlled release properties, notably triggered by α-amylase, facilitating targeted drug delivery to inflamed sites. In a mouse UC model induced by dextran sulfate sodium, the microspheres effectively accumulated in inflamed colons and both HC-MSs and DHC-MSs exhibited superior therapeutic efficacy in alleviating UC symptoms compared to free DEX. Moreover, mechanistic exploration uncovered the multifaceted therapeutic mechanisms of these formulations, encompassing anti-inflammatory actions, mitigation of spleen enlargement, and modulation of gut microbiota composition. These findings underscore the potential of HC-MSs and DHC-MSs as promising formulations for UC, with implications for advancing treatment modalities for various inflammatory bowel disorders.
Collapse
Affiliation(s)
- Da Huang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Yongming Wang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Chenlan Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Minglang Zou
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Yangcan Ming
- Department of Pediatrics, Wuhan NO.1 Hospital, Wuhan, Hubei 430022, China
| | - Fang Luo
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Zhenjin Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Ying Miao
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Na Wang
- Department of Pediatrics, Wuhan NO.1 Hospital, Wuhan, Hubei 430022, China.
| | - Zhenyu Lin
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Zuquan Weng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, Fujian 350108, China.
| |
Collapse
|
35
|
Vaseghi A, Parchin RA, Chamanie KR, Herb M, Maleki H, Sadeghizadeh M. Encapsulation of propolis extracted with methylal in the chitosan nanoparticles and its antibacterial and cell cytotoxicity studies. BMC Complement Med Ther 2024; 24:165. [PMID: 38641781 PMCID: PMC11027551 DOI: 10.1186/s12906-024-04472-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/11/2024] [Indexed: 04/21/2024] Open
Abstract
In this study we develop novel type of antibacterial chitosan-propolis NPs to improve theantimicrobial activity against various pathogens. To this aim, we primarily extracted propolis with methylal and ethanol as green solvents and its encapsulation with chitosan NPs. The developed propolis loaded chitosan NPs indicated antimicrobial and anti-biofilm properties against various gram positive and negative. FTIR revealed the successful encapsulation of the propolis extract with Ethanol (PE) and Methylal (PM) into the chitosan nano career matrix. HPLC and GC-MASS also confirmed the presence of flavonoids and phenols compounds of propolis extracted with both solvents. In addition, we confirmed the total phenolic and flavonoid compounds in propolis by calorimetric method of Folin-Ciocalteu and aluminum trichloride complex formation assays, respectively. PE-CH and PM-CH were optimized regarding physicochemical properties such as particle size, zeta potential, and poly dispersity index (PDI) index. DLS and SEM micrographs confirmed a spherical morphology in a range of 360-420 nm with Z potential values of 30-48 mV and PDI of 0.105-0.166 for PE-CH and PM-CH, respectively. The encapsulation efficiency was evaluated using colorimetric analysis, with median values ranging from 90 to 92%. The MIC values within the range of 2 to 230 µg/ml and MBC values between 3 to 346 μg/ml against both gram-positive and negative bacteria. While both PE and PM showed a significant reduction in the number of E. coli, S. aureus, and S. epidermidis, the use of PE-CH and PM-CH led to a statistically significant and greater reduction in number of E. coli, S. aureus, and S. epidermidis strains on the biofilm, pre-formed biofilm and planktonic phases. Besides, the DPPH assay showed significant antioxidant activity for these NPs within the range of 36 to 92%. MTT assay for MHFB-1, HFF, L929, MDF, and MCF-7 cells exhibited statistically significant differences in each other that show the IC50 between 60-160 µg/ml for normal cells and 20 for cancer cells. Finally the present study indicated that both PM and PM-CH greater than PE and PE-CH in which contain high flavonoid and phenolic contents with a high antioxidation potential antioxidant properties, which could be beneficial for cell proliferation and antibiotic and anticancer applications.
Collapse
Affiliation(s)
- Akbar Vaseghi
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reza Ashrafi Parchin
- Excir Faravaran Sabalan Company, Ardabil Science and Technology Park, Ardabil, Iran
| | | | - Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, 50935, Germany
| | - Hajar Maleki
- Department of Chemistry, Institute of Inorganic Chemistry, University of Cologne, Cologne, 50939, Germany
- Center for Molecular Medicine Cologne, CMMC Research Center, Cologne, 50931, Germany
| | - Majid Sadeghizadeh
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Jalal AleAhmad St, Tehran, Iran.
