1
|
Elkhashab M, Barreto G, Fauconnier M, Le Bourlout Y, Creemers LB, Nieminen HJ, Howard KA. ADAMTS5-specific gapmer release from an albumin biomolecular assembly and cartilage internalization triggered by ultrasound. Drug Deliv 2025; 32:2464921. [PMID: 39967271 PMCID: PMC11841101 DOI: 10.1080/10717544.2025.2464921] [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: 10/15/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVE Antisense oligonucleotides (ASOs) have reached the clinic; however, they lack tissue specificity. Albumin is a plasma-abundant macromolecule that has been shown to accumulate in inflamed tissues. In this work, we have designed a recombinant human albumin (rHA)-based biomolecular assembly incorporating a DNase-resistant phosphorothioate-based complementary oligonucleotide (cODN) and an anti-ADAMTS5 ASO for potential delivery to inflamed sites. Ultrasound (US) was used to trigger ASO release from the assembly and enhance internalization into articular cartilage. METHODS A phosphorothioate cODN was conjugated to rHA through a maleimide cross-linker after which, a therapeutic ADAMTS5-specific gapmer ASO was annealed to the cODN. ASO release was assessed after exposing the biomolecular assembly to different US conditions using an US-actuated medical needle operating at 32.2 kHz. Gene silencing efficiency of US-treated anti-ADAMTS5 ASO was assessed in human primary chondrocytes isolated from osteoarthritic patients. US-mediated ASO penetration into articular cartilage was assessed on ex vivo bovine articular cartilage. RESULTS ASO release was observed after exposure to US waves in continuous mode conditions that did not compromise ASO gene silencing efficiency in human chondrocytes. Furthermore, US increased ASO internalization into bovine articular cartilage after 30 min of application without detrimental effects on chondrocyte viability. CONCLUSION A medical needle driven by continuous US waves at 32.2 kHz has the capability of disassembling a duplex oligonucleotide and enhancing released ASOs internalization into articular cartilage. This work offers the potential delivery and the local triggered release of ASOs at the surface of articular cartilage providing potential benefits for the treatment of diverse cartilage pathologies.
Collapse
Affiliation(s)
- Marwa Elkhashab
- Interdisciplinary Nanoscience Center (iNANO) and Department of Molecular Biology, Aarhus University, Aarhus C, Denmark
| | - Goncalo Barreto
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
- Orton Orthopedic Hospital, Helsinki, Finland
| | - Maxime Fauconnier
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Yohann Le Bourlout
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Laura B. Creemers
- Department of Orthopedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Heikki J. Nieminen
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, Finland
| | - Kenneth A. Howard
- Interdisciplinary Nanoscience Center (iNANO) and Department of Molecular Biology, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
2
|
Wang H, Ren G, Xu Y, Deng R, Wang R, Zhou L. Novel erbium complex with anticancer activity against radiation resistant lung adenocarcinoma cells. J Inorg Biochem 2025; 269:112902. [PMID: 40132280 DOI: 10.1016/j.jinorgbio.2025.112902] [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: 07/20/2024] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
In this work, novel erbium complex with anticancer activity against radiation resistant lung adenocarcinoma cells was obtained and demonstrated. Firstly, stronger inhibitory effect of Er3+ on non-small cell lung cancer (NSCLC) cells and NSCLC- radiation resistant (RR) cells was experimentally confirmed. Then, by selecting highly biocompatible porphyrins as ligands, a novel erbium complex tetraphenylporphyrin erbium acetylacetonate (Er(acac)TPP) was synthesized and purified. Compared with Cisplatin, notably, Er(acac)TPP exhibits relatively higher inhibitory efficiency on NSCLC-RR cells. Moreover, the toxicities of Er(acac)TPP to normal cells are much lower than that of cancer cells. Subsequently, cell expansion, increased apoptosis, a decline in mitochondrial membrane potential (MMP), an accumulation of intracellular reactive oxygen species (ROS), increased Caspase-9 protein level and G2/M arrest were seen. These data all pointed to Er(acac)TPP as a possible candidate for more research and development as a chemotherapeutic drug.
Collapse
Affiliation(s)
- Hao Wang
- School of Materials Science and Engineering, Jilin Jianzhu University, Changchun 130118, China
| | - Guozhu Ren
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Yue Xu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Ruiping Deng
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Rui Wang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| | - Liang Zhou
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China.
| |
Collapse
|
3
|
Dar AI, Randhawa S, Verma M, Saini TC, Acharya A. Debugging the dynamics of protein corona: Formation, composition, challenges, and applications at the nano-bio interface. Adv Colloid Interface Sci 2025; 342:103535. [PMID: 40319752 DOI: 10.1016/j.cis.2025.103535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/07/2025]
Abstract
The intricate interplay between nanomaterials and the biological molecules has garnered considerable interest in understanding the dynamics of protein corona formation at the nano-bio interface. This review provides an in-depth exploration of protein-nanoparticle interactions, elucidating their structural dynamics and thermodynamics at the nano-Bio interface and further on emphasizing its formation, composition, challenges, and applications in the biomedical and nanotechnological domains, such as drug delivery, theranostics, and the translational medicine. We delve the nuanced mechanisms governing protein corona formation on nanoparticle surfaces, highlighting the influence of nanoparticle and biological factors, and review the impact of corona formation on the biological identity and functionality of nanoparticles. Notably, emerging applications of artificial intelligence and machine learning have begun to revolutionize this field, enabling accurate prediction of corona composition and related biological outcomes. These tools not only enhance efficiency over traditional experimental methods but also help decode complex protein-nanoparticle interaction patterns, offering new insights into corona-driven cellular responses and disease diagnostics. Additionally, it discusses recent advancements in the field of protein corona, particularly in translational nanomedicine and associated applications entailing current and future strategies which are aimed at mitigating the adverse effects of protein-nanoparticle interactions at the biological interface, for tailoring the protein coronas by engineering of the nanomaterials. This comprehensive assessment from chemical, technological, and biological aspects serves as a guiding beacon for the development of future nanomedicine, enabling the more effective emulation of the biological milieu and the design of protein-NP systems for enhanced biomedical applications.
Collapse
Affiliation(s)
- Aqib Iqbal Dar
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, HP 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shiwani Randhawa
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, HP 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mohini Verma
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, HP 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Trilok Chand Saini
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, HP 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Amitabha Acharya
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, HP 176061, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
4
|
Li Y, Du B, Yu L, Luo H, Rong H, Gao X, Yin J. Strategies and challenges of cytosolic delivery of proteins. J Drug Target 2025; 33:837-852. [PMID: 39862226 DOI: 10.1080/1061186x.2025.2458616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/11/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
The cytosolic delivery of therapeutic proteins represents a promising strategy for addressing diseases caused by protein dysfunction. Despite significant advances, efficient delivery remains challenging due to barriers such as cell membrane impermeability, endosomal sequestration and protein instability. This review summarises recent progress in protein delivery systems, including physical, chemical and biological approaches, with a particular focus on strategies that enhance endosomal escape and targeting specificity. We further discuss the clinical translatability of these approaches and propose future directions for improving delivery efficiency and safety, ultimately unlocking the therapeutic potential of intracellular proteins.
Collapse
Affiliation(s)
- Yuanyuan Li
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Baojie Du
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Lichao Yu
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Hong Luo
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haibo Rong
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Nanjing Medical University Affiliated Cancer Hospital, Nanjing, China
| | - Xiangdong Gao
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Yin
- School of Life Science and Technology, Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
5
|
Summer M, Hussain T, Ali S, Khan RRM, Muhammad G, Liaqat I. Exploring the underlying modes of organic nanoparticles in diagnosis, prevention, and treatment of cancer: a review from drug delivery to toxicity. INT J POLYM MATER PO 2025; 74:829-845. [DOI: 10.1080/00914037.2024.2375337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/28/2024] [Indexed: 08/04/2024]
Affiliation(s)
- Muhammad Summer
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, GC University Lahore
| | - Tauqeer Hussain
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, GC University Lahore
| | - Shaukat Ali
- Medical Toxicology and Biochemistry Laboratory, Department of Zoology, GC University Lahore
| | - Rana Rashad Mahmood Khan
- Department of Chemistry, Government College University Lahore, Faculty of Chemistry and Life Sciences
| | - Gulzar Muhammad
- Department of Chemistry, Government College University Lahore, Faculty of Chemistry and Life Sciences
| | - Iram Liaqat
- Microbiology Lab, Department of Zoology, Government College University Lahore
| |
Collapse
|
6
|
Abdullah KM, Sharma G, Singh AP, Siddiqui JA. Nanomedicine in Cancer Therapeutics: Current Perspectives from Bench to Bedside. Mol Cancer 2025; 24:169. [PMID: 40490771 DOI: 10.1186/s12943-025-02368-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Accepted: 05/26/2025] [Indexed: 06/11/2025] Open
Abstract
Cancer is among the leading causes of death worldwide, with projections indicating that it will claim 35 million lives by the year 2050. Conventional therapies, such as chemotherapy and immune modulation, have reduced cancer mortality to some extent; however, they have limited efficacy due to their broad mode of action, often resulting in cytotoxic effects on normal cells along with the malignant tissues, ultimately limiting their overall optimal therapeutic efficacy outcomes.Rapid advances in nanotechnology and an evolving understanding of cancer mechanisms have propelled the development of a diverse array of nanocarriers to vanquish the hurdles in achieving sophisticated drug delivery with reduced off-target toxicity. Nanoformulations can deliver the anti-cancer agents precisely to the tumor cell by integrating a multitarget approach that allows for tissue-, cell-, or organelle-specific delivery and internalization. Despite the immense interest and unmatched advancements in modern oncology equipped with nanomedicines, only a few nanoformulations have successfully translated into clinical settings. A major reason behind this shortcoming is the lack of a rationale design incorporating smart, responsive targeting features, leading to a compromised therapeutic window due to inefficient internalization or erroneous intracellular localization with unsuccessful payload release. This review aims to summarize the recent perspective of nanomedicine and its translation to clinical practice, with a particular focus on the evolution of strategies used in tumor targeting from traditional EPR-based passive mechanisms to advanced active and multi-stage approaches. We highlight the coupling of organelle-specific and stimuli-responsive nanocarriers, discuss the potential of biomimetic and cell-mediated delivery systems, and also shed light on technologies such as microfluidics, tumor-on-chip models, and AI-assisted synthesis. Finally, this review explores translational hurdles ranging from biological and manufacturing challenges to regulatory bottlenecks and outlines how innovative modeling systems and engineering solutions can bridge the gap from bench to bedside in cancer nanotherapeutics.
Collapse
Affiliation(s)
- K M Abdullah
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Gunjan Sharma
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Ajay P Singh
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, 39216, USA
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Jawed A Siddiqui
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, 39216, USA.
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, 39216, USA.
| |
Collapse
|
7
|
Koyra N, Yu H, Drummond CJ, Zhai J, Dyett B. Recent developments with pH-responsive lyotropic liquid crystalline lipid nanoparticles for targeted bioactive agent delivery. Expert Opin Drug Deliv 2025. [PMID: 40491048 DOI: 10.1080/17425247.2025.2518225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/30/2025] [Accepted: 06/06/2025] [Indexed: 06/11/2025]
Abstract
INTRODUCTION Lyotropic liquid crystalline lipid nanoparticles (LNPs) are a platform technology with broad-ranging potential in bioactive agent delivery applications. Their biomimetic properties impart the capacity to encapsulate large biomolecules and to overcome traditional biological barriers. AREAS COVERED The properties of lyotropic liquid crystalline LNPs can vary significantly between phases. We briefly introduce key concepts related to their formation and self-assembly and how ionization at the lipid-water interface, i.e. pH-responsiveness, can be leveraged to alter the properties of the nanoparticles. In this review, we summarize recent advances mainly from the past five years that highlight the role and impact of incorporating ionizable lipids, copolymers, and drug molecules in pH-responsive nanocarriers for the delivery of bioactive agents. EXPERT OPINION The development of pH-responsive lipid nanoparticles (pR_LNPs) is at the forefront of the new wave of mRNA therapeutics. The complexity of the biological journey faced by the nanoparticle and the broad spectrum of disease targets is sparking a surge in research activity. The accelerating development of new ionizable lipid materials to enhance mRNA delivery potential may benefit from closer consideration - or in tandem development - of self-assembly, interface ionization, and artificial intelligence integration.
Collapse
Affiliation(s)
- Natinael Koyra
- School of Science, STEM College, RMIT University, Melbourne, Victoria, Australia
| | - Haitao Yu
- School of Science, STEM College, RMIT University, Melbourne, Victoria, Australia
| | - Calum J Drummond
- School of Science, STEM College, RMIT University, Melbourne, Victoria, Australia
| | - Jiali Zhai
- School of Science, STEM College, RMIT University, Melbourne, Victoria, Australia
| | - Brendan Dyett
- School of Science, STEM College, RMIT University, Melbourne, Victoria, Australia
| |
Collapse
|
8
|
Lozano-García M, Dikici E, Bilbao D, Mohan P, Deo S, Daunert S. Multifunctional delivery strategies and nanoplatforms of SN-38 in cancer therapeutics. J Control Release 2025; 384:113937. [PMID: 40490199 DOI: 10.1016/j.jconrel.2025.113937] [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: 02/28/2025] [Revised: 06/03/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
SN-38 or 7-ethyl-10-hydroxycamptothecin is the active metabolite of irinotecan, a widely used chemotherapeutic agent for the treatment of colorectal, pancreatic, lung, breast, gastric, esophageal, hepatocellular, ovarian, brain, leukemia, and lymphoma malignancies. SN-38's antitumoral effect is 100 to 1000 times more potent than that of irinotecan. However, its clinical application is hindered by its poor solubility and chemical instability. To circumvent these challenges and avoid systemic toxicities, such as myelosuppression and diarrhea, several SN-38 delivery systems have been explored. In that regard, formulations based on targeted, controlled and tumor-responsive release of SN-38 have demonstrated to enhance its antitumoral effects and reduce the associated systemic toxicities by limiting the pharmacological activity to the desired tumor location. To this end, prodrugs, conjugates, nanoparticles, dendrimers, or lipid-based strategies for SN-38 delivery have been used. Most recently, multifunctional approaches have emerged as an attractive alternative to develop SN-38 delivery systems, combining several strategies in a single formulation, i.e., encapsulating nanocarriers, tumor-targeting ligands, stimuli-responsive elements, optimal linkers, drug combinations or bioimaging agents. Despite their therapeutic advantages, multifunctional delivery systems often face challenges concerning their clinical translation compared to conventional therapies, such as biocompatibility, scalability and cost-effectiveness issues. The aim of this work is to review the most recent progress that has been made in the development and assessment of multifunctional delivery systems for cancer treatment.
