1
|
Zhang Z, Cheng W, Li H, Li X, Zhang X, Li Y, Wang B. Lactate-depleted pillar[5]arene-based chiral supramolecular nanovesicles for L-glucose-mediated tumor-specific chemodynamic- and photodynamic-synergistic therapy. J Mater Chem B 2025; 13:5058-5069. [PMID: 40208309 DOI: 10.1039/d5tb00436e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
The distinct interactions of D/L-glucose with cells and biological systems have garnered significant attention. However, the impact of chiral glucose-modified nanomaterials on cancer diagnosis and treatment remains largely unexplored. Here, based on the host-guest interaction between D-/L-glucose-modified pillar[5]arene (D-/L-CP5) serving as the host molecule and Fe-porphyrin derivatives (FeTPPNHC) acting as the guest, an acid-responsive chiral supramolecular vesicle was constructed for transporting lactate oxidases (LOx) (denoted as LOx@D-/L-CP5⊃FeTPPNHC), aiming to enhance chirality-mediated tumor-specific cascade chemodynamic therapy (CDT) and photodynamic therapy (PDT) through the depletion of lactic acid (LA). Surprisingly, the L-glucose-mediated chiral vesicles exhibit remarkable chirality recognition and lactate depletion capabilities, which were higher than the D-glucose-mediated chiral vesicles. Once internalized by cancer cells, L-supramolecular nanomicelles can directly consume LA to generate a considerable amount of H2O2, which can then be converted into ˙OH and 1O2. In vitro and in vivo studies demonstrate the high tumor specificity and therapeutic efficacy of LOx@LCP5⊃FeTPPNHC. The findings suggest that chiral glucose-modified nanomaterials hold great potential in targeted cancer treatment, paving the way for the development of innovative cancer therapeutics based on their unique interactions with biological systems.
Collapse
Affiliation(s)
- Zefan Zhang
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Wenyuan Cheng
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Hui Li
- Department of Cardiovascular Medicine, Affiliated Hospital of Yan'an University, Yan'an, 716000, P. R. China
| | - Xinxin Li
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, China.
| | - Xuan Zhang
- Department of Cardiovascular Medicine, Affiliated Hospital of Yan'an University, Yan'an, 716000, P. R. China
| | - Yu Li
- Dr. Y. Li, Department of Magnetic Resonance, The First Clinical Medical College, Lanzhou University, Lanzhou, 730030, P. R. China
| | - Baodui Wang
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Lanzhou University, Lanzhou, Gansu 730000, China.
| |
Collapse
|
2
|
El-Naggar NEA, Elshafey N, Alafifi HI, Eltahy MA, Haikl RI, ElShazly HA, Ahmed YW, Hassan HI, Safo MM, Haroun SA, El-Sawah AA. Innovative strategy for chitosan nanoparticles biosynthesis using Gelidium amansii, statistical optimization, characterization, cytotoxicity and molecular docking against hepatocellular carcinoma. Int J Biol Macromol 2025:143687. [PMID: 40316091 DOI: 10.1016/j.ijbiomac.2025.143687] [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: 12/02/2024] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/04/2025]
Abstract
The demand for biosynthesized chitosan nanoparticles (chitosan-NPs) increases owing to their biocompatibility, versatile functional groups, mucoadhesive capabilities, antimicrobial activity, controlled drug-release, and anticancer efficacy. The current study presents an innovative green-synthesis strategy for chitosan-NPs using Gelidium amansii hot-water extract as a crosslinking and stabilizing agent. The UV-vis spectrum shows a strong absorption peak at 243 nm. The transmission electron microscopy analysis revealed that the biosynthesized chitosan-NPs were spherical and their sizes varied from 5.1 to 13.57 nm. Fourier-Transform Infra-Red analysis proved the presence of multiple functional groups, confirming the existence of capping agent on chitosan-NPs surface. The ζ-potential value on chitosan-NPs surface was +32.4 mV. To maximize chitosan-NPs yield, face-centered central composite design was used. The highest of 13.1 mg/mL was obtained using an initial pH of 8, a 24-h incubation time, and 1.5 % chitosan. Doxorubicin was successfully loaded into chitosan-NPs with encapsulation efficiency and drug loading of 65.42 ± 0.03 and 11.89 ± 0.01 %. The IC50 values for DOX, chitosan-NPs, and chitosan-NPs + DOX for HePG-2 cells were 7.32 ± 0.5, 21.17 ± 1.4, and 5.90 ± 0.3 μg/mL; respectively. Chitosan-NPs have a strong capacity to interact with the receptor proteins through numerous hydrogen bond interactions. These findings highlight future research on chitosan-NPs in biological and pharmacological applications requiring protein-ligand interactions.
Collapse
Affiliation(s)
- Noura El-Ahmady El-Naggar
- Department of Bioprocess Development, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City), Alexandria 21934, Egypt.
| | - Naglaa Elshafey
- Botany and Microbiology Department, Faculty of Science, Arish University, Al-Arish 45511, Egypt
| | - Hagar I Alafifi
- Biotechnology and Its Application Program, Department of Botany, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Manar A Eltahy
- Biotechnology and Its Application Program, Department of Botany, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Reem I Haikl
- Biotechnology and Its Application Program, Department of Botany, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Hagar A ElShazly
- Biotechnology and Its Application Program, Department of Botany, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Yasmin W Ahmed
- Biotechnology and Its Application Program, Department of Botany, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Hossam I Hassan
- Biotechnology and Its Application Program, Department of Botany, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed M Safo
- Biotechnology and Its Application Program, Department of Botany, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - S A Haroun
- Botany Department, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Asmaa A El-Sawah
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
3
|
Khoubafarin S, Nath P, Malla S, Desai D, Gorgas WD, Tiwari AK, Ray A. High-resolution multi-modal imaging of sub-cellular structures with low numerical aperture objective. JPHYS PHOTONICS 2025; 7:025021. [PMID: 40143999 PMCID: PMC11933920 DOI: 10.1088/2515-7647/adc04f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Imaging of subcellular structures, which underpins many of the advances in biological and medical sciences, requires microscopes with high numerical aperture (N.A.) objectives which are costly, complex, requires oil immersion and have very limited field-of-view, typically covering a handful of cells. Here, we leverage a low N.A. objective to simultaneously capture scattering, phase, and fluorescence images of subcellular structures in breast cancer cells (BT-20) and observe nanoparticle uptake, with sub-diffraction-limited resolution (<400 nm with a 0.25 N.A. objective) utilizing a 2-dimensional (2-D) microlens substrate. High resolution labeled and label-free images of subcellular components is made possible by implementing a specific configuration, wherein the sample is placed in close proximity to the microlens substrate, which results in efficient collection of the rapidly decaying evanescent waves that contains the high frequency information, thereby improving resolution and the light capture efficiency. The microlens-assisted imaging provides an easy-to-implement and cost-effective means of drastically improving the resolution of any microscope with low N.A. objective lenses, paving the way for the development of affordable, portable multi-modal imaging systems with high-resolution imaging capabilities. This technology has broad implications for various fields and could democratize access to high-quality microscopy, particularly for application in resource-limited settings.
Collapse
Affiliation(s)
- Somaiyeh Khoubafarin
- Department of Physics and Astronomy, University of Toledo, Toledo, OH 43606, United States of America
| | - Peuli Nath
- Department of Physics and Astronomy, University of Toledo, Toledo, OH 43606, United States of America
| | - Saloni Malla
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43606, United States of America
| | - Durgesh Desai
- Department of Physics and Astronomy, University of Toledo, Toledo, OH 43606, United States of America
| | - William D Gorgas
- Department of Electrical and Computer Science, University of Toledo, Toledo, OH 43606, United States of America
| | - Amit K Tiwari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas of Medical Sciences, Little Rock, AR, United States of America
| | - Aniruddha Ray
- Department of Physics and Astronomy, University of Toledo, Toledo, OH 43606, United States of America
| |
Collapse
|
4
|
Andongma AA, Whitten MMA, Chofong GN, Dyson PJ. The thrips gut pH and implications for symbiont-mediated RNAi. BULLETIN OF ENTOMOLOGICAL RESEARCH 2025:1-7. [PMID: 40235299 DOI: 10.1017/s0007485325000240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
The gut pH plays crucial roles in diet preference, habitat choice, insect fitness, and insect-microbial relationships. It significantly impacts enzyme activity efficiency, as well as the internalisation and efficacy of pesticides. Without a comprehensive understanding of the gut environment, potential pest management strategies cannot be fully optimised.This study investigates the gut pH of the globally invasive pest insect Western flower thrips Frankliniella occidentalis, and the effect its Gram-negative symbiotic gut bacterium BFo2 has on pH modulation. Indicator dyes were fed to F. occidentalis and the gut pH was found to vary between 6 and 7. In general, the larval and adult guts appear to have a pH of between 6 and 6.5; however, the posterior gut of some adults appears to be closer to 7. This almost neutral pH offers a favourable environment for the neutrophilic symbiotic BFo2. The ability of BFo2 isolates to buffer pH towards neutral was also observed during in vitro culture using broths at different pH values.This paper also discusses the implications of this gut environment on dsRNAi delivery. By laying the foundation for understanding how gut pH can be leveraged to enhance current pest management strategies, this study particularly benefits research aimed at optimising the delivery of lethal dsRNA through symbiont-mediated RNAi to Western flower thrips in pest management programs.
Collapse
Affiliation(s)
- Awawing A Andongma
- Applied Molecular Microbiology Group, Institute of Life Sciences, Swansea University School of Medicine, Swansea, UK
- Insect and Parasite Ecology Group, Lancaster Environment Centre, Lancaster University, Lancaster, UK
| | - Miranda M A Whitten
- Applied Molecular Microbiology Group, Institute of Life Sciences, Swansea University School of Medicine, Swansea, UK
| | - Gilbert N Chofong
- Julius Kühn Institute (JKI)-Federal Research Center for Cultivated Plants, Institute for Epidemiology and Pathogen Diagnostics, Braunschweig, Germany
| | - Paul J Dyson
- Applied Molecular Microbiology Group, Institute of Life Sciences, Swansea University School of Medicine, Swansea, UK
| |
Collapse
|
5
|
Ashoub MH, Golestani A, Amiri M, Razavi R, Farsinejad A. pH-sensitive sulfasalazine release from green-synthesized mesoporous Fe 3O 4@SiO 2 nanocomposites using Opuntia ficus-indica extract. J Pharm Sci 2025; 114:103792. [PMID: 40222721 DOI: 10.1016/j.xphs.2025.103792] [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: 12/02/2024] [Revised: 03/27/2025] [Accepted: 04/05/2025] [Indexed: 04/15/2025]
Abstract
This study presents the green synthesis of mesoporous Fe3O4@SiO2 nanocomposites using Opuntia ficus-indica extract for pH-sensitive delivery of sulfasalazine. The synthesized nanocomposites exhibited well-defined spherical morphology with particle sizes ranging from 80 to 120 nm and superior superparamagnetic properties with a saturation magnetization of 75.8 emu/g. X-ray diffraction analysis confirmed the formation of a pure magnetite phase with an average crystallite size of 17.37 nm, while nitrogen physisorption revealed a high specific surface area of 165.8 m²/g with uniform mesopores centered at 4.2 nm. The nanocomposites demonstrated exceptional drug loading characteristics with 81.1 % loading efficiency and 23.9 % loading capacity. pH-dependent release studies showed enhanced release under acidic conditions (82 % at pH 3.5) compared to physiological pH (50 % at pH 7.4), indicating potential for targeted drug delivery applications. Cytotoxicity studies using L929 cells and peripheral blood mononuclear cells (PBMCs) revealed remarkable biocompatibility at concentrations up to 200 µg/mL over 72 h. These findings establish the potential of green-synthesized mesoporous Fe3O4@SiO2 nanocomposites as efficient carriers for pH-sensitive drug delivery systems, offering an environmentally friendly approach to developing advanced therapeutic platforms.
Collapse
Affiliation(s)
- Muhammad Hossein Ashoub
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Amin Golestani
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Mahnaz Amiri
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran.
| | - Razieh Razavi
- Department of Chemistry, Faculty of Science, University of Jiroft, Jiroft, Iran.
| | - Alireza Farsinejad
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran; Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
6
|
Nguyen MN, Lipp P, Zucker I, Schäfer AI. Quantification of Nanoplastics and Inorganic Nanoparticles via Laser-Induced Breakdown Detection (LIBD). SMALL METHODS 2025:e2402060. [PMID: 40195877 DOI: 10.1002/smtd.202402060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/18/2025] [Indexed: 04/09/2025]
Abstract
Nanoparticles with diverse characteristics are difficult to quantify at low concentrations in the water environment (106-109 particles mL-1 for nanoplastics originating from the breakdown of plastic debris) for the evaluation of effective treatment methods. This study examines the sensitivity, or limit of detection (LOD), of laser-induced breakdown detection (LIBD) for the counting of nanoparticles, including nanoplastics. For polystyrene (PS) standards with sizes of 20-400 nm, LIBD shows relatively low LODs (for example, 2 × 106 particles mL-1 for 100 nm particles) compared with turbidity monitoring, UV-vis spectroscopy (both 6 × 108 particles mL-1), and nanoparticle tracking analysis (2 × 107 particles mL-1). For nanoplastics (PS, polypropylene, and polyethylene terephthalate), the detection limits are 104 - 105 particles mL-1, one to two orders of magnitude lower than the PS standards. LIBD can quantify inorganic nanoparticles, such as zeolite, titania, and hematite. The sensitivity increases (i.e., LOD reduces) with increasing particle density, while some particles are prone to artifacts. The low LODs make LIBD a robust technique for counting nanoparticles of various types and sizes, even at the concentrations found in the permeate of membrane-based water treatment systems. Given the high sensitivity, LIBD has the potential to be applied in membrane integrity monitoring and fundamental studies on membrane mechanisms.
