1
|
Xu S, Zhang Y, Li J, Zhang X, Wang W. External stimuli-responsive drug delivery to the posterior segment of the eye. Drug Deliv 2025; 32:2476140. [PMID: 40126105 PMCID: PMC11934192 DOI: 10.1080/10717544.2025.2476140] [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: 01/07/2025] [Revised: 02/24/2025] [Accepted: 02/28/2025] [Indexed: 03/25/2025] Open
Abstract
Posterior segment eye diseases represent the leading causes of vision impairment and blindness globally. Current therapies still have notable drawbacks, including the need for frequent invasive injections and the associated risks of severe ocular complications. Recently, the utility of external stimuli, such as light, ultrasound, magnetic field, and electric field, has been noted as a promising strategy to enhance drug delivery to the posterior segment of the eye. In this review, we briefly summarize the main physiological barriers against ocular drug delivery, focusing primarily on the recent advancements that utilize external stimuli to improve treatment outcomes for posterior segment eye diseases. The advantages of these external stimuli-responsive drug delivery strategies are discussed, with illustrative examples highlighting improved tissue penetration, enhanced control over drug release, and targeted drug delivery to ocular lesions through minimally invasive routes. Finally, we discuss the challenges and future perspectives in the translational research of external stimuli-responsive drug delivery platforms, aiming to bridge existing gaps toward clinical use.
Collapse
Affiliation(s)
- Shuting Xu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| | - Yaming Zhang
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| | - Jia Li
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| | - Xinyu Zhang
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| | - Weiping Wang
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Laboratory of Molecular Engineering and Nanomedicine, Dr. Li Dak-Sum Research Centre, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
2
|
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
|
3
|
Shinde S, Shah S, Famta P, Wagh S, Pandey G, Sharma A, Vambhurkar G, Jain A, Srivastava S. Next-Generation Transformable Nanomedicines: Revolutionizing Cancer Drug Delivery and Theranostics. Mol Pharm 2025. [PMID: 40317253 DOI: 10.1021/acs.molpharmaceut.4c01495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Nanomedicine has significantly advanced the treatment of various cancer phenotypes, addressing numerous challenges associated with conventional therapies. Researchers have extensively investigated the physicochemical properties of nanocarriers, such as charge, morphology, and surface chemistry, to optimize drug delivery systems. In the context of transformable nanomedicine, these properties are particularly critical for overcoming existing limitations, including suboptimal blood circulation times, sequestration by the reticuloendothelial system and mononuclear phagocyte system, and inefficient targeting of the tumor microenvironment (TME). Alterations in nanocarrier geometry, surface charge, and hydrophilicity have shown potential in mitigating these barriers, offering improved therapeutic outcomes and enhanced biomedical applications. This review explores controlled modulation of these properties in the context of anticancer therapy, offering an in-depth exploration of transformable strategies activated by both internal and external stimuli. We analyze the implications of these tunable characteristics on pharmacokinetics, biodistribution, and targeted delivery to the TME. Additionally, we address the current challenges in the clinical translation of these advanced nanocarriers and propose strategies to overcome these obstacles to enhance the clinical feasibility of nanomedicine-based cancer therapies.
Collapse
Affiliation(s)
- Swapnil Shinde
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Saurabh Shah
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Paras Famta
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Suraj Wagh
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Giriraj Pandey
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Abhishek Sharma
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Ganesh Vambhurkar
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Akshita Jain
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| | - Saurabh Srivastava
- Pharmaceutical Innovation and Translational Research Lab (PITRL), Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500037, India
| |
Collapse
|
4
|
Grayson W, Brown NM. Recent advances in the application of nanotechnology in joint arthroplasty: a narrative review. ANNALS OF JOINT 2025; 10:13. [PMID: 40385689 PMCID: PMC12082184 DOI: 10.21037/aoj-24-50] [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: 10/04/2024] [Accepted: 04/09/2025] [Indexed: 05/20/2025]
Abstract
Background and Objective Osteoarthritis (OA) is a multifactorial disease, involving biomechanical, inflammatory, and metabolic processes that ultimately impact the structure and function of a joint. Current therapeutic options can improve symptoms and prolong the time to surgery, yet they are not curative and are limited by their systemic side-effects and their inability to provide site-specific delivery. Nanomedicine takes advantage of the unique properties held by technology on the nanoscale (1-100 nm), including surface effects and quantum effects, that allow for novel mechanical, thermal, and magnetic functions. The primary aim of this narrative review is to summarize the recent advances made in nanotechnology and their uses in joint arthroplasty. Methods This narrative review was performed following a computerized search of the electronic database on PubMed in September 2024. Papers related to the use of nanotechnology in orthopaedic arthroplasty surgery were included for review. Key Context and Findings Nanotechnology holds the promise of optimizing OA treatment, refining the implants used during joint arthroplasty, and aiding in the diagnosis and treatment of post-operative joint infections. With the increasingly aging population and growing demand for joint replacement, this review aims to cover the novel applications of nanoparticles (NPs) within the realm of joint replacement surgery. Conclusions Future studies are needed to further investigate the clinical translation of NPs in joint arthroplasty. Additionally, the potential of NPs needs to be considered within their limitations and their safety profile that is still being defined.
Collapse
Affiliation(s)
- Whisper Grayson
- Department of Orthopaedic Surgery & Rehabilitation, Loyola University Health System, Maywood, IL, USA
| | - Nicholas M Brown
- Department of Orthopaedic Surgery & Rehabilitation, Loyola University Health System, Maywood, IL, USA
| |
Collapse
|
5
|
Mahata R, Manna S, Modak M, Choudhury SM. A review on the advancement of polydopamine (PDA)-based nanomaterials for cancer treatment. Med Oncol 2025; 42:165. [PMID: 40237855 DOI: 10.1007/s12032-025-02678-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/11/2025] [Indexed: 04/18/2025]
Abstract
The significance of cancer treatment research lies in addressing the high incidence of cancer, overcoming treatment challenges, and mitigating the harsh side effects of chemotherapeutic agents. Currently, nanotechnology is garnering significant attention for its potential applications in diagnostics and drug delivery, offering innovative solutions for disease detection and treatment. Among different types of nanoparticles (NPs), polymeric nanoparticles comprise biocompatible and biodegradable polymers that enhance drug pharmacokinetics and pharmacodynamics, minimize adverse effects, increase stability, and facilitate sustained drug release. These polymeric nanoparticle-based nanomedicines offer a versatile platform for various cancer treatments, notably enabling targeted drug delivery directly to tumors, tumor-imaging, hyperthermia, and photodynamic therapy. Being polymeric in nature polydopamine (PDA) nanomaterials are appeared as promising approaches in biology and medicine. This review article offers a concise summary of the latest developments in polydopamine-based cancer treatment, covering key findings, limitations, and emerging trend therapeutic approach of polydopamine nanomaterials, along with the properties and various methods of preparation. Physico-chemical properties of PDA-based nanomaterials in therapeutics have permitted several successful modifications in the field of cancer treatment.
Collapse
Affiliation(s)
- Rumi Mahata
- Department of Human Physiology, Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Sounik Manna
- Department of Human Physiology, Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Mrinmoyee Modak
- Department of Human Physiology, Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Sujata Maiti Choudhury
- Department of Human Physiology, Biochemistry, Molecular Endocrinology and Reproductive Physiology Laboratory, Vidyasagar University, Midnapore, West Bengal, 721102, India.
| |
Collapse
|
6
|
Xia Q, Zhou S, Zhou J, Zhao X, Saif MS, Wang J, Hasan M, Zhao M, Liu Q. Recent Advances and Challenges for Biological Materials in Micro/Nanocarrier Synthesis for Bone Infection and Tissue Engineering. ACS Biomater Sci Eng 2025; 11:1945-1969. [PMID: 40067283 DOI: 10.1021/acsbiomaterials.4c02118] [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: 04/15/2025]
Abstract
Roughly 1.71 billion people worldwide suffer from large bone abnormalities, which are the primary cause of disability. Traditional bone grafting procedures have several drawbacks that impair their therapeutic efficacy and restrict their use in clinical settings. A great deal of work has been done to create fresh, more potent strategies. Under these circumstances, a crucial technique for the regeneration of major lesions has emerged: bone tissue engineering (BTE). BTE involves the use of biomaterials that can imitate the natural design of bone. To yet, no biological material has been able to fully meet the parameters of the perfect implantable material, even though several varieties have been created and investigated for bone regeneration. Against this backdrop, researchers have focused a great deal of interest over the past few years on the subject of nanotechnology and the use of nanostructures in regenerative medicine. The ability to create nanoengineered particles that can overcome the current constraints in regenerative strategies─such as decreased cell proliferation and differentiation, insufficient mechanical strength in biological materials, and insufficient production of extrinsic factors required for effective osteogenesis has revolutionized the field of bone and tissue engineering. The effects of nanoparticles on cell characteristics and the application of biological materials for bone regeneration are the main topics of our review, which summarizes the most recent in vitro and in vivo research on the application of nanotechnology in the context of BTE.
Collapse
Affiliation(s)
- Qipeng Xia
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
| | - Shuyan Zhou
- School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, PR China
| | - Jingya Zhou
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- College of Acupuncture and Massage, Jiangxi University of Traditional Chinese Medicine, Nanchang 330004, PR China
| | - Xia Zhao
- Faculty of Medicine, Dalian University of Technology, Dalian 116024, PR China
| | - Muhammad Saqib Saif
- Department of Biochemistry, Faculty of Chemical and Biological Sciences, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Jianping Wang
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
| | - Murtaza Hasan
- Department of Biotechnology, Faculty of Chemical and Biological Sciences, The Islamia University of Bahawalpur, Bahawalpur 63100, Pakistan
| | - Min Zhao
- Yingtan People's Hospital, Nanchang University, Yingtan 335499, PR China
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
| | - Qiang Liu
- Medical Faculty of Dalian University of Technology-Yingtan People's Hospital Joint Research Center, Yingtan 335499, PR China
- Faculty of Medicine, Dalian University of Technology, Dalian 116024, PR China
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Wang W, Wang S, Xu S, Chai R, Yuan J, Zhang H, Li Y, Pu X, Li X, Sun J, He Z, Sun B. An assembly modules deformation strategy improved the chemical stability and self-assembly stability of docetaxel prodrugs nanoassemblies. NANOSCALE 2025; 17:7016-7029. [PMID: 39982137 DOI: 10.1039/d4nr05002a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Self-assembly prodrugs usually consist of drug modules, activation modules, and assembly modules. The selection of suitable modules to construct prodrug nanoassemblies with self-assembly stability and "intelligent" activation is a challenge. As a common assembly module, oleic acid can provide a driving force and steric hindrance for prodrugs self-assembly. However, the unsaturated double bond of oleic acid is readily oxidized and it affects its chemical stability. Herein, two docetaxel (DTX) prodrugs were designed using disulfide bonds as activation modules and two different fatty acids (isostearic acid and oleic acid) as assembly modules, respectively. Compared with oleic acid, isostearic acid had higher chemical stability. Simultaneously, the terminal propyl structure of isostearic acid compensated for the steric hindrance without a double bond. Overall, this structural deformation improved the self-assembly ability and chemical stability of the prodrug nanoassemblies, thus balancing the effectiveness and safety of the prodrugs. Our findings reveal the importance of the assembly modules and provide a guidance for the rational design of prodrug nanoassemblies.
Collapse
Affiliation(s)
- Wenjing Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, 103 Wenhua Road, Shenyang, 110016, China.
| | - Shuo Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, 103 Wenhua Road, Shenyang, 110016, China.
| | - Shengyao Xu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, 103 Wenhua Road, Shenyang, 110016, China.
| | - Rong Chai
- Peking Union Second Pharmaceutical Factory Ltd, Beijing 102600, China
| | - Jun Yuan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, 103 Wenhua Road, Shenyang, 110016, China.
| | - Hao Zhang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, 103 Wenhua Road, Shenyang, 110016, China.
| | - Yaqi Li
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, 103 Wenhua Road, Shenyang, 110016, China.
| | - Xiaohui Pu
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Xin Li
- Department of Respiratory Disease, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, 103 Wenhua Road, Shenyang, 110016, China.
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| | - Zhonggui He
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, 103 Wenhua Road, Shenyang, 110016, China.
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, N. Jinming Ave., Kaifeng 475004, China
| | - Bingjun Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, No. 59 Mailbox, 103 Wenhua Road, Shenyang, 110016, China.