Collapse
Affiliation(s)
- Mercedes Lozano-García
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami, Miami, FL 33136, USA
| | - Emre Dikici
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami, Miami, FL 33136, USA
| | - Daniel Bilbao
- Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Prasoon Mohan
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; Vascular and Interventional Radiology, University of Miami Health System, Miami, FL 33136, USA
| | - Sapna Deo
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
| | - Sylvia Daunert
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Dr. John T. Macdonald Foundation Biomedical Nanotechnology Institute (BioNIUM), University of Miami, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA; Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA.
| |
Collapse
|
9
|
Sharma R, Modi U, Kumar R, Sharma C, Srivastav AK, Bhatia D, Solanki R. Bio-inspired, programmable biomacromolecules based nanostructures driven cancer therapy. BIOMATERIALS ADVANCES 2025; 171:214235. [PMID: 39978287 DOI: 10.1016/j.bioadv.2025.214235] [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: 12/10/2024] [Revised: 01/28/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
Cancer remains a significant global health challenge, driving the development of advanced platforms for highly targeted and efficient drug delivery. Early-stage nanocarriers, such as synthetic polymeric and inorganic materials, face limitations in biocompatibility and biodegradability. In contrast, bioinspired nanocarriers derived from natural biomacromolecules mimic biological processes and present a promising alternative due to their biocompatibility, biodegradability and non-toxicity. The effectiveness of these drug delivery systems is influenced by factors such as size, shape, surface properties, morphology, functionalization, and preparation methods. Various biomacromolecule-inspired nanocarriers such as protein-based, lipid-based, carbohydrate-based and nucleic acid-based are now at the forefront of research. This review highlights the properties, advantages and limitations of different bioinspired materials. We also explore cutting-edge approaches for cancer therapy using these nanocarriers with recent in-vitro, in-vivo and patent evidence. Finally, we address the challenges and potential solutions associated with bioinspired nanocarriers, proposing future directions. Overall, this review explores nature-inspired drug delivery systems that have paved the way for advancements in cancer therapy.
Collapse
Affiliation(s)
- Rahul Sharma
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Unnati Modi
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | - Rahul Kumar
- Dr. B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Chirag Sharma
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India
| | | | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| | - Raghu Solanki
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382355, India.
| |
Collapse
|
10
|
Munusamy MA, Bharathi M, Alarfaj AA, Hussein-Al-Ali SH, Nagaiya R, Subbarayan S. Glutaraldehyde-crosslinked Naringenin-loaded Albumin Nanoparticles (GNANPs) induce antimicrobial properties and apoptosis in gastric cancer cells. Toxicol In Vitro 2025; 106:106037. [PMID: 40037499 DOI: 10.1016/j.tiv.2025.106037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 02/08/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
An assessment of the anticancer activity of Glutaraldehyde-crosslinked Naringenin-loaded Albumin Nanoparticles (GNANPs) against gastric cancer cells was the purpose of this study. The increasing prevalence of gastric cancer and the limitations of conventional therapies necessitate novel approaches that combine targeted drug delivery with therapeutic efficacy. Several techniques were used to characterize the synthesized GNANPs, including UV-visible spectroscopy, X-ray diffractometer (XRD), scanning electron microscope (SEM), transmission electron microscope (TEM), Fourier transform infrared (FT-IR), dynamic light scattering (DLS), and photoluminescence (PL). They were evaluated for their antimicrobial properties, cytotoxicity, ROS accumulation, apoptotic activity, and oxidative stress markers against AGS cells. The characterization analyses indicated the existence of Glutaraldehyde-crosslinked Naringenin-loaded Albumin Nanoparticles with an oval-shaped morphology and an average particle size of 127.80 nm. The existence of several elements and functional groups in the GNANPs was also detected using EDX and FT-IR analyses, respectively. The synthesized GNANPs have shown exceptional antibacterial activities by effectively inhibiting the growth of several infections. The treatment of GNANPs efficiently inhibited the growth of AGS cells. Fluorescence staining studies showed increased apoptosis and oxidative stress markers in AGS cells treated with synthesized Glutaraldehyde-crosslinked Naringenin-loaded Albumin Nanoparticles, indicating their potential as a viable cancer treatment option.
Collapse
Affiliation(s)
- Murugan Alwarkurichi Munusamy
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Chennai, Tamil Nadu 602 105, India
| | - Muruganantham Bharathi
- Centre for Bioinformatics, Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore-641021
| | - Abdullah A Alarfaj
- Department of Botany and Microbiology, College of Science, King Saud University, P. O. Box.2455, Riyadh 11451, Saudi Arabia
| | | | - Ravichandran Nagaiya
- Department of Life Science and Environmental Biochemistry, Pusan National University, Busan 46241, Republic of Korea
| | - Sarathbabu Subbarayan
- Muthayammal Centre for Advanced Research, Muthayammal College of Arts and Science, Rasipuram, Namakkal, Tamil Nadu 637408, India.
| |
Collapse
|
11
|
Zhang W, Wang F, Wang H, Xu T, Su H, Cui H. Balancing Chemical and Supramolecular Stability in OEGylated Supramolecular Polymers for Systemic Drug Delivery. J Am Chem Soc 2025; 147:17985-17993. [PMID: 40375654 DOI: 10.1021/jacs.5c03253] [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: 05/18/2025]
Abstract
The chemical conjugation of poly(ethylene glycol) (PEG) to therapeutic agents, known as PEGylation, is a well-established strategy for enhancing drug solubility, chemical stability, and pharmacokinetics. Here, we report on a class of supramolecular polymeric prodrugs by utilizing oligo(ethylene glycol) (OEG) to modify the hydrophobic anticancer drug camptothecin (CPT). These OEGylated prodrugs, despite their low molecular weight, spontaneously self-assemble into therapeutic supramolecular polymers (SPs) with a tubular morphology, featuring a dense OEG coating on the surface. By designing biodegradable linkers with varying chemical stabilities, we investigated how the release kinetics of CPT influence the in vitro and in vivo performance of these SPs. Our findings demonstrate that self-assembling prodrugs (SAPDs) with a self-immolative disulfanyl-ethyl carbonate (etcSS) linker exhibit a faster drug release rate than those with a reducible disulfanyl butyrate (buSS) linker, leading to higher potency and significantly improved antitumor efficacy. Notably, two stable tubular SPs, Tubustecan (TT) 1E and TT 7E, outperformed irinotecan─a clinically approved CPT prodrug─in a colon cancer model, achieving enhanced tumor growth inhibition and prolonged animal survival. These results highlight the potential of supramolecular OEGylation as an important strategy for engineering drug-based supramolecular polymers and underscore the critical role of chemical stability vs supramolecular stability in optimizing supramolecular prodrug design.
Collapse
Affiliation(s)
- Weijie Zhang
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Feihu Wang
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Han Wang
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Tian Xu
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Hao Su
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering and Institute for NanoBioTechnology, The Johns Hopkins University, 3400 North Charles Street, Baltimore, Maryland 21218, United States
- Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
12
|
Sarvepalli S, Vadarevu S. Role of artificial intelligence in cancer drug discovery and development. Cancer Lett 2025; 627:217821. [PMID: 40414522 DOI: 10.1016/j.canlet.2025.217821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/17/2025] [Accepted: 05/23/2025] [Indexed: 05/27/2025]
Abstract
The role of artificial intelligence (AI) in cancer drug discovery and development has garnered significant attention due to its potential to transform the traditionally time-consuming and expensive processes involved in bringing new therapies to market. AI technologies, such as machine learning (ML) and deep learning (DL), enable the efficient analysis of vast datasets, facilitate faster identification of drug targets, optimization of compounds, and prediction of clinical outcomes. This review explores the multifaceted applications of AI across various stages of cancer drug development, from early-stage discovery to clinical trial design, development. In early-stage discovery, AI-driven methods support target identification, virtual screening (VS), and molecular docking, offering precise predictions that streamline the identification of promising compounds. Additionally, AI is instrumental in de novo drug design and lead optimization, where algorithms can generate novel molecular structures and optimize their properties to enhance drug efficacy and safety profiles. Preclinical development benefits from AI's predictive modeling capabilities, particularly in assessing a drug's toxicity through in silico simulations. AI also plays a pivotal role in biomarker discovery, enabling the identification of specific molecular signatures that can inform patient stratification and personalized treatment approaches. In clinical development, AI optimizes trial design by leveraging real-world data (RWD), improving patient selection, and reducing the time required to bring new drugs to market. Despite its transformative potential, challenges remain, including issues related to data quality, model interpretability, and regulatory hurdles. Addressing these limitations is critical for fully realizing AI's potential in cancer drug discovery and development. As AI continues to evolve, its integration with other technologies, such as genomics and clustered regularly interspaced short palindromic repeats (CRISPR), holds promise for advancing personalized cancer therapies. This review provides a comprehensive overview of AI's impact on the cancer drug discovery and development and highlights future directions for this rapidly evolving field.
Collapse
Affiliation(s)
- Sruthi Sarvepalli
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA.
| | | |
Collapse
|
13
|
Zhou XH, Wang JW, You W, Gao F, Wang Z, Gao HJ, Shen AZ, Ou YH, Zhan X, Nie X, Tang LQ, You YZ. Targeting phosphatidylserine in tumor cell membranes with a zinc-containing molecule to efficiently combat tumor metastasis. J Nanobiotechnology 2025; 23:363. [PMID: 40394583 PMCID: PMC12090580 DOI: 10.1186/s12951-025-03418-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/27/2025] [Indexed: 05/22/2025] Open
Abstract
Metal drugs, such as platinum drugs, are widely used in tumor treatment. However, most traditional tumor treatments face the risk of failure due to the ineffective control over drug resistance and tumor metastasis. Targeting the cell membrane and disrupting its function to combat drug resistance and metastasis is a promising strategy. Nevertheless, membranolytic drugs always cause significant cytotoxicity. In this study, we developed a zinc-containing molecule to selectively kill tumor cells by targeting phosphatidylserine in the tumor cell membrane, which is commonly distributed in the outer cell membrane of tumor cells. Herein, a structurally optimized amphiphilic zinc-containing molecule, 2aZn, was developed by screening the appropriate hydrophobic tail and linker. This functional molecule can disrupt the tumor cell membrane to kill various types of tumor cells with minimal damage to normal tissue. After repeated stimulation, no obvious drug resistance was observed. Importantly, 2aZn could successfully combat tumor metastasis by destroying the cell membrane and reducing the capacity of cells to invade. As a result, zinc-containing molecules have the potential to overcome drug resistance and tumor metastasis in the treatment of tumors, providing a new perspective for the design of effective antitumour medications.
Collapse
Affiliation(s)
- Xiao-Hong Zhou
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Key Laboratory of Precision and Intelligent Chemistry and Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230026, China
| | - Jia-Wei Wang
- Center for Reproduction and Genetics, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Wei You
- Key Laboratory of Precision and Intelligent Chemistry and Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230026, China
| | - Fan Gao
- Key Laboratory of Precision and Intelligent Chemistry and Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230026, China
| | - Zhe Wang
- Key Laboratory of Precision and Intelligent Chemistry and Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230026, China
| | - Hong-Jie Gao
- Key Laboratory of Precision and Intelligent Chemistry and Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230026, China
| | - Ai-Zong Shen
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, Anhui, 230001, China
| | - Yang-Huan Ou
- Department of Vascular Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China.
| | - Xiang Zhan
- Department of Gastroenterology, The Second Affiliated Hospital of Anhui Province, Hefei, Anhui, 230601, China.
| | - Xuan Nie
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, Anhui, 230001, China.
| | - Li-Qin Tang
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Precision Pharmaceutical Preparations and Clinical Pharmacy, Hefei, Anhui, 230001, China.
| | - Ye-Zi You
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Key Laboratory of Precision and Intelligent Chemistry and Hefei National Laboratory for Physical Sciences at Microscale, University of Science and Technology of China, Hefei, 230026, China.
| |
Collapse
|
14
|
Alkhamach D, Khan SA, Greish K, Hassan HAFM, Haider M. Nanostructured lipid carriers in cancer therapy: Advances in passive and active targeting strategies. Int J Pharm 2025; 678:125736. [PMID: 40389069 DOI: 10.1016/j.ijpharm.2025.125736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 05/05/2025] [Accepted: 05/15/2025] [Indexed: 05/21/2025]
Abstract
Nanostructured lipid carriers (NLCs) have emerged as a promising drug delivery platform in cancer therapy, offering advantages such as enhanced drug solubility, stability, and controlled release. Recent efforts have focused on utilizing NLCs for passive and active tumor targeting to improve therapeutic outcomes. This review provides a comprehensive analysis of the role of NLCs in cancer therapy, with particular emphasis on their application in passive and active targeting strategies for precision oncology. Relevant studies were selected from recent literature, focusing on NLC formulation, targeting approaches, and therapeutic applications. NLCs enhance tumor-specific drug delivery through passive targeting via the enhanced permeability and retention (EPR) effect and active targeting via ligand-mediated mechanisms. Lymphatic-targeting NLCs enable improved drug delivery to metastatic niches, while stimuli-responsive NLCs facilitate site-specific release under tumor-associated conditions (e.g., pH, enzymatic activity, redox gradients). Advances in lipid composition, surfactant systems, and conjugation strategies significantly influence drug loading (DL), biodistribution, therapeutic efficacy, and clinical translation across various malignancies. NLCs represent a versatile and adaptable platform for precision cancer therapy. Continued optimization of formulation parameters, functionalization strategies, and clinical translation pathways is essential to fully realize their potential in targeted oncology applications.