| |
Collapse
|
36
|
Yao J, Chen Y, Zhang L, Cheng Y, Chen Z, Zhang Y, Zheng X, Lv Y, Wang S, Li Z, Zhao J. pH-responsive CuS/DSF/EL/PVP nanoplatform alleviates inflammatory bowel disease in mice via regulating gut immunity and microbiota. Acta Biomater 2024; 178:265-286. [PMID: 38417643 DOI: 10.1016/j.actbio.2024.02.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/19/2024] [Accepted: 02/21/2024] [Indexed: 03/01/2024]
Abstract
The clinical treatment of inflammatory bowel disease (IBD) is challenging. We developed copper sulfate (CuS)/disulfiram (DSF)/methacrylic acid-ethyl acrylate copolymer (EL)/polyvinylpyrrolidone (PVP) nanoplatform (CuS/DSF/EL/PVP) and evaluated its efficiency for treating IBD. After oral administration, the pH-sensitive EL protected the CuS/DSF/EL/PVP against degradation by acidic gastric juices. Once the colon was reached, EL was dissolved, releasing DSF and Cu2+. Further, the main in vivo metabolite of DSF can bind to Cu2+ and form copper (II) N, N-diethyldithiocarbamate (CuET), which significantly alleviated acute colitis in mice. Notably, CuS/DSF/EL/PVP outperformed CuS/EL/PVP and DSF/EL/PVP nanoplatforms in reducing colonic pathology and improving the secretion of inflammation-related cytokines (such as IL-4 and IL-10) in the colonic mucosa. RNA-seq analysis revealed that the nanoplatform reduced colonic inflammation and promoted intestinal mucosal repair by upregulating C-type lectin receptor (CLR)-related genes and signaling pathways. Furthermore, CuS/DSF/EL/PVP showed potential for improving colitis Th1/Th17 cells through innate immunity stimulation, down-regulation of inflammatory cytokines, and upregulation of anti-inflammatory cytokines. Additionally, the intervention with CuS/DSF/EL/PVP led to increased intestinal flora diversity, decreased Escherichia-Shigella abundance, and elevated levels of short-chain fatty acid (SCFA)-producing bacteria Prevotella, Lactobacillus, and Bifidobacterium, indicating their potential to modulate the dysregulated intestinal flora and suppress inflammation. STATEMENT OF SIGNIFICANCE: Our study introduces the CuS/DSF/EL/PVP nanoplatform as a therapeutic strategy for treating inflammatory bowel disease (IBD). This approach demonstrates significant efficacy in targeting the colon and alleviating acute colitis in mice. It uniquely modulates gut immunity and microbiota, exhibiting a notable impact on inflammation-related cytokines and promoting intestinal mucosal repair. The nanoplatform's ability to regulate gut flora diversity, combined with its cost-effective and scalable production, positions it as a potentially transformative treatment for IBD, offering new avenues for personalized medical interventions.
Collapse
Affiliation(s)
- Jinpeng Yao
- Department of Gastroenterology, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528244, PR China; National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Yu Chen
- Department of Gastroenterology, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528244, PR China
| | - Liang Zhang
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Yuancun Cheng
- School of Materials and Chemistry, the University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, PR China
| | - Zheng Chen
- School of Materials and Chemistry, the University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, PR China
| | - Yanhui Zhang
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Xiaoyi Zheng
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Yanwei Lv
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China
| | - Shige Wang
- School of Materials and Chemistry, the University of Shanghai for Science and Technology, No. 516 Jungong Road, Shanghai 200093, PR China.
| | - Zhaoshen Li
- Department of Gastroenterology, The Seventh Affiliated Hospital of Southern Medical University, Foshan 528244, PR China; National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China.
| | - Jiulong Zhao
- National Clinical Research Center for Digestive Diseases, Department of Gastroenterology, Changhai Hospital, Naval Medical University, No. 168 Changhai Road, Shanghai 200433, PR China.
| |
Collapse
|
37
|
Zhao C, Yang X, Fan M, Tian L, Sun T, Sun C, Jiang T. The investigation on sialic acid-modified pectin nanoparticles loaded with oxymatrine for orally targeting and inhibiting the of ulcerative colitis. Colloids Surf B Biointerfaces 2024; 236:113809. [PMID: 38447446 DOI: 10.1016/j.colsurfb.2024.113809] [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: 10/18/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
The aim of the study was to develop an oral targeting drug delivery system (OTDDS) of oxymatrine (OMT) to effectively treat ulcerative colitis (UC). The OTDDS of OMT (OMT/SA-NPs) was constructed with OMT, pectin, Ca2+, chitosan (CS) and sialic acid (SA). The obtained particles were characterized in terms of particle size, zeta potential, morphology, drug loading, encapsulation efficiency, drug release and stability. The average size of OMT/SA-NPs was 255.0 nm with a zeta potential of -12.4 mV. The loading content and encapsulation efficiency of OMT/SA-NPs were 14.65% and 84.83%, respectively. The particle size of OMT/SA-NPs changed slightly in the gastrointestinal tract. The nanoparticles can delivery most of the drug to the colon region. In vitro cell experiments showed that the SA-NPs had excellent biocompatibility and anti-inflammation, and the uptake of SA-NPs by RAW 264.7 cells was time and concentration-dependent. The conjugated SA can help the internalization of NPs into target cells. In vivo experiments showed that OMT/SA-NPs had a superior anti-inflammation effect and the effect of reducing UC, which was attributed to the delivery most of OMT to the colonic lumen, the specific targeting and retention in colitis site and the combined anti-inflammation of OMT and NPs.