Collapse
Affiliation(s)
- Minh N Nguyen
- Institute for Advanced Membrane Technology (IAMT), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Pia Lipp
- TZW: DVGW-Technologiezentrum Wasser, Karlsruher Str. 84, 76139, Karlsruhe, Germany
| | - Ines Zucker
- School of Mechanical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, 69978, Israel
| | - Andrea I Schäfer
- Institute for Advanced Membrane Technology (IAMT), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|
7
|
Liu X, Gao M, Bao J. Precisely Targeted Nanoparticles for CRISPR-Cas9 Delivery in Clinical Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:540. [PMID: 40214585 PMCID: PMC11990453 DOI: 10.3390/nano15070540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/31/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
Clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR-Cas9), an emerging gene-editing technology, has recently gained rapidly increasing attention. However, the lack of efficient delivery vectors to deliver CRISPR-Cas9 to specific cells or tissues has hindered the translation of this biotechnology into clinical applications. Chemically synthesized nanoparticles (NPs), as attractive non-viral delivery platforms for CRISPR-Cas9, have been extensively investigated because of their unique characteristics, such as controllable size, high stability, multi-functionality, bio-responsive behavior, biocompatibility, and versatility in chemistry. In this review, the key considerations for the precise design of chemically synthesized-based nanoparticles include efficient encapsulation, cellular uptake, the targeting of specific tissues and cells, endosomal escape, and controlled release. We discuss cutting-edge strategies to integrate chemical modifications into non-viral nanoparticles that guide the CRISPR-Cas9 genome-editing machinery to specific edits. We also highlighted the rationale of intelligent nanoparticle design. In particular, we have summarized promising functional groups and molecules that can effectively optimize carrier function. In addition, this review focuses on advances in the widespread application of NPs delivery in the biomedical fields to promote the development of safe, specific, and efficient NPs for delivering CRISPR-Cas9 systems, providing references for accelerating their clinical translational applications.
Collapse
Affiliation(s)
| | | | - Ji Bao
- Department of Pathology, Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, National Health Commission of China, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
8
|
Steffè A, Milano F, Reyes SG, Buco F, Leonetti R, Roque-Diaz Y, Zuffi S, Di Gianvincenzo P, Cortese AR, Ritacco H, Andreozzi P, Ortore MG, Moya SE, Marradi M. Supramolecular dextran/polyamine phosphate nanocapsules with smart responsiveness for encapsulation of therapeutics. J Colloid Interface Sci 2025; 683:620-630. [PMID: 39706081 DOI: 10.1016/j.jcis.2024.12.074] [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: 09/06/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024]
Abstract
The polyallylamine hydrochloride (PAH) polymer is here functionalized with branched and biocompatible polysaccharide dextran (DEX) molecules. Covalent conjugation of DEX to PAH has been achieved through a straightforward reductive amination approach, allowing for a controlled number of DEX chains per PAH polymer (PAH:DEXn, n = 0.1, 0.5, 1, 2, 5, 10). When exposed to phosphate buffer, PAH:DEXn polymers form supramolecular assemblies. Physico chemical characteristics and pH responsiveness of the assemblies are correlated with the number of dextran chains per PAH molecule. Nanocapsules (NCs) are formed when PAH:DEX ratio is 1. Capsule formation is explained by the branched nature of DEX and steric consideration ruling the organization of polyamine chains in phosphate buffer. NCs and glyconanoparticles formed with n < 1 are responsive to pH changes, being disassembled at endosomal pH < 6 and reassembled when 6 < pH < 9. Dynamic light Scattering (DLS), ζ-potential measurements, cryo-Electron Microscopy and Small Angle X-ray Scattering (SAXS) provided key information about their structure, morphology, size, polydispersity, surface charge, and stability over time. Protein entrapment into the NCs and pH-dependent release is demonstrated with bovine serum albumin (BSA) as model protein by diffusion measurements in fluorescence correlation spectroscopy (FCS), following changes in BSA conformation before and after triggering NC disassembly by circular dichroism (CD), and comparing NCs SAXS fingerprints with and without BSA. Our results show novel assemblies based on polyamine phosphate interactions with capacity of loading large molecules through the formation of capsules, which may find applications in the endosomal delivery of therapeutic proteins and enzymes.
Collapse
Affiliation(s)
- Aharon Steffè
- Department of Chemistry 'Ugo Schiff', University of Florence, via della Lastruccia 3-13, 50019 Sesto Fiorentino (FI), Italy
| | - Francesca Milano
- Department of Chemistry 'Ugo Schiff', University of Florence, via della Lastruccia 3-13, 50019 Sesto Fiorentino (FI), Italy
| | - Santiago Giménez Reyes
- Soft Matter Nanotechnology Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, 20014 Donostia-San Sebastián, Guipúzcoa, Spain; Instituto de Física del Sur (IFISUR), Departamento de Física, Universidad Nacional del Sur (UNS), CONICET, Av. Alem 1253, Bahía Blanca 8000, Argentina
| | - Francesca Buco
- Department of Chemistry 'Ugo Schiff', University of Florence, via della Lastruccia 3-13, 50019 Sesto Fiorentino (FI), Italy
| | - Riccardo Leonetti
- Department of Life and Environmental Sciences, Marche Polytechnic University, via Brecce Bianche, 60131 Ancona, Italy
| | - Yessica Roque-Diaz
- Department of Life and Environmental Sciences, Marche Polytechnic University, via Brecce Bianche, 60131 Ancona, Italy
| | - Sofia Zuffi
- Soft Matter Nanotechnology Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, 20014 Donostia-San Sebastián, Guipúzcoa, Spain; Molecular Oncology Laboratory, IIS BioGipuzkoa, P° Dr. Beguiristain s/n, 20014 San Sebastián, Gipuzkoa, Spain
| | - Paolo Di Gianvincenzo
- Soft Matter Nanotechnology Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, 20014 Donostia-San Sebastián, Guipúzcoa, Spain
| | - Angela Roberta Cortese
- Department of Chemistry 'Ugo Schiff', University of Florence, via della Lastruccia 3-13, 50019 Sesto Fiorentino (FI), Italy
| | - Hernan Ritacco
- Instituto de Física del Sur (IFISUR), Departamento de Física, Universidad Nacional del Sur (UNS), CONICET, Av. Alem 1253, Bahía Blanca 8000, Argentina
| | - Patrizia Andreozzi
- Department of Chemistry 'Ugo Schiff', University of Florence, via della Lastruccia 3-13, 50019 Sesto Fiorentino (FI), Italy
| | - Maria Grazia Ortore
- Department of Life and Environmental Sciences, Marche Polytechnic University, via Brecce Bianche, 60131 Ancona, Italy
| | - Sergio E Moya
- Soft Matter Nanotechnology Group, CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Paseo Miramón 182, 20014 Donostia-San Sebastián, Guipúzcoa, Spain.
| | - Marco Marradi
- Department of Chemistry 'Ugo Schiff', University of Florence, via della Lastruccia 3-13, 50019 Sesto Fiorentino (FI), Italy.
| |
Collapse
|
9
|
Firouzjaei AA, Mohammadi-Yeganeh S. Advancements in Targeted Therapies for Colorectal Cancer: Innovative Drug Formulation and Delivery Strategies. Arch Pharm (Weinheim) 2025; 358:e202400969. [PMID: 40259467 DOI: 10.1002/ardp.202400969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 04/23/2025]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality globally, with increasing incidence presenting significant treatment challenges. Traditional nontargeted therapies often result in high toxicity and limited efficacy, underscoring the need for improved treatment modalities. This review highlights recent advancements in drug delivery systems to enhance therapeutic outcomes for CRC. We examine innovative strategies, including computer-assisted pharmaceutical formulation, sustained-release matrices, and prodrugs, as well as targeted delivery mechanisms such as exosomes, liposomes, hydrogels, antibody-drug conjugates, and stimuli-responsive systems. These methodologies offer improved drug biodistribution, enhanced targeting of cancer cells, and reduced off-target effects, promising better clinical outcomes. Additionally, we discuss the development of novel formulations designed to optimize the delivery of therapeutic agents in advanced CRC. Ongoing clinical trials investigating these innovative systems signify a shift toward more effective patient treatment options. While challenges remain in the clinical application of these targeted therapies, continued research offers promising avenues for improving patient outcomes in CRC. This study aims to inform future strategies for managing this aggressive disease, ultimately enhancing survival rates and quality of life for affected individuals.
Collapse
Affiliation(s)
- Ali Ahmadizad Firouzjaei
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samira Mohammadi-Yeganeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Sun B, Gao W, Yu X, Zhang C, Du H, Luo Y, Zhu J, Yang P, Zhang M. Charge regulated pH/NIR dual responsive nanoplatforms centered on cuproptosis for enhanced cancer theranostics. Biomaterials 2025; 315:122907. [PMID: 39476451 DOI: 10.1016/j.biomaterials.2024.122907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 09/23/2024] [Accepted: 10/20/2024] [Indexed: 12/09/2024]
Abstract
Multifunctional nanoplatforms capable of simultaneously executing multimodal therapy and imaging functions are of great potentials for cancer theranostics. We present an elegantly designed, easy-to-fabricate poly(acrylic acid)/mesoporous calcium phosphate/mesoporous copper phosphate nanosphere (PAA/mCaP/mCuP NS) with outstanding pH/NIR-sensitive multimodal-synergic anti-tumor effects. Optimal porous PAA NS scaffolds were prepared at room temperature by balancing the intra-PAA polymer and polymer-solvents Lennard-Jones potentials in a water:isopropyl alcohol (IPA) mix-solvent. Subsequent sponging of Ca2+ and Cu2+, and adsorption of PO43- to the PAA template were achieved through exquisite electrostatic interactions among ions and the ionizable PAA side-chain in an aqueous environment. This forms the basis for the tumor microenvironment pH-triggered release of Cu2+ to induce cuproptosis, as well as the photothermal effect originating from CuP, while Ca2+ can enhance the nanoplatform's biocompatibility and can damage mitochondria when overloaded. Lastly, PAA/mCaP/mCuP NSs still exhibit high drug loading efficiency for doxorubicin (DOX), enabling chemotherapy. Satisfactory anti-tumor effects of these modalities, along with their synergistic effects, were verified both in vitro and in vivo, with the NSs demonstrating good biodegradation in the latter. The fabricated NS itself holds great promise as an anti-tumor nanomedicine, and the thorough mechanical insights into NS formation may inspire the design of next-generation multifunctional nanoplatforms.
Collapse
Affiliation(s)
- Bin Sun
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wei Gao
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xinyuan Yu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Chunpeng Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Haoyang Du
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yakun Luo
- National Health Commission Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, China
| | - Jiuxin Zhu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Piaoping Yang
- Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, China.
| | - Manjie Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), College of Pharmacy, Harbin Medical University, Harbin, China; Key Laboratory of Superlight Materials and Surface Technology, Ministry of Education, College of Materials Science and Chemical Engineering, Harbin Engineering University, Harbin, China.
| |
Collapse
|
11
|
Zhang G, Zhang X, Yue K, Zhong W. Mechanistic study of enhanced drug release in mixed pH-responsive peptide-loaded liposomes. J Biomol Struct Dyn 2025:1-15. [PMID: 40126078 DOI: 10.1080/07391102.2025.2481581] [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: 12/04/2024] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
Liposomes serving as nanocarriers offer significant advantages in drug delivery for tumor treatment. There still exists challenges in controlling drug release by disintegrating the liposome membrane for the improvement of therapeutic efficiency. In this paper, a novel method involving the mixture of short peptides with pH-responsive characteristics into the cargo has been introduced. This approach facilitates the release of doxorubicin (DOX) in the acidic tumor tissue environment. The efficacy of this improvement was elucidated through molecular dynamics simulations and experiments. Liposomes incorporating a 1:1 ratio of peptides-DOX exhibited pronounced pH sensitivity and an enhanced drug release profile. The underlying mechanism is attributed to the peptides entering tumor tissues and undergoing protonation in acidic conditions, which increases the hydrophilicity of the peptide-DOX clusters and the internal surface tension of the liposomes. This alteration disrupts the balance between the inner and outer surface tensions of the nanocarrier, causing the liposomes to structurally disintegrate and thus enhancing drug release. The results from both thermodynamic analysis results and experimental data confirm the augmented drug release efficiency of this method, offering valuable theoretical insights for nanoparticle design and determining the optimal mixing ratio for therapeutic applications.
Collapse
Affiliation(s)
- Genpei Zhang
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing, China
- Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province, China
| | - Xilong Zhang
- Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province, China
| | - Kai Yue
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing, China
- Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province, China
| | - Weishen Zhong
- School of Energy and Environmental Engineering, University of Science and Technology Beijing, Beijing, China
- Shunde Graduate School of University of Science and Technology Beijing, Shunde, Guangdong Province, China
| |
Collapse
|
12
|
Ngambenjawong C, Ko H, Samad T, Pishesha N, Ploegh HL, Bhatia SN. Nanobody-Targeted Conditional Antimicrobial Therapeutics. ACS NANO 2025; 19:9958-9970. [PMID: 40044143 PMCID: PMC11924319 DOI: 10.1021/acsnano.4c16007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/09/2025] [Accepted: 02/11/2025] [Indexed: 03/19/2025]
Abstract
Conditional therapeutics that rely on disease microenvironment-specific triggers for activation are a promising strategy to improve therapeutic cargos. Among the investigated triggers, protease activity is used most often because of its dysregulation in several diseases. How to optimally fine-tune protease activation for different therapeutic cargos remains a challenge. Here, we designed nanobody-targeted conditional antimicrobial therapeutics to deliver a model therapeutic peptide and protein to the site of bacterial infection. We explored several parameters that influence proteolytic activation. We report the use of targeting nanobodies to enhance the activation of therapeutics that are otherwise activated inefficiently despite extensive optimization of the cleavable linker. Specifically, the pairing of Ly6G/C or ADAM10-targeting nanobodies with ADAM10-cleavable linkers improved activation via proximity-enabled reactivity. This study demonstrates a distinct role of active targeting in conditional therapeutic activation. More broadly, this optimization framework provides a guideline for the development of conditional therapeutics to treat various diseases in which protease activity is dysregulated.