Collapse
Affiliation(s)
- Dana Alkhamach
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah 27272 Sharjah, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah 27272 Sharjah, United Arab Emirates
| | - Saeed Ahmad Khan
- Research Institute of Medical & Health Sciences, University of Sharjah 27272 Sharjah, United Arab Emirates
| | - Khaled Greish
- Department of Molecular Medicine, Princess Al-Jawhara Centre for Molecular Medicine, School of Medicine and Health Sciences Arabian Gulf University, Manama 328329, Bahrain
| | - Hatem A F M Hassan
- Medway School of Pharmacy, University of Kent, Chatham Maritime, Kent ME4 4TB, UK
| | - Mohamed Haider
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah 27272 Sharjah, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah 27272 Sharjah, United Arab Emirates.
| |
Collapse
|
15
|
Zhuang R, Chang X, Sha J, Yu Z, Shi E, Lu M, Liu J, Zhang G, Zhou D, Li L. Optoelectronic-Coupled-Driven Microrobot for Biological Cargo Transport in Conductive Isosmotic Glucose Solution. ACS APPLIED MATERIALS & INTERFACES 2025; 17:28425-28435. [PMID: 40299716 DOI: 10.1021/acsami.5c06042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
Electric field-driven micro/nanorobots, as micro/nanodevices with autonomous motion capability, have emerged as promising candidates for targeted cargo delivery in biomedical applications due to their advantages of label-free operation, selectivity, and controllability. In biological systems, many biological cargos need to be operated in conductive isosmotic solutions to ensure their viability. However, in the conductive solution, electric field-driven micro/nanorobots exhibit significantly reduced propulsion performance, despite retaining the capability to manipulate cargos by the dielectrophoretic force. This limitation restricts the wider applicability of electric field-driven micro/nanorobots in biomedical fields. This paper presents a novel optoelectronic-coupled-driven α-Fe2O3@aTiO2/Au microrobot, which exhibits significantly improved mobility and enables biological cargo transportation in the conductive isosmotic glucose solution. Benefiting from the flowerlike surface structure and composite photocatalytic material, the proposed microrobot exhibits enhanced photocatalytic capability, enabling efficient propulsion in glucose solution under light irradiation. In addition, the motion behavior of the microrobot under light, electric, and optoelectronic-coupled fields is investigated. It is found that the speed of the microrobot could exceed 300 μm/s under coupled fields, which is more than ten times faster than that of previously reported electric field-driven micro/nanorobots. Due to the magnetic property, the proposed microrobot can be precisely navigated under the guidance of an external uniform magnetic field. Furthermore, the proposed microrobot can achieve the transportation of various biological cargos in a conductive isosmotic glucose solution. The proposed microrobot opens a new avenue for targeted delivery and holds great potential for applications in the biological and pharmaceutical fields.
Collapse
Affiliation(s)
- Rencheng Zhuang
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
- Chongqing Research Institute, Harbin Institute of Technology, Chongqing 400722, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Xiaocong Chang
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
- Chongqing Research Institute, Harbin Institute of Technology, Chongqing 400722, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Jinrui Sha
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
- Chongqing Research Institute, Harbin Institute of Technology, Chongqing 400722, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Zehao Yu
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
- Chongqing Research Institute, Harbin Institute of Technology, Chongqing 400722, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Enbo Shi
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
- Chongqing Research Institute, Harbin Institute of Technology, Chongqing 400722, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Minqiao Lu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Junmin Liu
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
- Chongqing Research Institute, Harbin Institute of Technology, Chongqing 400722, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Guangyu Zhang
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Dekai Zhou
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
- Chongqing Research Institute, Harbin Institute of Technology, Chongqing 400722, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| | - Longqiu Li
- State Key Laboratory of Robotics and System, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
- Zhengzhou Research Institute, Harbin Institute of Technology, Zhengzhou 450000, China
| |
Collapse
|
16
|
Li Z, Xiao C, Yang X, Li Z. Progress in the mechanical properties of nanoparticles for tumor-targeting delivery. Chem Soc Rev 2025. [PMID: 40341776 DOI: 10.1039/d3cs00912b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Cancer nanomedicines have attracted significant attention in the past several decades, and the physicochemical properties, such as the size, shape, composition, surface charge, hydrophobicity, and mechanical properties, of nanoparticles have been optimized for potent cancer therapy. Since publishing our 2020 tutorial review "Influence of nanomedicine mechanical properties on tumor targeting delivery" in Chemical Society Reviews, substantial advancements have been made in understanding the role of mechanical properties in cancer nanomedicine. Notably, in vivo transport processes that are dependent on the mechanical properties of nanomedicine, including long circulation, tumor accumulation, and deep penetration, have been extensively studied using various nano-drug delivery systems. These studies have demonstrated that leveraging these mechanical properties can significantly enhance the antitumor efficacy of nanomedicine. In this review, we categorize the advancements in the mechanical properties of cancer nanomedicine into three distinct themes: the interactions between nanoparticles with varied mechanical properties and cells (2002 - present), the impact of these properties on in vivo delivery processes (2007 - present), and the strategic use of mechanical properties to boost cancer therapy (2023 - present). We analyze how different mechanical properties of organic, inorganic, hybrid, and biological nanoparticles affect their delivery processes at the macroscopic level, i.e., in tissues, organs and cells. At the microscopic level, their biological and physical interactions with biological barriers, physiological structures, cell membranes, organelles, and other structures reveal the potential mechanism of nanoparticles' mechanical properties in determining their antitumor efficacy. Furthermore, we address the current challenges and future prospects in the mechanical properties of cancer nanomedicine, as well as the clinical translation potential of nanoparticles with diverse mechanical characteristics.
Collapse
Affiliation(s)
- Zheng Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
| | - Chen Xiao
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
| | - Xiangliang Yang
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| | - Zifu Li
- Department of Nanomedicine and Biopharmaceuticals, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China.
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Key Laboratory of Molecular Biophysics of Ministry of Education, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medical, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan, 430074, P. R. China
| |
Collapse
|
17
|
Jaiswal N, Mahata N, Chanda N. Nanogold-albumin conjugates: transformative approaches for next-generation cancer therapy and diagnostics. NANOSCALE 2025; 17:11191-11220. [PMID: 40237258 DOI: 10.1039/d4nr05279j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Nanogold-albumin conjugates have garnered significant attention as a highly adaptable theranostic platform, capable of delivering a wide range of therapeutics, from small-molecule drugs to larger biomolecules, while offering promising applications for monitoring and managing cancer. The remarkable theranostic capabilities of these conjugates stem from the combined strengths of gold and albumin, which provide low toxicity, a large surface area, customizable surface chemistry, and unique optical properties, all contributing to their potential in cancer therapy. This review delves into the design and development of two primary types of nanogold-albumin conjugate: supramolecular albumin-coated gold nanoparticles (GNP-BSA/HSA) and albumin-templated ultra-small gold nanoclusters (GNC-BSA/HSA). Each strategy offers distinct advantages, enabling the fine-tuning of conjugate properties to optimize therapeutic delivery and facilitate cancer-specific bio-sensing. The integration of gold and albumin further improves biocompatibility, extends circulation time, and enhances tumor targeting, making these conjugates an attractive option for cancer treatment. The review also focuses on the refinement of surface chemistry to achieve precise targeting of cancer cells, as well as the challenges and future prospects for advancing nanogold-albumin systems in clinical applications.
Collapse
Affiliation(s)
- Namita Jaiswal
- Human Centered Robotics and Cybernetics Group, CSIR-Central Mechanical Engineering Research Institute, Durgapur, India.
- Department of Biotechnology, National Institute of Technology (NIT), Durgapur, India
| | - Nibedita Mahata
- Department of Biotechnology, National Institute of Technology (NIT), Durgapur, India
| | - Nripen Chanda
- Human Centered Robotics and Cybernetics Group, CSIR-Central Mechanical Engineering Research Institute, Durgapur, India.
| |
Collapse
|
18
|
Wang R, Wang K, Li Z, Long H, Zhang D, Li Y, Xia Z, Guo X, Chen W, Cao F, Jiang F. Blood-Brain Barrier-Permeable, Reactive Oxygen Species-Producing, and Mitochondria-Targeting Nanosystem Amplifies Glioblastoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27434-27447. [PMID: 40289326 DOI: 10.1021/acsami.5c02384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Gemcitabine (GTB), a clinically approved nucleoside analogue for cancer treatment, faces therapeutic limitations due to rapid enzymatic deactivation by cytidine deaminase (CDA) in tumor microenvironments. Over 90% of systemically administered GTB undergoes catalytic conversion to inactive 2'-deoxy-2',2'-difluorouracil metabolites through CDA-mediated deamination. To address this pharmacological challenge, we developed a multifunctional codelivery nanosystem through strategic engineering of reactive oxygen species (ROS)-generating, mitochondria-targeting CPUL1-TPP (CT) nanoaggregates. These self-assembling CT/GTB complexes were further optimized with DSPE-MPEG2k (DP) and Angiopep-2-conjugated DSPE-MPEG2k (Ang-DP) to create blood-brain barrier (BBB)-penetrating Ang-DP@CT/GTB nanoparticles, enhancing both physiological stability and low-density lipoprotein receptor-related protein 1 (LRP1)-mediated glioma targeting. Comparative analyses revealed that Ang-DP@CT/GTB nanoparticles significantly enhanced GTB's antiglioblastoma efficacy compared to free drug administration in both in vitro and in vivo models. Mechanistic investigations demonstrated that the nanosystem upregulates heme oxygenase-1 (HO-1), subsequently downregulating CDA expression to mitigate GTB metabolism. This coordinated molecular modulation prolongs GTB's therapeutic activity while leveraging the ROS-generating capacity of CT components for synergistic tumor suppression. The BBB-permeable codelivery platform exemplifies a rational design paradigm for multifunctional carrier-free pure nanodrugs (PNDs), demonstrating how clinical drug reformulation can overcome inherent pharmacokinetic limitations. This nanotechnology-driven approach provides critical insights for optimizing chemotherapeutic performance through metabolic pathway regulation and targeted delivery engineering.
Collapse
Affiliation(s)
- Rong Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Ke Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Zhuolin Li
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Haoping Long
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Dongyu Zhang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Yanting Li
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Zhuolu Xia
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Xindong Guo
- Beijing Laboratory of Biomedical Materials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Wei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Feng Cao
- Department of Pharmaceutical, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, P. R. China
| | - Feng Jiang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, P. R. China
| |
Collapse
|
19
|
Khoury R, Longobardi G, Barnatan TT, Venkert D, García Alvarado A, Yona A, Green Buzhor M, Shahar S, Wang Q, Acúrcio RC, Guedes RC, Florindo HF, Zhao JJ, Satchi-Fainaro R. Radiation-guided nanoparticles enhance the efficacy of PARP inhibitors in primary and metastatic BRCA1-deficient tumors via immunotherapy. J Control Release 2025; 383:113812. [PMID: 40319918 DOI: 10.1016/j.jconrel.2025.113812] [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: 12/17/2024] [Revised: 04/06/2025] [Accepted: 05/01/2025] [Indexed: 05/07/2025]
Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPi) have revolutionized the treatment landscape for patients suffering from BRCA1-mutated breast and ovarian cancers. However, responses are not durable. We demonstrate that treatment with PARPi, niraparib, increases programmed death-ligand 1 (PD-L1) expression in BRCA1-deficient cancer cells, contributing to immune evasion. To circumvent this, we developed P-selectin-targeted poly (lactic-co-glycolic) acid (PLGA)-poly (ethylene glycol) (PEG)-based nanoparticles (NPs) encapsulating PARP and PD-L1 inhibitors at a synergistic ratio. To further enhance tumor targeting, we leveraged radiation-induced P-selectin upregulation in BRCA1-deficient cancer cells and their associated angiogenic endothelial cells, improving NP accumulation in the primary tumors and hard-to-target metastatic sites, including brain metastasis. Using a combination of traditional 2-dimensional (2D) cell cultures, advanced 3-dimensional (3D) spheroids, tumor-on-a-chip platforms, and in vivo models, we demonstrate the enhanced accumulation and efficacy of the radiation-guided P-selectin-targeted NPs in primary and brain-metastatic BRCA1-deficient tumors.
Collapse
Affiliation(s)
- Rami Khoury
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Giuseppe Longobardi
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tania T Barnatan
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dana Venkert
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| | - América García Alvarado
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Adi Yona
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Marina Green Buzhor
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shir Shahar
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Qiwei Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Rita C Acúrcio
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Rita C Guedes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Helena F Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Jean J Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medical and Health Sciences, School of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; Tel Aviv University, Center for Nanoscience and Nanotechnology, Tel Aviv 6997801, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel.
| |
Collapse
|
20
|
Hyun H, Sun B, Yazdimamaghani M, Wielgus A, Wang Y, Montgomery SA, Zhang T, Cheng J, Serody JS, Wang AZ. Tumor-specific surface marker-independent targeting of tumors through nanotechnology and bioorthogonal glycochemistry. J Clin Invest 2025; 135:e184964. [PMID: 40067370 PMCID: PMC12043094 DOI: 10.1172/jci184964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 03/05/2025] [Indexed: 05/02/2025] Open
Abstract
Biological targeting is crucial for effective cancer treatment with reduced toxicity but is limited by the availability of tumor surface markers. To overcome this, we developed a nanoparticle-based (NP-based), tumor-specific surface marker-independent (TRACER) targeting approach. Utilizing the unique biodistribution properties of NPs, we encapsulated Ac4ManNAz (Maz) to selectively label tumors with azide-reactive groups. Surprisingly, while NP-delivered Maz was cleared by the liver, it did not label macrophages, potentially reducing off-target effects. To exploit this tumor-specific labeling, we functionalized anti-4-1BB Abs with dibenzocyclooctyne to target azide-labeled tumor cells and activate the immune response. In syngeneic B16F10 melanoma and orthotopic 4T1 breast cancer models, TRACER enhanced the therapeutic efficacy of anti-4-1BB, increasing the median survival time. Immunofluorescence analyses revealed increased tumor infiltration of CD8+ T and NK cells with TRACER. Importantly, TRACER reduced the hepatotoxicity associated with anti-4-1BB, resulting in normal serum ALT and AST levels and decreased CD8+ T cell infiltration into the liver. Quantitative analysis confirmed a 4.5-fold higher tumor-to-liver ratio of anti-4-1BB accumulation with TRACER compared with conventional anti-4-1BB Abs. Our work provides a promising approach for developing targeted cancer therapies that circumvent limitations imposed by the paucity of tumor-specific markers, potentially improving efficacy and reducing off-target effects to overcome the liver toxicity associated with anti-4-1BB.