- Joint International Research Laboratory of Intelligent Drug Delivery Systems, Ministry of Education, China
| |
Collapse
|
10
|
Ding H, Su L, Xie Z, Castano AD, Li Y, Perez LR, Chen J, Luo K, Tian X, Battaglia G. Morphological insights in oxidative sensitive nanocarrier pharmacokinetics, targeting, and photodynamic therapy. J Mater Chem B 2025; 13:3852-3863. [PMID: 39946164 DOI: 10.1039/d4tb02194k] [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: 03/17/2025]
Abstract
Nanoparticle (NP) morphology holds significant importance in nanomedicine, particularly concerning its implications for biological responses. This study investigates the impact of synthesizing polymers with varying degrees of methionine (MET) polymerization on three distinct drug delivery systems: spherical micelles, worm-like micelles, and vesicles, all loaded with the photosensitizer chlorin e6 (Ce6). We analyzed their distribution at both cellular and animal levels, revealing how NP morphology influences cellular uptake, subcellular localization, penetration of multicellular spheroids, blood half-life, and biodistributions across major organs. Employing a physiologically based pharmacokinetic (PBPK) model enabled us to simulate diverse distribution patterns and quantify the targeting efficiency of NPs toward tumors. Our investigation elucidates that spherical micelles exhibit lower accumulation levels within the reticuloendothelial system, potentially mitigating adverse side effects despite their higher glomerular filtration rate. This nuanced understanding underscores the complex interplay between NP morphology and biological responses, providing valuable insights into optimizing therapeutic efficacy while minimizing undesirable effects. We thus report the integration of experimental analyses with PBPK modeling to elucidate the topological characteristics of NP, thereby shedding light on their distribution patterns, therapeutic efficacy, and potential side effects.
Collapse
Affiliation(s)
- Haitao Ding
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
| | - Liping Su
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
| | - Zhendong Xie
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
- Institute for Bioengineering of Catalunya (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I Reixac, 08028, Barcelona, Spain.
- Department of Electronic and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
| | - Aroa Duro Castano
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK
| | - Yunkun Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
| | - Lorena Ruiz Perez
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
- Institute for Bioengineering of Catalunya (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I Reixac, 08028, Barcelona, Spain.
- Department of Applied Physics, University of Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
| | - Junyang Chen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
- Institute for Bioengineering of Catalunya (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I Reixac, 08028, Barcelona, Spain.
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, 610041, China
| | - Xiaohe Tian
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
| | - Giuseppe Battaglia
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, Sichuan Province, China.
- Institute for Bioengineering of Catalunya (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona (Spain), Carrer Baldiri I Reixac, 08028, Barcelona, Spain.
- Department of Electronic and Biomedical Engineering, University of Barcelona, Martí i Franquès 1, 08028, Barcelona, Spain
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK
- Catalan Institution for Research and Advanced Studies (ICREA), Passeig de Lluís Companys, 23, 08010, Barcelona, Spain
| |
Collapse
|
11
|
Yu N, Xu Y, Sun Q, Ge Y, Guo Y, Chen M, Shan H, Zheng M, Chen Z, Zhao S, Chen X. Size-specific clonidine-loaded liposomes: Advancing melanoma microenvironment suppression with safety and precision. J Control Release 2025; 379:120-134. [PMID: 39756687 DOI: 10.1016/j.jconrel.2025.01.001] [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: 09/02/2024] [Revised: 11/26/2024] [Accepted: 01/01/2025] [Indexed: 01/07/2025]
Abstract
The immunosuppressive tumor microenvironment (TME) plays a crucial role in the progression and treatment resistance of melanoma. Modulating the TME is thus a key strategy for enhancing therapeutic outcomes. Previousstudies have identified clonidine (CLD), an α2-adrenergic receptor agonist, as a promising agent that enhances T lymphocyte infiltration and reduces myeloid-derived suppressor cells within the TME, thereby promoting antitumor immune responses. In this study, we discovered that CLD reshaped the melanoma immune microenvironment, facilitating T-cell activation and exerting antitumor effects. However, the high doses of CLD required for effective TME modulation pose significant toxicity concerns, limiting its clinical applicability. To address this, we employed the controllable cavitation-on-a-chip (CCC) platform to formulate CLD-loaded liposomes and optimize their size. This approach aimed to enhance the precision and efficacy of drug delivery while reducing systemic side effects. Our results demonstrated that size-specific CLD liposomes, particularly those at 50 nm, significantly improved tumor growth inhibition and immune cell infiltration within the TME. Moreover, these optimized liposomes mitigate adverse effects associated with high-dose CLD treatment. This study indicates the potential of CCC-optimized CLD liposomes as a safer and more effective melanoma therapy, highlighting the critical interplay between liposome size control and therapeutic outcomes in cancer treatment.
Collapse
Affiliation(s)
- Nianzhou Yu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yantao Xu
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qi Sun
- Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China; School of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Yi Ge
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yeye Guo
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Maike Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Han Shan
- Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China; School of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Mingde Zheng
- Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China; School of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China
| | - Zeyu Chen
- Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China; School of Mechanical and Electrical Engineering, Central South University, Changsha 410083, China.
| | - Shuang Zhao
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China.
| | - Xiang Chen
- Department of Dermatology, Hunan Engineering Research Center of Skin Health and Disease, Hunan Key Laboratory of Skin Cancer and Psoriasis, Xiangya Hospital, Central South University, Changsha, Hunan, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China; Furong Laboratory (Precision Medicine), Changsha 410008, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
12
|
Son Phan K, Nghi Do H, Thuy Doan B, Thu Huong Le T, Thu Trang Mai T, Bao Ngoc Nguyen Q, Nham Dong T, Hung Bui Ha B, Dung Dang V, Dang LH, Quyen Tran N, Thu Ha P. The Influence of Cyanine 5.5 and Doxorubicin on Cell Cycle Arrest, Magnetic Resonance, and Near-Infrared Fluorescence Optical Imaging for Fe 3O 4-Encapsulated PLA-TPGS Nanoparticles. ChemMedChem 2025; 20:e202400586. [PMID: 39568159 DOI: 10.1002/cmdc.202400586] [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: 07/30/2024] [Revised: 11/06/2024] [Accepted: 11/20/2024] [Indexed: 11/22/2024]
Abstract
The combination of magnetic resonance imaging (MRI)/near-infrared (NIR) fluorescence signals and chemotherapy agents has been developed for cancer diagnosis and treatment. In this work, we investigated the impacts of Cyanine 5.5 and Doxorubicin on cell cycle arrest, magnetic resonance, and NIR fluorescence optical imaging for Fe3O4-encapsulated nanosystems based on poly(lactide)-tocopheryl polyethylene glycol succinate (PLA-TPGS) copolymer. Although Cyanine 5.5 and Fe3O4 nanoparticles (NPs) are less cytotoxic than Doxorubicin, they present a cytostatic effect, inducing cell cycle arrest at the G2/M phase in human brain adenocarcinoma (CCF-STTG1) cells. For MRI applications, the permeability of the PLA-TPGS copolymer coating layer to water molecules might lengthen the translational diffusion time (τ D ${{{{\bf\tau}}}_{{\bf D}}}$ ), causing the higher relaxivity ratio (r2/r1) compared to bare Fe3O4 NPs under an applied magnetic field (7 Tesla). Notably, the chemical structures of Cyanine 5.5 and Doxorubicin significantly contribute to the enhancement of the T2 relaxivities of Fe3O4 NPs through π-π and ρ-π conjugation. Furthermore, the radiance ratio and signal-to-noise ratio enhancement and a slight blue shift in the optimal excitation and emission wavelengths were recorded. These findings show the potential for in vivo MRI and NIR bioimaging experiments of the nanoparticles.
Collapse
Affiliation(s)
- Ke Son Phan
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Huu Nghi Do
- Institute of Natural Products Chemistry, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Bich Thuy Doan
- The Institute I-CLeHS Institute of Chemistry for Life and Health Sciences, ENSCP Chimie ParisTech, PSL Université, CNRS UMR, 8060, Paris, France
| | - Thi Thu Huong Le
- Faculty of Natural Resources and Environment, Vietnam National University of Agriculture, Trau Quy, Gia Lam District, Hanoi, Vietnam
| | - Thi Thu Trang Mai
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Quynh Bao Ngoc Nguyen
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Thi Nham Dong
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Bao Hung Bui Ha
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Viet Dung Dang
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| | - Le Hang Dang
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, HCMC, VietNam
| | - Ngoc Quyen Tran
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, HCMC, VietNam
| | - Phuong Thu Ha
- Institute of Materials Science, Vietnam Academy of Science and Technology, 18 Hoang Quoc Viet Road, Cau Giay District, Hanoi, Vietnam
| |
Collapse
|
13
|
Sivakumar PM, Zarepour A, Akhter S, Perumal G, Khosravi A, Balasekar P, Zarrabi A. Anionic polysaccharides as delivery carriers for cancer therapy and theranostics: An overview of significance. Int J Biol Macromol 2025; 294:139211. [PMID: 39732249 DOI: 10.1016/j.ijbiomac.2024.139211] [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: 07/24/2024] [Revised: 11/13/2024] [Accepted: 12/24/2024] [Indexed: 12/30/2024]
Abstract
Recently, cancer therapy has witnessed remarkable advancements with a growing focus on precision medicine and targeted drug delivery strategies. The application of anionic polysaccharides has gained traction in various drug delivery systems. Anionic polysaccharides have emerged as promising delivery carriers in cancer therapy and theranostics, offering numerous advantages such as biocompatibility, low toxicity, and the ability to encapsulate and deliver therapeutic agents to tumor sites with high specificity. This review underscores the significance of anionic polysaccharides as essential components of the evolving landscape of cancer therapy and theranostics. These polymers can be tailored to carry a wide range of therapeutic cargo, including chemotherapeutic agents, nucleic acids, and imaging agents. Their negative charge enables electrostatic interactions with positively charged drugs and facilitates the formation of stable nanoparticles, liposomes, or hydrogels for controlled drug release. Additionally, their hydrophilic nature aids in prolonging circulation time, reducing drug degradation, and minimizing off-target effects. Besides, some of them could act as targeting agents or therapeutic compounds that lead to improved therapeutic performance. This review offers valuable information for researchers, clinicians, and biomedical engineers. It provides insights into the recent progress in the applications of anionic polysaccharide-based delivery platforms in cancer theranostics to transform patient outcomes.
Collapse
Affiliation(s)
- Ponnurengam Malliappan Sivakumar
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam; School of Medicine and Pharmacy, Duy Tan University, Da Nang, Vietnam
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India.
| | - Sohail Akhter
- New Product Development, Global R&D, Sterile ops, TEVA Pharmaceutical Industries Ltd., Aston Ln N, Halton, Preston Brook, Runcorn WA7 3FA, UK.
| | - Govindaraj Perumal
- Department of Biomedical Engineering, School of Dental Medicine, University of Connecticut (UConn) Health, Farmington, CT 06030, USA.
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Türkiye
| | - Premkumar Balasekar
- Department of Pharmacology, K.K. College of Pharmacy, Affiliated to The Tamilnadu Dr. M.G.R. Medical University, Gerugambakkam 600128, India
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Türkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan.
| |
Collapse
|
14
|
Malinovskaya J, Kovshova T, Melnikov P, Li Z, Dhakal N, Knoll J, Valikhov M, Ermolenko Y, Chernysheva A, Gurina O, Chekhonin V, Wacker MG, Gelperina S. The second phase of tumor invasion driven by immune cells: A study on doxorubicin-loaded PLG nanoparticles. J Control Release 2025; 378:750-762. [PMID: 39724952 DOI: 10.1016/j.jconrel.2024.12.056] [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: 10/22/2024] [Revised: 12/01/2024] [Accepted: 12/21/2024] [Indexed: 12/28/2024]
Abstract
Poly(lactide-co-glycolide) (PLG) nanoparticles loaded with doxorubicin have reached phase-I clinical trials for treating advanced solid tumors. This study explores cell hitchhiking, where nanoparticles associate with blood cells and investigates the impact on pharmacokinetics and tumor migration. Previous findings highlighted the early post-injection phase dominated by nonspecific nanoparticle-cell interactions and burst release. In contrast, this study examines the subsequent phase of tumor invasion, emphasizing the role of immune cells, mostly neutrophils, in redistributing the carrier to the tumor site via blood cell hitchhiking. We provide a detailed investigation of nanoparticle extravasation kinetics and mechanisms, showing qualitative and quantitative evidence of increased nanoparticle association with immune cells over time. By 30 min post-injection, approximately 15 % of monocytes and 15-19 % of neutrophils tested positive for nanoparticles, with significant differences observed between ex vivo and in vivo experiments, and between healthy and tumor-bearing animals. This study underscores the ambiguous role of immune cell-mediated tumor targeting. While the total accumulation of the carrier rises, this fraction is partially trapped in immune cells without any chance to escape.