Collapse
Affiliation(s)
- Chayanon Ngambenjawong
- Koch Institute
for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute
for Medical Engineering and Science, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- School of
Biomolecular Science and Engineering, Vidyasirimedhi
Institute of Science and Technology (VISTEC), Rayong 21210, Thailand
| | - Henry Ko
- Koch Institute
for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute
for Medical Engineering and Science, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Tahoura Samad
- Koch Institute
for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute
for Medical Engineering and Science, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Novalia Pishesha
- Koch Institute
for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Division
of Immunology, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Hidde L. Ploegh
- Program in
Cellular and Molecular Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Sangeeta N. Bhatia
- Koch Institute
for Integrative Cancer Research, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Institute
for Medical Engineering and Science, Massachusetts
Institute of Technology, Cambridge, Massachusetts 02139, United States
- Howard
Hughes
Medical Institute, Cambridge, Massachusetts 02139, United States
- Department
of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department
of Medicine, Brigham and Women’s
Hospital and Harvard Medical School, Boston, Massachusetts 02115, United States
- Broad
Institute
of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
13
|
Noury H, Rahdar A, Romanholo Ferreira LF, Jamalpoor Z. AI-driven innovations in smart multifunctional nanocarriers for drug and gene delivery: A mini-review. Crit Rev Oncol Hematol 2025; 210:104701. [PMID: 40086770 DOI: 10.1016/j.critrevonc.2025.104701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/07/2025] [Accepted: 03/07/2025] [Indexed: 03/16/2025] Open
Abstract
The convergence of artificial intelligence (AI) and nanomedicine has revolutionized the design of smart multifunctional nanocarriers (SMNs) for drug and gene delivery, offering unprecedented precision, efficiency, and personalization in therapeutic applications. AI-driven approaches enhance the development of these nanocarriers by accelerating their design, optimizing drug loading and release kinetics, improving biocompatibility, and predicting interactions with biological barriers. This review explores the transformative role of AI in the fabrication and functionalization of SMNs, emphasizing its impact on overcoming challenges in targeted drug delivery, controlled release, and theranostics. We discuss the integration of AI with advanced nanomaterials-such as polymeric, lipidic, and inorganic nanoparticles-highlighting their potential in oncology and hematology. Furthermore, we examine recent clinical and preclinical case studies demonstrating AI-assisted nanocarrier development for personalized medicine. The synergy between AI and nanotechnology paves the way for next-generation precision therapeutics, addressing critical limitations in traditional drug delivery systems. However, data standardization, regulatory compliance, and translational scalability challenges remain. This review underscores the need for interdisciplinary collaboration to unlock AI's potential in nanomedicine fully, ultimately advancing the clinical application of SMNs for more effective and safer patient care.
Collapse
Affiliation(s)
- Hamid Noury
- Health Research Center, Chamran Hospital, Tehran, Iran
| | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol 538-98615, Iran.
| | | | - Zahra Jamalpoor
- Trauma and Surgery Research Center, Aja University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Hauck AV, Komforth P, Erlenbusch J, Stickdorn J, Radacki K, Braunschweig H, Besenius P, Van Herck S, Nuhn L. Aliphatic polycarbonates with acid degradable ketal side groups as multi-pH-responsive immunodrug nanocarriers. Biomater Sci 2025; 13:1414-1425. [PMID: 39575699 DOI: 10.1039/d4bm00949e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Pharmacokinetics and biodistribution profiles of active substances are crucial aspects for their safe and successful administration. Since many immunogenic compounds do not meet all requirements for safe and effective administration, well-defined drug nanocarrier systems are necessary with a stimuli-responsive drug-release profile. For this purpose, a novel pH-responsive aliphatic cyclic carbonate is introduced with benzyl ketal side chains and polymerized onto a poly(ethylene glycol) macroinitiator. The resulting block copolymers could be formulated via a solvent-evaporation method into well-defined polymeric micelles. The hydrophobic carbonate block was equipped with an acid degradable ketal side group that served as an acid-responsive functional group. Already subtle pH alternations led to micelle disassembly and the release of the active cargo. Furthermore, basic carbonate backbone degradation assured the pH responsiveness of the nanocarriers in both acidic and basic conditions. To investigate the delivery capacity of polymeric micelles, the model small molecule compound CL075, which serves as an immunotherapeutic TLR7/8 agonist, was encapsulated. Incubation studies with human blood plasma revealed the absence of undesirable protein adsorption on the drug-loaded nanoparticles. Furthermore, in vitro applications confirmed cell uptake of the nanodrug formulations by macrophages and the induction of payload-mediated immune stimulation. Altogether, these results underline the huge potential of the developed multi-pH-responsive polymeric nanocarrier for immunodrug delivery.
Collapse
Affiliation(s)
- Adrian V Hauck
- Chair of Macromolecular Chemistry, Institute of Functional Materials and Biofabrication, Julius-Maximilians-Universität Würzburg, 97070 Würzburg, Germany.
| | - Patric Komforth
- Max Planck Institute for Polymer Research, 55128 Mainz, Germany
| | - Jessica Erlenbusch
- Department of Chemistry, Johannes-Gutenberg-Universität Mainz, 55122 Mainz, Germany
| | | | - Krzysztof Radacki
- Institute for Sustainable Chemistry and Catalysis with Boron, Julius-Maximilians-Universität Würzburg, 97074 Würzburg, Germany
| | - Holger Braunschweig
- Institute for Sustainable Chemistry and Catalysis with Boron, Julius-Maximilians-Universität Würzburg, 97074 Würzburg, Germany
| | - Pol Besenius
- Department of Chemistry, Johannes-Gutenberg-Universität Mainz, 55122 Mainz, Germany
| | - Simon Van Herck
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, 14850, USA
| | - Lutz Nuhn
- Chair of Macromolecular Chemistry, Institute of Functional Materials and Biofabrication, Julius-Maximilians-Universität Würzburg, 97070 Würzburg, Germany.
| |
Collapse
|
15
|
Raoufi A, Soleimani Samarkhazan H, Nouri S, Khaksari MN, Abbasi Sourki P, Sargazi Aval O, Baradaran B, Aghaei M. Macrophages in graft-versus-host disease (GVHD): dual roles as therapeutic tools and targets. Clin Exp Med 2025; 25:73. [PMID: 40048037 PMCID: PMC11885342 DOI: 10.1007/s10238-025-01588-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/03/2025] [Indexed: 03/09/2025]
Abstract
Graft-versus-host disease remains one of the most formidable barriers to the complete success of hematopoietic stem cell transplantation that has emerged as the curative approach for many hematopoietic malignancies because it affects quality of life and overall survival. Macrophages are among the important members of the immune system, which perform dual roles in GVHD as both therapeutic tools and targets. This review epitomizes the multifunctional role of macrophages in the pathophysiology of both acute and chronic GVHD. Macrophages play an important role in the early phase of GVHD because of their recruitment and infiltration into target organs. Furthermore, they polarize into two functionally different phenotypes, including M1 and M2. In the case of acute GVHD, most macrophages express the M1 phenotype characterized by the production of pro-inflammatory cytokines that contribute to tissue damage. In contrast, in chronic GVHD, macrophages tend toward the M2 phenotype associated with the repair of tissues and fibrosis. A critical balance among these phenotypes is central to the course and severity of GVHD. Further interactions of macrophages with other lymphocytes such as T cells, B cells, and fibroblast further determine the course of GVHD. Macrophage interaction associated with alloreactive T cells promotes inflammation. This is therefore important in inducing injuries of tissues during acute GVHD. Interaction of macrophages, B cell, fibroblast, and CD4+ T cells promotes fibrosis during chronic GVHD and, hence, the subsequent dysfunction of organs. These are some insights, while several challenges remain. First, the impact of the dominant cytokines in GVHD on the polarization of macrophages is incompletely characterized and sometimes controversial. Second, the development of targeted therapies able to modulate macrophage function without systemic side effects remains an area of ongoing investigation. Future directions involve the exploration of macrophage-targeted therapies, including small molecules, antibodies, and nanotechnology, which modulate macrophage behavior and improve patient outcomes. This underlines the fact that a profound understanding of the dual role of macrophages in GVHD is essential for developing new and more effective therapeutic strategies. Targeting macrophages might represent one avenue for decreasing the incidence and severity of GVHD and improving the success and safety of HSCT.
Collapse
Affiliation(s)
- Atieh Raoufi
- Department of Immunology, Student Research Committee, School of Medicine, Zanjan University of Medical Science, Zanjan, Iran
| | - Hamed Soleimani Samarkhazan
- Student Research Committee, Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sina Nouri
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Mohammad Navid Khaksari
- Department of Hematology and Blood Banking, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvaneh Abbasi Sourki
- Department of Hematology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran
| | - Omolbanin Sargazi Aval
- Department of Hematology, Faculty of Allied Medical Sciences, Zabol University of Medical Sciences, Zabol, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Daneshghah Ave, Tabriz, Iran.
| | - Mojtaba Aghaei
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
16
|
Marvi PK, Das P, Jafari A, Hassan S, Savoji H, Srinivasan S, Rajabzadeh AR. Multifunctional Carbon Dots In Situ Confined Hydrogel for Optical Communication, Drug Delivery, pH Sensing, Nanozymatic Activity, and UV Shielding Applications. Adv Healthc Mater 2025; 14:e2403876. [PMID: 39757485 PMCID: PMC11874666 DOI: 10.1002/adhm.202403876] [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: 10/07/2024] [Revised: 11/29/2024] [Indexed: 01/07/2025]
Abstract
Inspired by the emerging potential of photoluminescent hydrogels, this work unlocks new avenues for advanced biosensing, bioimaging, and drug delivery applications. Carbon quantum dots (CDs) are deemed particularly promising among various optical dyes, for enhancing polymeric networks with superior physical and chemical properties. This study presents the synthesis of CDs derived from Prunella vulgaris, a natural plant resource, through a single-step hydrothermal process, followed by their uniform integration into hydrogel matrices via an in situ free radical graft polymerization. The resulting CD-integrated hydrogels exhibit multifunctionality in biomedical applications, featuring a diffusion-controlled drug release mechanism, permit concurrent delivery of photoluminescent CDs and therapeutic agents, enabling real-time monitoring over 32 h. In addition, these hydrogels function as a broad-range optical pH sensor (pH 3-11), provide robust ultraviolet (UV) shielding, and demonstrate nanozyme-like peroxidase activity. Critically, biocompatibility tests confirm their non-cytotoxicity toward fibroblast cells, establishing these hydrogels as promising candidates for diverse biomedical applications. These include advanced wound dressings that monitor the healing process and detect infection through pH sensing, and promote healing through the nanozymatic activity, all while maintaining a moist wound microenvironment. These hydrogels demonstrate exceptional suitability for advanced smart drug delivery, effective UV-blocking, and as innovative platforms for in vivo sensing and bioimaging.
Collapse
Affiliation(s)
- Parham Khoshbakht Marvi
- School of Biomedical EngineeringMcMaster University1280 Main Street West HamiltonHamiltonOntarioL8S 4L7Canada
| | - Poushali Das
- School of Biomedical EngineeringMcMaster University1280 Main Street West HamiltonHamiltonOntarioL8S 4L7Canada
| | - Arman Jafari
- Institute of Biomedical EngineeringDepartment of Pharmacology and PhysiologyFaculty of MedicineUniversity of MontrealMontrealQCH3T 1J4Canada
- Research CenterCentre Hospitalier Universitaire Sainte‐JustineMontrealQCH3T 1C5Canada
- Montreal TransMedTech InstituteMontrealQCH3T 1J4Canada
| | - Shiza Hassan
- School of Engineering Practice and TechnologyMcMaster University1280 Main Street West HamiltonHamiltonOntarioL8S 4L7Canada
| | - Houman Savoji
- Institute of Biomedical EngineeringDepartment of Pharmacology and PhysiologyFaculty of MedicineUniversity of MontrealMontrealQCH3T 1J4Canada
- Research CenterCentre Hospitalier Universitaire Sainte‐JustineMontrealQCH3T 1C5Canada
- Montreal TransMedTech InstituteMontrealQCH3T 1J4Canada
- Center for Applied Research On Polymers and Composites (CREPEC)MontrealQCH3A 0C3Canada
| | - Seshasai Srinivasan
- School of Biomedical EngineeringMcMaster University1280 Main Street West HamiltonHamiltonOntarioL8S 4L7Canada
- School of Engineering Practice and TechnologyMcMaster University1280 Main Street West HamiltonHamiltonOntarioL8S 4L7Canada
| | - Amin Reza Rajabzadeh
- School of Biomedical EngineeringMcMaster University1280 Main Street West HamiltonHamiltonOntarioL8S 4L7Canada
- School of Engineering Practice and TechnologyMcMaster University1280 Main Street West HamiltonHamiltonOntarioL8S 4L7Canada
| |
Collapse
|
17
|
Zheng K, Ouyang X, Xie H, Peng S. Responsive Zwitterionic Materials for Enhanced Drug Delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:3744-3756. [PMID: 39907524 DOI: 10.1021/acs.langmuir.4c04809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Zwitterionic materials have traditionally been recognized as exceptional antifouling agents, imparting nanocarriers with extended circulation times in vivo. Despite much studies on antifouling ability, the responsive zwitterionic materials that change physicochemical properties stimulated by mild signals are much less explored. As is known, there are multiple biological barriers in antitumor drug delivery, including the blood circulation barrier, non-specific organ distribution, elevated tumor interstitial pressure, tumor cytomembrane barrier, and lysosomal barrier. Multiple biological barriers restrict the delivery efficiency of nanocarriers to tumors, leading to a reduced therapeutic effect and increased side effects. Therefore, it is far from satisfactory to overcome the blood circulation barrier alone for classical zwitterionic antitumor materials. To address this challenge, recently developed responsive zwitterionic materials have been engineered to overcome multiple biological barriers, thereby enabling more effective antitumor drug delivery. Furthermore, responsive zwitterionic materials could respond to signals by themselves without the need of incorporating extra stimuli-responsive groups, which maintains the simplicity of the molecular structure. In this mini-review, the recent progress of antitumor zwitterionic materials responding to pH, temperature, enzyme, or reactive oxygen species is summarized. Furthermore, prospects and challenges of responsive zwitterionic materials are provided to promote better development of this field.