Collapse
Affiliation(s)
- Hyesun Hyun
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill (UNC), Chapel Hill, North Carolina, USA
| | - Bo Sun
- College of Pharmacy, Skaggs Pharmaceutical Sciences Center, University of Arizona, Tucson, Arizona, USA
| | - Mostafa Yazdimamaghani
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill (UNC), Chapel Hill, North Carolina, USA
| | - Albert Wielgus
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill (UNC), Chapel Hill, North Carolina, USA
| | | | - Stephanie Ann Montgomery
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill (UNC), Chapel Hill, North Carolina, USA
- Department of Pathology and Laboratory Medicine, and
- Division of Comparative Medicine, UNC, Chapel Hill, North Carolina, USA
| | - Tian Zhang
- Department of Medicine and Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jianjun Cheng
- School of Engineering, Westlake University; Hangzhou, Zhejiang, China
| | - Jonathan S. Serody
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill (UNC), Chapel Hill, North Carolina, USA
- Department of Medicine and
- Department of Immunology and Microbiology, UNC, Chapel Hill, North Carolina, USA
| | - Andrew Z. Wang
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
21
|
Lin MZ, Li M, Zhang H, Li WX, Li Z, Wang LZ, Chen C, Li XL, Fan HJ, Jiang GB, Song K. Overcoming chemoresistance in acute myeloid leukemia via co-delivery of siGLUT1 and hydroxycamptothecin using hyaluronic acid-conjugated nanocarriers. Int J Biol Macromol 2025; 308:142492. [PMID: 40147647 DOI: 10.1016/j.ijbiomac.2025.142492] [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: 07/03/2024] [Revised: 03/08/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Multi-drug resistance (MDR) presents a major challenge in the treatment of acute myeloid leukemia (AML). Combining chemotherapy and gene therapy offers a promising strategy to improve drug sensitivity in resistant AML cells. However, designing an effective delivery system for co-administration of multiple agents while maintaining biosafety remains challenging. In this study, we developed a biocompatible co-delivery system that incorporates hydroxycamptothecin (HCPT) and glucose transporter 1 (GLUT1) small interfering RNA (siRNA). HCPT was loaded onto gold nanoparticles through crystallization, ensuring drug stability and safety. The branched configuration of self-branched chitosan contributed to improved gene delivery efficiency. The hyaluronic acid-conjugated nanocarrier specifically targeted CD44 receptors expressed on AML cells, while the pH-sensitive properties of self-branched chitosan promoted localized drug and gene release. This system effectively delivered the therapeutic agents to tumor sites, improving cellular uptake and synergistically inhibiting DNA synthesis by downregulating glycolysis and P-glycoprotein expression in leukemic cells. Both in vitro and in vivo experiments demonstrated strong antitumor efficacy and excellent biosafety. This co-delivery system offers a promising strategy for overcoming drug resistance in AML and holds potential for clinical translation.
Collapse
MESH Headings
- Hyaluronic Acid/chemistry
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Humans
- Drug Resistance, Neoplasm/drug effects
- Animals
- Camptothecin/analogs & derivatives
- Camptothecin/pharmacology
- Camptothecin/chemistry
- Camptothecin/administration & dosage
- Mice
- RNA, Small Interfering/genetics
- RNA, Small Interfering/administration & dosage
- Drug Carriers/chemistry
- Cell Line, Tumor
- Nanoparticles/chemistry
Collapse
Affiliation(s)
- Min-Zhao Lin
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Min Li
- Department of Pharmy, The First Affiliated Hospital of Jishou University, Jishou, Hunan 416000, China
| | - Hongyan Zhang
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Wei-Xiong Li
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Zenglin Li
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Ling-Zhi Wang
- Department of Pharmy, The First Affiliated Hospital of Jishou University, Jishou, Hunan 416000, China
| | - Chao Chen
- Department of Pharmy, The First Affiliated Hospital of Jishou University, Jishou, Hunan 416000, China
| | - Xiao-Lan Li
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou, Hunan 416000, China
| | - Hong-Jie Fan
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou, Hunan 416000, China
| | - Gang-Biao Jiang
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China.
| | - Kui Song
- Department of Hematology, The First Affiliated Hospital of Jishou University, Jishou, Hunan 416000, China.
| |
Collapse
|
22
|
Fu Y, Sun J, Yang C, Li W, Wang Y. Diversified nanocarrier design to optimize glucose oxidase-mediated anti-tumor therapy: Strategy and progress. Int J Biol Macromol 2025; 306:141581. [PMID: 40023419 DOI: 10.1016/j.ijbiomac.2025.141581] [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: 12/20/2024] [Revised: 02/08/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Given the inherent complexity and heterogeneity of tumors, current therapeutic approaches often fall short in meeting prognostic requirements. Starvation therapy (ST) utilizing glucose oxidase (GOx) has emerged as a promising strategy, specifically targeting tumor glucose consumption to disrupt nutrient supply. However, the therapeutic potential of GOx is significantly hampered by its inherent limitations as a protein, particularly its poor stability and short in vivo half-life. In recent years, the development of nanocarriors has provided an effective platform for intravenous and local tumor delivery of GOx. This review systematically examines three key strategies in GOx delivery: stimulus-response, biofilm modification, and local delivery. The progress in various carrier systems for GOx-mediated tumor therapy is comprehensively summarized, providing valuable insights for nanocarrier design. Furthermore, the existing challenges and future directions to advance the development of GOx-based tumor therapies are critically analyzed.
Collapse
Affiliation(s)
- Yuhan Fu
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China
| | - Jialin Sun
- Department of medicine, Heilongjiang Minzu College, Harbin, Heilongjiang Province, China
| | - Chunyu Yang
- Department of Pathology, Harbin 242 Hospital, Harbin, Heilongjiang Province, China
| | - Weinan Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China.
| | - Yanhong Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China; Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, Heilongjiang Province, China.
| |
Collapse
|
23
|
Gautam S, Joshi S, Jindal P, Patel P, Pal RR, Kumar M, Gupta GD, Kurmi BD. Recent doxorubicin-conjugates in cancer drug delivery: Exploring conjugation strategies for enhanced efficacy and reduced toxicity. Int J Pharm 2025; 675:125556. [PMID: 40187698 DOI: 10.1016/j.ijpharm.2025.125556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Doxorubicin is a first-line treatment of cancer that works on the mechanism of DNA intercalation and topoisomerase II poisoning. Since the 20th century, Doxorubicin has been used as a promising drug to treat several types of cancer, both solid or metastatic, including breast, thyroid, bladder, ovarian, or gastric cancer, etc. Even though it shows promising effects on cancer cells, it also shows its effects on healthy cells with cancerous cells, which leads to several severe side effects, such as cardiomyopathy, phlebitis, congestive heart failure (CHF), etc., which limits its usage in chemotherapy. Several research has focused on the targeted delivery of doxorubicin to cancerous cells to reduce side effects and improve efficacy. To optimize its anticancer potential, scientists have recently been developing nano-formulations and investigating various conjugations. The structure of doxorubicin consists of two primary functional groups that can be employed for conjugation with a variety of biomolecules, The first is the primary amine group in a sugar moiety, and the other one is the primary hydroxyl group in the aliphatic chain ring. In this paper, we have mentioned several conjugations of doxorubicin such as antibodies, nanoparticles, polymers, and phytochemical conjugations. Different studies regarding these conjugations are also mentioned, which represent promising strategies to optimize cancer treatment by minimizing side effects.
Collapse
Affiliation(s)
- Shreastha Gautam
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Sachin Joshi
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Priya Jindal
- Department of Pharmaceutical Quality Assurance, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road Moga-142001, Punjab, India.
| | - Ravi Raj Pal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Ghanshyam Das Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga-142001, Punjab, India.
| |
Collapse
|
24
|
Salum KCR, Miranda GBA, Dias AL, Carneiro JRI, Bozza PT, da Fonseca ACP, Silva T. The endocannabinoid system in cancer biology: a mini-review of mechanisms and therapeutic potential. Oncol Rev 2025; 19:1573797. [PMID: 40370489 PMCID: PMC12075236 DOI: 10.3389/or.2025.1573797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 04/18/2025] [Indexed: 05/16/2025] Open
Abstract
The Endocannabinoid System (ECS) plays a critical role in maintaining physiological homeostasis, influencing a range of processes such as neuroprotection, inflammation, energy metabolism, and immune responses. Comprising cannabinoid receptors (CB1 and CB2), endogenous ligands (endocannabinoids), and the enzymes responsible for their synthesis and degradation, the ECS has attracted increasing attention in cancer research. Cannabinoid receptor activation has been associated with the regulation of cancer-related processes, including cell proliferation, apoptosis, and angiogenesis, suggesting that the ECS may have a role in tumor progression and cancer treatment. Preclinical studies have shown that cannabinoids, through their interaction with CB1 and CB2 receptors, can inhibit tumor cell growth, induce programmed cell death, and suppress the formation of new blood vessels in various cancer models. Despite these encouraging findings, the clinical translation of ECS-targeted therapies remains in its early stages. The complexity of tumor heterogeneity, the variability in patient responses, and the challenges associated with the pharmacokinetics of cannabinoids are significant obstacles to the broader application of these findings in clinical settings. This review provides an overview of the current understanding of the ECS's involvement in cancer biology, focusing on key mechanisms by which it may influence carcinogenesis. Additionally, we discuss the therapeutic potential of targeting the ECS in cancer treatment, while highlighting the limitations and uncertainties that need to be addressed through ongoing research.
Collapse
Affiliation(s)
- Kaio Cezar Rodrigues Salum
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Gabriel Brendo Alves Miranda
- Postgraduate Program in Translational Biomedicine - UNIGRANRIO/AFYA, Duque de Caxias, Brazil
- Genetics Laboratory - UNIGRANRIO/AFYA, Duque de Caxias, Brazil
| | - Alessandra Lima Dias
- Postgraduate Program in Translational Biomedicine - UNIGRANRIO/AFYA, Duque de Caxias, Brazil
- Genetics Laboratory - UNIGRANRIO/AFYA, Duque de Caxias, Brazil
| | - João Regis Ivar Carneiro
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Patrícia Torres Bozza
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Ana Carolina Proença da Fonseca
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Postgraduate Program in Translational Biomedicine - UNIGRANRIO/AFYA, Duque de Caxias, Brazil
- Genetics Laboratory - UNIGRANRIO/AFYA, Duque de Caxias, Brazil
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Tamara Silva
- Postgraduate Program in Translational Biomedicine - UNIGRANRIO/AFYA, Duque de Caxias, Brazil
- Genetics Laboratory - UNIGRANRIO/AFYA, Duque de Caxias, Brazil
| |
Collapse
|
25
|
Lei S, Li J, Zhu M, Zhou W, Fu X, Wu S, Chen X, Zhang J, Duan X, Wang W, Men K. Chimeric Antigen Receptor-Engineered Cell Membrane-Coated Nanoparticles Promote Dual-Targeted mRNA-Based Cancer Gene Therapy. ACS NANO 2025; 19:15668-15684. [PMID: 40227438 DOI: 10.1021/acsnano.4c18153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
Gene therapy using mRNA has facilitated progress in cancer therapy. However, its application is hindered by a limited tumor-targeted delivery approach, leading to off-target effects and safety concerns. Chimeric antigen receptor (CAR) molecules enable T cells to recognize specific antigens in a major histocompatibility complex-unrestricted manner. CAR approaches provide an "off-the-shelf" solution for introducing additional targeting functionality to a cell membrane. Cancer cell membrane-coated nanoparticles with homotypic tumor-targeted properties provide a readily accessible platform for gene engineering and membrane extraction. Herein, we demonstrate a CAR-inspired cancer cell membrane-coated platform for delivering an mRNA formulation through a dual tumor-targeted mechanism. The simplified human epidermal growth factor receptor 2 (HER2)-specific CAR molecule (comprising an extracellular HER2-binding domain, a hinge, and a transmembrane domain) was engineered on the cell membrane of cancer cells to establish CAR-CT26 cells. The extracted CAR-CT26 membrane (CARM) was subsequently coated onto the lipid nanoparticle (LNP)-mRNA surface to form a CARM@LNP-mRNA complex. In vitro, the CARM-coated nanoparticles exhibited enhanced mRNA transfection efficiency toward CT26 cells overexpressing target HER2 antigens. Systemic administration of the CARM@LNP-mRNA formulation resulted in stronger tumor-targeting ability and tumor suppression in HER2+ CT26 subcutaneous tumors and peritoneal cavity metastasis models than that observed with the CT26 cell membrane-coated version. Our data suggest that CARM@LNP is a feasible choice for mRNA-based gene therapy. These results provide evidence for the systemic administration of CARM@LNP-mRNA as a promising tumor-targeted therapeutic strategy.
Collapse
Affiliation(s)
- Sibei Lei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Pharmacy, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming 650032, China
| | - Jingmei Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Manfang Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Weilin Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xizi Fu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shan Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiayu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Wei Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ke Men
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
26
|
Huang D, Huang W, Liu M, Chen J, Xiao D, Peng Z, He H, Shen H, Jin Q, Chen L, Rao D, Zhao M, Huang J. Progress of mesenchymal stem cell-derived exosomes in targeted delivery of antitumor drugs. Cancer Cell Int 2025; 25:169. [PMID: 40301903 PMCID: PMC12042352 DOI: 10.1186/s12935-025-03795-x] [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: 02/10/2025] [Accepted: 04/17/2025] [Indexed: 05/01/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are currently being used in clinical trials for the treatment of a wide range of diseases and have a wide range of applications in the fields of tissue engineering and regeneration. Exosomes are extracellular vesicles containing a variety of components such as proteins, nucleic acids and lipids, which are widely present in biological fluids and have the functions of participating in intercellular information transfer, immune response and tissue repair, and can also be used as carriers to target and deliver tumors to improve therapeutic effects. Mesenchymal stem cell-derived Exosomes (MSC-Exos), which have the advantages of low immunogenicity and high tumor homing ability, have attracted much attention in targeted drug delivery. Here, we review the current knowledge on the involvement of MSC-Exos in tumor progression and their potential as drug delivery systems in targeted therapies. It also discusses the advantages and prospects of MSC-Exos as a drug carrier and the challenges that still need to be overcome.