Collapse
Affiliation(s)
- Julia Malinovskaya
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047, Moscow, Russia
| | - Tatyana Kovshova
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047, Moscow, Russia
| | - Pavel Melnikov
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034, Moscow, Russia
| | - Zhuoxuan Li
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, 4 Science Drive 2, 117544, Singapore
| | - Namrata Dhakal
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, 4 Science Drive 2, 117544, Singapore
| | - Julian Knoll
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, 4 Science Drive 2, 117544, Singapore
| | - Marat Valikhov
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034, Moscow, Russia
| | - Yulia Ermolenko
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047, Moscow, Russia
| | - Anastasia Chernysheva
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034, Moscow, Russia
| | - Olga Gurina
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034, Moscow, Russia
| | - Vladimir Chekhonin
- Department of Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, 119034, Moscow, Russia
| | - Matthias G Wacker
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, 4 Science Drive 2, 117544, Singapore..
| | - Svetlana Gelperina
- D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, 125047, Moscow, Russia.
| |
Collapse
|
15
|
Echterhof A, Dharmaraj T, Blankenberg P, Targ B, Bollyky PL, Smith NM, Blankenberg F. Whole-body Bacteriophage Distribution Characterized by a Physiologically based Pharmacokinetic Model. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.06.636931. [PMID: 39975270 PMCID: PMC11839030 DOI: 10.1101/2025.02.06.636931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
In 2019 there were over 2.8 million cases of antibiotic-resistant bacterial infection in the US with gram negative organisms having up to a 6% rate of mortality. Bacteriophage (phage) therapy holds great promise to treat such infections. However, the biologic features which influence the pharmacokinetics (PK) of phage have been difficult to characterize due to a lack of standardized protocols of phage purification, tissue assay, and labeling. Here we present robust methods for ultrapure phage preparation as well as non-destructive highly stable attachment of radio-iodide to phage using the well described Sulfo-SHPP linker. We purified and radiolabeled the phage strains, PAML-31-1, OMKO1, and Luz24 lytic to drug-resistant Pseudomonas aeruginosa for biodistribution assay in normal young adult CD-1 mice injected via penile vein. Groups of 5 mice were euthanized and tissues/organs removed for weighing and scintillation well counting of I-125 activity at 30 min, 1h, 2h, 4h, 8h, and 24h. A physiologically based PK (PBPK) model was then constructed focusing on compartments describing blood, lung, muscle, bone, liver, stomach, spleen, small intestines, large intestines, and kidney. Model permeability coefficient (PS) was estimated across all organs as being 0.0227. Tissue partition coefficients (KP) were estimated for high perfusion organs (lung and kidney) as 0.000138, GI organs (liver, spleen, and stomach) as 0.627, and all other organs as 0.220. Elimination was governed by MPS-mediated elimination (TMPS,deg) and active secretion at epithelial barriers (CLActive), which were estimated as 0.00301 h and 0.0145 L/h/kg, respectively. Monte Caro simulations showed that the rapid elimination phage in humans is expected, resulting in phage blood concentrations being lower than 102 PFU/mL (limit of quantification by plaque assay) by 12 hours. As such, multi-dose regimens and continuous infusion regimens were the only strategies that allowed continuously detectible phage concentrations. Evaluation of different dose levels showed that at a maximum dose of 1012 PFU, phage concentrations are expected to be approximately 107 PFU/g. Our physiologically based PK model of phage represents the first rigorous pre-clinical assessment of phage PK utilizing contemporary pharmacometric approaches amenable to both pre-clinical and clinical study design.
Collapse
Affiliation(s)
- Arne Echterhof
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
- Institute of Medical Microbiology, University Hospital of Muenster, Muenster, Germany
| | - Tejas Dharmaraj
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Patrick Blankenberg
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Bobby Targ
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Nicholas M Smith
- Division of Clinical and Translational Therapeutics, School of Pharmacy & Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Francis Blankenberg
- Division of Pediatric Radiology and Nuclear Medicine, Department of Radiology, Lucile Packard Children's Hospital, Stanford, California, USA
| |
Collapse
|
16
|
Dey S, Ghosh M, Dev A. Signalling and molecular pathways, overexpressed receptors of colorectal cancer and effective therapeutic targeting using biogenic silver nanoparticles. Gene 2025; 936:149099. [PMID: 39557372 DOI: 10.1016/j.gene.2024.149099] [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: 08/15/2024] [Revised: 10/18/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Increasing morbidity and mortality in CRC is a potential threat to human health. The major challenges for better treatment outcomes are the heterogeneity of CRC cases, complicated molecular pathway cross-talks, the influence of gut dysbiosis in CRC, and the lack of multimodal target-specific drug delivery. The overexpression of many receptors in CRC cells may pave the path for targeting them with multiple ligands. The design of a more target-specific drug-delivery device with multiple ligand-functionalized, green-synthesized silver nanoparticles is highly promising and may also deliver other approved chemotherapeutic agents. This review presents the various aspects of colorectal cancer and over-expressed receptors that can be targeted with appropriate ligands to enhance the specific drug delivery potency of green synthesised silver nanoparticles. This review aims to broaden further research into this multi-ligand functionalised, safer and effective silver nano drug delivery system.
Collapse
Affiliation(s)
- Sandip Dey
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Manik Ghosh
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India
| | - Abhimanyu Dev
- Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Jharkhand, India.
| |
Collapse
|
17
|
Ito Y, Kasuya H, Kataoka M, Nakamura N, Yoshikawa T, Nakashima T, Zhang H, Li Y, Matsukawa T, Inoue S, Oneyama C, Ohta S, Kagoya Y. Plasma membrane-coated nanoparticles and membrane vesicles to orchestrate multimodal antitumor immunity. J Immunother Cancer 2025; 13:e010005. [PMID: 39864848 PMCID: PMC11784344 DOI: 10.1136/jitc-2024-010005] [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: 07/02/2024] [Accepted: 12/30/2024] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND A number of immunotherapeutic approaches have been developed and are entering the clinic. Bispecific antibodies (BsAbs) are one of these modalities and induce robust efficacy by endogenous T cells in several hematological malignancies. However, most of the treated patients experience only a temporary benefit. Currently available BsAbs provide only anti-CD3 antibody-mediated T-cell stimulation, but not the costimulation or cytokine signaling essential for full T-cell activation. Here, we hypothesized that the simultaneous input of more comprehensive signals would elicit more robust and durable effector T-cell functions. METHODS We genetically engineered the leukemia cell line K562 to express BsAbs, costimulatory ligands, cytokines, and blocking antibodies against immune checkpoint molecules on the cell surface, from which we obtained plasma membrane fractions by mechanical homogenization and subsequent isolation steps. Plasma membranes were reconstituted on the poly (lactic-co-glycolic acid) surface to generate membrane-coated nanoparticles (NPs). Alternatively, nano-sized membrane vesicles (MVs) were generated by ultrasonic dispersion of the isolated membranes. The antitumor function of NPs and MVs loaded with various immunomodulatory factors was evaluated in vitro and in vivo. RESULTS Both membrane-coated NPs and MVs induced BsAb-mediated antigen-specific cytotoxic activity in non-specific T cells, with MVs inducing a slightly better response in vivo. Importantly, T-cell activation was elicited only in the presence of target tumor cells, providing a safety advantage for clinical use. NPs and MVs expressing costimulatory molecules (CD80/4-1BBL) and cytokines (interleukin (IL)-7/IL-15) further enhanced effector T-cell function and induced therapeutic efficacy in vivo. In addition, MVs expressing immune checkpoint antibodies and inflammatory cytokines IL-12 and IL-18 induced objective antitumor responses in solid tumor models partly by converting immunosuppressive macrophages to proinflammatory phenotypes and inducing cytotoxic T-cell infiltration into the tumor. Finally, we showed that MVs were also engineered to activate natural killer (NK) cells by loading multiple ligands. MVs loaded with BsAbs, 4-1BBL, IL-15, and IL-21 induced NK-cell cytotoxic activity in an antigen-specific manner. CONCLUSIONS We developed antitumor NPs and MVs that efficiently induced antitumor immune responses in vivo by simultaneously delivering multiple immunostimulatory signals to endogenous T cells. This platform enables the delivery of desired combinations of antitumor immune signals into T cells and NK cells.
Collapse
Affiliation(s)
- Yusuke Ito
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Hitomi Kasuya
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Mirei Kataoka
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
| | - Noriko Nakamura
- Institute of Engineering Innovation, The University of Tokyo, Tokyo, Japan
| | - Toshiaki Yoshikawa
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Takahiro Nakashima
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Hematology and Oncology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Haosong Zhang
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cellular Oncology, Department of Cancer Diagnostics and Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yang Li
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
- Division of Cellular Oncology, Department of Cancer Diagnostics and Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuya Matsukawa
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Inoue
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Chitose Oneyama
- Division of Cancer Cell Regulation, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Seiichi Ohta
- Institute of Engineering Innovation, The University of Tokyo, Tokyo, Japan
| | - Yuki Kagoya
- Division of Tumor Immunology, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan
- Division of Immune Response, Aichi Cancer Center Research Institute, Nagoya, Japan
| |
Collapse
|
18
|
Spada A, Gerber-Lemaire S. Surface Functionalization of Nanocarriers with Anti-EGFR Ligands for Cancer Active Targeting. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:158. [PMID: 39940134 PMCID: PMC11820047 DOI: 10.3390/nano15030158] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/17/2025] [Accepted: 01/18/2025] [Indexed: 02/14/2025]
Abstract
Active cancer targeting consists of the selective recognition of overexpressed biomarkers on cancer cell surfaces or within the tumor microenvironment, enabled by ligands conjugated to drug carriers. Nanoparticle (NP)-based systems are highly relevant for such an approach due to their large surface area which is amenable to a variety of chemical modifications. Over the past decades, several studies have debated the efficiency of passive targeting, highlighting active targeting as a more specific and selective approach. The choice of conjugation chemistry for attaching ligands to nanocarriers is critical to ensure a stable and robust system. Among the panel of cancer biomarkers, the epidermal growth factor receptor (EGFR) stands as one of the most frequently overexpressed receptors in different cancer types. The design and development of nanocarriers with surface-bound anti-EGFR ligands are vital for targeted therapy, relying on their facilitated capture by EGFR-overexpressing tumor cells and enabling receptor-mediated endocytosis to improve drug accumulation within the tumor microenvironment. In this review, we examine several examples of the most recent and significant anti-EGFR nanocarriers and explore the various conjugation strategies for NP functionalization with anti-EGFR biomolecules and small molecular ligands. In addition, we also describe some of the most common characterization techniques to confirm and analyze the conjugation patterns.
Collapse
Affiliation(s)
| | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland;
| |
Collapse
|
19
|
Munyayi TA, Crous A. Advancing Cancer Drug Delivery with Nanoparticles: Challenges and Prospects in Mathematical Modeling for In Vivo and In Vitro Systems. Cancers (Basel) 2025; 17:198. [PMID: 39857980 PMCID: PMC11763932 DOI: 10.3390/cancers17020198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/30/2024] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Mathematical models are crucial for predicting the behavior of drug conjugate nanoparticles and optimizing drug delivery systems in cancer therapy. These models simulate interactions among nanoparticle properties, tumor characteristics, and physiological conditions, including drug resistance and targeting specificity. However, they often rely on assumptions that may not accurately reflect in vivo conditions. In vitro studies, while useful, may not fully capture the complexities of the in vivo environment, leading to an overestimation of nanoparticle-based therapy effectiveness. Advancements in mathematical modeling, supported by preclinical data and artificial intelligence, are vital for refining nanoparticle-based therapies and improving their translation into effective clinical treatments.