Collapse
Affiliation(s)
- Chunying Zhao
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China
| | - Xin Yang
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China
| | - Mengyao Fan
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China
| | - Linan Tian
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China
| | - Tongtong Sun
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China
| | - Changshan Sun
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China.
| | - Tongying Jiang
- Shenyang Pharmaceutical University, Benxi, Liaoning 110016, PR China.
| |
Collapse
|
38
|
Huang D, Zou M, Xu C, Wang Y, Xu Z, Zhang W, Tang S, Weng Z. Colon-Targeted Oral Delivery of Hydroxyethyl Starch-Curcumin Microcapsules Loaded with Multiple Drugs Alleviates DSS-Induced Ulcerative Colitis in Mice. Macromol Biosci 2024; 24:e2300465. [PMID: 38111343 DOI: 10.1002/mabi.202300465] [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: 10/16/2023] [Revised: 12/05/2023] [Indexed: 12/20/2023]
Abstract
Combination therapy through colon-targeted oral delivery of multiple drugs presents a promising approach for effectively treating ulcerative colitis (UC). However, the codelivery of drugs with diverse physicochemical properties in a single formulation remains a formidable challenge. Here, microcapsules are designed based on hydroxyethyl starch-curcumin (HES─CUR) conjugates to enable the simultaneous delivery of hydrophobic dexamethasone acetate (DA) and hydrophilic cefazolin sodium (CS), yielding multiple drug-loaded microcapsules (CS/DA-loaded HES─CUR microcapsules, CDHC-MCs) tailored for colon-targeted therapy of UC. Thorough characterization confirms the successful synthesis and exceptional biocompatibility of CDHC-MCs. Biodistribution studies demonstrate that the microcapsules exhibit an impressive inflammatory targeting effect, accumulating preferentially in inflamed colons. In vivo experiments employing a dextran-sulfate-sodium-induced UC mouse model reveal that CDHC-MCs not only arrest UC progression but also facilitate the restoration of colon length and alleviate inflammation-related splenomegaly. These findings highlight the potential of colon-targeted delivery of multiple drugs within a single formulation as a promising strategy to enhance UC treatment, and the CDHC-MCs developed in this study hold great potential in developing novel oral formulations for advanced UC therapy.
Collapse
Affiliation(s)
- Da Huang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, China
| | - Minglang Zou
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, China
| | - Chenlan Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, China
| | - Yongming Wang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, China
| | - Zhenjin Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, China
| | - Wancong Zhang
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515051, China
- Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong, 515051, China
- Shantou Plastic surgery Clinical Research Center, Shantou, Guangdong, 515051, China
| | - Shijie Tang
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong, 515051, China
- Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong, 515051, China
- Shantou Plastic surgery Clinical Research Center, Shantou, Guangdong, 515051, China
| | - Zuquan Weng
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian, 350108, China
| |
Collapse
|
39
|
Wang N, Chen L, Huang W, Gao Z, Jin M. Current Advances of Nanomaterial-Based Oral Drug Delivery for Colorectal Cancer Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:557. [PMID: 38607092 PMCID: PMC11013305 DOI: 10.3390/nano14070557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 03/10/2024] [Accepted: 03/20/2024] [Indexed: 04/13/2024]
Abstract
Colorectal cancer (CRC) is a common malignant tumor, and traditional treatments include surgical resection and radiotherapy. However, local recurrence, distal metastasis, and intestinal obstruction are significant problems. Oral nano-formulation is a promising treatment strategy for CRC. This study introduces physiological and environmental factors, the main challenges of CRC treatment, and the need for a novel oral colon-targeted drug delivery system (OCDDS). This study reviews the research progress of controlled-release, responsive, magnetic, targeted, and other oral nano-formulations in the direction of CRC treatment, in addition to the advantages of oral colon-targeted nano-formulations and concerns about the oral delivery of related therapeutic agents to inspire related research.