Collapse
Affiliation(s)
- Defa Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Wenlong Huang
- Department of General Medicine, First People's Hospital of Zunyi (Third Affiliated Hospital of Zunyi Medical University), Zunyi, 563000, China
| | - Meijin Liu
- People's Hospital of Ganzhou Economic Development Zone, Ganzhou, 341000, China
| | - Jie Chen
- Department of Laboratory Medicine, the Affiliated Yongchuan Hospital of Chongqing Medical University, Yongchuan, Chongqing, 402177, China
| | - Dewang Xiao
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
| | - Zongbo Peng
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, 341000, China
| | - Haoquan He
- Department of General Practice, Ditian Community health centre, Jinhua jindong, xiaoshun, 321000, China
| | - Haibin Shen
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Qing Jin
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China
| | - Linli Chen
- Laboratory Medicine, Guizhou Aerospace Hospital, Zunyi, 563100, China
| | - Dingyu Rao
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| | - Minghong Zhao
- Laboratory Medicine, Guizhou Aerospace Hospital, Zunyi, 563100, China.
| | - Junyun Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
27
|
Chakroborty S, Nath N, Sahoo S, Singh BP, Bal T, Tiwari K, Hailu YK, Singh S, Kumar P, Chakraborty C. A review of emerging trends in nanomaterial-driven AI for biomedical applications. NANOSCALE ADVANCES 2025:d5na00032g. [PMID: 40370571 PMCID: PMC12071765 DOI: 10.1039/d5na00032g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/18/2025] [Indexed: 05/16/2025]
Abstract
The field of artificial intelligence (AI) is expanding quickly. To mimic the structure and biological evolution of the human brain, AI was developed to enable computers to acquire knowledge and manipulate their surroundings. There have been notable developments in the use of AI in healthcare; it can enhance diagnosis and treatment in various medical specialties. The cost of prompt diagnosis and treatment is hampered by the absence of efficient, dependable, and reasonably priced detection and real-time monitoring. Smart health tracking systems integrating AI and nanoscience are an emerging frontier that solves these obstacles. Targeted delivery of drug systems, biosensing, imaging, and other diagnostic and therapeutic fields can widely benefit abundantly from nanoscience in healthcare. AI technology has the potential to expand biomedical applications by analyzing and interpreting biological data, speeding up drug discovery, and identifying novel molecules with predictive behavior. This review outlines the current obstacles and potential opportunities for delivering personal healthcare using AI-assisted clinical decision support systems.
Collapse
Affiliation(s)
- Subhendu Chakroborty
- School of Basic Sciences, Department of Chemistry, Chandigarh University Uttar Pradesh Unnao India
| | - Nibedita Nath
- Department of Chemistry, D. S. Degree College Laida Sambalpur 768214 Odisha India
| | - Sameeta Sahoo
- School of Basic Sciences, Department of Chemistry, Chandigarh University Uttar Pradesh Unnao India
| | - Bhanu Pratap Singh
- Department of Computing & Information Sciences, Chandigarh University Uttar Pradesh Unnao India
| | - Trishna Bal
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology Mesra Ranchi 835215 India
| | - Karunesh Tiwari
- Department of Physics, Mai Nefhi College of Science, Eritrean Institute of Technology Mai Nefhi Eritrea
| | - Yosief Kasshun Hailu
- Department of Physics, Mai Nefhi College of Science, Eritrean Institute of Technology Mai Nefhi Eritrea
| | - Sunita Singh
- IES College of Education, IES University Bhopal Madhya Pradesh 462044 India
| | - Pravin Kumar
- Inter University Accelerator Centre New Delhi 110067 India
| | - Chandra Chakraborty
- Department of Allied Sciences, Graphic Era (Deemed to be University) Clement Town 248002 Dehradun India
| |
Collapse
|
28
|
Alemi PS, Mohamadali M, Arabahmadi S, Irani S, Sharifi F. Carboxymethyl Chitosan and Chitosan as a Bioactive Delivery System: A Review. Biotechnol Appl Biochem 2025:e2758. [PMID: 40275440 DOI: 10.1002/bab.2758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/16/2025] [Indexed: 04/26/2025]
Abstract
The functionality and mechanism of bioactive agents (BA) in treating various diseases have been studied as a progressive route. Designing an effective delivery system for transferring these molecules and components is a major challenge. For that reason, a wide range of biomaterials has been introduced to deliver BA to the target tissue or cells. Chitosan (CTS) is a nontoxic, biocompatible, biodegradable, and notable point low-cost polymer, and, as a result, can be effectively utilized in the formulation of diverse delivery systems, in biomedical applications. However, CTS has some limitations, such as poor solubility in aqueous and alkaline media, rapid swelling and degradation, and consequence fast release agent. The CTS derivative carboxymethyl chitosan (CMC) is an acceptable candidate for overcoming these limitations. CMC is a high-impact grade for pharmaceutical and biomedical applications because of its nontoxic, biocompatible, biodegradable, gelation, mucoadhesive, antibacterial, and antifungal. CMC bioactivity potentials are related to carboxyl and methyl groups added through chemical modification in the CTS backbone. In this review, the physical and chemical properties of CTS and CMC have been introduced and discussed. Afterward, its biomedical applications with delivery approaches for various BA (drugs, genes, proteins), microfluidic, and cancer have been considered.
Collapse
Affiliation(s)
- Parinaz Sadat Alemi
- Department of Biological Sciences, University of Texas at Dallas, Richardson, Texas, USA
| | - Marjan Mohamadali
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Samira Arabahmadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Fereshteh Sharifi
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
- Soft Tissue Engineering Research Center, Tissue Engineering and Regenerative Medicine Institute, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
29
|
Borah ST, Mondal A, Das B, Saha S, Das Sarma J, Gupta P. β-Cyclodextrin Encapsulated Platinum(II)-Based Nanoparticles: Photodynamic Therapy and Inhibition of the NF-κB Signaling Pathway in Glioblastoma. ACS APPLIED BIO MATERIALS 2025; 8:3331-3342. [PMID: 40148119 DOI: 10.1021/acsabm.5c00103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
This study explores cell death through photodynamic therapy (PDT) with β-cyclodextrin-encapsulated platinum(II)-based nanoparticles (Pt-NPs) and the effect on the NF-κB and stress pathways in glioblastoma. The encapsulation of the cyclometalated Pt(II) complex Pt(LL') within β-cyclodextrin (β-CD) enhances its biocompatibility, improves cellular penetration, and boosts emission, thereby increasing the effectiveness of PDT. Both Pt(LL') and Pt-NPs show minimal toxicity in the dark; however, Pt-NPs significantly increase toxicity toward glioblastoma Kr158 cells upon irradiation at 390 nm. The PDT-induced cell death is further validated through apoptosis assays and the modulation of some key survival pathways like NF-κB/p65, DJ-1, and ERp29. This is the first report of β-cyclodextrin-encapsulated platinum(II)-based nanoparticles designed to target glioblastoma cells through PDT, offering a promising strategy for enhancing therapeutic efficacy.
Collapse
Affiliation(s)
- Sakira Tabassum Borah
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, India 741246
| | - Anushka Mondal
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, India 741246
| | - Bishnu Das
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, India 741246
| | - Sanchari Saha
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, India 741246
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research, Kolkata, India 741246
| | - Parna Gupta
- Department of Chemical Sciences, Indian Institute of Science Education and Research, Kolkata, India 741246
| |
Collapse
|
30
|
Yao W, Lin Y, Xu N, Xi Q, Liu Y, Li L. Laminarin-coated Genexol-PM pH sensitive nanomicelles targeting miR-620/IRF2BP2 axis for inhibition of cell proliferation and induction of apoptosis in Invitro thyroid carcinoma. Int J Biol Macromol 2025; 310:143198. [PMID: 40246117 DOI: 10.1016/j.ijbiomac.2025.143198] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 04/19/2025]
Abstract
This study explores the efficacy of Laminarin-coated Genexol-PM pH-sensitive nanomicelles targeting the miR-620/IRF2BP2 axis for cancer therapy using Dextran (BP), BP@PLGA, and BP@PLGA/PLA drug delivery systems. Among these, BP@PLGA/PLA demonstrated the highest cytotoxic potential in AGS cells, as confirmed by MTT assays, due to its advanced dual-polymer composition, which enhances drug encapsulation, stability, and targeted release in acidic tumor microenvironments. AO-EB and DAPI nuclear staining further validated these findings, showing significant apoptotic activity in BP@PLGA/PLA-treated cells, characterized by chromatin condensation, nuclear fragmentation, and apoptotic body formation. Additionally, ROS detection using carboxy-H2DCFDA staining indicated that BP@PLGA/PLA induced the highest oxidative stress levels, further driving apoptosis and disrupting cancer cell viability. In contrast, Dextran (BP) exhibited minimal cytotoxicity, and BP@PLGA showed moderate effectiveness, highlighting the superior therapeutic efficacy of BP@PLGA/PLA. The pH-sensitive nature of Laminarin-coated Genexol-PM micelles further enhanced the targeted inhibition of the miR-620/IRF2BP2 axis, improving specificity while minimizing off-target effects. By leveraging both oxidative stress mechanisms and apoptosis induction, BP@PLGA/PLA offers a promising approach for overcoming limitations in conventional chemotherapy. These findings underscore the potential of pH-responsive nanomicelles in precision oncology, offering improved drug delivery, enhanced therapeutic index, and a more effective strategy for combating drug-resistant cancers.
Collapse
Affiliation(s)
- Wei Yao
- Department of General Surgery, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang 110004, Liaoning, China.
| | - Yuhe Lin
- Department of Oncology, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang 110004, Liaoning, China.
| | - Nan Xu
- Department of Plastic Surgery, 1st. Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning, China.
| | - Qi Xi
- The First Hospital of China Medical University, Department of Pain Medicine Shenyang, Liaoning, 110001, China
| | - Yan Liu
- The First Hospital of China Medical University, Department of Pain Medicine Shenyang, Liaoning, 110001, China
| | - Li Li
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, No.36, Sanhao Street, Shenyang 110004, Liaoning, China.
| |
Collapse
|
31
|
George S, Saju H, Jaikumar T, Raj R, Nisarga R, Sontakke S, Sangshetti J, Paul MK, Arote RB. Deciphering a crosstalk between biological cues and multifunctional nanocarriers in lung cancer therapy. Int J Pharm 2025; 674:125395. [PMID: 40064384 DOI: 10.1016/j.ijpharm.2025.125395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/08/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025]
Abstract
In recent years, the utilization of nanocarriers has significantly broadened across a diverse spectrum of biomedical applications. However, the clinical translation of these tiny carriers is limited and encounters hurdles, particularly in the intricate landscape of the tumor microenvironment. Lung cancer poses unique hurdles for nanocarrier design. Multiple physiological barriers hinder the efficient drug delivery to the lungs, such as the complex anatomy of the lung, the presence of mucus, immune responses, and rapid clearance mechanisms. Overcoming these obstacles necessitates a targeted approach that minimizes off-target effects while effectively penetrating nanoparticles/cargo into specific lung tissues or cells. Furthermore, understanding the cellular uptake mechanisms of these nano carriers is also essential. This knowledge aids in developing nanocarriers that efficiently enter cells and transfer their payload for the most effective therapeutic outcome. Hence, a thorough understanding of biological cues becomes crucial in designing multifunctional nanocarriers tailored for treating lung cancer. This review explores the essential biological cues critical for developing a flexible nanocarrier specifically intended to treat lung cancer. Additionally, it discusses advancements in nanotheranostics in lung cancer.
Collapse
Affiliation(s)
- Sharon George
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Hendry Saju
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Tharun Jaikumar
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Reshma Raj
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - R Nisarga
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Samruddhi Sontakke
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Jaiprakash Sangshetti
- Y. B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Rauza Baugh, Aurangabad 431001, India
| | - Manash K Paul
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India; Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), 90095 CA, USA.
| | - Rohidas B Arote
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India; Dental Research Institute, School of Dentistry, Seoul National University, Gwanak-ku, Seoul 08826, Republic of Korea.
| |
Collapse
|
32
|
Han W, Xiong N, Huang L. Probiotics and nanoparticle-mediated nutrient delivery in the management of transfusion-supported diseases. Front Cell Infect Microbiol 2025; 15:1575798. [PMID: 40292219 PMCID: PMC12021914 DOI: 10.3389/fcimb.2025.1575798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Bone marrow is vital for hematopoiesis, producing blood cells essential for oxygen transport, immune defense, and clotting. However, disorders like leukemia, lymphoma, aplastic anemia, and myelodysplastic syndromes can severely disrupt its function, leading to life-threatening complications. Traditional treatments, including chemotherapy and stem cell transplants, have significantly improved patient outcomes but are often associated with severe side effects and limitations, necessitating the exploration of safer, more targeted therapeutic strategies. Nanotechnology has emerged as a promising approach for addressing these challenges, particularly in the delivery of nutraceuticals-bioactive compounds derived from food sources with potential therapeutic benefits. Despite their promise, nutraceuticals often face clinical limitations due to poor bioavailability, instability, and inefficient delivery to target sites. Nanoparticles offer a viable solution by enhancing the stability, absorption, and targeted transport of nutraceuticals to bone marrow while minimizing systemic side effects. This study explores a range of bone marrow disorders, conventional treatment modalities, and the potential of nanoparticles to enhance nutraceutical-based therapies. By improving targeted delivery and therapeutic efficacy, nanoparticles could revolutionize bone marrow disease management, providing patients with more effective and less invasive treatment options. These advancements represent a significant step toward safer and more efficient therapeutic approaches, ultimately improving patient prognosis and overall health.
Collapse
Affiliation(s)
- Wendao Han
- Department of Blood Transfusion, Meizhou People’s Hospital, Meizhou Academy of
Medical Sciences, Meizhou, China
| | | | | |
Collapse
|
33
|
Nagao K, Paniagua EV, Lei K, Beckham JL, Worthington P, Manthey M, Ye M, Koehler F, Kim YJ, Malkin E, Onoda M, Kent N, Michida S, Guerra EC, Macfarlane RJ, Anikeeva P. Adeno-associated viruses escort nanomaterials to specific cells and tissues. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647267. [PMID: 40291644 PMCID: PMC12026743 DOI: 10.1101/2025.04.04.647267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
The delivery of nanotherapeutics to specific tissues relies on bespoke targeting strategies or invasive surgeries. Conversely, adeno-associated viruses (AAVs) can target specific tissues following intravenous injections. Here we show that cell-targeting properties of AAVs could be broadly conferred to nanomaterials. We develop a strategy to couple AAV capsids to nanoparticles that is invariant of viral serotype or nanomaterial chemistry and permits control over stoichiometry of the AAV-nanoparticle chimeras. The chimeras selectively escort nanoparticles into cell classes governed by AAV serotypes. When applied to magnetic nanoparticles, the AAV-nanoparticle chimeras enable magnetically localized gene delivery. In vivo, we show that leveraging the brain-targeting AAV serotype CAP-B10 achieves nanoparticle delivery to the parenchyma with ∼10% efficiency (% injected dose/g [brain] ) while avoiding accumulation in the liver. The enhanced delivery efficiency and tissue specificity highlight the potential of AAV-chimeras as a versatile strategy to escort broad classes of nanotherapeutics to the brain and beyond.