Collapse
Affiliation(s)
| | - Anine Crous
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, P.O. Box 17011, Doornfontein 2028, South Africa
| |
Collapse
|
20
|
Ullah A, Khan M, Zhang Y, Shafiq M, Ullah M, Abbas A, Xianxiang X, Chen G, Diao Y. Advancing Therapeutic Strategies with Polymeric Drug Conjugates for Nucleic Acid Delivery and Treatment. Int J Nanomedicine 2025; 20:25-52. [PMID: 39802382 PMCID: PMC11717654 DOI: 10.2147/ijn.s429279] [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: 09/13/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
The effective clinical translation of messenger RNA (mRNA), small interfering RNA (siRNA), and microRNA (miRNA) for therapeutic purposes hinges on the development of efficient delivery systems. Key challenges include their susceptibility to degradation, limited cellular uptake, and inefficient intracellular release. Polymeric drug conjugates (PDCs) offer a promising solution, combining the benefits of polymeric carriers and therapeutic agents for targeted delivery and treatment. This comprehensive review explores the clinical translation of nucleic acid therapeutics, focusing on polymeric drug conjugates. It investigates how these conjugates address delivery obstacles, enhance systemic circulation, reduce immunogenicity, and provide controlled release, improving safety profiles. The review delves into the conjugation strategies, preparation methods, and various classes of PDCs, as well as strategic design, highlighting their role in nucleic acid delivery. Applications of PDCs in treating diseases such as cancer, immune disorders, and fibrosis are also discussed. Despite significant advancements, challenges in clinical adoption persist. The review concludes with insights into future directions for this transformative technology, underscoring the potential of PDCs to advance nucleic acid-based therapies and combat infectious diseases significantly.
Collapse
Affiliation(s)
- Aftab Ullah
- School of Medicine, Huaqiao University, Quanzhou, Fujian, People’s Republic of China
| | - Marina Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science and Technology, Kohat, Pakistan
| | - Yibang Zhang
- School of Pharmacy, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Muhammad Shafiq
- Research Institute of Clinical Pharmacy, Department of Pharmacology, Shantou University Medical College, Shantou, Guangdong, People’s Republic of China
| | - Mohsan Ullah
- School of Medicine, Huaqiao University, Quanzhou, Fujian, People’s Republic of China
| | - Azar Abbas
- Institute of Medicine, Shenzhen Institute of Advanced Technology, Shenzhen, Guangdong, People’s Republic of China
| | - Xu Xianxiang
- School of Medicine, Huaqiao University, Quanzhou, Fujian, People’s Republic of China
| | - Gang Chen
- School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, Shandong, People’s Republic of China
- Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao central Medical Group), Qingdao, Shandong, People’s Republic of China
| | - Yong Diao
- School of Medicine, Huaqiao University, Quanzhou, Fujian, People’s Republic of China
| |
Collapse
|
21
|
Gupta DS, Suares D. Uncovering the Emerging Prospects of Lipid-based Nanoparticulate Vehicles in Lung Cancer Management: A Recent Perspective. Pharm Nanotechnol 2025; 13:155-170. [PMID: 38468532 DOI: 10.2174/0122117385286781240228060152] [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: 10/11/2023] [Revised: 12/31/2023] [Accepted: 02/15/2024] [Indexed: 03/13/2024]
Abstract
Lung cancer, a leading cause of cancer-related deaths globally, is gaining research interest more than ever before. Owing to the burden of pathogenesis on the quality of life of patients and subsequently the healthcare system, research efforts focus on its management and amelioration. In an effort to improve bioavailability, enhance stability, minimize adverse effects and reduce the incidence of resistance, nanotechnological platforms have been harnessed for drug delivery and improving treatment outcomes. Lipid nanoparticles, in particular, offer an interesting clinical opportunity with respect to the delivery of a variety of agents. These include synthetic chemotherapeutic agents, immunotherapeutic molecules, as well as phytoconstituents with promising anticancer benefits. In addition to this, these systems are being studied for their usage in conjunction with other treatment strategies. However, their applications remain limited owing to a number of challenges, chiefly clinical translation. There is a need to address the scalability of such technologies, in order to improve accessibility. The authors aim to offer a comprehensive understanding of the evolution of lipid nanoparticles and their application in lung cancer, the interplay of disease pathways and their mechanism of action and the potential for delivery of a variety of agents. Additionally, a discussion with respect to results from preclinical studies has also been provided. The authors have also provided a well-rounded insight into the limitations and future perspectives. While the possibilities are endless, there is a need to undertake focused research to expedite clinical translation and offer avenues for wider applications in disease management.
Collapse
Affiliation(s)
- Dhruv Sanjay Gupta
- Department of Pharmaceutical Sciences, Shobhaben Pratapbhai Patel School of Pharmacy & Technology, Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India
| | - Divya Suares
- Department of Pharmaceutical Sciences, Shobhaben Pratapbhai Patel School of Pharmacy & Technology, Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai, 400056, India
| |
Collapse
|
22
|
Muhammad FA, Altalbawy FMA, Mandaliya V, Saraswat SK, Rekha MM, Aulakh D, Chahar M, Mahdi MS, Jaber MA, Alhadrawi M. Targeting breast tumor extracellular matrix and stroma utilizing nanoparticles. Clin Transl Oncol 2024:10.1007/s12094-024-03793-x. [PMID: 39692807 DOI: 10.1007/s12094-024-03793-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024]
Abstract
Breast cancer is a complicated malignancy and is known as the most common cancer in women. Considerable experiments have been devoted to explore the basic impacts of the tumor stroma, particularly the extracellular matrix (ECM) and stromal components, on tumor growth and resistance to treatment. ECM is made up of an intricate system of proteins, glycosaminoglycans, and proteoglycans, and maintains structural support and controls key signaling pathways involved in breast tumors. ECM can block different drugs such as chemotherapy and immunotherapy drugs from entering the tumor stroma. Furthermore, the stromal elements, such as cancer-associated fibroblasts (CAFs), immune cells, and blood vessels, have crucial impacts on tumor development and therapeutic resistance. Recently, promising outcomes have been achieved in using nanotechnology for delivering drugs to tumor stroma and crossing ECM in breast malignancies. Nanoparticles have various benefits for targeting the breast tumor stroma, such as improved permeability and retention, extended circulation time, and the ability to actively target the area. This review covers the latest developments in nanoparticle therapies that focus on breast tumor ECM and stroma. We will explore different approaches using nanoparticles to target the delivery of anticancer drugs like chemotherapy, small molecule drugs, various antitumor products, and other specific synthetic therapeutic agents to the breast tumor stroma. Furthermore, we will investigate the utilization of nanoparticles in altering the stromal elements, such as reprogramming CAFs and immune cells, and also remodeling ECM.
Collapse
Affiliation(s)
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, 12613, Egypt.
| | - Viralkumar Mandaliya
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | | | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Damanjeet Aulakh
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology Chitkara University, Rajpura, Punjab, 140401, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering and Technology, NIMS University Rajasthan, Jaipur, India
| | | | | | - Merwa Alhadrawi
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
23
|
Bevilacqua G, Corvino R, Capriotti AL, Montone CM, Moriconi M, Salciccia S, Brunelli V, Santarelli V, Sciarra B, Laganà A, Santini D, Sciarra A, Gentilucci A. The Protein Corona on Nanoparticles for Tumor Targeting in Prostate Cancer-A Review of the Literature and Experimental Trial Protocol. BIOLOGY 2024; 13:1024. [PMID: 39765691 PMCID: PMC11672965 DOI: 10.3390/biology13121024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
The National Cancer Institute (NCI) recognizes the potential of technologies based on the use of nanoparticles (NPs) in revolutionizing clinical approaches to the diagnosis and prognosis of cancer. Recent research suggests that once NPs come into contact with the biological fluid of cancer patients, they are covered by proteins, forming a "protein corona" composed of hundreds of plasma proteins. The concept of a personalized, disease-specific protein corona, demonstrating substantial differences in NP corona profiles between patients with and without cancer, has been introduced. We developed the design of an experimental prospective single-center study with patients allocated in a 1:1:1 ratio of one of three arms: untreated patients with benign prostatic hyperplasia (BPH), untreated patients with non-metastatic prostate cancer (PCa), and metastatic prostate cancer patients starting systemic therapies with new androgen-targeted agents or taxanes. The protocol aims to develop and implement sensitive nanotools with two distinct objectives: First, to design NPs capable of selectively binding and detecting biomarkers in order to build a predictive diagnostic model to effectively discriminate between patient sera affected by BPH and PCa. Secondly, within the population with PCa, in the case of initial advanced metastatic diagnosis, the objective is to find biomarkers capable of predicting the response to systemic treatments to improve the precision and efficiency of monitoring treatment outcomes. For protein and metabolite corona experiments, we developed a cross-reactive sensor array platform with cancer detection capacity made of three liposomal formulations with different surface charges. For proteomic-NP studies, proteins were identified and quantified using nano-high-performance LC (nanoHPLC) coupled with MS/MS (nanoHPLC-MS/MS). Metabolites were instead analyzed using an untargeted metabolomic approach. Compared with previous review articles, the novelty of this review is represented by the analysis of the possible clinical applications of protein corona NPs focused on PCa and the presentation of a new clinical protocol in the metastatic phase of PCa.
Collapse
Affiliation(s)
- Giulio Bevilacqua
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, 00161 Rome, Italy; (G.B.); (R.C.); (M.M.); (S.S.); (V.B.); (V.S.); (A.G.)
| | - Roberta Corvino
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, 00161 Rome, Italy; (G.B.); (R.C.); (M.M.); (S.S.); (V.B.); (V.S.); (A.G.)
| | - Anna Laura Capriotti
- Department of Chemistry, University Sapienza, 00161 Rome, Italy; (A.L.C.); (C.M.M.); (A.L.)
| | - Carmela Maria Montone
- Department of Chemistry, University Sapienza, 00161 Rome, Italy; (A.L.C.); (C.M.M.); (A.L.)
| | - Martina Moriconi
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, 00161 Rome, Italy; (G.B.); (R.C.); (M.M.); (S.S.); (V.B.); (V.S.); (A.G.)
| | - Stefano Salciccia
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, 00161 Rome, Italy; (G.B.); (R.C.); (M.M.); (S.S.); (V.B.); (V.S.); (A.G.)
| | - Valentina Brunelli
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, 00161 Rome, Italy; (G.B.); (R.C.); (M.M.); (S.S.); (V.B.); (V.S.); (A.G.)
| | - Valerio Santarelli
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, 00161 Rome, Italy; (G.B.); (R.C.); (M.M.); (S.S.); (V.B.); (V.S.); (A.G.)
| | - Beatrice Sciarra
- Department of Pharmaceutic Chemistry, University Sapienza, 00161 Rome, Italy;
| | - Aldo Laganà
- Department of Chemistry, University Sapienza, 00161 Rome, Italy; (A.L.C.); (C.M.M.); (A.L.)
| | - Daniele Santini
- Department of Oncology, University Sapienza, 00161 Rome, Italy;
| | - Alessandro Sciarra
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, 00161 Rome, Italy; (G.B.); (R.C.); (M.M.); (S.S.); (V.B.); (V.S.); (A.G.)
| | - Alessandro Gentilucci
- Department “Materno Infantile e Scienze Urologiche”, University Sapienza, 00161 Rome, Italy; (G.B.); (R.C.); (M.M.); (S.S.); (V.B.); (V.S.); (A.G.)
| |
Collapse
|
24
|
Öztürk K, Kaplan M, Çalış S. Effects of nanoparticle size, shape, and zeta potential on drug delivery. Int J Pharm 2024; 666:124799. [PMID: 39369767 DOI: 10.1016/j.ijpharm.2024.124799] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/16/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
Nanotechnology has brought about a significant revolution in drug delivery, and research in this domain is increasingly focusing on understanding the role of nanoparticle (NP) characteristics in drug delivery efficiency. First and foremost, we center our attention on the size of nanoparticles. Studies have indicated that NP size significantly influences factors such as circulation time, targeting capabilities, and cellular uptake. Secondly, we examine the significance of nanoparticle shape. Various studies suggest that NPs of different shapes affect cellular uptake mechanisms and offer potential advantages in directing drug delivery. For instance, cylindrical or needle-like NPs may facilitate better cellular uptake compared to spherical NPs. Lastly, we address the importance of nanoparticle charge. Zeta potential can impact the targeting and cellular uptake of NPs. Positively charged NPs may be better absorbed by negatively charged cells, whereas negatively charged NPs might perform more effectively in positively charged cells. This review provides essential insights into understanding the role of nanoparticles in drug delivery. The properties of nanoparticles, including size, shape, and charge, should be taken into consideration in the rational design of drug delivery systems, as optimizing these characteristics can contribute to more efficient targeting of drugs to the desired tissues. Thus, research into nanoparticle properties will continue to play a crucial role in the future of drug delivery.