Collapse
Affiliation(s)
- Nuoya Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (N.W.); (L.C.); (W.H.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
- Department of Pharmacy, Yanbian University, Yanji 133000, China
| | - Liqing Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (N.W.); (L.C.); (W.H.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (N.W.); (L.C.); (W.H.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhonggao Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (N.W.); (L.C.); (W.H.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Mingji Jin
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China; (N.W.); (L.C.); (W.H.)
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulations, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
40
|
Chauhan S, Harwansh RK. Recent advances in nanocarrier systems for ulcerative colitis: A new era of targeted therapy and biomarker integration. J Drug Deliv Sci Technol 2024; 93:105466. [DOI: 10.1016/j.jddst.2024.105466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
41
|
Mehrotra S, Kalyan BG P, Nayak PG, Joseph A, Manikkath J. Recent Progress in the Oral Delivery of Therapeutic Peptides and Proteins: Overview of Pharmaceutical Strategies to Overcome Absorption Hurdles. Adv Pharm Bull 2024; 14:11-33. [PMID: 38585454 PMCID: PMC10997937 DOI: 10.34172/apb.2024.009] [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: 11/18/2022] [Revised: 07/04/2023] [Accepted: 08/16/2023] [Indexed: 04/09/2024] Open
Abstract
Purpose Proteins and peptides have secured a place as excellent therapeutic moieties on account of their high selectivity and efficacy. However due to oral absorption limitations, current formulations are mostly delivered parenterally. Oral delivery of peptides and proteins (PPs) can be considered the need of the hour due to the immense benefits of this route. This review aims to critically examine and summarize the innovations and mechanisms involved in oral delivery of peptide and protein drugs. Methods Comprehensive literature search was undertaken, spanning the early development to the current state of the art, using online search tools (PubMed, Google Scholar, ScienceDirect and Scopus). Results Research in oral delivery of proteins and peptides has a rich history and the development of biologics has encouraged additional research effort in recent decades. Enzyme hydrolysis and inadequate permeation into intestinal mucosa are the major causes that result in limited oral absorption of biologics. Pharmaceutical and technological strategies including use of absorption enhancers, enzyme inhibition, chemical modification (PEGylation, pro-drug approach, peptidomimetics, glycosylation), particulate delivery (polymeric nanoparticles, liposomes, micelles, microspheres), site-specific delivery in the gastrointestinal tract (GIT), membrane transporters, novel approaches (self-nanoemulsifying drug delivery systems, Eligen technology, Peptelligence, self-assembling bubble carrier approach, luminal unfolding microneedle injector, microneedles) and lymphatic targeting, are discussed. Limitations of these strategies and possible innovations for improving oral bioavailability of protein and peptide drugs are discussed. Conclusion This review underlines the application of oral route for peptide and protein delivery, which can direct the formulation scientist for better exploitation of this route.
Collapse
Affiliation(s)
- Sonal Mehrotra
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Pavan Kalyan BG
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | - Pawan Ganesh Nayak
- Department of Pharmacology,Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| | | | - Jyothsna Manikkath
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal 576104, Karnataka State, India
| |
Collapse
|
42
|
Valachová K, Hassan ME, Šoltés L. Hyaluronan: Sources, Structure, Features and Applications. Molecules 2024; 29:739. [PMID: 38338483 PMCID: PMC10856924 DOI: 10.3390/molecules29030739] [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: 12/06/2023] [Revised: 01/28/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Hyaluronan (HA) is a non-sulfated glycosaminoglycan that is present in a variety of body tissues and organs. Hyaluronan has a wide range of biological activities that are frequently influenced by molar mass; however, they also depend greatly on the source, purity, and kind of impurities in hyaluronan. High-molar-mass HA has anti-inflammatory, immunosuppressive, and antiangiogenic properties, while low-molar-mass HA has opposite properties. A number of chemical modifications have been performed to enhance the stability of HA and its applications in medical practice. Hyaluronan is widely applied in medicine, such as viscosupplementation, ophthalmology, otolaryngology, wound healing, cosmetics, and drug delivery. In this review, we summarized several medical applications of polymers based on the hyaluronan backbone.