Collapse
Affiliation(s)
- Ke Zheng
- School of Materials Science and Engineering, Dongguan University of Technology, Dongguan, Guangdong 523808, China
| | - Xumei Ouyang
- Zhuhai People's Hospital (Zhuhai Clinical Medical College of Jinan University), Zhuhai, Guangdong 519000, China
| | - Hong Xie
- Department of Veterinary Medicine, Faculty of Animal Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Shaojun Peng
- Department of Biology, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, Guangdong 519087, China
| |
Collapse
|
18
|
Zhao S, Yu N, Han H, Guo S, Murthy N. Advances in acid-degradable and enzyme-cleavable linkers for drug delivery. Curr Opin Chem Biol 2025; 84:102552. [PMID: 39642424 PMCID: PMC11788058 DOI: 10.1016/j.cbpa.2024.102552] [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: 09/03/2024] [Revised: 10/23/2024] [Accepted: 11/13/2024] [Indexed: 12/08/2024]
Abstract
Drug delivery vectors have the potential to improve the efficacy of therapeutics, including small molecules and nucleic acid-based drugs. However, challenges remain in developing linkages that enable the precise and efficient release of therapeutic cargo in response to mildly acidic environments or lysosomal enzymes. This review highlights recent advances in acid-degradable acetal/ketal and enzyme-cleavable linkages for endolysosomal release. These innovations include the developments of azido-acetal linkers with improved stability and hydrolysis kinetics, organocatalytic trans-isopropenylation for synthesizing asymmetric ketals and their applications in drug delivery, and enzyme-cleavable linkers activated by cathepsin B or β-galactosidase.
Collapse
Affiliation(s)
- Sheng Zhao
- Department of Bioengineering and Innovative Genomics Institute, University of California Berkeley, 2151 Berkeley Way, Berkeley, CA 94720, USA
| | - Na Yu
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Hesong Han
- Department of Bioengineering and Innovative Genomics Institute, University of California Berkeley, 2151 Berkeley Way, Berkeley, CA 94720, USA
| | - Shutao Guo
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for New Organic Matter, College of Chemistry, Nankai University, Tianjin, 300071, China.
| | - Niren Murthy
- Department of Bioengineering and Innovative Genomics Institute, University of California Berkeley, 2151 Berkeley Way, Berkeley, CA 94720, USA.
| |
Collapse
|
19
|
Elsayed N, How CW, Foo JB. Development and characterization of pH-sensitive zerumbone-encapsulated liposomes for lung fibrosis via inhalation route. Eur J Pharm Biopharm 2025; 207:114599. [PMID: 39617356 DOI: 10.1016/j.ejpb.2024.114599] [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: 07/09/2024] [Revised: 10/30/2024] [Accepted: 11/29/2024] [Indexed: 01/28/2025]
Abstract
Zerumbone (ZER), a compound derived from the rhizome of Zingiber Zerumbet (L.) Smith, has demonstrated anti-inflammatory properties but suffers from poor water solubility, limiting its clinical application. While ZER's effects on lung inflammation are known, its role in lung fibrosis remains unexplored. Herein, ZER was encapsulated in pH-sensitive liposomes formulated with oleic acid, dipalmitoylphosphatidylcholine, and cholesterol to enhance ZER solubility and delivery to the acidic environment of lung fibrosis. The liposomes were optimized using Box-Behnken design, resulting in an average diameter of 87.8 ± 3.5 nm, a polydispersity index of 0.16 ± 0.2, and a zeta potential of -24 ± 0.32 mV. ZER release from the carrier followed zero-order kinetics and showed higher release in acidic settings. Cascade impactor and HPLC analyses confirmed that ZER liposome powder produced by freeze-drying reached stage 7, indicating effective delivery to deep lung regions. The uptake of ZER liposomes was concentration and pH-dependent, being higher in acidic conditions and greater in MRC-5 cells compared to A549 cells. Notably, ZER liposomes reduced cell migration and downregulated fibrotic markers such as fibronectin, MMP-2, and α-SMA in MRC-5 and A549 cells. This study suggests that ZER liposomes hold promise for treating lung fibrosis and merit further investigation.
Collapse
Affiliation(s)
- Nourhan Elsayed
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Chee Wun How
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Malaysia
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health & Medical Sciences, Taylor's University, Subang Jaya, Malaysia; Digital Health and Medical Advancements Impact Lab, Taylor's University, Subang Jaya, Malaysia.
| |
Collapse
|
20
|
Li K, Wang Z. Degradable Semi-Cycloaliphatic Epoxy Resin for Recyclable Carbon Fiber-Reinforced Composite Materials. Polymers (Basel) 2025; 17:293. [PMID: 39940497 PMCID: PMC11820351 DOI: 10.3390/polym17030293] [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: 12/13/2024] [Revised: 01/02/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
The development of an energy-saving method to recycle expensive carbon fibers (CFs) from end-of-life thermosetting resin-based CF-reinforced composites (CFRCs) is strongly desired because of the environmental and economic issues. The replacement of traditional thermosetting matrixes with controllably degradable epoxy resins provides a promising solution to this challenging task. In this work, a liquid acetal-containing semi-cycloaliphatic epoxy resin (H-ER) is designed and synthesized. After curing, H-ER shows simultaneously increased thermal stability, shearing strength, flexural strength, strain at break, and critical stress intensity factors by 126%, 26.5%, 17.0%, and 29.5%, respectively, in comparison with ERL-4221. Particularly, the cured H-ER is sufficiently resistant to organic solvents, bases, and weak acids but degrades rapidly in a modestly strong acidic aqueous solution, and the rate of degradation is controlled by modulating the acidity. GC-MS and FTIR spectra demonstrate that the degradation is indeed due to the cleavage of acetal linkages in the network, and the degradation-generated benzaldehyde may be reused as a raw material for the synthesis of the H-ER resin. More importantly, for the CFRCs using H-ER as a matrix, the CFs are readily recovered without detectable damage and are able to be recycled for CFRC fabrication.
Collapse
Affiliation(s)
| | - Zhonggang Wang
- Department of Polymer Science and Materials, School of Chemical Engineering, Dalian University of Technology, Dalian 116024, China
| |
Collapse
|
21
|
Mishra AP, Kumar R, Harilal S, Nigam M, Datta D, Singh S, Waranuch N, Chittasupho C. Demystifying the management of cancer through smart nano-biomedicine via regulation of reactive oxygen species. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:497-532. [PMID: 39480523 DOI: 10.1007/s00210-024-03469-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/17/2024] [Indexed: 11/02/2024]
Abstract
Advancements in therapeutic strategies and combinatorial approaches for cancer management have led to the majority of cancers in the initial stages to be regarded as treatable and curable. However, certain high-grade cancers in the initial stages are still regarded as chronic and difficult to manage, requiring novel therapeutic strategies. In this era of targeted and precision therapy, novel strategies for targeted delivery of drug and synergistic therapies, integrating nanotherapeutics, polymeric materials, and modulation of the tumor microenvironment are being developed. One such strategy is the study and utilization of smart-nano biomedicine, which refers to stimuli-responsive polymeric materials integrated with the anti-cancer drug that can modulate the reactive oxygen species (ROS) in the tumor microenvironment or can be ROS responsive for the mitigation as well as management of various cancers. The article explores in detail the ROS, its types, and sources; the antioxidant system, including scavengers and their role in cancer; the ROS-responsive targeted polymeric materials, including synergistic therapies for the treatment of cancer via modulating the ROS in the tumor microenvironment, involving therapeutic strategies promoting cancer cell death; and the current landscape and future prospects.
Collapse
Affiliation(s)
- Abhay Prakash Mishra
- Cosmetics and Natural Products Research Centre, Department of Pharmaceutical Technology, Naresuan University, Phitsanulok, 65000, Thailand
| | - Rajesh Kumar
- Faculty of Pharmaceutical Sciences, Kerala University of Health Sciences, Kerala, 680596, India.
| | - Seetha Harilal
- Faculty of Pharmaceutical Sciences, Kerala University of Health Sciences, Kerala, 680596, India
| | - Manisha Nigam
- Department of Biochemistry, Hemvati Nandan Bahuguna Garhwal University, Srinagar Garhwal, Uttarakhand, 246174, India
| | - Deepanjan Datta
- Department of Pharmaceutics, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal Karnataka, 576104, India
| | - Sudarshan Singh
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Neti Waranuch
- Cosmetics and Natural Products Research Centre, Department of Pharmaceutical Technology, Naresuan University, Phitsanulok, 65000, Thailand
| | - Chuda Chittasupho
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| |
Collapse
|
22
|
Banerjee A, Biswas M, Shukla P, Pathak RK. Emerging Roles of Gossypol in Therapy: Innovations in Prodrug Design and Nanoformulation Frontiers. ChemMedChem 2024; 19:e202400309. [PMID: 39136592 DOI: 10.1002/cmdc.202400309] [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: 04/28/2024] [Accepted: 07/31/2024] [Indexed: 10/08/2024]
Abstract
Stimuli activatable systems have the potential to deliver drugs to targeted areas by releasing therapeutic agents in response to diseased specific microenvironments such as the acidic environment commonly found in diseased tissues. This review article focuses on gossypol, a bioactive compound with inherent toxicity attributed to various factors, including the presence of its formyl groups. It highlights the potential of imine-linked gossypol-based prodrugs and nanoparticle formulations for targeted delivery and controlled release. The unique presence of polyphenolic cores on gossypol can be utilized to prepare nanoparticles. This review offers valuable insights into designing safer and more effective drug delivery systems by elucidating the masking effect and stimuli-responsive release mechanisms. Numerous examples demonstrate the conversion of formyl groups to imines, creating prodrugs that mask reactive functionalities and offer pH-responsive release. This insight can guide the design of combination therapeutics, where a second drug with an amine terminal group can form imine-linked prodrugs. Additionally, the second part discusses the use of polyphenolic moieties to create stable nanoparticles from infinite polymeric networks. Through a comprehensive examination of gossypol's properties and applications, this review emphasizes the broader implications of such a masking strategy for optimizing the therapeutic benefits of many similar bioactive compounds while minimizing adverse effects.
Collapse
Affiliation(s)
- Arka Banerjee
- Department of Chemical Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Transit Campus, Industrial Training Institute (ITI), Berhampur Engineering School Road, Berhampur, 760010, Odisha, India
| | - Megha Biswas
- Department of Chemical Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Transit Campus, Industrial Training Institute (ITI), Berhampur Engineering School Road, Berhampur, 760010, Odisha, India
| | - Prakash Shukla
- Department of Chemical Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Transit Campus, Industrial Training Institute (ITI), Berhampur Engineering School Road, Berhampur, 760010, Odisha, India
| | - Rakesh Kumar Pathak
- Department of Chemical Sciences, Indian Institute of Science Education and Research Berhampur (IISER Berhampur), Transit Campus, Industrial Training Institute (ITI), Berhampur Engineering School Road, Berhampur, 760010, Odisha, India
| |
Collapse
|
23
|
Zhao S, Gao K, Han H, Stenzel M, Yin B, Song H, Lawanprasert A, Nielsen JE, Sharma R, Arogundade OH, Pimcharoen S, Chen YJ, Paul A, Tuma J, Collins MG, Wyle Y, Cranick MG, Burgstone BW, Perez BS, Barron AE, Smith AM, Lee HY, Wang A, Murthy N. Acid-degradable lipid nanoparticles enhance the delivery of mRNA. NATURE NANOTECHNOLOGY 2024; 19:1702-1711. [PMID: 39179796 DOI: 10.1038/s41565-024-01765-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 07/19/2024] [Indexed: 08/26/2024]
Abstract
Lipid nanoparticle (LNP)-mRNA complexes are transforming medicine. However, the medical applications of LNPs are limited by their low endosomal disruption rates, high toxicity and long tissue persistence times. LNPs that rapidly hydrolyse in endosomes (RD-LNPs) could solve the problems limiting LNP-based therapeutics and dramatically expand their applications but have been challenging to synthesize. Here we present an acid-degradable linker termed 'azido-acetal' that hydrolyses in endosomes within minutes and enables the production of RD-LNPs. Acid-degradable lipids composed of polyethylene glycol lipids, anionic lipids and cationic lipids were synthesized with the azido-acetal linker and used to generate RD-LNPs, which significantly improved the performance of LNP-mRNA complexes in vitro and in vivo. Collectively, RD-LNPs delivered mRNA more efficiently to the liver, lung, spleen and brains of mice and to haematopoietic stem and progenitor cells in vitro than conventional LNPs. These experiments demonstrate that engineering LNP hydrolysis rates in vivo has great potential for expanding the medical applications of LNPs.