Collapse
|
34
|
Zhang H, Chen H, Guo G, Lin J, Chen X, Huang P, Lin C, Lin H, Lu Y, Lin J, Li X, Zhang W. Nanotechnology in prostate cancer: a bibliometric analysis from 2004 to 2023. Discov Oncol 2025; 16:451. [PMID: 40175778 PMCID: PMC11965044 DOI: 10.1007/s12672-025-02265-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/28/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Prostate cancer (PC) contributes to male mortality worldwide. The objective of this study is to comprehensively depict the scientific accomplishments and research trends in nanotechnology for PC applications. METHODS Utilizing the Web of Science Core Collection database, publications were gathered on the basis of inclusion and selection criteria. The publications were analyzed and visualized using VOSviewer, R-studio and CiteSpace software tools. RESULTS A total of 1949 studies were incorporated. Farokhzad was the most productive author. The United States and China released 58.13% of the total publications. The Chinese Academy of Sciences was the most influential institution, and the International Journal of Nanomedicine stood out as a prominent journal in this field. The most frequently referenced publication and research subject category were identified. The most extensively investigated area was nanoparticle-based drug delivery, while recent research has focused on anticancer with novel nanocarriers. CONCLUSION A bibliometric analysis in the PC and nanotechnology was conducted between 2004 and 2023. The overview and characteristics of the publications were identified. We discussed the application and restrictions faced by nanotechnology in PC management. The study of nanotechnology in PC treatment needs to be further studied.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Hongpeng Chen
- Department of Oncology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Gaowei Guo
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Jinming Lin
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Xiaosheng Chen
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Peidong Huang
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Chuqi Lin
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Huirong Lin
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Yong Lu
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Jieming Lin
- Department of Operating Room, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China
| | - Xinji Li
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China.
| | - Wei Zhang
- Department of Urology, Jieyang People's Hospital, Jieyang, 522000, Guangdong, People's Republic of China.
| |
Collapse
|
35
|
Linderman SW, DeRidder L, Sanjurjo L, Foote MB, Alonso MJ, Kirtane AR, Langer R, Traverso G. Enhancing immunotherapy with tumour-responsive nanomaterials. Nat Rev Clin Oncol 2025; 22:262-282. [PMID: 40050505 DOI: 10.1038/s41571-025-01000-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/05/2025] [Indexed: 03/09/2025]
Abstract
The targeted delivery of immunotherapies to tumours using tumour-responsive nanomaterials is a promising area of cancer research with the potential to address the limitations of systemic administration such as on-target off-tumour toxicities and a lack of activity owing to the immunosuppressive tumour microenvironment (TME). Attempts to address these challenges include the design and functionalization of nanomaterials capable of releasing their cargoes in response to specific TME characteristics, thus facilitating the targeted delivery of immune-checkpoint inhibitors, cytokines, mRNAs, vaccines and, potentially, chimaeric antigen receptors as well as of agents that modulate the extracellular matrix and induce immunogenic cell death. In this Review, we describe these various research efforts in the context of the dynamic properties of the TME, such as pH, reductive conditions, reactive oxygen species, hypoxia, specific enzymes, high levels of ATP and locoregional aspects, which can be leveraged to enhance the specificity and efficacy of nanomaterial-based immunotherapies. Highlighting preclinical successes and ongoing clinical trials, we evaluate the current landscape and potential of these innovative approaches. We also consider future research directions as well as the most important barriers to successful clinical translation, emphasizing the transformative potential of tumour-responsive nanomaterials in overcoming the barriers that limit the activity of traditional immunotherapies, thus improving patient outcomes.
Collapse
Affiliation(s)
- Stephen W Linderman
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Hospital Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Louis DeRidder
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard-MIT Division of Health Science Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Lucía Sanjurjo
- Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Michael B Foote
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
- Department of Pharmacy and Pharmaceutical Technology, University of Santiago de Compostela, Santiago de Compostela, Spain
- IMDEA Nanosciences Institute, Madrid, Spain
| | - Ameya R Kirtane
- Department of Pharmaceutics, University of Minnesota, Minneapolis, MN, USA
| | - Robert Langer
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Giovanni Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
36
|
Nguyen VN, Nguyen Cao TG, Jeong H, Truong Hoang Q, Pham BTT, Bang J, Koh CW, Kang JH, Lee JH, Wu X, Rhee WJ, Ko YT, Swamy KMK, Park S, Park J, Shim MS, Yoon J. Tumor-Targeted Exosome-Based Heavy Atom-Free Nanosensitizers With Long-Lived Excited States for Safe and Effective Sono-Photodynamic Therapy of Solid Tumors. Adv Healthc Mater 2025:e2500927. [PMID: 40165690 DOI: 10.1002/adhm.202500927] [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: 02/19/2025] [Revised: 03/17/2025] [Indexed: 04/02/2025]
Abstract
Theranostic nanosensitizers with combined near-infrared (NIR) fluorescence imaging and sono-photodynamic effects have great potential for use in the personalized treatment of deep-seated tumors. However, developing effective nanosensitizers for NIR fluorescence image-guided sono-photodynamic therapy remains a considerable challenge, including the low generation efficacy of reactive oxygen species (ROS), poor photostability, and the absence of cancer specificity. Herein, a novel heavy atom-free nanosensitizer is developed, which exhibits intense NIR fluorescence, high ROS generation efficiency, and improved aqueous stability. By conjugating a bulky and electron-rich group, 4-(1,2,2-triphenylvinyl)-1,1'-biphenyl (TPE), to the IR820 backbone, the resulting IR820 bearing TPE (IR820-TPE) effectively generates ROS via type I and II photochemical mechanisms under 808 nm laser irradiation. Moreover, TPE conjugation considerably increases the sono-photodynamic performance of IR820. To improve the intracellular delivery and tumor-targeting ability of IR820-TPE, biotin-conjugated exosome (B-Exo) is used as a natural nanocarrier. In vitro studies demonstrate the outstanding therapeutic performance of IR820-TPE-loaded B-Exo (IR820-TPE@B-Exo) in synergistic sono-photodynamic cancer therapy. In vivo studies reveal that IR820-TPE@B-Exo shows enhanced tumor accumulation, strong fluorescence signals, and effective sono-photodynamic therapeutic activity with high biosafety. This work demonstrates that IR820-TPE@B-Exo is a promising sono-phototheranostic agent for safe and targeted cancer therapy and NIR fluorescence imaging.
Collapse
Affiliation(s)
- Van-Nghia Nguyen
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
- Department of Chemistry, School of Chemistry and Life Sciences, Hanoi University of Science and Technology, Ha Noi, 100000, Vietnam
| | - Thuy Giang Nguyen Cao
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Hyunsun Jeong
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Quan Truong Hoang
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Binh T T Pham
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Jieun Bang
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Chang Woo Koh
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Ji Hee Kang
- College of Pharmacy, Gachon University, Incheon, 21936, Republic of Korea
| | - Jeong Hyun Lee
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Xiaofeng Wu
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Won Jong Rhee
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
- Research Center for Bio Materials & Process Development, Incheon National University, Incheon, 22012, Republic of Korea
| | - Young Tag Ko
- College of Pharmacy, Gachon University, Incheon, 21936, Republic of Korea
| | - K M K Swamy
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Sungnam Park
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - JaeHong Park
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, 03760, Republic of Korea
| |
Collapse
|
37
|
George M, Boukherroub R, Sanyal A, Szunerits S. Treatment of lung diseases via nanoparticles and nanorobots: Are these viable alternatives to overcome current treatments? Mater Today Bio 2025; 31:101616. [PMID: 40124344 PMCID: PMC11930446 DOI: 10.1016/j.mtbio.2025.101616] [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: 12/05/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Challenges Respiratory diseases remain challenging to treat, with current efforts primarily focused on managing symptoms rather than maintaining overall lung health. Traditional treatment methods, such as oral or parenteral administration of antiviral, antibacterial, and anti-inflammatory drugs, face limitations. These include difficulty in delivering therapeutic agents to pathogens residing deep in the airways and the risk of severe side effects due to high systemic drug concentrations. The growing threat of drug-resistant pathogens further complicates infection management. Advancements The lung's large surface area offers an attractive target for inhalation-based drug delivery. Nanoparticles (NP) enable uniform and sustained drug distribution across the alveolar network, overcoming challenges posed by complex lung anatomy. Recent breakthroughs in nanorobots (NR) have demonstrated precise navigation through biological environments, delivering therapies directly to affected lung areas with enhanced accuracy. Nanotechnology has also shown promise in treating lung cancer, with nanoparticles engineered to overcome biological barriers, improve drug solubility, and enable controlled drug release. Future scope This review explores the progress of NP and NR in addressing challenges in pulmonary drug delivery. These innovations allow targeted delivery of nucleic acids, drugs, or peptides to the pulmonary epithelium with unprecedented accuracy, offering significant potential for improving therapeutic effectiveness in respiratory disorders.
Collapse
Affiliation(s)
- Meekha George
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University (DPU), Viktor-Kaplan-Straße 2, Geb. E, 2700, Wiener Neustadt, Austria
| | - Rabah Boukherroub
- Univ. Lille, CNRS, Univ. Polytechnique, Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| | - Amitav Sanyal
- Department of Chemistry, Bogazici University, Bebek, 34342, Istanbul, Turkey
| | - Sabine Szunerits
- Laboratory for Life Sciences and Technology (LiST), Faculty of Medicine and Dentistry, Danube Private University (DPU), Viktor-Kaplan-Straße 2, Geb. E, 2700, Wiener Neustadt, Austria
- Univ. Lille, CNRS, Univ. Polytechnique, Hauts-de-France, UMR 8520 - IEMN, F-59000, Lille, France
| |
Collapse
|
38
|
Vetter VC, Yazdi M, Gialdini I, Pöhmerer J, Seidl J, Höhn M, Lamb DC, Wagner E. Ionic Coating of siRNA Polyplexes with cRGD-PEG-Hyaluronic Acid To Modulate Serum Stability and In Vivo Performance. Biochemistry 2025; 64:1509-1529. [PMID: 40102188 DOI: 10.1021/acs.biochem.4c00650] [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: 03/20/2025]
Abstract
Efficient delivery of siRNA-based polyplexes to tumors remains a major challenge. Nonspecific interactions in the bloodstream, limited circulation time, and nontargeted biodistribution hamper sufficient tumor accumulation. To address these challenges, we developed an ionic hyaluronic acid (HA) coating to shield sequence-defined oligoaminoamide-based polyplexes. This coating should shield the positive polyplex surface charge, thus reducing nonspecific interactions and enhancing serum stability. Additionally, we modified the HA coating with the cyclic RGDfK (cRGD) peptide to specifically target tumor endothelial cells (TECs). Optionally, a polyethylene glycol (PEG) spacer was also introduced to improve ligand presentation on the polyplex surface. The HA-coated polyplexes exhibited favorable physicochemical properties, including a negative zeta potential and effective siRNA retention within the polyplex, which was not adversely affected by PEG or cRGD modification. In vitro analyses revealed that these polyplexes not only enhanced tumor cell association and preserved the high transfection efficiency of plain cationic polyplexes but also exhibited coating-dependent cellular internalization, as evidenced by a competitive inhibition experiment. Even in the presence of serum, the HA-coated polyplexes encapsulated siRNA effectively, exhibited suitable particle sizes, and maintained a high gene silencing efficiency. In vivo studies involving intravenous administration into Neuro2a tumor-bearing mice showed that the HA coating, particularly when modified with PEG and cRGD, significantly increased the tumor accumulation of polyplexes. HA-PEG-cRGD-shielded polyplexes exhibited significantly enhanced in vivo gene silencing in tumors compared with plain polyplexes. Collectively, our results indicate a superior performance of HA-coated polyplexes in terms of stability and cellular uptake, both in vitro and in vivo.
Collapse
Affiliation(s)
- Victoria C Vetter
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
| | - Mina Yazdi
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
| | - Irene Gialdini
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, Munich 81377, Germany
| | - Jana Pöhmerer
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
| | - Johanna Seidl
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
| | - Miriam Höhn
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
| | - Don C Lamb
- Department of Chemistry, Ludwig Maximilians-Universität München, Butenandtstraße 5-13, Munich 81377, Germany
- Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
- Center for NanoScience (CeNS), Ludwig-Maximilians-Universität, Butenandtstraße 5-13, Munich 81377, Germany
- CNATM─Cluster for Nucleic Acid Therapeutics, Würmtalstr. 201, Munich 81377, Germany
| |
Collapse
|
39
|
Vigo M, Placci M, Muro S. Isoform-specific vs. isoform-universal drug targeting: a new targeting paradigm illustrated by new anti-ICAM-1 antibodies. J Drug Target 2025; 33:562-574. [PMID: 39639798 DOI: 10.1080/1061186x.2024.2438884] [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: 10/21/2024] [Revised: 11/20/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Drug targeting can be achieved by coupling drugs or their carriers to affinity molecules, mostly antibodies (Abs), which recognise specific protein targets. However, most proteins are not expressed in an exclusive configuration but as various isoforms. Hence, selected targeting molecules may fail to target with enough efficiency in clinical trials, which is overlooked. We illustrate this by targeting intercellular adhesion molecule 1 (ICAM-1), a cell-surface protein overexpressed in many pathologies. Most ICAM-1 targeting studies used Ab R6.5, which binds ICAM-1 domain 2 (D2). Yet, literature and our data show that D2 is frequently absent among ICAM-1 isoforms. We thus produced a battery of five new Abs (B4, B6, B11, C12 and G2) and tested their ability to recognise both full-length and -D2 ICAM-1. In solution, all Abs recognised both ICAM-1 forms (from 5.3 × 1011 to 4.2 × 1012 sum intensity/well). Coating them on nanocarriers (NCs) rendered G2 specific against -D2 ICAM-1 (4.2 × 106 NCs/well) while other Abs kept their dual recognition (from 6.4 × 106 to 2.2 × 107 NCs/well). All Abs induced NC intracellular uptake in respective cells (from 42% to 85%) and displayed good cross-species reactivity (from 4.4 × 1011 to 2.6 × 1012 sum intensity/well). These Abs represent valuable tools to target ICAM-1 and illustrate a new targeting paradigm that may improve classical strategies.