Collapse
Affiliation(s)
- Kıvılcım Öztürk
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Türkiye
| | - Meryem Kaplan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Türkiye; Department of Pharmaceutical Technology, Faculty of Pharmacy, Süleyman Demirel University, 32260 Isparta, Türkiye
| | - Sema Çalış
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Türkiye.
| |
Collapse
|
25
|
Nemakhavhani L, Abrahamse H, Kumar SSD. A review on dendrimer-based nanoconjugates and their intracellular trafficking in cancer photodynamic therapy. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2024; 52:384-398. [PMID: 39101753 DOI: 10.1080/21691401.2024.2368033] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 08/06/2024]
Abstract
Nanotechnology-based cancer treatment has received considerable attention, and these treatments generally use drug-loaded nanoparticles (NPs) to target and destroy cancer cells. Nanotechnology combined with photodynamic therapy (PDT) has demonstrated positive outcomes in cancer therapy. Combining nanotechnology and PDT is effective in targeting metastatic cancer cells. Nanotechnology can also increase the effectiveness of PDT by targeting cells at a molecular level. Dendrimer-based nanoconjugates (DBNs) are highly stable and biocompatible, making them suitable for drug delivery applications. Moreover, the hyperbranched structures in DBNs have the capacity to load hydrophobic compounds, such as photosensitizers (PSs) and chemotherapy drugs, and deliver them efficiently to tumour cells. This review primarily focuses on DBNs and their potential applications in cancer treatment. We discuss the chemical design, mechanism of action, and targeting efficiency of DBNs in tumour metastasis, intracellular trafficking in cancer treatment, and DBNs' biocompatibility, biodegradability and clearance properties. Overall, this study will provide the most recent insights into the application of DBNs and PDT in cancer therapy.
Collapse
Affiliation(s)
- Lufuno Nemakhavhani
- Laser Research Centre, University of Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, University of Johannesburg, Johannesburg, South Africa
| | | |
Collapse
|
26
|
Chen X, Wan H, Lu L, Li R, Sun B, Ren J. PLGA-PEG-c(RGDfK)- Kushenol E Micelles With a Therapeutic Potential for Targeting Ovarian Cancer. IET Nanobiotechnol 2024; 2024:7136323. [PMID: 39649540 PMCID: PMC11623995 DOI: 10.1049/nbt2/7136323] [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: 12/28/2023] [Revised: 09/30/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024] Open
Abstract
Background: As a naturally derived inhibitor of autophagy, Kushenol E (KE) is a biprenylated flavonoid and is isolated from Sophora flavescens, which has been used for the treatment of cancer, hepatitis, and skin diseases. However, KE, as a poorly soluble drug, exhibited strong autophagy regulating activity in in vitro cancer cell lines, but no related studies have reported its antiovarian cancer property. Therefore, it is very beneficial to enhance the antineoplastic properties of KE by establishing an ovarian tumor-targeting nanoparticle system modified with tumor-homing c(RGDfK) peptides. Materials and Methods: In the current study, poly(lactic-co-glycolic acid)-poly(ethylene glycol)-modified with cyclic RGDfK peptide (PLGA-PEG-c(RGDfK))-KE micelles (PPCKM) were prepared to overcome the poor water solubility of KE to meet the requirement of tumor-active targeting. The effect of PPCKM on ovarian cancer was evaluated on SKOV-3 cells and xenograft models in BALB/c nude mice. Results: The PPCKM showed a higher drug cumulative release ratio (82.16 ± 7.69% vs. 34.96 ± 3.05%, at 1.5 h) with good morphology, particle size (93.41 ± 2.84 nm), and entrapment efficiency (89.7% ± 1.3%). The cell viability, migration, and apoptosis analysis of SKOV-3 cells demonstrated that PPCKM retained potent antitumor effects and promoted apoptosis at early and advanced stages with concentration-dependent. Based on the establishment of xenograft models in BALB/c nude mice, we discovered that PPCKM reduced tumor volume and weight, inhibited proliferating cell nuclear antigen (PCNA) and Ki67 expression, as well as promoted apoptosis by targeting the tumor site. Conclusion: The findings in this study suggest that PPCKM may serve as an effective therapeutic option for ovarian cancer.
Collapse
Affiliation(s)
- Xue Chen
- Department of Traditional Chinese Medicine, Nanxiang Branch of Ruijin Hospital, Shanghai 201802, China
| | - Haopeng Wan
- Department of Traditional Chinese Medicine, Nanxiang Branch of Ruijin Hospital, Shanghai 201802, China
| | - Lijuan Lu
- Department of Gynecology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215000, China
| | - Ran Li
- School of Pharmacy, Jiangsu University, Zhenjiang 212001, China
| | - Bo Sun
- Department of Gynecology, Fangta Traditional Chinese Medicine Hospital of Songjiang District of Shanghai, Shanghai 201699, China
| | - Juan Ren
- Clinical Medical Center of Oncology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200241, China
| |
Collapse
|
27
|
Pangua C, Espuelas S, Simón JA, Álvarez S, Martínez-Ohárriz C, Collantes M, Peñuelas I, Calvo A, Irache JM. Enhancing bevacizumab efficacy in a colorectal tumor mice model using dextran-coated albumin nanoparticles. Drug Deliv Transl Res 2024:10.1007/s13346-024-01734-3. [PMID: 39455507 DOI: 10.1007/s13346-024-01734-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 10/28/2024]
Abstract
Bevacizumab is a monoclonal antibody (mAb) that prevents the growth of new blood vessels and is currently employed in the treatment of colorectal cancer (CRC). However, like other mAb, bevacizumab shows a limited penetration in the tumors, hampering their effectiveness and inducing adverse reactions. The aim of this work was to design and evaluate albumin-based nanoparticles, coated with dextran, as carriers for bevacizumab in order to promote its accumulation in the tumor and, thus, improve its antiangiogenic activity. These nanoparticles (B-NP-DEX50) displayed a mean size of about 250 nm and a payload of about 110 µg/mg. In a CRC mice model, these nanoparticles significantly reduced tumor growth and increased tumor doubling time, tumor necrosis and apoptosis more effectively than free bevacizumab. At the end of study, bevacizumab plasma levels were higher in the free drug group, while tumor levels were higher in the B-NP-DEX50 group (2.5-time higher). In line with this, the biodistribution study revealed that nanoparticles accumulated in the tumor core, potentially improving therapeutic efficacy while reducing systemic exposure. In summary, B-NP-DEX can be an adequate alternative to improve the therapeutic efficiency of biologically active molecules, offering a more specific biodistribution to the site of action.
Collapse
Affiliation(s)
- Cristina Pangua
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain
| | - Socorro Espuelas
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Jon Ander Simón
- Program in Solid Tumors, CIMA of the University of Navarra, Pamplona, 31008, Spain
| | - Samuel Álvarez
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain
| | | | - María Collantes
- Radiopharmacy Unit, Clinica Universidad de Navarra, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Iván Peñuelas
- Radiopharmacy Unit, Clinica Universidad de Navarra, Pamplona, 31008, Spain
- Translational Molecular Imaging Unit (UNIMTRA), Department of Nuclear Medicine, Clinica Universidad de Navarra, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Alfonso Calvo
- Program in Solid Tumors, CIMA of the University of Navarra, Pamplona, 31008, Spain
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain
| | - Juan M Irache
- NANO-VAC Research Group, Department of Pharmaceutical Sciences, School of Pharmacy and Nutrition, University of Navarra, C/ Irunlarrea 1, Pamplona, 31008, Spain.
- Institute for Health Research (IdiSNA), Pamplona, 31008, Spain.
| |
Collapse
|
28
|
Sun X, Lin Y, Zhong X, Fan C, Liu Z, Chen X, Luo Z, Wu J, Tima S, Zhang Z, Jiang J, Du X, Zhou X, Zhong Z. Alendronate-functionalized polymeric micelles target icaritin to bone for mitigating osteoporosis in a rat model. J Control Release 2024; 376:37-51. [PMID: 39368708 DOI: 10.1016/j.jconrel.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 10/07/2024]
Abstract
Formulating drugs into nanoparticles that target sites of disease can lead to strong therapeutic effects with lower doses of drugs and lower rates of off-target adverse effects. Few ways to target drugs to bone have been described, hampering the treatment of osteoporosis. Here we exploit the ability of alendronate to bind tightly to hydroxyapatite in bone as a tactic to target polymeric micelles loaded with the plant flavonoid icaritin to osteoporotic lesions. The traditional Chinese medicine icaritin, from Herba Epimedii, has previously been shown to inhibit adipogenesis and enhance osteogenesis by bone mesenchymal stem cells, but the compound on its own persists only briefly in the bloodstream. Our delivery system led to stronger inhibition of adipogenesis and activation of osteogenesis in a rat model of osteoporosis than when the icaritin-loaded micelles lacked alendronate. These results establish the feasibility of using alendronate to target osteogenic phytomolecules to sites of bone injury, which may guide the development of effective therapies against osteoporosis and, by extension, other bone disorders.
Collapse
Affiliation(s)
- Xiaoduan Sun
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Yan Lin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xingyue Zhong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Chao Fan
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zhen Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xin Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Zaiyi Luo
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Jili Wu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Singkome Tima
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Zhirong Zhang
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China
| | - Xingjie Du
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Xiangyu Zhou
- Department of General Surgery (Thyroid Surgery), the Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan 646000, China.
| | - Zhirong Zhong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan 646000, China; Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, Sichuan 646000, China.
| |
Collapse
|
29
|
Heshmati N, Chakka LRJ, Zhang Y, Maniruzzaman M. Fabrication of mRNA encapsulated lipid nanoparticles using state of the art SMART-MaGIC technology and transfection in vitro. Sci Rep 2024; 14:22714. [PMID: 39349578 PMCID: PMC11442764 DOI: 10.1038/s41598-024-73804-y] [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: 09/20/2024] [Indexed: 10/02/2024] Open
Abstract
The messenger ribose nucleic acid (mRNA) in the form of Corona virus of 2019 (COVID-19) vaccines were effectively delivered through lipid nanoparticles (LNP) proving its use as effective carriers in clinical applications. In the present work, mRNA (erythropoietin (EPO)) encapsulated LNPs were prepared using a next generation state-of-the-art patented, Sprayed Multi Absorbed-droplet Reposing Technology (SMART) coupled with Multi-channeled and Guided Inner-Controlling printheads (MaGIC) technologies. The LNP-mRNA were synthesized at different N/P ratios and the particles were characterized for particle size and zeta potential (Zetasizer), encapsulation or complexation (gel retardation assay) and transfection (Fluorescence microscopy and ELISA) in MG63 sarcoma cells in vitro. The results showed a narrow distribution of mRNA-lipid particles of 200 nm when fabricated with SMART alone and then the size was reduced to approximately 50 nm with the combination of SMART-MaGIC technologies. The gel retardation assay showed that the N/P > 1 exhibited strong encapsulation of mRNA with lipid. The in vitro results showed the toxicity profile of the lipids where N/P ratio of 5 was optimized with > 50% cell viability. It can be concluded that the functional LNP-mRNA prepared and analyzed with SMART-MaGIC technologies, could be a potential new fabrication method of mRNA loaded LNPs for point-of-service or distributed manufacturing.
Collapse
Affiliation(s)
- Niloofar Heshmati
- Department of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Leela Raghava Jaidev Chakka
- Department of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Yu Zhang
- Department of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| | - Mohammed Maniruzzaman
- Department of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, 78712, USA.
- Pharmaceutical Engineering and 3D Printing (PharmE3D) Lab, Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University, MS, 38677, USA.
| |
Collapse
|
30
|
Zhang LZ, Du RJ, Wang D, Qin J, Yu C, Zhang L, Zhu HD. Enteral Route Nanomedicine for Cancer Therapy. Int J Nanomedicine 2024; 19:9889-9919. [PMID: 39351000 PMCID: PMC11439897 DOI: 10.2147/ijn.s482329] [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: 06/11/2024] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
With the in-depth knowledge of the pathological and physiological characteristics of the intestinal barrier-portal vein/intestinal lymphatic vessels-systemic circulation axis, oral targeted drug delivery is frequently being renewed. With many advantages, such as high safety, convenient administration, and good patient compliance, many researchers have begun to explore targeted drug delivery from intravenous injections to oral administration. Over the past few decades, the fields of materials science and nanomedicine have produced various drug delivery platforms that hold great potential in overcoming the multiple barriers associated with oral drug delivery. However, the oral transport of particles into the systemic circulation is extremely difficult due to immune rejection and biochemical invasion in the intestine, which limits absorption and entry into the bloodstream. The feasibility of the oral delivery of targeted drugs to sites outside the gastrointestinal tract (GIT) is unknown. This article reviews the biological barriers to drug absorption, the in vivo fate and transport mechanisms of drug carriers, the theoretical basis for oral administration, and the impact of carrier structural evolution on oral administration to achieve this goal. Finally, this article reviews the characteristics of different nano-delivery systems that can enhance the bioavailability of oral therapeutics and highlights their applications in the efficient creation of oral anticancer nanomedicines.