Collapse
Affiliation(s)
- Sheng Zhao
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Kewa Gao
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Hesong Han
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA.
| | - Michael Stenzel
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Boyan Yin
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Hengyue Song
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Atip Lawanprasert
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Josefine Eilsø Nielsen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
- Department of Bioengineering, School of Medicine, Stanford University, Stanford, CA, USA
| | - Rohit Sharma
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Opeyemi H Arogundade
- Department of Bioengineering and Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Sopida Pimcharoen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Yu-Ju Chen
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
| | - Abhik Paul
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jan Tuma
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Pathophysiology, Faculty of Medicine in Pilsen, Charles University, Plzen, Czech Republic
| | - Michael G Collins
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yofiel Wyle
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Matileen Grace Cranick
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA
| | - Benjamin W Burgstone
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Barbara S Perez
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA
| | - Annelise E Barron
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
| | - Andrew M Smith
- Department of Bioengineering and Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Hye Young Lee
- Department of Cellular and Integrative Physiology, University of Texas, Health Science Center at San Antonio, San Antonio, TX, USA
| | - Aijun Wang
- Department of Surgery, Department of Biomedical Engineering and Institute for Pediatric Regenerative Medicine/Shriners Children's, University of California, Davis, Sacramento, CA, USA.
| | - Niren Murthy
- Department of Bioengineering and Innovative Genomics Institute, University of California, Berkeley, CA, USA.
| |
Collapse
|
24
|
Yu S, Webber MJ. Engineering disease analyte response in peptide self-assembly. J Mater Chem B 2024; 12:10757-10769. [PMID: 39382032 DOI: 10.1039/d4tb01860e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
A need to enhance the precision and specificity of therapeutic nanocarriers inspires the development of advanced nanomaterials capable of sensing and responding to disease-related cues. Self-assembled peptides offer a promising nanocarrier platform with versatile use to create precisely defined nanoscale materials. Disease-relevant cues can range from large biomolecules, such as enzymes, to ubiquitous small molecules with varying concentrations in healthy versus diseased states. Notably, pH changes (i.e., H+ concentration), redox species (e.g., H2O2), and glucose levels are significant spatial and/or temporal indicators of therapeutic need. Self-assembled peptides respond to these cues by altering their solubility, modulating electrostatic interactions, or facilitating chemical transformations through dynamic or labile bonds. This review explores the design and construction of therapeutic nanocarriers using self-assembled peptides, focusing on how peptide sequence engineering along with the inclusion of non-peptidic components can link the assembly state of these nanocarriers to the presence of disease-relevant small molecules.
Collapse
Affiliation(s)
- Sihan Yu
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| | - Matthew J Webber
- Department of Chemical & Biomolecular Engineering, University of Notre Dame, Notre Dame, IN 46556, USA.
| |
Collapse
|
25
|
Santra S, Molla MR. Small molecule-based core and shell cross-linked nanoassemblies: from self-assembly and programmed disassembly to biological applications. Chem Commun (Camb) 2024; 60:12101-12117. [PMID: 39301871 DOI: 10.1039/d4cc03515a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Supramolecular assemblies of stimuli-responsive amphiphilic molecules have been of utmost interest in targeted drug delivery applications, owing to their capability of sequestering drug molecules in one set of conditions and releasing them in another. To minimize undesired disassembly and stabilize noncovalently encapsulated drug molecules, the strategy of core or shell cross-linking has become a fascinating approach to constructing cross-linked polymeric or small molecule-based nanoassemblies. In this article, we discuss the design and synthetic strategies for cross-linked nanoassemblies from small molecule-based amphiphiles, with robust stability and enhanced drug encapsulation capability. We highlight their potential biomedical applications, particularly in drug or gene delivery, and cell imaging. This feature article offers a comprehensive overview of the recent developments in the application of small molecule-based covalently cross-linked nanocarriers for materials and biomedical applications, which may inspire the use of these materials as a potential drug delivery system for future chemotherapeutic applications.
Collapse
Affiliation(s)
- Subrata Santra
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata-700009, India.
| | - Mijanur Rahaman Molla
- Department of Chemistry, University of Calcutta, 92 A. P. C. Road, Kolkata-700009, India.
| |
Collapse
|
26
|
Al-Shaeli M, Benkhaya S, Al-Juboori RA, Koyuncu I, Vatanpour V. pH-responsive membranes: Mechanisms, fabrications, and applications. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 946:173865. [PMID: 38880142 DOI: 10.1016/j.scitotenv.2024.173865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/31/2024] [Accepted: 06/07/2024] [Indexed: 06/18/2024]
Abstract
Understanding the mechanisms of pH-responsiveness allows researchers to design and fabricate membranes with specific functionalities for various applications. The pH-responsive membranes (PRMs) are particular categories of membranes that have an amazing aptitude to change their properties such as permeability, selectivity and surface charge in response to changes in pH levels. This review provides a brief introduction to mechanisms of pH-responsiveness in polymers and categorizes the applied polymers and functional groups. After that, different techniques for fabricating pH-responsive membranes such as grafting, the blending of pH-responsive polymers/microgels/nanomaterials, novel polymers and graphene-layered PRMs are discussed. The application of PRMs in different processes such as filtration membranes, reverse osmosis, drug delivery, gas separation, pervaporation and self-cleaning/antifouling properties with perspective to the challenges and future progress are reviewed. Lastly, the development and limitations of PRM fabrications and applications are compared to provide inclusive information for the advancement of next-generation PRMs with improved separation and filtration performance.
Collapse
Affiliation(s)
- Muayad Al-Shaeli
- Paul Wurth Chair, Faculty of Science, Technology and Medicine, University of Luxembourg, Avenue de l'Universit'e, L-4365 Esch-sur-Alzette, Luxembourg
| | - Said Benkhaya
- Department of Civil and Environmental Engineering, Shantou University, Shantou, Guangdong 515063, China
| | - Raed A Al-Juboori
- NYUAD Water Research Center, New York University Abu Dhabi, P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Ismail Koyuncu
- National Research Center on Membrane Technologies, Istanbul Technical University, Maslak, Turkey; Department of Environmental Engineering, Istanbul Technical University, Maslak, Istanbul 34469, Turkey
| | - Vahid Vatanpour
- Department of Environmental Engineering, Istanbul Technical University, Maslak, Istanbul 34469, Turkey; Department of Applied Chemistry, Faculty of Chemistry, Kharazmi University, 15719-14911 Tehran, Iran.
| |
Collapse
|
27
|
Verma VS, Pandey A, Jha AK, Badwaik HKR, Alexander A, Ajazuddin. Polyethylene Glycol-Based Polymer-Drug Conjugates: Novel Design and Synthesis Strategies for Enhanced Therapeutic Efficacy and Targeted Drug Delivery. Appl Biochem Biotechnol 2024; 196:7325-7361. [PMID: 38519751 DOI: 10.1007/s12010-024-04895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2024] [Indexed: 03/25/2024]
Abstract
Due to their potential to enhance therapeutic results and enable targeted drug administration, polymer-drug conjugates that use polyethylene glycol (PEG) as both the polymer and the linker for drug conjugation have attracted much research. This study seeks to investigate recent developments in the design and synthesis of PEG-based polymer-drug conjugates, emphasizing fresh ideas that fill in existing knowledge gaps and satisfy the increasing need for more potent drug delivery methods. Through an extensive review of the existing literature, this study identifies key challenges and proposes innovative strategies for future investigations. The paper presents a comprehensive framework for designing and synthesizing PEG-based polymer-drug conjugates, including rational molecular design, linker selection, conjugation methods, and characterization techniques. To further emphasize the importance and adaptability of PEG-based polymer-drug conjugates, prospective applications are highlighted, including cancer treatment, infectious disorders, and chronic ailments.
Collapse
Affiliation(s)
- Vinay Sagar Verma
- Faculty of Pharmaceutical Sciences, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai, Durg, Chhattisgarh, 490023, India
| | - Aakansha Pandey
- Faculty of Pharmaceutical Sciences, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India
| | - Arvind Kumar Jha
- Shri Shankaracharya Professional University, Junwani, Bhilai, 490020, Chhattisgarh, India
| | - Hemant Kumar Ramchandra Badwaik
- Shri Shankaracharya College of Pharmaceutical Sciences, Junwani, Bhilai, 490020, Chhattisgarh, India.
- Shri Shankaracharya Institute of Pharmaceutical Sciences and Research, Shri Shankaracharya Technical Campus, Junwani, Bhilai, 490020, Chhattisgarh, India.
| | - Amit Alexander
- Department of Pharmaceuticals, National Institute of Pharmaceutical Education and Research, Ministry of Chemical and Fertilizers, Guwahati, 781101, Assam, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Kohka, Bhilai, Durg, Chhattisgarh, 490023, India.
| |
Collapse
|
28
|
Mukkukada Ravi R, Mani A, Rahim S, Anirudhan TS. A Self-Skin Permeable Doxorubicin Loaded Nanogel Composite as a Transdermal Device for Breast Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:50407-50429. [PMID: 39259941 DOI: 10.1021/acsami.4c11373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Modern drug delivery research focuses on developing biodegradable nanopolymer systems. The present study proposed a polymer-based composite nanogel as a transdermal drug delivery system for the pH-responsive targeted and controlled delivery of anticancer drug doxorubicin (DOX). Nanogels have properties of both hydrogels and nanomaterials. The β-cyclodextrin-based nanogels can enhance the loading capacity of poorly soluble drugs and promote a sustained drug release. The β-cyclodextrin-grafted methacrylic acid conjugated hyaluronic acid composite nanogel was successfully synthesized. β-Cyclodextrin was first grafted onto methacrylic acid. The composite nanogel-based drug carrier was prepared by controlled radical polymerization (CRP) of β-cyclodextrin-grafted methacrylic acid with hyaluronic acid. The doxorubicin-loaded carrier was characterized by Fourier transform infrared (FTIR) spectroscopy, nuclear magnetic resonance (NMR) spectroscopy, ultraviolet-visible (UV-vis) spectroscopy, zeta potential analysis, dynamic light scattering (DLS), scanning electron microscopy (SEM), and transmission electron microscopy (TEM). The drug loading and release efficiencies were carried out at different pH levels. The maximum drug loading and encapsulation efficiencies of the synthesized final nanogel composite material at pH 8.0 were 86.44 ± 2.12 and 96.07 ± 2.01%, respectively. The DOX-loaded final material showed a 90.0 ± 2.6% release percentage of DOX at pH 5.5, whereas at pH 7.4, the release percentage of DOX was observed to be only 35.0 ± 0.3%. In vitro swelling, degradation, hemocompatibility, drug release kinetics, cytotoxicity, apoptosis, cell colocalization, skin irritation, and skin permeation studies, along with in vivo pharmacokinetic studies, were performed to prove the efficacy of the synthesized nanogel composite as a transdermal carrier for doxorubicin.
Collapse
Affiliation(s)
- Rajeev Mukkukada Ravi
- Department of Chemistry, School of Physical and Mathematical Sciences, Research Centre, University of Kerala, Kariavattom, Trivandrum 695 581, India
| | - Athira Mani
- Department of Chemistry, School of Physical and Mathematical Sciences, Research Centre, University of Kerala, Kariavattom, Trivandrum 695 581, India
| | - Suriya Rahim
- Department of Chemistry, School of Physical and Mathematical Sciences, Research Centre, University of Kerala, Kariavattom, Trivandrum 695 581, India
| | - Thayyath Sreenivasan Anirudhan
- Department of Chemistry, School of Physical and Mathematical Sciences, Research Centre, University of Kerala, Kariavattom, Trivandrum 695 581, India
| |
Collapse
|
29
|
Unagolla JM, Das S, Flanagan R, Oehler M, Menon JU. Targeting chronic liver diseases: Molecular markers, drug delivery strategies and future perspectives. Int J Pharm 2024; 660:124381. [PMID: 38917958 PMCID: PMC11246230 DOI: 10.1016/j.ijpharm.2024.124381] [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: 01/28/2024] [Revised: 06/10/2024] [Accepted: 06/22/2024] [Indexed: 06/27/2024]
Abstract
Chronic liver inflammation, a pervasive global health issue, results in millions of annual deaths due to its progression from fibrosis to the more severe forms of cirrhosis and hepatocellular carcinoma (HCC). This insidious condition stems from diverse factors such as obesity, genetic conditions, alcohol abuse, viral infections, autoimmune diseases, and toxic accumulation, manifesting as chronic liver diseases (CLDs) such as metabolic dysfunction-associated steatotic liver disease (MASLD), metabolic dysfunction-associated steatohepatitis (MASH), alcoholic liver disease (ALD), viral hepatitis, drug-induced liver injury, and autoimmune hepatitis. Late detection of CLDs necessitates effective treatments to inhibit and potentially reverse disease progression. However, current therapies exhibit limitations in consistency and safety. A potential breakthrough lies in nanoparticle-based drug delivery strategies, offering targeted delivery to specific liver cell types, such as hepatocytes, Kupffer cells, and hepatic stellate cells. This review explores molecular targets for CLD treatment, ongoing clinical trials, recent advances in nanoparticle-based drug delivery, and the future outlook of this research field. Early intervention is crucial for chronic liver disease. Having a comprehensive understanding of current treatments, molecular biomarkers and novel nanoparticle-based drug delivery strategies can have enormous impact in guiding future strategies for the prevention and treatment of CLDs.
Collapse
Affiliation(s)
- Janitha M Unagolla
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Subarna Das
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Riley Flanagan
- Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881, USA
| | - Marin Oehler
- Department of Biomedical Engineering, College of Engineering, University of Rhode Island, Kingston, RI 02881, USA
| | - Jyothi U Menon
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA; Department of Chemical Engineering, University of Rhode Island, Kingston, RI 02881, USA.
| |
Collapse
|
30
|
Atrooz OM, Reihani N, Mozafari MR, Salawi A, Taghavi E. Enhancing hair regeneration: Recent progress in tailoring nanostructured lipid carriers through surface modification strategies. ADMET AND DMPK 2024; 12:431-462. [PMID: 39091900 PMCID: PMC11289513 DOI: 10.5599/admet.2376] [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: 05/13/2024] [Revised: 07/17/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND AND PURPOSE Hair loss is a prevalent problem affecting millions of people worldwide, necessitating innovative and efficient regrowth approaches. Nanostructured lipid carriers (NLCs) have become a hopeful option for transporting bioactive substances to hair follicles because of their compatibility with the body and capability to improve drug absorption. REVIEW APPROACH Recently, surface modification techniques have been used to enhance hair regeneration by improving the customization of NLCs. These techniques involve applying polymers, incorporating targeting molecules, and modifying the surface charge. KEY RESULTS The conversation focuses on how these techniques enhance stability, compatibility with the body, and precise delivery to hair follicles within NLCs. Moreover, it explains how surface-modified NLCs can improve the bioavailability of hair growth-promoting agents like minoxidil and finasteride. Furthermore, information on how surface-modified NLCs interact with hair follicles is given, uncovering their possible uses in treating hair loss conditions. CONCLUSION This review discusses the potential of altering the surface of NLCs to customize them for enhanced hair growth. It offers important information for upcoming studies on hair growth.