Collapse
Affiliation(s)
- Marco Vigo
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Biomedicine Doctorate Program, University of Barcelona, Barcelona, Spain
| | - Marina Placci
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Biotechnology Doctorate Program, University of Barcelona, Barcelona, Spain
| | - Silvia Muro
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute for Science and Technology (BIST), Barcelona, Spain
- Institution of Catalonia for Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
40
|
Adhikari A, Chen IA. Antibody-Nanoparticle Conjugates in Therapy: Combining the Best of Two Worlds. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409635. [PMID: 40051146 PMCID: PMC12001320 DOI: 10.1002/smll.202409635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/02/2025] [Indexed: 04/17/2025]
Abstract
Monoclonal antibodies (mAbs) and antibody fragments have revolutionized medicine as highly specific binding agents and inhibitors. At the same time, several types of nanomaterials, including liposomes, lipid nanoparticles (NPs), polymersomes, metal and metal oxide NPs, and protein nanostructures, are increasingly utilized and explored for therapeutic potential due to their versatility, chemical and physical properties, and tunability. However, nanomaterials alone often lack specificity, leading to relatively low efficacy and/or high toxicity. To address this problem, a rapidly emerging area is antibody-nanomaterial conjugates (ANCs), which combine the precise targeting specificity of antibodies with the effector functionality of the nanomaterial. In this review, we give a brief introduction to mAbs and major conjugation techniques, describe major classes of nanomaterials being studied for therapeutic potential, and review the literature on ANCs of each class. Special focus is given to emerging applications including ANCs addressing the blood-brain barrier, ANCs delivering nucleic acids, and light-activated ANCs. While many disease targets are related to cancer, ANCs are also under development to address autoimmune, neurological, and infectious diseases. While important challenges remain, ANCs are poised to become a next-generation therapeutic technology.
Collapse
Affiliation(s)
- Aniruddha Adhikari
- Department of Chemical and Biomolecular EngineeringDepartment of Chemistry and BiochemistryUniversity of CaliforniaLos AngelesCA90049USA
| | - Irene A. Chen
- Department of Chemical and Biomolecular EngineeringDepartment of Chemistry and BiochemistryUniversity of CaliforniaLos AngelesCA90049USA
| |
Collapse
|
41
|
Tran V, Nguyen N, Renkes S, Nguyen KT, Nguyen T, Alexandrakis G. Current and Near-Future Technologies to Quantify Nanoparticle Therapeutic Loading Efficiency and Surface Coating Efficiency with Targeted Moieties. Bioengineering (Basel) 2025; 12:362. [PMID: 40281721 PMCID: PMC12025210 DOI: 10.3390/bioengineering12040362] [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: 01/09/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 04/29/2025] Open
Abstract
Active targeting nanoparticles are a new generation of drug and gene delivery systems with the potential for greatly improved therapeutics delivery compared to conventional nanomedicine approaches. Despite their potential, the translation of active targeting nanoparticles faces challenges in production scale-up and batch consistency. Accurate quality control methods for nanoparticle therapeutic payload and coating characterization are critical for attaining the desired levels of batch repeatability, drug/gene loading efficiency, targeting molecule coating effectiveness, and safety profiles. Current limitations in nanoparticle characterization technologies, such as relying on ensemble-average analysis, pose challenges in assessing the drug/gene content and surface modification heterogeneity, which can greatly affect therapeutic outcomes. Single-molecule analysis technologies have emerged as a promising alternative, offering rich information on heterogeneity and stochastic variations between nanoparticle batches. This review first evaluates and identifies the challenges of traditional nanoparticle characterization tools that rely on indirect, bulk solution quantification methods. Subsequently, newly emerging characterization technologies are introduced for the quantification of therapeutic loading and targeted moiety coating efficiencies with single-nanoparticle resolution, to help guide researchers towards advancing the translation of active targeting nanoparticles into the clinical setting.
Collapse
Affiliation(s)
| | | | | | | | - Tam Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA; (V.T.); (N.N.); (S.R.); (K.T.N.)
| | - George Alexandrakis
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA; (V.T.); (N.N.); (S.R.); (K.T.N.)
| |
Collapse
|
42
|
Zhang W, Wang Q, Zhai F, Fan X, Meng F, Shen G, Zhu Y, Cao J, Yu F. Core-Shell Magnetic Nanocarriers: Fe 3O 4-Hydroxyapatite/Polysuccinimide Hybrids for Enhanced Oral Bioavailability of Fluorouracil. Int J Nanomedicine 2025; 20:3671-3695. [PMID: 40130197 PMCID: PMC11932132 DOI: 10.2147/ijn.s507458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/08/2025] [Indexed: 03/26/2025] Open
Abstract
Objective This study pioneers a pH-responsive core-shell nanoplatform integrating magnetic Fe3O4-hydroxyapatite (Fe/HAP) with polysuccinimide (PSI) polymer, engineered to enhance tumor-targeted delivery of fluorouracil (5-FU) for liver cancer therapy. Methods The individual components-hydroxyapatite (HAP), magnetite (F3O4), iron-doped hydroxyapatite (Fe/HAP), and polysuccinimide (PSI)-were synthesized and systematically characterized through Fourier-transform infrared (FTIR) spectroscopy and scanning electron microscopy (SEM). Through a combination of single-factor experiments and Box-Behnken design (BBD) response surface methodology, the formulation parameters were optimized for two nanoparticle systems: (1) non-magnetic 5-FU-loaded PSI-HAP (designated as 5-FU@DC, where DC denotes "drug carrier") and (2) magnetic-targeted formulations 5-FU@PSI-Fe/HAP with varying iron content (5-FU@FeDC20, 5-FU@FeDC30, 5-FU@FeDC40). The engineered nanoparticles were thoroughly characterized for their morphological characteristics, hydrodynamic properties (particle size distribution and zeta potential), magnetic responsiveness (vibrating sample magnetometry), and pH-dependent drug release profiles. Nile Red was used to label the drug-loaded nanoparticles, and small animal imaging technology was employed to track their distribution in mice in vivo. Furthermore, in vitro studies examined the effects of these formulations on the proliferation, apoptosis, and migration of Huh-7 liver cancer cells. Results The formulations (5-FU@DC and 5-FU@FeDC) were found to form uniform spherical or near-spherical nanoparticles. Vibrating sample magnetometer (VSM) analysis confirmed that the 5-FU@FeDC formulations displayed paramagnetic properties. Zeta potential measurements showed that all prepared systems had negative charges, similar to human biological membranes. All nanoparticles gradually released the drug at pH levels above 5, with the release rate increasing as the pH increased. Compared to the non-magnetic 5-FU@DC formulation, the magnetic 5-FU@FeDC formulations showed significantly longer distribution and retention times in liver tissue and more effectively inhibited the proliferation of Huh-7 cells. Conclusion The current study developed a magnetic targeting nano-delivery system using PSI and Fe/HAP as formulation excipients. The system offers uniform particle size, a simple preparation process, and a cost-effective method for targeted drug delivery. It is not only suitable for liver-targeted drug delivery but also applicable for drug delivery to other tissues in the body for anti-tumor drugs.
Collapse
Affiliation(s)
- Wenhui Zhang
- School of Pharmacy, Mudanjiang Medical University, Mudanjiang, 157011, People’s Republic of China
| | - Qiang Wang
- Hongqi People Hospital, Mudanjiang Medical University, Mudanjiang, 157011, People’s Republic of China
| | - Fengguo Zhai
- School of Pharmacy, Mudanjiang Medical University, Mudanjiang, 157011, People’s Republic of China
| | - Xingjun Fan
- School of Public Health, Mudanjiang Medical University, Mudanjiang, 157011, People’s Republic of China
| | - Fanqin Meng
- School of Pharmacy, Mudanjiang Medical University, Mudanjiang, 157011, People’s Republic of China
| | - Guangzhi Shen
- School of Pharmacy, Mudanjiang Medical University, Mudanjiang, 157011, People’s Republic of China
| | - Ying Zhu
- School of Pharmacy, Mudanjiang Medical University, Mudanjiang, 157011, People’s Republic of China
| | - Jingdan Cao
- School of Pharmacy, Mudanjiang Medical University, Mudanjiang, 157011, People’s Republic of China
| | - Fengbo Yu
- School of Pharmacy, Mudanjiang Medical University, Mudanjiang, 157011, People’s Republic of China
| |
Collapse
|
43
|
Lee CE, Noh KM, Kim S, Hong J, Kim K. Recent Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand Engineering Strategies for Precise Strike Therapy against Tumor. Biomater Res 2025; 29:0170. [PMID: 40110051 PMCID: PMC11922527 DOI: 10.34133/bmr.0170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/23/2025] [Accepted: 03/02/2025] [Indexed: 03/22/2025] Open
Abstract
Effective drug delivery relies on the selection of suitable carriers, which is crucial for protein-based therapeutics such as tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). One of the key advantages of TRAIL is its ability to selectively induce apoptosis in cancer cells excluding healthy tissues by binding to death receptors DR4 and DR5, which are highly expressed in various cancer cells. Despite this promise, the clinical application of TRAIL has been limited by its short half-life, limited stability, and inefficient delivery to tumor sites. To overcome currently available clinical and engineering approaches, a series of sophisticated strategies is required: (a) the design of biomaterial-mediated carriers for enhanced targeting efficacy, particularly via optimizing selected materials, composition, formulation, and surface modulation. Moreover, (b) development of genetically modified cellular products for augmented TRAIL secretion toward tumor microenvironments and (c) cell surface engineering techniques for TRAIL immobilization onto infusible cell populations are also discussed in the present review. Among these approaches, living cell-based carriers offer the distinct advantage of systemically administered TRAIL-functionalized cells capturing circulating tumor cells in the bloodstream, thereby preventing secondary tumor formation. This review provides insight into the development of novel TRAIL delivery platforms, discusses considerations for clinical translation, and suggests future directions and complementary strategies to advance the field of TRAIL-based cancer therapeutics.
Collapse
Affiliation(s)
- Chae Eun Lee
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Kyung Mu Noh
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Sungjun Kim
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| | - Jiyeon Hong
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyobum Kim
- Department of Chemical and Biochemical Engineering, Dongguk University, Seoul 04620, Republic of Korea
| |
Collapse
|
44
|
Zhu Q, Yu C, Chen Y, Luo W, Li M, Zou J, Xiao F, An S, Saiding Q, Tao W, Kong N, Xie T. Dual mRNA nanoparticles strategy for enhanced pancreatic cancer treatment and β-elemene combination therapy. Proc Natl Acad Sci U S A 2025; 122:e2418306122. [PMID: 40067898 PMCID: PMC11929461 DOI: 10.1073/pnas.2418306122] [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: 09/09/2024] [Accepted: 01/27/2025] [Indexed: 03/25/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is notoriously immune-resistant, limiting the clinical efficacy of single-agent immune modulators and thereby necessitating the exploration of multimodal immunotherapy combinations. Traditional approaches combining conventional immune checkpoint inhibitors with neoantigen vaccines have shown some promise in treating PDAC but are often compromised by intratumoral T lymphocyte exhaustion and systemic toxicity. Hence, novel approaches are needed to address these challenges. Herein, we demonstrate that mRNA polymeric nanoparticles encoding anti-PD-1 antibodies in situ at the tumor site enhance the therapeutic efficacy of neoantigen-based mRNA vaccine for PDAC. This mRNA-based, in situ anti-PD-1 antibody production strategy also protects tumor-infiltrating T cells from PD-1 inhibition, potentially reducing the toxicities induced by systemic checkpoint inhibition. Our study may provide an innovative dual mRNA nanoparticle strategy for effective tumor neoantigen immunotherapy, as well as an mRNA cancer combination therapy strategy with other clinically approved drugs (e.g., β-elemene).
Collapse
Affiliation(s)
- Qianru Zhu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang311121, China
| | - Chuao Yu
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang311121, China
| | - Yiquan Chen
- Liangzhu Laboratory, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Eye Center of The Second Affliated Hospital, Zhejiang University, Hangzhou, Zhejiang311121, China
| | - Wei Luo
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang311121, China
| | - Meng Li
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang311121, China
| | - Jianhua Zou
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang311121, China
| | - Fan Xiao
- Liangzhu Laboratory, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Eye Center of The Second Affliated Hospital, Zhejiang University, Hangzhou, Zhejiang311121, China
- Center for Nanomedicine and Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Soohwan An
- Center for Nanomedicine and Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University, Zhejiang Provincial Key Lab of Ophthalmology, Eye Center of The Second Affliated Hospital, Zhejiang University, Hangzhou, Zhejiang311121, China
- Center for Nanomedicine and Department of Anesthesiology, Perioperative, and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA02115
| | - Tian Xie
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang311121, China
| |
Collapse
|
45
|
Liu X, Shao Y, Li Y, Chen Z, Shi T, Tong Q, Zou X, Ju L, Pan J, Zhuang R, Pan X. Extensive Review of Nanomedicine Strategies Targeting the Tumor Microenvironment in PDAC. Int J Nanomedicine 2025; 20:3379-3406. [PMID: 40125427 PMCID: PMC11927507 DOI: 10.2147/ijn.s504503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers in the world, mainly because of its powerful pro-connective tissue proliferation matrix and immunosuppressive tumor microenvironment (TME), which promote tumor progression and metastasis. In addition, the extracellular matrix leads to vascular collapse, increased interstitial fluid pressure, and obstruction of lymphatic return, thereby hindering effective drug delivery, deep penetration, and immune cell infiltration. Therefore, reshaping the TME to enhance tumor perfusion, increase deep drug penetration, and reverse immune suppression has become a key therapeutic strategy. Traditional therapies for PDAC, including surgery, radiation, and chemotherapy, face significant limitations. Surgery is challenging due to tumor location and growth, while chemotherapy and radiation are hindered by the dense extracellular matrix and immunosuppressive TME. In recent years, the advancement of nanotechnology has provided new opportunities to improve drug efficacy. Nanoscale drug delivery systems (NDDSs) provide several advantages, including improved drug stability in vivo, enhanced tumor penetration, and reduced systemic toxicity. However, the clinical translation of nanotechnology in PDAC therapy faces several challenges. These include the need for precise targeting and control over drug release, potential immune responses to the nanocarriers, and the scalability and cost-effectiveness of production. This article provides an overview of the latest nanobased methods for achieving better therapeutic outcomes and overcoming drug resistance. We pay special attention to TME-targeted therapy in the context of PDAC, discuss the advantages and limitations of current strategies, and emphasize promising new developments. By emphasizing the enormous potential of NDDSs in improving the treatment outcomes of patients with PDAC, while critically discussing the limitations of traditional therapies and the challenges faced by nanotechnology in achieving clinical breakthroughs, our review paves the way for future research in this rapidly developing field.