Collapse
Affiliation(s)
- Katarína Valachová
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dúbravská cesta 9, 84104 Bratislava, Slovakia
| | - Mohamed E. Hassan
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dúbravská cesta 9, 84104 Bratislava, Slovakia
- Centre of Excellence, Encapsulation & Nanobiotechnology Group, Chemistry of Natural and Microbial Products Department, National Research Centre, El Behouth Street, Cairo 12622, Egypt
| | - Ladislav Šoltés
- Centre of Experimental Medicine, Institute of Experimental Pharmacology and Toxicology, Slovak Academy of Sciences, Dúbravská cesta 9, 84104 Bratislava, Slovakia
| |
Collapse
|
43
|
Rabeh ME, Vora LK, Moore JV, Bayan MF, McCoy CP, Wylie MP. Dual stimuli-responsive delivery system for self-regulated colon-targeted delivery of poorly water-soluble drugs. BIOMATERIALS ADVANCES 2024; 157:213735. [PMID: 38154402 DOI: 10.1016/j.bioadv.2023.213735] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 12/04/2023] [Accepted: 12/14/2023] [Indexed: 12/30/2023]
Abstract
Inflammatory bowel disease (IBD) are chronic inflammatory conditions which cause significant patient morbidity. Local drug delivery to the colon can improve treatment efficacy and reduce side effects associated with IBD treatment. Smart drug delivery systems are designed to regulate the release of therapeutic agents at the desired site of action. pH-responsive drug carriers have been previously utilised for improved oral drug delivery beyond stomach harsh conditions. Additionally, the colon possesses a diverse microbiome secreting bioactive molecules e.g., enzymes, that can be exploited for targeted drug delivery. We herein synthesised and characterised a 2-hydroxyethyl methacrylate and methacrylic acid copolymer, crosslinked with an azobenzyl crosslinker, that displayed pH- and enzyme-responsive properties. The swelling and drug release from hydrogel were analysed in pH 1.2, 6.5 and 7.4 buffers, and in the presence of rat caecal matter using metronidazole and mesalamine as model BCS Class I and IV drugs, respectively. Swelling studies displayed pH-responsive swelling behaviour, where swelling was maximum at pH 7.4 and minimum at pH 1.2 (69 % versus 32 %). Consequently, drug release was limited in gastric and small intestinal conditions but increased significantly when exposed to colonic conditions containing caecal matter. This system displays promising capacity for achieving colon-targeted drug delivery with enhanced dissolution of poorly water-soluble drugs for local treatment of IBD and other colon-targeted therapies.
Collapse
Affiliation(s)
- Mohmmad E Rabeh
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | | | - Jessica V Moore
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK
| | - Mohammad F Bayan
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK; Faculty of Pharmacy, Philadelphia University, P.O Box 1, Amman 19392, Jordan
| | - Colin P McCoy
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK.
| | - Matthew P Wylie
- School of Pharmacy, Queen's University Belfast, Belfast BT9 7BL, UK.
| |
Collapse
|
44
|
Harwansh RK, Bhati H, Deshmukh R. Recent Updates on the Therapeutics Benefits, Clinical Trials, and Novel Delivery Systems of Chlorogenic Acid for the Management of Diseases with a Special Emphasis on Ulcerative Colitis. Curr Pharm Des 2024; 30:420-439. [PMID: 38299405 DOI: 10.2174/0113816128295753240129074035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/08/2024] [Accepted: 01/16/2024] [Indexed: 02/02/2024]
Abstract
Ulcerative colitis (UC) is a multifactorial disorder of the large intestine, especially the colon, and has become a challenge globally. Allopathic medicines are primarily available for the treatment and prevention of UC. However, their uses are limited due to several side effects. Hence, an alternative therapy is of utmost importance in this regard. Herbal medicines are considered safe and effective for managing human health problems. Chlorogenic acid (CGA), the herbal-derived bioactive, has been reported for pharmacological effects like antiinflammatory, immunomodulatory, antimicrobial, hepatoprotective, antioxidant, anticancer, etc. This review aims to understand the antiinflammatory and chemopreventive potential of CGA against UC. Apart from its excellent therapeutic potential, it has been associated with low absorption and poor oral bioavailability. In this context, colon-specific novel drug delivery systems (NDDS)are pioneering to overcome these problems. The pertinent literature was compiled from a thorough search on various databases such as ScienceDirect, PubMed, Google Scholar, etc., utilizing numerous keywords, including ulcerative colitis, herbal drugs, CGA, pharmacological activities, mechanism of actions, nanoformulations, clinical updates, and many others. Relevant publications accessed till now were chosen, whereas non-relevant papers, unpublished data, and non-original articles were excluded. The present review comprises recent studies on pharmacological activities and novel drug delivery systems of CGA for managing UC. In addition, the clinical trials of CGA against UC have been discussed.