Collapse
Affiliation(s)
- Lin-Zhu Zhang
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, Nanjing, People's Republic of China
| | - Rui-Jie Du
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, Nanjing, People's Republic of China
| | - Duo Wang
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, Nanjing, People's Republic of China
| | - Juan Qin
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, Nanjing, People's Republic of China
| | - Chao Yu
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, Nanjing, People's Republic of China
| | - Lei Zhang
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, Nanjing, People's Republic of China
| | - Hai-Dong Zhu
- Center of Interventional Radiology & Vascular Surgery, Department of Radiology, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Medical School, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
31
|
Einafshar E, Javid H, Amiri H, Akbari-Zadeh H, Hashemy SI. Curcumin loaded β-cyclodextrin-magnetic graphene oxide nanoparticles decorated with folic acid receptors as a new theranostic agent to improve prostate cancer treatment. Carbohydr Polym 2024; 340:122328. [PMID: 38857995 DOI: 10.1016/j.carbpol.2024.122328] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/12/2024]
Abstract
This article presents a novel approach to treating prostate cancer using a nanocarrier composed of folic acid (FA), β-cyclodextrin (β-CD), and magnetic graphene oxide (MGO) as a theranostic agent. The carrier is designed to improve the solubility and bioavailability of curcumin, a potential therapeutic substance against prostate cancer. Folic acid receptors overexpressed on the surface of solid tumors, including prostate cancer, may facilitate targeted drug delivery to tumor cells while avoiding nonspecific effects on healthy tissues. The anticancer efficacy of Folic acid-curcumin@β-CD-MGO in vitro was also examined on LNCaP (an androgen-dependent) and PC3 (an androgen-independent) prostate cancer cells. The relaxivity of nanoparticles in MRI images was also investigated as a diagnostic factor. The results showed a concentration-dependent inhibitory effect on cell proliferation, induction of oxidative damage, and apoptotic effects. Also, nanoparticle relaxometry shows that this agent can be used as a negative contrast agent in MRI images. Overall, this study represents a promising theranostic agent to improve the delivery and trace of curcumin and enhance its therapeutic potential in the treatment of prostate cancer.
Collapse
Affiliation(s)
- Elham Einafshar
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Javid
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran
| | - Hamed Amiri
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hadi Akbari-Zadeh
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Isaac Hashemy
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
32
|
Tiwari S, Kumar R, Devi S, Sharma P, Chaudhary NR, Negi S, Tandel N, Marepally S, Pied S, Tyagi RK. Biogenically synthesized green silver nanoparticles exhibit antimalarial activity. DISCOVER NANO 2024; 19:136. [PMID: 39217276 PMCID: PMC11365884 DOI: 10.1186/s11671-024-04098-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
The suboptimal efficacies of existing anti-malarial drugs attributed to the emergence of drug resistance dampen the clinical outcomes. Hence, there is a need for developing novel drug and drug targets. Recently silver nanoparticles (AgNPs) constructed with the leaf extracts of Euphorbia cotinifolia were shown to possess antimalarial activity. Therefore, the synthesized AgNPs from Euphorbia cotinifolia (EcAgNPs) were tested for their parasite clearance activity. We determined the antimalarial activity in the asexual blood stage infection of 3D7 (laboratory strain) P. falciparum. EcAgNPs demonstrated the significant inhibition of parasite growth (EC50 of 0.75 µg/ml) in the routine in vitro culture of P. falciparum. The synthesized silver nanoparticles were seen to induce apoptosis in P. falciparum through increased reactive oxygen species (ROS) ROS production and activated programmed cell death pathways characterized by the caspase-3 and calpain activity. Also, altered transcriptional regulation of Bax/Bcl-2 ratio indicated the enhanced apoptosis. Moreover, inhibited expression of PfLPL-1 by EcAgNPs is suggestive of the dysregulated host fatty acid flux via parasite lipid storage. Overall, our findings suggest that EcAgNPs are a non-toxic and targeted antimalarial treatment, and could be a promising therapeutic approach for clearing malaria infection.
Collapse
Affiliation(s)
- Savitri Tiwari
- School of Biological and Life Sciences, Galgotias University, Gautam Buddha Nagar, Greater Noida, 201310, India
| | - Reetesh Kumar
- Faculty of Agricultural Sciences, Institute of Applied Sciences and Humanities, GLA University, Mathura, 281406, India
| | - Sonia Devi
- Biomedical Parasitology and Translational-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh, 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - Prakriti Sharma
- Biomedical Parasitology and Translational-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh, 160036, India
| | - Neil Roy Chaudhary
- Biomedical Parasitology and Translational-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh, 160036, India
| | - Sushmita Negi
- Biomedical Parasitology and Translational-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh, 160036, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India
| | - Nikunj Tandel
- Institute of Science, Nirma University, Ahmedabad, Gujarat, India
- Malaria Research Lab, CSIR-Centre for Cellular and Molecular Biology (CCMB), Habsiguda, Hyderabad, Telangana, 500007, India
| | - Srujan Marepally
- Centre for Stem Cell Research (a Unit of inStem, Bengaluru), Christian Medical College Campus, Bagayam, Vellore, Tamil Nadu, 632002, India
| | - Sylviane Pied
- CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-9 CIIL, Institut Pasteur de Lille, University of Lille, 59019, Lille, France
| | - Rajeev K Tyagi
- Biomedical Parasitology and Translational-Immunology Lab, Division of Cell Biology and Immunology, CSIR-Institute of Microbial Technology (IMTECH), Sec-39A, Chandigarh, 160036, India.
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
33
|
Park S, Nguyen VP, Wang X, Paulus YM. Gold Nanoparticles for Retinal Molecular Optical Imaging. Int J Mol Sci 2024; 25:9315. [PMID: 39273264 PMCID: PMC11395175 DOI: 10.3390/ijms25179315] [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: 06/29/2024] [Revised: 08/03/2024] [Accepted: 08/16/2024] [Indexed: 09/15/2024] Open
Abstract
The incorporation of gold nanoparticles (GNPs) into retinal imaging signifies a notable advancement in ophthalmology, offering improved accuracy in diagnosis and patient outcomes. This review explores the synthesis and unique properties of GNPs, highlighting their adjustable surface plasmon resonance, biocompatibility, and excellent optical absorption and scattering abilities. These features make GNPs advantageous contrast agents, enhancing the precision and quality of various imaging modalities, including photoacoustic imaging, optical coherence tomography, and fluorescence imaging. This paper analyzes the unique properties and corresponding mechanisms based on the morphological features of GNPs, highlighting the potential of GNPs in retinal disease diagnosis and management. Given the limitations currently encountered in clinical applications of GNPs, the approaches and strategies to overcome these limitations are also discussed. These findings suggest that the properties and efficacy of GNPs have innovative applications in retinal disease imaging.
Collapse
Affiliation(s)
- Sumin Park
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA;
| | - Van Phuc Nguyen
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA;
- Department of Ophthalmology, Johns Hopkins University, Baltimore, MD 21287, USA
| | - Xueding Wang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA;
| | - Yannis M. Paulus
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48105, USA;
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI 48105, USA;
- Department of Ophthalmology, Johns Hopkins University, Baltimore, MD 21287, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21287, USA
| |
Collapse
|
34
|
Li Y, Shen X, Ding H, Zhang Y, Pan D, Su L, Wu Y, Fang Z, Zhou J, Gong Q, Luo K. Dendritic nanomedicine enhances chemo-immunotherapy by disturbing metabolism of cancer-associated fibroblasts for deep penetration and activating function of immune cells. Acta Pharm Sin B 2024; 14:3680-3696. [PMID: 39220877 PMCID: PMC11365400 DOI: 10.1016/j.apsb.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/21/2024] [Accepted: 02/28/2024] [Indexed: 09/04/2024] Open
Abstract
Inefficient drug penetration hurdled by the stroma in the tumor tissue leads to a diminished therapeutic effect for drugs and a reduced infiltration level of immune cells. Herein, we constructed a PEGylated dendritic epirubicin (Epi) prodrug (Epi-P4D) to regulate the metabolism of cancer-associated fibroblasts (CAFs), thus enhancing Epi penetration into both multicellular tumor spheroids (MTSs) and tumor tissues in mouse colon cancer (CT26), mouse breast cancer (4T1) and human breast cancer (MDA-MB-231) models. Enhanced cytotoxicity against CT26 MTSs and remarkable antitumor efficacy of Epi-P4D were ascribed to reduced fibronectin, α-SMA, and collagen secretion. Besides, thinning of the tumor tissue stroma and efficient eradication of tumor cells promoted the immunogenic cell death effect for dendritic cell (DC) maturation and subsequent immune activation, including elevating the CD4+ T cell population, reducing CD4+ and CD8+ T cell hyperactivation and exhaustion, and amplifying the natural killer (NK) cell proportion and effectively activating them. As a result, this dendritic nanomedicine thinned the stroma of tumor tissues to enhance drug penetration and facilitate immune cell infiltration for elevated antitumor efficacy.
Collapse
Affiliation(s)
- Yunkun Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoding Shen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haitao Ding
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuxin Zhang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dayi Pan
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liping Su
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yahui Wu
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zaixiang Fang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Zhou
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, and Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
35
|
Martino A, Terracciano R, Milićević B, Milošević M, Simić V, Fallon BC, Carcamo-Bahena Y, Royal ALR, Carcamo-Bahena AA, Butler EB, Willson RC, Kojić M, Filgueira CS. An Insight into Perfusion Anisotropy within Solid Murine Lung Cancer Tumors. Pharmaceutics 2024; 16:1009. [PMID: 39204354 PMCID: PMC11360231 DOI: 10.3390/pharmaceutics16081009] [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: 06/25/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 09/04/2024] Open
Abstract
Blood vessels are essential for maintaining tumor growth, progression, and metastasis, yet the tumor vasculature is under a constant state of remodeling. Since the tumor vasculature is an attractive therapeutic target, there is a need to predict the dynamic changes in intratumoral fluid pressure and velocity that occur across the tumor microenvironment (TME). The goal of this study was to obtain insight into perfusion anisotropy within lung tumors. To achieve this goal, we used the perfusion marker Hoechst 33342 and vascular endothelial marker CD31 to stain tumor sections from C57BL/6 mice harboring Lewis lung carcinoma tumors on their flank. Vasculature, capillary diameter, and permeability distribution were extracted at different time points along the tumor growth curve. A computational model was generated by applying a unique modeling approach based on the smeared physical fields (Kojic Transport Model, KTM). KTM predicts spatial and temporal changes in intratumoral pressure and fluid velocity within the growing tumor. Anisotropic perfusion occurs within two domains: capillary and extracellular space. Anisotropy in tumor structure causes the nonuniform distribution of pressure and fluid velocity. These results provide insights regarding local vascular distribution for optimal drug dosing and delivery to better predict distribution and duration of retention within the TME.
Collapse
Affiliation(s)
- Antonio Martino
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
- Department of Materials Science and Engineering, University of Houston, Houston, TX 77024, USA
| | - Rossana Terracciano
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
- Department of Electronics and Telecommunications, Politecnico di Torino, 10129 Torino, Italy
| | - Bogdan Milićević
- Bioengineering Research and Development Center (BioIRC), 34000 Kragujevac, Serbia; (B.M.); (M.M.); (V.S.)
- Faculty of Engineering, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Miljan Milošević
- Bioengineering Research and Development Center (BioIRC), 34000 Kragujevac, Serbia; (B.M.); (M.M.); (V.S.)
- Institute for Information Technologies, University of Kragujevac, 34000 Kragujevac, Serbia
- Faculty of Information Technology, Belgrade Metropolitan University, 11000 Belgrade, Serbia
| | - Vladimir Simić
- Bioengineering Research and Development Center (BioIRC), 34000 Kragujevac, Serbia; (B.M.); (M.M.); (V.S.)