Collapse
Affiliation(s)
- Omar M. Atrooz
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Al-Ahliyya Amman University, Amman 19328, Jordan
- Department of Biological Sciences, Mutah University, Mutah, Jordan
| | - Nasim Reihani
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, Victoria 3800, Australia
| | - M. R. Mozafari
- Australasian Nanoscience and Nanotechnology Initiative (ANNI), Monash University LPO, Clayton, Victoria 3800, Australia
| | - Ahmad Salawi
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, 45142, Saudi Arabia
| | - Elham Taghavi
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu, 21030 Kuala Nerus, Terengganu, Malaysia
| |
Collapse
|
31
|
Chen Y, Feng J, Chen Y, Xia C, Yao M, Ding W, Li X, Fu X, Zheng S, Ma Y, Zou J, Lan M, Gao F. ROS-responsive nano-medicine for navigating autophagy to enhance targeted therapy of inflammatory bowel disease. Int J Pharm 2024; 659:124117. [PMID: 38615805 DOI: 10.1016/j.ijpharm.2024.124117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Inflammatory bowel disease (IBD) is a chronic gastrointestinal disorder characterized by immune dysregulation and intestinal inflammation. Rapamycin (Ra), an mTORC1 pathway inhibitor, has shown promise for autophagy induction in IBD therapy but is associated with off-target effects and toxicity. To address these issues, we developed an oral liposome responsive to reactive oxygen species (ROS) using lipids and amphiphilic materials. We combined ketone thiol (TK) for ROS responsive and hyaluronic acid (HA) with high affinity for CD44 receptors to prepare rapamycin-loaded nanoparticle (Ra@TH). Owing to its ROS responsive characteristic, Ra@TH can achieve inflammatory colonic targeting. Additionally, Ra@TH can induce autophagy by inhibiting the mTORC1 pathway, leading to the clearance of damaged organelles, pathogenic microorganisms and oxidative stress products. Simultaneously, it also collaboratively inhibits the NF-κB pathway suppressed by the removal of ROS resulting from TK cleavage, thereby mediating the expression of inflammatory factors. Furthermore, Ra@TH enhances the expression of typical tight junction proteins, synergistically restoring intestinal barrier function. Our research not only expands the understanding of autophagy in IBD treatment but also introduces a promising therapeutic approach for IBD patients.
Collapse
Affiliation(s)
- You Chen
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Juewen Feng
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yang Chen
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Chuanhe Xia
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Min Yao
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Wenxing Ding
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xiang Li
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xiuzhi Fu
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Shulei Zheng
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yin Ma
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jiafeng Zou
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China; Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China
| | - Minbo Lan
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Feng Gao
- Shanghai Frontier Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, China; Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China; Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China; Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China; Engineering Research Center of Pharmaceutical Process Chemistry, Ministry of Education, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
32
|
Huang Y, Zhang Q, Lam CYK, Li C, Yang C, Zhong Z, Zhang R, Yan J, Chen J, Yin B, Wong SHD, Yang M. An Aggregation-Induced Emission-Based Dual Emitting Nanoprobe for Detecting Intracellular pH and Unravelling Metabolic Variations in Differentiating Lymphocytes. ACS NANO 2024; 18:15935-15949. [PMID: 38833531 DOI: 10.1021/acsnano.4c03796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Monitoring T lymphocyte differentiation is essential for understanding T cell fate regulation and advancing adoptive T cell immunotherapy. However, current biomarker analysis methods necessitate cell lysis, leading to source depletion. Intracellular pH (pHi) can be affected by the presence of lactic acid (LA), a metabolic mediator of T cell activity such as glycolysis during T cell activation; therefore, it is a potentially a good biomarker of T cell state. In this work, a dual emitting enhancement-based nanoprobe, namely, AIEgen@F127-AptCD8, was developed to accurately detect the pHi of T cells to "read" the T cell differentiation process. The nanocore of this probe comprises a pair of AIE dyes, TPE-AMC (pH-sensitive moiety) and TPE-TCF, that form a donor-acceptor pair for sensitive detection of pHi by dual emitting enhancement analysis. The nanoprobe exhibits a distinctly sensitive narrow range of pHi values (from 6.0 to 7.4) that can precisely distinguish the differentiated lymphocytes from naïve ones based on their distinct pHi profiles. Activated CD8+ T cells demonstrate lower pHi (6.49 ± 0.09) than the naïve cells (7.26 ± 0.11); Jurkat cells exhibit lower pHi (6.43 ± 0.06) compared to that of nonactivated ones (7.29 ± 0.09) on 7 days post-activation. The glycolytic product profiles in T cells strongly correlate with their pHi profiles, ascertaining the reliability of probing pHi for predicting T cell states. The specificity and dynamic detection capabilities of this nanoprobe make it a promising tool for indirectly and noninvasively monitoring T cell activation and differentiation states.
Collapse
Affiliation(s)
- Yingying Huang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Qin Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Ching Ying Katherine Lam
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Chuanqi Li
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Chen Yang
- Department of Applied Physics, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Zhiming Zhong
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Ruolin Zhang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Jiaxiang Yan
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Jiareng Chen
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Bohan Yin
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Siu Hong Dexter Wong
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237, China
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
- Research Institute for Sports Science and Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
| | - Mo Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
- Research Institute for Sports Science and Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong 999077, China
- The Hong Kong Polytechnic University Shenzhen Research Institute, Shenzhen 518000, China
| |
Collapse
|
33
|
Shano LB, Karthikeyan S, Kennedy LJ, Chinnathambi S, Pandian GN. MOFs for next-generation cancer therapeutics through a biophysical approach-a review. Front Bioeng Biotechnol 2024; 12:1397804. [PMID: 38938982 PMCID: PMC11208718 DOI: 10.3389/fbioe.2024.1397804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 05/20/2024] [Indexed: 06/29/2024] Open
Abstract
Metal-organic frameworks (MOFs) have emerged as promising nanocarriers for cancer treatment due to their unique properties. Featuring high porosity, extensive surface area, chemical stability, and good biocompatibility, MOFs are ideal for efficient drug delivery, targeted therapy, and controlled release. They can be designed to target specific cellular organelles to disrupt metabolic processes in cancer cells. Additionally, functionalization with enzymes mimics their catalytic activity, enhancing photodynamic therapy and overcoming apoptosis resistance in cancer cells. The controllable and regular structure of MOFs, along with their tumor microenvironment responsiveness, make them promising nanocarriers for anticancer drugs. These carriers can effectively deliver a wide range of drugs with improved bioavailability, controlled release rate, and targeted delivery efficiency compared to alternatives. In this article, we review both experimental and computational studies focusing on the interaction between MOFs and drug, explicating the release mechanisms and stability in physiological conditions. Notably, we explore the relationship between MOF structure and its ability to damage cancer cells, elucidating why MOFs are excellent candidates for bio-applicability. By understanding the problem and exploring potential solutions, this review provides insights into the future directions for harnessing the full potential of MOFs, ultimately leading to improved therapeutic outcomes in cancer treatment.
Collapse
Affiliation(s)
- Leon Bernet Shano
- Department of Physics, School of Advanced Sciences, Vellore Institute of Technology (VIT), Chennai, Tamil Nadu, India
| | - Subramani Karthikeyan
- Centre for Healthcare Advancement, Innovation and Research, Vellore Institute of Technology (VIT), Chennai, Tamil Nadu, India
| | - Lourdusamy John Kennedy
- Department of Physics, School of Advanced Sciences, Vellore Institute of Technology (VIT), Chennai, Tamil Nadu, India
| | - Shanmugavel Chinnathambi
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto, Japan
| | - Ganesh N. Pandian
- Institute for Integrated Cell-Material Sciences, Institute for Advanced Study, Kyoto University, Kyoto, Japan
| |
Collapse
|
34
|
Zhao H, Guo J, Huai J, Li R, Han H, Huang X, Jiang Y, Shuang S. A novel pH-sensitive hemi-cyanine containing tetrahydropyridine ring near-infrared fluorescence probe with lysosome-targeting ability. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 314:124162. [PMID: 38522377 DOI: 10.1016/j.saa.2024.124162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/23/2024] [Accepted: 03/13/2024] [Indexed: 03/26/2024]
Abstract
In recent years, hemi-cyanine dyes have been widely used as biological probes due to their red-light emission characteristics and high fluorescence quantum yield. In this study, we synthesized a novel hemi-cyanine dye containing a tetrahydropyridine ring. A lysosomal target was introduced into its structure to create a new pH-sensitive near-infrared fluorescent probe that successfully targeted lysosomes. The results showed that when the probe solution was excited at the absorption wavelength of 650 nm, its fluorescence emission wavelength was about 700 nm, and the peak intensity changed with different pH values in a wide range. Therefore, this probe enabled non-invasive detection of changes in the acidic environment of lysosomes in living organisms and showed good imaging capabilities. Moreover, the probe displays high sensitivity and good stability. The theoretical calculation of a probe structure has also been completed to discuss the relationship between structure and property.
Collapse
Affiliation(s)
- Hongwei Zhao
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China.
| | - Jingrong Guo
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Jiameng Huai
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Ruyue Li
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Hui Han
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Xiao Huang
- Shanxi Science and Technology Resources and Large-Scale Instrument Open-Sharing Center, Taiyuan, 03006, China
| | - Yuna Jiang
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| | - Shaomin Shuang
- School of Chemistry and Chemical Engineering, Institute of Environmental Science, Shanxi University, Taiyuan, 030006, China
| |
Collapse
|
35
|
Yang F, Gong S, Hu D, Chen L, Wang W, Cheng B, Yang J, Li B, Wang X. The biological response of pH-switch-based gold nanoparticle-composite polyamino acid embolic material. NANOSCALE 2024; 16:10448-10457. [PMID: 38752569 DOI: 10.1039/d4nr00989d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
With continuous advances in medical technology, non-invasive embolization has emerged as a minimally invasive treatment, offering new possibilities in cancer therapy. Fluorescent labeling can achieve visualization of therapeutic agents in vivo, providing technical support for precise treatment. This paper introduces a novel in situ non-invasive embolization composite material, Au NPs@(mPEG-PLGTs), created through the electrostatic combination of L-cysteine-modified gold nanoparticles (Au NPs) and methoxy polyethylene glycol amine-poly[(L-glutamic acid)-(L-tyrosine)] (mPEG-PLGTs). Experiments were undertaken to confirm the biocompatibility, degradability, stability and performance of this tumor therapy. The research results demonstrated a reduction in tumor size as early as the fifth day after the initial injection, with a significant 90% shrinkage in tumor volume observed after a 20-day treatment cycle, successfully inhibiting tumor growth and exhibiting excellent anti-tumor effects. Utilizing near-infrared in vivo imaging, Au NPs@(mPEG-PLGTs) displayed effective fluorescence tracking within the bodies of nude BALB-c mice. This study provides a novel direction for the further development and innovation of in situ non-invasive embolization in the field, highlighting its potential for rapid, significant therapeutic effects with minimal invasiveness and enhanced safety.
Collapse
Affiliation(s)
- Feng Yang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P.R.China.
- Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P.R.China
| | - Shiwen Gong
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P.R.China.
- Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P.R.China
| | - Die Hu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P.R.China.
- Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P.R.China
| | - Lihua Chen
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P.R.China.
- Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P.R.China
| | - Wenyuan Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P.R.China.
- Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P.R.China
| | - Bo Cheng
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan 430060, P.R.China
| | - Jing Yang
- School of Foreign Languages, Wuhan University of Technology, Wuhan 430070, P.R.China
| | - Binbin Li
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P.R.China.
- Hainan Institute, Wuhan University of Technology, Sanya 572000, P.R.China
- Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P.R.China
| | - Xinyu Wang
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P.R.China.
- Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu hydrogen Valley, Foshan 528200, P.R.China
- Hainan Institute, Wuhan University of Technology, Sanya 572000, P.R.China
- Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan 430070, P.R.China
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan 430060, P.R.China
| |
Collapse
|
36
|
Lin C, Chen L, He Y, Xiang W, Nie Y, Cai B, Guo Z. Injectable, self-healing and degradable dynamic hydrogels with tunable mechanical properties and stability by thermal-induced micellization. RSC Adv 2024; 14:16207-16217. [PMID: 38769971 PMCID: PMC11103349 DOI: 10.1039/d4ra02480j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/02/2024] [Indexed: 05/22/2024] Open
Abstract
Dynamic hydrogels possessing injectable, degradable and self-healing abilities have attracted considerable attention in the biomedical field in recent years, but it is difficult to tune the mechanical properties and stability of conventional dynamic hydrogels. In this work, we synthesized ABA-triblock copolymers via RAFT polymerization, where the A block consisted of thermo-sensitive poly(N-isopropylacrylamide-co-diacetone acrylamide) and the B block was hydrophilic poly(acrylamide). Subsequently, dynamic hydrogels were obtained based on the acylhydrazone bonds between the triblock copolymers and adipic acid dihydrazide (ADH). The obtained hydrogels exhibited injectable and self-healable abilities. In response to the thermal-induced micellization of their temperature-responsive blocks, the mechanical strength of the hydrogels not only increased, but also they exhibited high stability even at pH 2.0. Moreover, the hydrogel in the stable state could be degraded by the fracture of its trithiocarbonate groups. In addition, the hydrogels exhibited good cytocompatibility and controlled release behavior for doxorubicin (DOX). Considering these attractive tunable properties, these dynamic hydrogels show various potential applications in the biomedical field, such as drug carriers and cell or tissue engineering scaffolds.