Collapse
Affiliation(s)
- Ranjit K Harwansh
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India
| | - Hemant Bhati
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India
| | - Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura 281406, India
| |
Collapse
|
45
|
Zhang Q, Fu H, Zhang Y, Li L, Yan G. Rapidly degradable konjac glucomannan hydrogels cross-linked with olsalazine for colonic drug release. Biomed Mater Eng 2024; 35:125-137. [PMID: 37718772 DOI: 10.3233/bme-230066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
BACKGROUND Polysaccharide hydrogel is one of the most important materials for the colon target drug release system. However, the degradation time of polysaccharide hydrogel is much longer than the retention time in the colon. The drugs are expelled from the body before being released. OBJECTIVE In order to match the degradation of drug carriers and their retention time in the colon, a rapidly degradable konjac glucomannan (KGM) hydrogel was designed for colon target drug release. METHODS A crosslinker containing azo bond, olsalazine, was used to prepare the rapidly degradable KGM hydrogel. The degradation and drug release of the hydrogels with different crosslinking densities in the normal buffer and the human fecal medium were studied to evaluate the efficiency of colon drug release. RESULTS More than 50% of the KGM hydrogel by weight was degraded and more than 60% of the 5-fluorouracil (5-Fu) was released within 48 h in 5% w/v human fecal medium. CONCLUSION The drug was released more rapidly in a simulated colon environment than in a normal buffer. Furthermore, the drug release was controlled by the degradation of the hydrogel. The KGM hydrogel containing azo crosslinker has great potential for colon drug release.
Collapse
Affiliation(s)
- Qiao Zhang
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Huili Fu
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Yunfei Zhang
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Liang Li
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, China
| | - Guoping Yan
- School of Materials Science and Engineering, Wuhan Institute of Technology, Wuhan, China
| |
Collapse
|
46
|
Tajik E, Vaezi Z, Tabarsa M, Hekmat A, Naderi-Manesh H. Grafting of sinapic acid onto glucosamine nanoparticle as a potential therapeutic drug with enhanced anti-inflammatory activities in osteoarthritis treatment. Int J Biol Macromol 2023; 253:127454. [PMID: 37844822 DOI: 10.1016/j.ijbiomac.2023.127454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/25/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023]
Abstract
Glucosamine (Glu) is a cartilage and joint fluid matrix precursor that modulates osteoarthritic joint changes. To improve the enzymatic stability, glucosamine was developed into nanoglucosamine by the ionic gelation method through sodium tripolyphosphate (TPP) as cross-linking agent. The optimized mass ratio of Glu:TPP was (3:1) with the particle size 163 ± 25 nm and surface charge -5 mV. Then Sinapic acid (SA) as a natural phenolic acid with strong antioxidant and antimicrobial activities has been grafted onto glucosamine nanoparticles (GluNPs) with grafting efficiency (73 ± 6 %). The covalent insertion of SA was confirmed by UV-Vis, FTIR, 1HNMR, XRD, and FESEM analyses and the other physicochemical properties were also characterized. SA-g-GluNPs showed spherical shape with a mean diameter of 255 ± 20 nm and zeta potential +16 mV. The in vitro release profile of SA-g-GluNPs exhibited the sustained and pH-dependent drug release property. SA-g-GluNPs had a more pronounced effect on reducing the elevated levels of LPS-induced oxidative stress and pro-inflammatory cytokines than free SA in the human chondrocyte C28/I2 cell line. Furthermore, the antibacterial properties against E. coli and S. aureus were also improved by SA-g-GluNPs. This study demonstrated the potential of phenolic acid grafted GluNPs in therapeutic drug applications for chondroprotection and food industries.
Collapse
Affiliation(s)
- Ehteram Tajik
- Department of Biophysics, Science and Research branch, Islamic Azad University, Tehran, Iran
| | - Zahra Vaezi
- Department of Bioactive Compounds, Faculty of Interdisciplinary Science and Technologies, Tarbiat Modares University, Tehran, Iran
| | - Mehdi Tabarsa
- Department of Bioactive Compounds, Faculty of Interdisciplinary Science and Technologies, Tarbiat Modares University, Tehran, Iran; Department of Seafood Processing, Faculty of Marine Sciences, Tarbiat Modares University, Noor, Iran
| | - Azadeh Hekmat
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Hossein Naderi-Manesh
- Department of Bioactive Compounds, Faculty of Interdisciplinary Science and Technologies, Tarbiat Modares University, Tehran, Iran; Department of Biophysics, Faculty of Biological Science, Tarbiat Modares University, Postal codes: 14115-154 Tehran, Iran.