- Institute for Information Technologies, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Blake C. Fallon
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
| | - Yareli Carcamo-Bahena
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
| | - Amber Lee R. Royal
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
| | - Aileen A. Carcamo-Bahena
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
| | - Edward Brian Butler
- Department of Radiation Oncology, Houston Methodist Research Institute, Houston, TX 77030, USA;
| | - Richard C. Willson
- Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX 77024, USA;
| | - Miloš Kojić
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
- Bioengineering Research and Development Center (BioIRC), 34000 Kragujevac, Serbia; (B.M.); (M.M.); (V.S.)
- Serbian Academy of Sciences and Arts, 11000 Belgrade, Serbia
| | - Carly S. Filgueira
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX 77030, USA; (A.M.); (R.T.); (B.C.F.); (Y.C.-B.); (A.L.R.R.); (A.A.C.-B.); (M.K.)
- Department of Cardiovascular Surgery, Houston Methodist Research Institute, Houston, TX 77030, USA
| |
Collapse
|
36
|
Grancharova T, Zagorchev P, Pilicheva B. Iron Oxide Nanoparticles: Parameters for Optimized Photoconversion Efficiency in Synergistic Cancer Treatment. J Funct Biomater 2024; 15:207. [PMID: 39194645 DOI: 10.3390/jfb15080207] [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: 07/02/2024] [Revised: 07/22/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Photothermal therapy (PTT) can overcome cancer treatment resistance by enhancing the cell membrane permeability, facilitating drug accumulation, and promoting drug release within the tumor tissue. Iron oxide nanoparticles (IONPs) have emerged as effective agents for PTT due to their unique properties and biocompatibility. Approved for the treatment of anemia, as MRI contrast agents, and as magnetic hyperthermia mediators, IONPs also offer excellent light-to-heat conversion and can be manipulated using external magnetic fields for targeted accumulation in specific tissue. Optimizing parameters such as the laser wavelength, power density, shape, size, iron oxidation state, functionalization, and concentration is crucial for IONPs' effectiveness. In addition to PTT, IONPs enhance other cancer treatment modalities. They improve tumor oxygenation, enhancing the efficacy of radiotherapy and photodynamic therapy. IONPs can also trigger ferroptosis, a programmed cell death pathway mediated by iron-dependent lipid peroxidation. Their magneto-mechanical effect allows them to exert a mechanical force on cancer cells to destroy tumors, minimizing the damage to healthy tissue. This review outlines strategies for the management of the photothermal performance and PTT efficiency with iron oxide nanoparticles, as well as synergies with other cancer therapies.
Collapse
Affiliation(s)
- Tsenka Grancharova
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Plamen Zagorchev
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Bissera Pilicheva
- Research Institute, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| |
Collapse
|
37
|
Guo T, Wang Y, Chen D, Cui S, Guo S, Feng Y, Zhu J, Chang L, Zhang J, Gao X, Wei X. Dual-Drug Loaded Nanobubbles Combined with Sonodynamic and Chemotherapy for Hepatocellular Carcinoma Therapy. Int J Nanomedicine 2024; 19:7367-7381. [PMID: 39050872 PMCID: PMC11268764 DOI: 10.2147/ijn.s460329] [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: 01/23/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
Purpose Chemotherapy remains the primary therapeutic approach for advanced Hepatocellular Carcinoma (HCC). The therapeutic effect of chemotherapy is limited and the toxic side effects are serious. The aim of this study is to develop a nanobubble that is ultrasonically responsive to reduce the toxic side effects of direct chemotherapy. Methods We developed curcumin/doxorubicin-cis-aconitic anhydride-polyethylene glycol nanobubble (C/DCNB) surface modified with acid-sensitive polyethylene glycol (PEG). And it is loaded with curcumin (CUR) and doxorubicin (DOX), as liposomes at the nanoscale for diagnosis and therapy of tumors. Results In this study, the acid-sensitive PEG on the surface layer of nanobubbles serves to stabilize them in the blood circulatory system and in normal tissues, while peeling off in the acidic tumor microenvironment (pH 6.8). C/DCNB can identify tumor sites through contrast-enhanced ultrasound (CEUS). And ultrasound-mediated nanobubbles promote permeability of the tumor vascular, thus improving the enhanced permeability and retention (EPR) effects in the tumor, leading to the accumulation of nanobubbles in the tumor. After endocytosis of nanobubbles, drugs are released and curcumin generates reactive oxygen species (ROS) under ultrasound conditions. CUR can enhance the sensitivity of tumor cells to DOX by inhibiting the expression of P-glycoprotein. In vitro and vivo experiments demonstrate that C/DCNB can facilitate contrast-enhanced ultrasound imaging while simultaneously delivering drugs, enabling both imaging and treatment. Conclusion The combination of C/DCNB and ultrasound provides an effective strategy for improving the efficiency of HCC therapy and imaging.
Collapse
Affiliation(s)
- Tiantian Guo
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, People’s Republic of China
| | - Yao Wang
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, People’s Republic of China
| | - Dixuan Chen
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, 300070, People’s Republic of China
| | - Sifan Cui
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, 300070, People’s Republic of China
| | - Shuyue Guo
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, People’s Republic of China
| | - Yixing Feng
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, People’s Republic of China
| | - Jialin Zhu
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, People’s Republic of China
| | - Luchen Chang
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, People’s Republic of China
| | - Jiawei Zhang
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, 300070, People’s Republic of China
| | - Xiujun Gao
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, 300070, People’s Republic of China
| | - Xi Wei
- Department of Diagnostic and Therapeutic Ultrasonography, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University, Tianjin, 300060, People’s Republic of China
| |
Collapse
|
38
|
Sun X, He Z, Lu R, Liu Z, Chiampanichayakul S, Anuchapreeda S, Jiang J, Tima S, Zhong Z. Hyaluronic acid-modified liposomes Potentiated in-vivo anti-hepatocellular carcinoma of icaritin. Front Pharmacol 2024; 15:1437515. [PMID: 39055490 PMCID: PMC11270019 DOI: 10.3389/fphar.2024.1437515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction: Icaritin (ICT), a promising anti-hepatocellular carcinoma (HCC) prenylated flavonoid, is hindered from being applied due to its low water solubility and high lipophilicity in poorly differentiated HCC which is associated with upregulation of CD44 isoforms. Thus, hyaluronic acid (HA), a natural polysaccharide with high binding ability to CD44 receptors, was used to formulate a modified liposome as a novel targeted ICT-delivery system for HCC treatment. Methods: The ICT-Liposomes (Lip-ICT) with and without HA were prepared by a combined method of thin-film dispersion and post-insertion. The particle size, polydispersity (PDI), zeta potential, encapsulation efficacy (%EE), drug loading content (%DLC), and in vitro drug release profiles were investigated for physicochemical properties, whereas MTT assay was used to assess cytotoxic effects on HCC cells, HepG2, and Huh7 cells. Tumor bearing nude mice were used to evaluate the inhibitory effect of HA-Lip-ICT and Lip-ICT in vivo. Results: Lip-ICT and HA-Lip-ICT had an average particle size of 171.2 ± 1.2 nm and 208.0 ± 3.2 nm, with a zeta potential of -13.9 ± 0.83 and -24.8 ± 0.36, respectively. The PDI resulted from Lip-ICT and HA-Lip-ICT was 0.28 ± 0.02 and 0.26 ± 0.02, respectively. HA-Lip-ICT demonstrated higher in vitro drug release when pH was dropped from 7.4 to 5.5, The 12-h release rate of ICT from liposomes increased from 30% at pH7.4 to more than 60% at pH5.5. HA-Lip-ICT displayed higher toxicity than Lip-ICT in both HCC cells, especially Huh7with an IC50 of 34.15 ± 2.11 μM. The in vivo tissue distribution and anti-tumor experiments carried on tumor bearing nude mice indicated that HA-Lip- ICT exhibited higher tumor accumulation and achieved a tumor growth inhibition rate of 63.4%. Discussion: The nano-sized Lip-ICT was able to prolong the drug release time and showed long-term killing HCC cells ability. Following conjugation with HA, HA-Lip-ICT exhibited higher cytotoxicity, stronger tumor targeting, and tumor suppression abilities than Lip-ICT attributed to HA-CD44 ligand-receptor interaction, increasing the potential of ICT to treat HCC.
Collapse
Affiliation(s)
- Xiaoduan Sun
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zhenzhen He
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Ruilin Lu
- Suining First People’s Hospital, Suining, China
| | - Zhongbing Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Sawitree Chiampanichayakul
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Cancer Research Unit of Associated Medical Sciences (AMS-CRU), Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | - Songyot Anuchapreeda
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Cancer Research Unit of Associated Medical Sciences (AMS-CRU), Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | - Jun Jiang
- Department of General Surgery (Thyroid Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Singkome Tima
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Cancer Research Unit of Associated Medical Sciences (AMS-CRU), Chiang Mai University, Chiang Mai, Thailand
- Center of Excellence in Pharmaceutical Nanotechnology, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | - Zhirong Zhong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
- Central Nervous System Drug Key Laboratory of Sichuan Province, Luzhou, China
| |
Collapse
|
39
|
Alharbi HM. Exploring the Frontier of Biopolymer-Assisted Drug Delivery: Advancements, Clinical Applications, and Future Perspectives in Cancer Nanomedicine. Drug Des Devel Ther 2024; 18:2063-2087. [PMID: 38882042 PMCID: PMC11178098 DOI: 10.2147/dddt.s441325] [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: 11/06/2023] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
The burgeoning global mortality rates attributed to cancer have precipitated a critical reassessment of conventional therapeutic modalities, most notably chemotherapy, due to their pronounced adverse effects. This reassessment has instigated a paradigmatic shift towards nanomedicine, with a particular emphasis on the potentialities of biopolymer-assisted drug delivery systems. Biopolymers, distinguished by their impeccable biocompatibility, versatility, and intrinsic biomimetic properties, are rapidly ascending as formidable vectors within the cancer theragnostic arena. This review endeavors to meticulously dissect the avant-garde methodologies central to biopolymer-based nanomedicine, exploring their synthesis, functional mechanisms, and subsequent clinical ramifications. A key focus of this analysis is the pioneering roles and efficacies of lipid-based, polysaccharide, and composite nano-carriers in enhancing drug delivery, notably amplifying the enhanced permeation and retention effect. This examination is further enriched by referencing flagship nano formulations that have received FDA endorsement, thereby underscoring the transformative potential and clinical viability of biopolymer-based nanomedicines. Furthermore, this discourse illuminates groundbreaking advancements in the realm of photodynamic therapy and elucidates the implications of advanced imaging techniques in live models. Conclusively, this review not only synthesizes current research trajectories but also delineates visionary pathways for the integration of cutting-edge biomaterials in cancer treatment. It charts a course for future explorations within the dynamic domain of biopolymer-nanomedicine, thereby contributing to a deeper understanding and enhanced application of these novel therapeutic strategies.
Collapse
Affiliation(s)
- Hanan M Alharbi
- Department of Pharmaceutical Sciences, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
40
|
Amulya E, Bahuguna D, Negi M, Phatale V, Sikder A, Vambhurkar G, Katta CB, Dandekar MP, Madan J, Srivastava S. Lipid engineered nanomaterials: A novel paradigm shift for combating stroke. APPLIED MATERIALS TODAY 2024; 38:102194. [DOI: 10.1016/j.apmt.2024.102194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
41
|
Al-Thamarani S, Gad S, Abdel Fattah IO, Hammadi SH, Hammady TM. Comparative analysis of oral and local intraovarian administration of metformin and nanoparticles (NPs11) in alleviating testosterone-induced polycystic ovary syndrome in rats. Tissue Cell 2024; 88:102394. [PMID: 38663112 DOI: 10.1016/j.tice.2024.102394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 06/17/2024]
Abstract
Polycystic ovary syndrome (PCOS) is an endocrine and metabolic dysfunction. This study aims to compare the oral and local treatments of metformin or its nanoparticles (NPs11) for ameliorating PCOS in rats. Rats were divided into 4 groups: the control group with no drug treatment; the PCOS group, where subcutaneous testosterone was given (10 mg/kg/day) for 28 days; the MET group, where metformin was administered orally or locally; and the NP group, where metformin NPs11 were also administered orally or locally. Oral administrations were for 21 days, while local injection was performed once surgically. After 7 weeks, all rats were sacrificed; blood glucose and serum hormonal levels and lipid profile were estimated, and the ovaries were assessed by histopathological, Ki-67 immunohistochemical, and histomorphometric evaluations. Blood glucose levels were significantly decreased in groups of orally administered metformin or NPs11 only, while the most efficient option for modulating PCOS-induced hormonal and lipid profile changes was intraovarian injection of NPs11. The ovaries of PCOS rats demonstrated large follicular cysts, massive collagen depositions, and attenuated Ki-67 immunoexpression. Also, the PCOS group revealed a significant decrease in the count of all stages of growing follicles, corpora lutea, granulosa cell layer thickness, and surface area of corpora lutea, in addition to an increase in the number of atretic follicles and follicular cysts, theca cell layer thickness, and surface area of the follicular cysts. All these parameters were recovered with metformin or their NPs11 treatments in different degrees, while local injection of NPs11 was the best option.