Collapse
Affiliation(s)
- Xing Liu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 311400, People’s Republic of China
| | - Yidan Shao
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Yunjiang Li
- Radiology Department, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Zuhua Chen
- Radiology Department, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Tingting Shi
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Qiao Tong
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Xi Zou
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Liping Ju
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Jinming Pan
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Rangxiao Zhuang
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| | - Xuwang Pan
- Department of Pharmaceutical Preparation, Affiliated Hangzhou Xixi Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310023, People’s Republic of China
| |
Collapse
|
46
|
Zhang J, Yang Z, Liu Y, Liu Y, Qu J, Pan X. Recent Advances in Smart Linkage Strategies for Developing Drug Conjugates for Targeted Delivery. Top Curr Chem (Cham) 2025; 383:13. [PMID: 40080285 DOI: 10.1007/s41061-025-00497-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/16/2025] [Indexed: 03/15/2025]
Abstract
Targeted drug delivery systems effectively solve the problem of off-target toxicity of chemotherapeutic drugs by combining chemotherapeutic drugs with antibodies or peptides, thereby promoting drug targeting to the tumor site and bringing further hope for cancer treatment. The development of stimulus-responsive smart linkage technologies has led to the emergence of drug conjugates. Linkage technologies play a crucial role in the design, synthesis, and in vivo circulation of drug conjugates, as they determine the release of cytotoxic drugs from the conjugates and their subsequent therapeutic efficacy. This article reviews some of the smart linkage strategies used in designing drug conjugates, with a focus on the tumor microenvironment and exogenous stimuli as conditions influencing controlled drug release. This review introduces linker classifications and cleavage mechanisms, discusses modular linkers that promote the efficient synthesis of conjugates, and discusses the differences between linkage strategies. Furthermore, this article focuses on the implementation of self-assembly in drug conjugates, which is currently of great interest. Related concepts are introduced and relevant examples of their applications are provided. Furthermore, a comprehensive discourse is presented on the challenges that may arise in the research and clinical implementation of diverse linkage strategies, along with the associated enhancement measures. Finally, the factors that should be considered when designing linkage strategies for drug conjugates are summarized, offering strategies and ideas for scientists involved in drug conjugate research. It is particularly noteworthy that appropriate linkage strategies allow for the intracellular release of drugs after internalization of the conjugates, thereby maximizing their tumor cell-killing effect.
Collapse
Affiliation(s)
- Jie Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zeyu Yang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yu Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yuying Liu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jingkun Qu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
47
|
Fan S, Wang W, Che W, Xu Y, Jin C, Dong L, Xia Q. Nanomedicines Targeting Metabolic Pathways in the Tumor Microenvironment: Future Perspectives and the Role of AI. Metabolites 2025; 15:201. [PMID: 40137165 PMCID: PMC11943624 DOI: 10.3390/metabo15030201] [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: 01/30/2025] [Revised: 02/19/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Tumor cells engage in continuous self-replication by utilizing a large number of resources and capabilities, typically within an aberrant metabolic regulatory network to meet their own demands. This metabolic dysregulation leads to the formation of the tumor microenvironment (TME) in most solid tumors. Nanomedicines, due to their unique physicochemical properties, can achieve passive targeting in certain solid tumors through the enhanced permeability and retention (EPR) effect, or active targeting through deliberate design optimization, resulting in accumulation within the TME. The use of nanomedicines to target critical metabolic pathways in tumors holds significant promise. However, the design of nanomedicines requires the careful selection of relevant drugs and materials, taking into account multiple factors. The traditional trial-and-error process is relatively inefficient. Artificial intelligence (AI) can integrate big data to evaluate the accumulation and delivery efficiency of nanomedicines, thereby assisting in the design of nanodrugs. Methods: We have conducted a detailed review of key papers from databases, such as ScienceDirect, Scopus, Wiley, Web of Science, and PubMed, focusing on tumor metabolic reprogramming, the mechanisms of action of nanomedicines, the development of nanomedicines targeting tumor metabolism, and the application of AI in empowering nanomedicines. We have integrated the relevant content to present the current status of research on nanomedicines targeting tumor metabolism and potential future directions in this field. Results: Nanomedicines possess excellent TME targeting properties, which can be utilized to disrupt key metabolic pathways in tumor cells, including glycolysis, lipid metabolism, amino acid metabolism, and nucleotide metabolism. This disruption leads to the selective killing of tumor cells and disturbance of the TME. Extensive research has demonstrated that AI-driven methodologies have revolutionized nanomedicine development, while concurrently enabling the precise identification of critical molecular regulators involved in oncogenic metabolic reprogramming pathways, thereby catalyzing transformative innovations in targeted cancer therapeutics. Conclusions: The development of nanomedicines targeting tumor metabolic pathways holds great promise. Additionally, AI will accelerate the discovery of metabolism-related targets, empower the design and optimization of nanomedicines, and help minimize their toxicity, thereby providing a new paradigm for future nanomedicine development.
Collapse
Affiliation(s)
| | | | | | | | | | - Lei Dong
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (S.F.); (W.W.); (W.C.); (Y.X.); (C.J.)
| | - Qin Xia
- State Key Laboratory of Molecular Medicine and Biological Diagnosis and Treatment (Ministry of Industry and Information Technology), Aerospace Center Hospital, School of Life Science, Beijing Institute of Technology, Beijing 100081, China; (S.F.); (W.W.); (W.C.); (Y.X.); (C.J.)
| |
Collapse
|
48
|
Chen D, Du Y, Wang X, Li H, Wu X, Kuang X, Li C, Zhao J, Xiong Y, Sun M, Tu J, Liu S, Sun C. Phase-separating Pt(IV)-graft-glycopeptides sequentially sensing pH and redox for deep tumor penetration and targeting chemotherapy. J Control Release 2025; 379:743-756. [PMID: 39832748 DOI: 10.1016/j.jconrel.2025.01.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/23/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025]
Abstract
Active-targeting nanomedicines have been widely employed in cancer treatment for increasing therapeutic index. However, the limited permeability caused by the binding site barrier (BSB) and size hindrances compromises their clinical antitumor efficacy in patients. Herein, learning from the liquid-liquid phase separation (LLPS) of bio-macromolecules, we report phase-separating glycopeptides (HEP) from polyhistidine (PHis) grafted hyaluronic acid (HA), which can sense the tumor extracellular pH and concomitantly overcome size and BSB dilemmas for enhanced tumor penetration. HEP aggregates into nanodroplets in solution at neutral pH. Upon reaching the acidic extracellular environment of tumors, the pH-responsive PHis triggers a phase separation, converting the coacervate nanodroplets into monomeric glycopeptides. This enables HEP conjugated with the platinum prodrug (HEPPt) to deeply penetrate into tumors by overcoming the BSB effect arising from the interaction between nanodroplets and cluster of differentiation 44 (CD44), as well as resolving the size challenges. Moreover, HEPPt in monomeric states exhibits promoted cellular uptake after pH-triggered phase separation, attributed to the transmembrane effect of exposed PHis. Subsequently, the rapid release of Pt(II), triggered by tumor intracellular reducing environment, exerts excellent antitumor activity. The phase-separating glycopeptides represent a promising platform for improving tumor penetration and intracellular delivery of therapeutic agents.
Collapse
Affiliation(s)
- Dali Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; Department of Pharmaceutics, School of Pharmacy, 639 Longmian Avenue, Nanjing 211198, China
| | - Yunai Du
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; Department of Pharmaceutics, School of Pharmacy, 639 Longmian Avenue, Nanjing 211198, China
| | - Xitong Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; Department of Pharmaceutics, School of Pharmacy, 639 Longmian Avenue, Nanjing 211198, China
| | - Huihong Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; Department of Pharmaceutics, School of Pharmacy, 639 Longmian Avenue, Nanjing 211198, China
| | - Xinjiao Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; Department of Pharmaceutics, School of Pharmacy, 639 Longmian Avenue, Nanjing 211198, China
| | - Xiaoqin Kuang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; Department of Pharmaceutics, School of Pharmacy, 639 Longmian Avenue, Nanjing 211198, China
| | - Chunjiayu Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; Department of Pharmaceutics, School of Pharmacy, 639 Longmian Avenue, Nanjing 211198, China
| | - Jianing Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; Department of Pharmaceutics, School of Pharmacy, 639 Longmian Avenue, Nanjing 211198, China
| | - Yerong Xiong
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; Department of Pharmaceutics, School of Pharmacy, 639 Longmian Avenue, Nanjing 211198, China
| | - Minjie Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; Department of Pharmaceutics, School of Pharmacy, 639 Longmian Avenue, Nanjing 211198, China.
| | - Jiasheng Tu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; Department of Pharmaceutics, School of Pharmacy, 639 Longmian Avenue, Nanjing 211198, China.
| | - Siyan Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; Research Center for Traceability and Standardization of TCMs, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China.
| | - Chunmeng Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 639 Longmian Avenue, Nanjing 211198, China; NMPA Key Laboratory for Research and Evaluation of Pharmaceutical Preparations and Excipients, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China; Department of Pharmaceutics, School of Pharmacy, 639 Longmian Avenue, Nanjing 211198, China.
| |
Collapse
|
49
|
Roser SM, Munarin F, Polucha C, Minor AJ, Choudhary G, Coulombe KLK. Customized Heparinized Alginate and Collagen Hydrogels for Tunable, Local Delivery of Angiogenic Proteins. ACS Biomater Sci Eng 2025; 11:1612-1628. [PMID: 39945764 DOI: 10.1021/acsbiomaterials.4c01823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2025]
Abstract
Therapeutic protein delivery has ushered in a promising new generation of disease treatment, garnering more recognition for its clinical potential than ever. However, proteins' limited stability, extremely short average half-lives, and evidenced toxicity following systemic delivery continue to undercut their efficacy. Biomaterial-based protein delivery, however, demonstrates the potential to overcome these obstacles. To this end, we have developed a heparinized alginate and collagen hydrogel for the local, sustained delivery of therapeutic proteins. In an effort to match this ubiquitous application of protein delivery to various disease states and target tissues with sufficient versatility, we identified three distinct delivery modes as design targets. A shear-thinning, low-viscosity injectable for minimal tissue damage, a higher-viscosity gel plug for subcutaneous injection, and a submillimeter-thickness film for solid-form implantation were optimized and characterized in this work. In vitro assessments confirmed feasible injection control, mechanical stability for up to 6 h of unsubmerged storage, and isotropic early collagen fibril assembly. Release kinetics were assessed both in vitro and in vivo, demonstrating up to 14 days of functional vascular endothelial growth factor delivery. Rodent models of pulmonary hypertension, subcutaneous injection, and myocardial infarction, three promising applications of protein therapeutics, were used to assess the feasible delivery and biocompatibility of the injectable gel, gel plug, and film, respectively. Histological evaluation of the delivered materials and surrounding tissue showed high biocompatibility with cell and blood vessel infiltration, remodeling, and integration with the host tissue. Our successful customization of the biomaterial to heterogeneous delivery modes demonstrates its versatile capacity for the local, sustained delivery of therapeutic proteins for a diverse array of regenerative medicine applications.
Collapse
Affiliation(s)
- Stephanie M Roser
- School of Engineering, Institute for Biology, Engineering, and Medicine, Brown University, Providence, Rhode Island 02912, United States
| | - Fabiola Munarin
- School of Engineering, Institute for Biology, Engineering, and Medicine, Brown University, Providence, Rhode Island 02912, United States
| | - Collin Polucha
- School of Engineering, Institute for Biology, Engineering, and Medicine, Brown University, Providence, Rhode Island 02912, United States
| | - Alicia J Minor
- School of Engineering, Institute for Biology, Engineering, and Medicine, Brown University, Providence, Rhode Island 02912, United States
| | - Gaurav Choudhary
- Division of Cardiology, Providence VA Medical Center, Providence, Rhode Island 02908, United States
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island 02903, United States
| | - Kareen L K Coulombe
- School of Engineering, Institute for Biology, Engineering, and Medicine, Brown University, Providence, Rhode Island 02912, United States
- Cardiovascular Research Center, Cardiovascular Institute, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, Rhode Island 02903, United States
| |
Collapse
|
50
|
Akhtar S, Zuhair F. Advancing Nanomedicine Through Electron Microscopy: Insights Into Nanoparticle Cellular Interactions and Biomedical Applications. Int J Nanomedicine 2025; 20:2847-2878. [PMID: 40078651 PMCID: PMC11899938 DOI: 10.2147/ijn.s500978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/16/2025] [Indexed: 03/14/2025] Open
Abstract
Nanomedicine has revolutionized cancer treatment by the development of nanoparticles (NPs) that offer targeted therapeutic delivery and reduced side effects. NPs research in nanomedicine significantly focuses on understanding their cellular interactions and intracellular mechanisms. A precise understanding of nanoparticle interactions at the subcellular level is crucial for their effective application in cancer therapy. Electron microscopy has proven essential, offering high-resolution insights into nanoparticle behavior within biological systems. This article reviews the role of electron microscopy in elucidating the cellular uptake and intracellular interactions of NPs. Transmission electron microscopy (TEM) provides imaging capabilities, such as cryo three-dimensional tomography, which offer in-depth insights into nanoparticle localization, endocytosis pathways, and subcellular interactions, while high resolution-TEM is primarily used for studying the atomic structure of isolated NPs rather than nanoparticles within cells or tissues. On the other hand, scanning electron microscopy (SEM) is ideal for examining larger surface areas and provides a broader perspective on the morphology and topography of the samples. The review highlights the advantages of electron microscopy in visualizing nanoparticle interactions with cellular structures and tracking their mechanisms of action. It also addresses the challenges associated with electron microscopy characterization, such as tedious sample preparation, static imaging limitations, and a restricted field of view. By examining various nanoparticle uptake pathways, and cellular destination of NPs with examples, the article emphasizes the importance of these pathways to optimize nanoparticle design and enhance therapeutic efficacy. This review underscores the need for continued advancement in electron microscopy techniques to improve the effectiveness of nanomedicine and address existing challenges. In summary, electron microscopy is a key tool for advancing our understanding of nanoparticle behavior in biological contexts, aiding in the design and optimization of nanomedicines by providing insights into nanoparticle cellular dynamics, uptake mechanisms, and therapeutic applications.
Collapse
Affiliation(s)
- Sultan Akhtar
- Department of Biophysics Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - Fatimah Zuhair
- Department of Infection Control, Alzahra General Hospital, Qatif, 31911, Saudi Arabia
| |
Collapse
|