| |
Collapse
|
47
|
Zhang Y, Wu Y, Yan Y, Ma Y, Tu L, Shao J, Tang X, Chen L, Liang G, Yin L. Dual-Targeted Nanoparticle-in-Microparticle System for Ulcerative Colitis Therapy. Adv Healthc Mater 2023; 12:e2301518. [PMID: 37660262 DOI: 10.1002/adhm.202301518] [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: 05/11/2023] [Revised: 08/01/2023] [Indexed: 09/04/2023]
Abstract
Conventional oral therapy for ulcerative colitis (UC) is associated with premature release or degradation of drugs in the harsh gastrointestinal environment, resulting in reduced therapeutic effectiveness. Consequently, the present study aims to develop a dual-targeted delivery system with a nanoparticle-in-microparticle (nano-in-micro) structure. The prepared Asiatic Acid-loaded delivery system (AA/CDM-BT-ALG) has pH-sensitive properties. Cellular uptake evaluation confirms that nanoparticles exhibit targeted absorption by macrophages and Caco-2 cells through mannose (Man) receptor and biotin-mediated endocytosis, respectively. Therefore, this mechanism effectively enhances intracellular drug concentration. Additionally, the biodistribution study conducted on the gastrointestinal tract of mice indicates that the colon of the microspheres group shows higher fluorescence intensity with longer duration than the other groups. This finding indicates that the microspheres exhibit selective accumulation in areas of colon inflammation. In vivo experiments in colitis mice showed that AA/CDM-BT-ALG significantly alleviates the histopathological characteristics of the colon, reduced neutrophil, and macrophage infiltration, and decreases pro-inflammatory cytokine expression. Furthermore, the effect of AA/CDM-BT-ALG on colitis is validated to be closely related to the TLR4/MyD88/NF-κB signaling pathway. The present findings suggest that the development of a dual-targeted delivery system is accomplished effectively, with the potential to serve as a drug-controlled release system for treating UC.
Collapse
Affiliation(s)
- Yawen Zhang
- School of Pharmacy, Hangzhou Medical College, 182 Tianmushan Rd, Hangzhou, 310013, China
| | - Yue Wu
- School of Pharmacy, Hangzhou Medical College, 182 Tianmushan Rd, Hangzhou, 310013, China
| | - Yuping Yan
- School of Pharmacy, Hangzhou Medical College, 182 Tianmushan Rd, Hangzhou, 310013, China
| | - Yijing Ma
- School of Pharmacy, Hangzhou Medical College, 182 Tianmushan Rd, Hangzhou, 310013, China
| | - Linglan Tu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, 310013, China
| | - Jingjing Shao
- School of Pharmacy, Wenzhou Medical University, Wenzhou, 325000, China
| | - Xuanyu Tang
- School of Pharmacy, Hangzhou Medical College, 182 Tianmushan Rd, Hangzhou, 310013, China
| | - Lingfeng Chen
- School of Pharmacy, Hangzhou Medical College, 182 Tianmushan Rd, Hangzhou, 310013, China
| | - Guang Liang
- School of Pharmacy, Hangzhou Medical College, 182 Tianmushan Rd, Hangzhou, 310013, China
| | - Lina Yin
- School of Pharmacy, Hangzhou Medical College, 182 Tianmushan Rd, Hangzhou, 310013, China
| |
Collapse
|
48
|
Shewaiter MA, Selim AA, Rashed HM, Moustafa YM, Gad S. Niosomal formulation of mefenamic acid for enhanced cancer targeting; preparation, characterization and biodistribution study using radiolabeling technique. J Cancer Res Clin Oncol 2023; 149:18065-18080. [PMID: 37982828 PMCID: PMC10725351 DOI: 10.1007/s00432-023-05482-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/18/2023] [Indexed: 11/21/2023]
Abstract
BACKGROUND This work aimed to prepare niosomal formulations of an anticancer agent [mefenamic acid (MEF)] to enhance its cancer targeting. 131I was utilized as a radiolabeling isotope to study the radio-kinetics of MEF niosomes. METHODS niosomal formulations were prepared by the ether injection method and assessed for entrapment efficiency (EE%), zeta potential (ZP), polydispersity index (PDI) and particle size (PS). MEF was labeled with 131I by direct electrophilic substitution reaction through optimization of radiolabeling-related parameters. In the radio-kinetic study, the optimal 131I-MEF niosomal formula was administered intravenously (I.V.) to solid tumor-bearing mice and compared to I.V. 131I-MEF solution as a control. RESULTS the average PS and ZP values of the optimal formulation were 247.23 ± 2.32 nm and - 28.3 ± 1.21, respectively. The highest 131I-MEF labeling yield was 98.7 ± 0.8%. The biodistribution study revealed that the highest tumor uptake of 131I-MEF niosomal formula and 131I-MEF solution at 60 min post-injection were 2.73 and 1.94% ID/g, respectively. CONCLUSION MEF-loaded niosomes could be a hopeful candidate in cancer treatment due to their potent tumor uptake. Such high targeting was attributed to passive targeting of the nanosized niosomes and confirmed by radiokinetic evaluation.