Collapse
Affiliation(s)
- Chunqing Lin
- College of Chemistry, Chemical Engineering and Environmental Science, Minnan Normal University Zhangzhou 363000 PR China
| | - Leniu Chen
- College of Chemistry, Chemical Engineering and Environmental Science, Minnan Normal University Zhangzhou 363000 PR China
| | - Yuan He
- College of Chemistry, Chemical Engineering and Environmental Science, Minnan Normal University Zhangzhou 363000 PR China
| | - Wenlong Xiang
- College of Chemistry, Chemical Engineering and Environmental Science, Minnan Normal University Zhangzhou 363000 PR China
| | - Yujing Nie
- College of Chemistry, Chemical Engineering and Environmental Science, Minnan Normal University Zhangzhou 363000 PR China
| | - Baixue Cai
- Chongqing Academy of Metrology and Quality Inspection Chongqing 401120 PR China
| | - Zanru Guo
- College of Chemistry, Chemical Engineering and Environmental Science, Minnan Normal University Zhangzhou 363000 PR China
| |
Collapse
|
37
|
Lin W, Hu S, Li K, Shi Y, Pan C, Xu Z, Li D, Wang H, Li B, Chen H. Breaking Osteoclast-Acid Vicious Cycle to Rescue Osteoporosis via an Acid Responsive Organic Framework-Based Neutralizing and Gene Editing Platform. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307595. [PMID: 38126648 DOI: 10.1002/smll.202307595] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/14/2023] [Indexed: 12/23/2023]
Abstract
In the osteoporotic microenvironment, the acidic microenvironment generated by excessive osteoclasts not only causes irreversible bone mineral dissolution, but also promotes reactive oxygen species (ROS) production to induce osteoblast senescence and excessive receptor activator of nuclear factor kappa-B ligand (RANKL) production, which help to generate more osteoclasts. Hence, targeting the acidic microenvironment and RANKL production may break this vicious cycle to rescue osteoporosis. To achieve this, an acid-responsive and neutralizing system with high in vivo gene editing capacity is developed by loading sodium bicarbonate (NaHCO3) and RANKL-CRISPR/Cas9 (RC) plasmid in a metal-organic framework. This results showed ZIF8-NaHCO3@Cas9 (ZNC) effective neutralized acidic microenvironment and inhibited ROS production . Surprisingly, nanoparticles loaded with NaHCO3 and plasmids show higher transfection efficiency in the acidic environments as compared to the ones loaded with plasmid only. Finally, micro-CT proves complete reversal of bone volume in ovariectomized mice after ZNC injection into the bone remodeling site. Overall, the newly developed nanoparticles show strong effect in neutralizing the acidic microenvironment to achieve bone protection through promoting osteogenesis and inhibiting osteolysis in a bidirectional manner. This study provides new insights into the treatment of osteoporosis for biomedical and clinical therapies.
Collapse
Affiliation(s)
- Wenzheng Lin
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, 225009, P. R. China
- Jiangsu Key laboratory of integrated traditional Chinese and Western Medicine for prevention and treatment of Senile Diseases, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Sihan Hu
- Orthopedic Institute, Department of Orthopedic Surgery, First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215006, P. R. China
| | - Ke Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, 225009, P. R. China
| | - Yu Shi
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, 225009, P. R. China
| | - Chun Pan
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Zhuobin Xu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Dandan Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Huihui Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
- Jiangsu Key laboratory of integrated traditional Chinese and Western Medicine for prevention and treatment of Senile Diseases, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Bin Li
- Orthopedic Institute, Department of Orthopedic Surgery, First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, 215006, P. R. China
| | - Hao Chen
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, 225009, P. R. China
- Jiangsu Key laboratory of integrated traditional Chinese and Western Medicine for prevention and treatment of Senile Diseases, Yangzhou University, Yangzhou, 225001, P. R. China
| |
Collapse
|
38
|
Szulc A, Woźniak M. Targeting Pivotal Hallmarks of Cancer for Enhanced Therapeutic Strategies in Triple-Negative Breast Cancer Treatment-In Vitro, In Vivo and Clinical Trials Literature Review. Cancers (Basel) 2024; 16:1483. [PMID: 38672570 PMCID: PMC11047913 DOI: 10.3390/cancers16081483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 04/07/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
This literature review provides a comprehensive overview of triple-negative breast cancer (TNBC) and explores innovative targeted therapies focused on specific hallmarks of cancer cells, aiming to revolutionize breast cancer treatment. TNBC, characterized by its lack of expression of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), presents distinct features, categorizing these invasive breast tumors into various phenotypes delineated by key elements in molecular assays. This article delves into the latest advancements in therapeutic strategies targeting components of the tumor microenvironment and pivotal hallmarks of cancer: deregulating cellular metabolism and the Warburg effect, acidosis and hypoxia, the ability to metastasize and evade the immune system, aiming to enhance treatment efficacy while mitigating systemic toxicity. Insights from in vitro and in vivo studies and clinical trials underscore the promising effectiveness and elucidate the mechanisms of action of these novel therapeutic interventions for TNBC, particularly in cases refractory to conventional treatments. The integration of targeted therapies tailored to the molecular characteristics of TNBC holds significant potential for optimizing clinical outcomes and addressing the pressing need for more effective treatment options for this aggressive subtype of breast cancer.
Collapse
Affiliation(s)
| | - Marta Woźniak
- Department of Clinical and Experimental Pathology, Division of General and Experimental Pathology, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| |
Collapse
|
39
|
Liu J, Cabral H, Mi P. Nanocarriers address intracellular barriers for efficient drug delivery, overcoming drug resistance, subcellular targeting and controlled release. Adv Drug Deliv Rev 2024; 207:115239. [PMID: 38437916 DOI: 10.1016/j.addr.2024.115239] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/16/2024] [Accepted: 02/27/2024] [Indexed: 03/06/2024]
Abstract
The cellular barriers are major bottlenecks for bioactive compounds entering into cells to accomplish their biological functions, which limits their biomedical applications. Nanocarriers have demonstrated high potential and benefits for encapsulating bioactive compounds and efficiently delivering them into target cells by overcoming a cascade of intracellular barriers to achieve desirable therapeutic and diagnostic effects. In this review, we introduce the cellular barriers ahead of drug delivery and nanocarriers, as well as summarize recent advances and strategies of nanocarriers for increasing internalization with cells, promoting intracellular trafficking, overcoming drug resistance, targeting subcellular locations and controlled drug release. Lastly, the future perspectives of nanocarriers for intracellular drug delivery are discussed, which mainly focus on potential challenges and future directions. Our review presents an overview of intracellular drug delivery by nanocarriers, which may encourage the future development of nanocarriers for efficient and precision drug delivery into a wide range of cells and subcellular targets.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Peng Mi
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
40
|
Hunt NJ, Lockwood GP, Heffernan SJ, Daymond J, Ngu M, Narayanan RK, Westwood LJ, Mohanty B, Esser L, Williams CC, Kuncic Z, McCourt PAG, Le Couteur DG, Cogger VC. Oral nanotherapeutic formulation of insulin with reduced episodes of hypoglycaemia. NATURE NANOTECHNOLOGY 2024; 19:534-544. [PMID: 38168926 PMCID: PMC11026164 DOI: 10.1038/s41565-023-01565-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 11/02/2023] [Indexed: 01/05/2024]
Abstract
Injectable insulin is an extensively used medication with potential life-threatening hypoglycaemic events. Here we report on insulin-conjugated silver sulfide quantum dots coated with a chitosan/glucose polymer to produce a responsive oral insulin nanoformulation. This formulation is pH responsive, is insoluble in acidic environments and shows increased absorption in human duodenum explants and Caenorhabditis elegans at neutral pH. The formulation is sensitive to glucosidase enzymes to trigger insulin release. It is found that the formulation distributes to the liver in mice and rats after oral administration and promotes a dose-dependent reduction in blood glucose without promoting hypoglycaemia or weight gain in diabetic rodents. Non-diabetic baboons also show a dose-dependent reduction in blood glucose. No biochemical or haematological toxicity or adverse events were observed in mice, rats and non-human primates. The formulation demonstrates the potential to orally control blood glucose without hypoglycaemic episodes.
Collapse
Affiliation(s)
- Nicholas J Hunt
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia.
- Sydney Nano Institute, The University of Sydney, Camperdown, New South Wales, Australia.
- Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia.
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia.
| | - Glen P Lockwood
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia
| | - Scott J Heffernan
- Royal Prince Alfred Hospital, SLHD, Camperdown, New South Wales, Australia
| | - Jarryd Daymond
- Sydney Nano Institute, The University of Sydney, Camperdown, New South Wales, Australia
- Sydney Business School, The University of Sydney, Camperdown, New South Wales, Australia
| | - Meng Ngu
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia
- Department of Gastroenterology, Concord Repatriation General Hospital, SLHD, Concord, New South Wales, Australia
| | - Ramesh K Narayanan
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia
| | - Lara J Westwood
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Sydney Nano Institute, The University of Sydney, Camperdown, New South Wales, Australia
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia
| | - Biswaranjan Mohanty
- Sydney Analytical Core Research Facility, The University of Sydney, Camperdown, New South Wales, Australia
| | - Lars Esser
- CSIRO Manufacturing, Clayton, Victoria, Australia
| | | | - Zdenka Kuncic
- Sydney Nano Institute, The University of Sydney, Camperdown, New South Wales, Australia
- School of Physics, The University of Sydney, Camperdown, New South Wales, Australia
| | - Peter A G McCourt
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia
- Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - David G Le Couteur
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia
| | - Victoria C Cogger
- Faculty of Medicine and Health, The University of Sydney, Camperdown, New South Wales, Australia.
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney Local Health District (SLHD), Concord, New South Wales, Australia.
| |
Collapse
|
41
|
Soni S, Kori SK, Sahu P, Kashaw V, Dahiya R, Iyer AK, Soni V, Kashaw SK. Herbal nanogels: Revolutionizing skin cancer therapy through nanotechnology and natural remedies. EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY REPORTS 2024; 10:100126. [DOI: 10.1016/j.ejmcr.2023.100126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
42
|
Hu R, Lan J, Zhang D, Shen W. Nanotherapeutics for prostate cancer treatment: A comprehensive review. Biomaterials 2024; 305:122469. [PMID: 38244344 DOI: 10.1016/j.biomaterials.2024.122469] [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: 09/27/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/22/2024]
Abstract
Prostate cancer (PCa) is the most prevalent solid organ malignancy and seriously affects male health. The adverse effects of prostate cancer therapeutics can cause secondary damage to patients. Nanotherapeutics, which have special targeting abilities and controlled therapeutic release profiles, may serve as alternative agents for PCa treatment. At present, many nanotherapeutics have been developed to treat PCa and have shown better treatment effects in animals than traditional therapeutics. Although PCa nanotherapeutics are highly attractive, few successful cases have been reported in clinical practice. To help researchers design valuable nanotherapeutics for PCa treatment and avoid useless efforts, herein, we first reviewed the strategies and challenges involved in prostate cancer treatment. Subsequently, we presented a comprehensive review of nanotherapeutics for PCa treatment, including their targeting methods, controlled release strategies, therapeutic approaches and mechanisms. Finally, we proposed the future prospects of nanotherapeutics for PCa treatment.
Collapse
Affiliation(s)
- Ruimin Hu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jin Lan
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Dinglin Zhang
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Wenhao Shen
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
43
|
Chauhan A, Chaudhury S. Multivalent Salt-Induced Self-Assembly of Amphiphilic Polyelectrolytes of Different Charge Fractions: A Coarse-Grained Molecular Dynamics Simulation Study. J Phys Chem B 2024; 128:2037-2044. [PMID: 38359799 DOI: 10.1021/acs.jpcb.3c07886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Amphiphilic polymers with both hydrophobic and hydrophilic blocks are of great interest for their potential applications in drug delivery. Their self-assembly behavior in response to environmental factors like ion charge and multivalent salt concentration has been the subject of recent investigation. Our study utilizes coarse-grained molecular dynamics simulations to investigate the aggregation behavior of amphiphilic copolymers upon introducing tetravalent salt at varying charge fractions. We identify a critical concentration, Cs*, where the aggregation number reaches its maximum for each charge fraction, followed by a subsequent decrease at the excessive salt regime. This study reveals distinct morphological transitions in response to increasing salt concentration and decreasing charged fractions, namely, (i) stable dispersed micelles, (ii) a singular micelle comprising all copolymer chains, and (iii) redispersed micelles, particularly evident at lower charged fractions. Our study highlights the significant influence of tetravalent salt and charge fractions of polyelectrolyte chains on the self-assembly behavior of polyelectrolyte copolymers.
Collapse
Affiliation(s)
- Akshay Chauhan
- Department of Chemistry, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pune, Maharashtra 411008, India
| | - Srabanti Chaudhury
- Department of Chemistry, Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pune, Maharashtra 411008, India
| |
Collapse
|
44
|
Özcan Z, Hazar Yoruç AB. Vinorelbine-loaded multifunctional magnetic nanoparticles as anticancer drug delivery systems: synthesis, characterization, and in vitro release study. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:256-269. [PMID: 38440320 PMCID: PMC10910576 DOI: 10.3762/bjnano.15.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/16/2024] [Indexed: 03/06/2024]
Abstract
In this study, a multifunctional therapeutic agent combining chemotherapy and photothermal therapy on a single platform has been developed in the form of vinorelbine-loaded polydopamine-coated iron oxide nanoparticles. Vinorelbine (VNB) is loaded on the surface of iron oxide nanoparticles produced by a solvothermal technique after coating with polydopamine (PDA) with varying weight ratios as a result of dopamine polymerisation and covalent bonding of thiol-polyethylene glycol (SH-PEG). The VNB/PDA/Fe3O4 nanoparticles have a saturation magnetisation value of 60.40 emu/g in vibrating sample magnetometry, which proves their magnetisation. Vinorelbine, which is used as an effective cancer therapy agent, is included in the nanocomposite structure, and in vitro drug release studies under different pH conditions (pH 5.5 and 7.4) and photothermal activity at 808 nm NIR laser irradiation are investigated. The comprehensive integration of precise multifunctional nanoparticles design, magnetic response, and controlled drug release with photothermal effect brings a different perspective to advanced cancer treatment research.