Collapse
Affiliation(s)
- Mona A Shewaiter
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Sinai University, Kantara, Egypt
| | - Adli A Selim
- Labeled Compounds Department, Hot Laboratories Center, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Hassan M Rashed
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Sinai University, Kantara, Egypt.
- Labeled Compounds Department, Hot Laboratories Center, Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Yasser M Moustafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt
| | - Shadeed Gad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt.
| |
Collapse
|
49
|
Min K, Sahu A, Jeon SH, Tae G. Emerging drug delivery systems with traditional routes - A roadmap to chronic inflammatory diseases. Adv Drug Deliv Rev 2023; 203:115119. [PMID: 37898338 DOI: 10.1016/j.addr.2023.115119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 10/30/2023]
Abstract
Inflammation is prevalent and inevitable in daily life but can generally be accommodated by the immune systems. However, incapable self-healing and persistent inflammation can progress to chronic inflammation, leading to prevalent or fatal chronic diseases. This review comprehensively covers the topic of emerging drug delivery systems (DDSs) for the treatment of chronic inflammatory diseases (CIDs). First, we introduce the basic biology of the chronic inflammatory process and provide an overview of the main CIDs of the major organs. Next, up-to-date information on various DDSs and the associated strategies for ensuring targeted delivery and stimuli-responsiveness applied to CIDs are discussed extensively. The implementation of traditional routes of drug administration to maximize their therapeutic effects against CIDs is then summarized. Finally, perspectives on future DDSs against CIDs are presented.
Collapse
Affiliation(s)
- Kiyoon Min
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Abhishek Sahu
- Department of Biotechnology, National Institute of Pharmaceutical Education & Research (NIPER), Hajipur, 844102, India
| | - Sae Hyun Jeon
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Giyoong Tae
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea.
| |
Collapse
|
50
|
Zhao Y, He R, Zang J, Yin W, Su R, Xiong W, Xu W, Zhang J, Liu Y, Ren T, Huang Y, Li Y. Pathologically catalyzed physical coating restores the intestinal barrier for inflammatory bowel disease therapy. J Nanobiotechnology 2023; 21:444. [PMID: 37996883 PMCID: PMC10668504 DOI: 10.1186/s12951-023-02227-0] [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: 09/30/2023] [Accepted: 11/19/2023] [Indexed: 11/25/2023] Open
Abstract
Intestinal epithelia impairment of inflammatory bowel disease (IBD) leads to the leakage of bacteria and antigens and the consequent persistent immune imbalance. Restoring the epithelial barrier is a promising therapeutic target but lacks effective and safe clinical interventions. By identifying the catalase (CAT) presence in the IBD pathological environment, we herein develop a CAT-catalyzed pathologically coating on the damaged epithelial barrier to inhibit intestinal leakage for IBD therapy. With the codelivery of CaO2 (a CAT substrate) and dopamine, the nanosystem can enable CAT-catalyzed oxygen (O2) production and in-situ polymerization of dopamine and then yield a thin and integrative polydopamine (PDA) coating on the intestinal barrier due to the highly adhesive property of PDA. In vivo study demonstrates that PDA coating provides not only a protective barrier by restricting intestinal leakage but also a favorable anti-inflammation effect. Beyond drug management, this work provides a physical repair strategy via catalyzed coating for IBD therapy.
Collapse
Affiliation(s)
- Yuge Zhao
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO), School of Medicine, Tongji University, Shanghai, 200092, China
| | - Ruiqing He
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO), School of Medicine, Tongji University, Shanghai, 200092, China
| | - Jie Zang
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO), School of Medicine, Tongji University, Shanghai, 200092, China
| | - Weimin Yin
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO), School of Medicine, Tongji University, Shanghai, 200092, China
| | - Runping Su
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO), School of Medicine, Tongji University, Shanghai, 200092, China
| | - Wei Xiong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weihua Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jiaxin Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yiqiong Liu
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO), School of Medicine, Tongji University, Shanghai, 200092, China
| | - Tianbin Ren
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO), School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yongzhuo Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Yongyong Li
- Shanghai Tenth People's Hospital, The Institute for Biomedical Engineering & Nano Science (iNANO), School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|