Collapse
Affiliation(s)
- Zeynep Özcan
- Yildiz Technical University, Faculty of Chemistry and Metallurgy, Department of Metallurgical and Materials Engineering, 34210, Istanbul, Turkey
| | - Afife Binnaz Hazar Yoruç
- Yildiz Technical University, Faculty of Chemistry and Metallurgy, Department of Metallurgical and Materials Engineering, 34210, Istanbul, Turkey
| |
Collapse
|
45
|
Miller HA, Zhang Y, Smith BR, Frieboes HB. Anti-tumor effect of pH-sensitive drug-loaded nanoparticles optimized via an integrated computational/experimental approach. NANOSCALE 2024; 16:1999-2011. [PMID: 38193595 DOI: 10.1039/d3nr06414j] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
The acidic pH of tumor tissue has been used to trigger drug release from nanoparticles. However, dynamic interactions between tumor pH and vascularity present challenges to optimize therapy to particular microenvironment conditions. Despite recent development of pH-sensitive nanomaterials that can accurately quantify drug release from nanoparticles, tailoring release to maximize tumor response remains elusive. This study hypothesizes that a computational modeling-based platform that simulates the heterogeneously vascularized tumor microenvironment can enable evaluation of the complex intra-tumoral dynamics involving nanoparticle transport and pH-dependent drug release, and predict optimal nanoparticle parameters to maximize the response. To this end, SPNCD nanoparticles comprising superparamagnetic cores of iron oxide (Fe3O4) and a poly(lactide-co-glycolide acid) shell loaded with doxorubicin (DOX) were fabricated. Drug release was measured in vitro as a function of pH. A 2D model of vascularized tumor growth was calibrated to experimental data and used to evaluate SPNCD effect as a function of drug release rate and tissue vascular heterogeneity. Simulations show that pH-dependent drug release from SPNCD delays tumor regrowth more than DOX alone across all levels of vascular heterogeneity, and that SPNCD significantly inhibit tumor radius over time compared to systemic DOX. The minimum tumor radius forecast by the model was comparable to previous in vivo SPNCD inhibition data. Sensitivity analyses of the SPNCD pH-dependent drug release rate indicate that slower rates are more inhibitory than faster rates. We conclude that an integrated computational and experimental approach enables tailoring drug release by pH-responsive nanomaterials to maximize the tumor response.
Collapse
Affiliation(s)
- Hunter A Miller
- Department of Bioengineering, University of Louisville, Louisville, KY, USA.
| | - Yapei Zhang
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Bryan Ronain Smith
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Hermann B Frieboes
- Department of Bioengineering, University of Louisville, Louisville, KY, USA.
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, USA
- James Graham Brown Cancer Center, University of Louisville, Louisville, KY, USA
- Center for Predictive Medicine, University of Louisville, Louisville, KY, USA
| |
Collapse
|
46
|
Hughes KA, Misra B, Maghareh M, Samart P, Nguyen E, Hussain S, Geldenhuys WJ, Bobbala S. Flash nanoprecipitation allows easy fabrication of pH-responsive acetalated dextran nanoparticles for intracellular release of payloads. DISCOVER NANO 2024; 19:4. [PMID: 38175336 PMCID: PMC10766584 DOI: 10.1186/s11671-023-03947-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 12/21/2023] [Indexed: 01/05/2024]
Abstract
Acetalated dextran (Ac-Dex) nanoparticles are currently of immense interest due to their sharp pH-responsive nature and high biodegradability. Ac-Dex nanoparticles are often formulated through single- or double-emulsion methods utilizing polyvinyl alcohol as the stabilizer. The emulsion methods utilize toxic organic solvents such as dichloromethane or chloroform and require multi-step processing to form stable Ac-Dex nanoparticles. Here, we introduce a simple flash nanoprecipitation (FNP) approach that utilizes a confined impinging jet mixer and a non-toxic solvent, ethanol, to form Ac-Dex nanoparticles rapidly. Ac-Dex nanoparticles were stabilized using nonionic PEGylated surfactants, D-α-Tocopherol polyethylene glycol succinate (TPGS), or Pluronic (F-127). Ac-Dex nanoparticles formed using FNP were highly monodisperse and stably encapsulated a wide range of payloads, including hydrophobic, hydrophilic, and macromolecules. When lyophilized, Ac-Dex TPGS nanoparticles remained stable for at least one year with greater than 80% payload retention. Ac-Dex nanoparticles were non-toxic to cells and achieved intracellular release of payloads into the cytoplasm. In vivo studies demonstrated a predominant biodistribution of Ac-Dex TPGS nanoparticles in the liver, lungs, and spleen after intravenous administration. Taken together, the FNP technique allows easy fabrication and loading of Ac-Dex nanoparticles that can precisely release payloads into intracellular environments for diverse therapeutic applications. pH-responsive Acetalateddextran can be formulated using nonionic surfactants, such as TPGS or F-127, for intracellular release of payloads. Highly monodisperse and stable nanoparticles can be created through the simple, scalable flash nanoprecipitation technique, which utilizes a confined impingement jet mixer.
Collapse
Affiliation(s)
- Krystal A Hughes
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26505, USA
| | - Bishal Misra
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26505, USA
| | - Maryam Maghareh
- Department of Clinical Pharmacy, West Virginia University School of Pharmacy, Morgantown, WV, 26505, USA
| | - Parinya Samart
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26505, USA
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ethan Nguyen
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26505, USA
| | - Salik Hussain
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, Morgantown, WV, 26505, USA
- Department of Physiology, Pharmacology and Toxicology, West Virginia University, Morgantown, WV, 26505, USA
| | - Werner J Geldenhuys
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26505, USA
- Department of Neuroscience, West Virginia University School of Medicine, Morgantown, WV, 26505, USA
| | - Sharan Bobbala
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26505, USA.
| |
Collapse
|
47
|
Pandey P, Garg A, Singh V, Rai G, Mishra N. Clinical Trials and Future Prospects of Autophagy and ROS in Cancer. CANCER DRUG DISCOVERY AND DEVELOPMENT 2024:337-369. [DOI: 10.1007/978-3-031-66421-2_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
48
|
Mahajan K, Bhattacharya S. The Advancement and Obstacles in Improving the Stability of Nanocarriers for Precision Drug Delivery in the Field of Nanomedicine. Curr Top Med Chem 2024; 24:686-721. [PMID: 38409730 DOI: 10.2174/0115680266287101240214071718] [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: 10/13/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/28/2024]
Abstract
Nanocarriers have emerged as a promising class of nanoscale materials in the fields of drug delivery and biomedical applications. Their unique properties, such as high surface area- tovolume ratios and enhanced permeability and retention effects, enable targeted delivery of therapeutic agents to specific tissues or cells. However, the inherent instability of nanocarriers poses significant challenges to their successful application. This review highlights the importance of nanocarrier stability in biomedical applications and its impact on biocompatibility, targeted drug delivery, long shelf life, drug delivery performance, therapeutic efficacy, reduced side effects, prolonged circulation time, and targeted delivery. Enhancing nanocarrier stability requires careful design, engineering, and optimization of physical and chemical parameters. Various strategies and cutting-edge techniques employed to improve nanocarrier stability are explored, with a focus on their applications in drug delivery. By understanding the advances and challenges in nanocarrier stability, this review aims to contribute to the development and implementation of nanocarrier- based therapies in clinical settings, advancing the field of nanomedicine.
Collapse
Affiliation(s)
- Kalpesh Mahajan
- Department of Quality Assurence, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, India
| | - Sankha Bhattacharya
- Department of Pharmaceutics, School of Pharmacy and Technology Management, SVKMS NMIMS Maharashtra, Shirpur, 425405, India
| |
Collapse
|
49
|
Ghosh A, Himaja A, Biswas S, Kulkarni O, Ghosh B. Advances in the Delivery and Development of Epigenetic Therapeutics for the Treatment of Cancer. Mol Pharm 2023; 20:5981-6009. [PMID: 37899551 DOI: 10.1021/acs.molpharmaceut.3c00610] [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: 10/31/2023]
Abstract
Gene expression at the transcriptional level is altered by epigenetic modifications such as DNA methylation, histone methylation, and acetylation, which can upregulate, downregulate, or entirely silence genes. Pathological dysregulation of epigenetic processes can result in the development of cancer, neurological problems, metabolic disorders, and cardiovascular diseases. It is of promising therapeutic interest to find medications that target these epigenetic alterations. Despite the enormous amount of work that has been done in this area, very few molecules have been approved for clinical purposes. This article provides a comprehensive review of recent advances in epigenetic therapeutics for cancer, with a specific focus on emerging delivery and development strategies. Various delivery systems, including pro-drugs, conjugated molecules, nanoparticles (NPs), and liposomes, as well as remedial strategies such as combination therapies, and epigenetic editing, are being investigated to improve the efficacy and specificity of epigenetic drugs (epi-drugs). Furthermore, the challenges associated with available epi-drugs and the limitations of their translation into clinics have been discussed. Target selection, isoform selectivity, physiochemical properties of synthesized molecules, drug screening, and scalability of epi-drugs from preclinical to clinical fields are the major shortcomings that are addressed. This Review discusses novel strategies for the identification of new biomarkers, exploration of the medicinal chemistry of epigenetic modifiers, optimization of the dosage regimen, and design of proper clinical trials that will lead to better utilization of epigenetic modifiers over conventional therapies. The integration of these approaches holds great potential for improving the efficacy and precision of epigenetic treatments, ultimately benefiting cancer patients.
Collapse
Affiliation(s)
- Aparajita Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science- Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
- Pharmacology Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Ambati Himaja
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science- Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Swati Biswas
- Nanomedicine Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Onkar Kulkarni
- Pharmacology Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science-Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| | - Balaram Ghosh
- Epigenetic Research Laboratory, Department of Pharmacy, Birla Institute of Technology & Science- Pilani, Hyderabad Campus, Jawahar Nagar, Medchal, Hyderabad 500078, Telangana, India
| |
Collapse
|
50
|
Jia Y, Taledaohan A, Jia R, Wang X, Jia Y, Liu J, Wang Y. Chitosan nanomedicine containing RGD peptide and PAD4 inhibitor based on phenyl boronate coupling inhibition of primary tumor growth and lung metastasis. Biomed Pharmacother 2023; 168:115826. [PMID: 37931514 DOI: 10.1016/j.biopha.2023.115826] [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: 09/03/2023] [Revised: 10/25/2023] [Accepted: 10/31/2023] [Indexed: 11/08/2023] Open
Abstract
Stimulus-responsive nanodrugs have been extensively studied and their structural changes in the cells are important for controlled intracellular drug release. Histone citrullination of peptidylarginine deiminase 4 (PAD4) regulates the expression of tumor suppressor genes. In our previous study, compounds such as YW3-56 (356) were developed as potent PAD4 inhibitors with excellent anti-tumor activity in vitro and in vivo. To enhance the antitumor activity and improve the bioavailability, we further optimized the structure by modifying the phenylboronic acid moiety to the PAD4 inhibitor (4B). Taking advantage of the oxidative stress responsiveness of the phenylboronic acid moiety, in this study, we covalently attached 4B to RGD sequence peptide modified chitosan (K-CRGDV) to construct this new oxidative stress responsive nanodrug (K-CRGDV-4B). The modification of RGD sequence peptide conferred the nanodrug the ability to actively target tumors. The release mechanism was verified by UV-Vis spectroscopy, NMR. The anti-tumor and anti-metastatic properties of K-CRGDV-4B were demonstrated by in vitro cytotoxicity assay and in vivo mouse Lewis lung cancer metastasis model. In addition, K-CRGDV-4B modulates the ratio of immune cells in LLC tumor-bearing mice. Immunosuppressive proteins such as PD1 were inhibited, while IFN-γ and IFN-β, which are stimulators of tumor immune responses, were upregulated. Overall, K-CRGDV-4B is a stimulus-responsive nanodrug that responds to the tumor microenvironment by inhibiting PAD4 activity, blocking the formation of neutrophil extracellular traps (NETs), and improving the tumor immune microenvironment.
Collapse
Affiliation(s)
- Yijiang Jia
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences of Capital Medical University, 10 Xi Tou Tiao, You An Men, Beijing 100069, People's Republic of China; Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, People's Republic of China
| | - Ayijiang Taledaohan
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences of Capital Medical University, 10 Xi Tou Tiao, You An Men, Beijing 100069, People's Republic of China; Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, People's Republic of China
| | - Renbo Jia
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences of Capital Medical University, 10 Xi Tou Tiao, You An Men, Beijing 100069, People's Republic of China; Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, People's Republic of China
| | - Xin Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences of Capital Medical University, 10 Xi Tou Tiao, You An Men, Beijing 100069, People's Republic of China; Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, People's Republic of China
| | - Yunshu Jia
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences of Capital Medical University, 10 Xi Tou Tiao, You An Men, Beijing 100069, People's Republic of China; Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, People's Republic of China
| | - Jiawang Liu
- Medicinal Chemistry Core, Office of Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Yuji Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences of Capital Medical University, 10 Xi Tou Tiao, You An Men, Beijing 100069, People's Republic of China; Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, People's Republic of China.
| |
Collapse
|