Collapse
Affiliation(s)
- Sadeq Al-Thamarani
- Department of Pharmacy, Faculty of Medicine and Health Sciences, Thamar University, Dhamar 87246, Yemen
| | - Shadeed Gad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| | - Islam Omar Abdel Fattah
- Department of Human Anatomy and Embryology, Faculty of Medicine, Suez Canal University, Ismailia 41522, Egypt.
| | - Sami H Hammadi
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria 21526, Egypt
| | - Taha M Hammady
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
42
|
Mendes BB, Zhang Z, Conniot J, Sousa DP, Ravasco JMJM, Onweller LA, Lorenc A, Rodrigues T, Reker D, Conde J. A large-scale machine learning analysis of inorganic nanoparticles in preclinical cancer research. NATURE NANOTECHNOLOGY 2024; 19:867-878. [PMID: 38750164 DOI: 10.1038/s41565-024-01673-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 04/10/2024] [Indexed: 06/21/2024]
Abstract
Owing to their distinct physical and chemical properties, inorganic nanoparticles (NPs) have shown promising results in preclinical cancer therapy, but designing and engineering them for effective therapeutic purposes remains a challenge. Although a comprehensive database of inorganic NP research is not currently available, it is crucial for developing effective cancer therapies. In this context, machine learning (ML) has emerged as a transformative tool, but its adaptation to nanomedicine is hindered by inexistent or small datasets. Here we assembled a large database of inorganic NPs, comprising experimental datasets from 745 preclinical studies in cancer nanomedicine. Using descriptive statistics and explainable ML models we mined this database to gain knowledge of inorganic NP design patterns and inform future NP research for cancer treatment. Our analyses suggest that NP shape and therapy type are prominent features in determining in vivo efficacy, measured as a percentage of tumour reduction. Moreover, our database provides a large-scale open-access resource for discriminative ML that the broader nanotechnology community can utilize. Our work blueprints data mining for translational cancer research and offers evidence for standardizing NP reporting to accelerate and de-risk inorganic NP-based drug delivery, which may help to improve patient outcomes in clinical settings.
Collapse
Affiliation(s)
- Bárbara B Mendes
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Zilu Zhang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - João Conniot
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Diana P Sousa
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - João M J M Ravasco
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Lauren A Onweller
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Andżelika Lorenc
- Instituto de Investigação do Medicamento (iMed), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
- Department of Biopharmacy, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Tiago Rodrigues
- Instituto de Investigação do Medicamento (iMed), Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal.
| | - Daniel Reker
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA.
| | - João Conde
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas (NMS|FCM), Universidade NOVA de Lisboa, Lisbon, Portugal.
| |
Collapse
|
43
|
Jiang X, Xu S, Miao Y, Huang K, Wang B, Ding B, Zhang Z, Zhao Z, Zhang X, Shi X, Yu M, Tian F, Gan Y. Curvature-mediated rapid extravasation and penetration of nanoparticles against interstitial fluid pressure for improved drug delivery. Proc Natl Acad Sci U S A 2024; 121:e2319880121. [PMID: 38768353 PMCID: PMC11145294 DOI: 10.1073/pnas.2319880121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 04/12/2024] [Indexed: 05/22/2024] Open
Abstract
Elevated interstitial fluid pressure (IFP) within pathological tissues (e.g., tumors, obstructed kidneys, and cirrhotic livers) creates a significant hindrance to the transport of nanomedicine, ultimately impairing the therapeutic efficiency. Among these tissues, solid tumors present the most challenging scenario. While several strategies through reducing tumor IFP have been devised to enhance nanoparticle delivery, few approaches focus on modulating the intrinsic properties of nanoparticles to effectively counteract IFP during extravasation and penetration, which are precisely the stages obstructed by elevated IFP. Herein, we propose an innovative solution by engineering nanoparticles with a fusiform shape of high curvature, enabling efficient surmounting of IFP barriers during extravasation and penetration within tumor tissues. Through experimental and theoretical analyses, we demonstrate that the elongated nanoparticles with the highest mean curvature outperform spherical and rod-shaped counterparts against elevated IFP, leading to superior intratumoral accumulation and antitumor efficacy. Super-resolution microscopy and molecular dynamics simulations uncover the underlying mechanisms in which the high curvature contributes to diminished drag force in surmounting high-pressure differentials during extravasation. Simultaneously, the facilitated rotational movement augments the hopping frequency during penetration. This study effectively addresses the limitations posed by high-pressure impediments, uncovers the mutual interactions between the physical properties of NPs and their environment, and presents a promising avenue for advancing cancer treatment through nanomedicine.
Collapse
Affiliation(s)
- Xiaohe Jiang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sai Xu
- University of Chinese Academy of Sciences, Beijing 100049, China
- Chinese Academy of Sciences Center for Excellence in Nanoscience National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yunqiu Miao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Kang Huang
- University of Chinese Academy of Sciences, Beijing 100049, China
- Chinese Academy of Sciences Center for Excellence in Nanoscience National Center for Nanoscience and Technology, Beijing 100190, China
| | - Bingqi Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bingwen Ding
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhuan Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Zitong Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, Henan University, Kaifeng 475004, China
| | - Xinxin Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xinghua Shi
- University of Chinese Academy of Sciences, Beijing 100049, China
- Chinese Academy of Sciences Center for Excellence in Nanoscience National Center for Nanoscience and Technology, Beijing 100190, China
| | - Miaorong Yu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Falin Tian
- University of Chinese Academy of Sciences, Beijing 100049, China
- Chinese Academy of Sciences Center for Excellence in Nanoscience National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yong Gan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- National Medical Products Administration Key Laboratory for Quality Research and Evaluation of Pharmaceutical Excipients, National Institutes for Food and Drug Control, Beijing 100050, China
| |
Collapse
|
44
|
Aebisher D, Rogóż K, Myśliwiec A, Dynarowicz K, Wiench R, Cieślar G, Kawczyk-Krupka A, Bartusik-Aebisher D. The use of photodynamic therapy in medical practice. Front Oncol 2024; 14:1373263. [PMID: 38803535 PMCID: PMC11129581 DOI: 10.3389/fonc.2024.1373263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024] Open
Abstract
Cancer therapy, especially for tumors near sensitive areas, demands precise treatment. This review explores photodynamic therapy (PDT), a method leveraging photosensitizers (PS), specific wavelength light, and oxygen to target cancer effectively. Recent advancements affirm PDT's efficacy, utilizing ROS generation to induce cancer cell death. With a history spanning over decades, PDT's dynamic evolution has expanded its application across dermatology, oncology, and dentistry. This review aims to dissect PDT's principles, from its inception to contemporary medical applications, highlighting its role in modern cancer treatment strategies.
Collapse
Affiliation(s)
- David Aebisher
- Department of Photomedicine and Physical Chemistry, Medical College of The Rzeszów University, Rzeszów, Poland
| | - Kacper Rogóż
- English Division Science Club, Medical College of The Rzeszów University, Rzeszów, Poland
| | - Angelika Myśliwiec
- Center for Innovative Research in Medical and Natural Sciences, Medical College of The University of Rzeszów, Rzeszów, Poland
| | - Klaudia Dynarowicz
- Center for Innovative Research in Medical and Natural Sciences, Medical College of The University of Rzeszów, Rzeszów, Poland
| | - Rafał Wiench
- Department of Periodontal Diseases and Oral Mucosa Diseases, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Zabrze, Poland
| | - Grzegorz Cieślar
- Department of Internal Medicine, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Medical University of Silesia, Bytom, Poland
| | - Aleksandra Kawczyk-Krupka
- Department of Internal Medicine, Angiology and Physical Medicine, Center for Laser Diagnostics and Therapy, Medical University of Silesia, Bytom, Poland
| | - Dorota Bartusik-Aebisher
- Department of Biochemistry and General Chemistry, Medical College of The Rzeszów University, Rzeszów, Poland
| |
Collapse
|
45
|
Karpov TE, Darwish A, Mitusova K, Postovalova AS, Akhmetova DR, Vlasova OL, Shipilovskikh SA, Timin AS. Controllable synthesis of barium carbonate nano- and microparticles for SPECT and CT imaging. J Mater Chem B 2024; 12:4232-4247. [PMID: 38601990 DOI: 10.1039/d3tb02480f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
The design and synthesis of nano- and microcarriers for preclinical and clinical imaging are highly attractive due to their unique features, for example, multimodal properties. However, broad translation of these carriers into clinical practice is postponed due to the unknown biological reactivity of the new components used for their synthesis. Here, we have developed microcarriers (∼2-3 μm) and nanocarriers (<200 nm) made of barium carbonate (BaCO3) for multiple imaging applications in vivo. In general, barium in the developed carriers can be used for X-ray computed tomography, and the introduction of a diagnostic isotope (99mTc) into the BaCO3 structure enables in vivo visualization using single-photon emission computed tomography. The bioimaging has shown that the radiolabeled BaCO3 nano- and microcarriers had different biodistribution profiles and tumor accumulation efficiencies after intratumoral and intravenous injections. In particular, in the case of intratumoral injection, all the types of used carriers mostly remained in the tumors (>97%). For intravenous injection, BaCO3 microcarriers were mainly localized in the lung tissues. However, BaCO3 NPs were mainly accumulated in the liver. These results were supported by ex vivo fluorescence imaging, direct radiometry, and histological analysis. The BaCO3-based micro- and nanocarriers showed negligible in vivo toxicity towards major organs such as the heart, lungs, liver, kidneys, and spleen. This study provides a simple strategy for the design and fabrication of the BaCO3-based carriers for the development of dual bioimaging.
Collapse
Affiliation(s)
- Timofey E Karpov
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg 195251, Russian Federation.
- Granov Russian Research Center of Radiology & Surgical Technologies, Leningradskaya Street 70 Pesochny, St. Petersburg 197758, Russian Federation
| | - Aya Darwish
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg 195251, Russian Federation.
| | - Ksenia Mitusova
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg 195251, Russian Federation.
| | - Alisa S Postovalova
- Granov Russian Research Center of Radiology & Surgical Technologies, Leningradskaya Street 70 Pesochny, St. Petersburg 197758, Russian Federation
- ITMO University, Lomonosova 9, St. Petersburg 191002, Russian Federation
| | - Darya R Akhmetova
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg 195251, Russian Federation.
- ITMO University, Lomonosova 9, St. Petersburg 191002, Russian Federation
| | - Olga L Vlasova
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg 195251, Russian Federation.
| | | | - Alexander S Timin
- Peter The Great St. Petersburg Polytechnic University, Polytechnicheskaya 29, St. Petersburg 195251, Russian Federation.
| |
Collapse
|
46
|
Cooley MB, Wegierak D, Exner AA. Using imaging modalities to predict nanoparticle distribution and treatment efficacy in solid tumors: The growing role of ultrasound. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1957. [PMID: 38558290 PMCID: PMC11006412 DOI: 10.1002/wnan.1957] [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: 04/26/2023] [Revised: 12/22/2023] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
Nanomedicine in oncology has not had the success in clinical impact that was anticipated in the early stages of the field's development. Ideally, nanomedicines selectively accumulate in tumor tissue and reduce systemic side effects compared to traditional chemotherapeutics. However, this has been more successful in preclinical animal models than in humans. The causes of this failure to translate may be related to the intra- and inter-patient heterogeneity of the tumor microenvironment. Predicting whether a patient will respond positively to treatment prior to its initiation, through evaluation of characteristics like nanoparticle extravasation and retention potential in the tumor, may be a way to improve nanomedicine success rate. While there are many potential strategies to accomplish this, prediction and patient stratification via noninvasive medical imaging may be the most efficient and specific strategy. There have been some preclinical and clinical advances in this area using MRI, CT, PET, and other modalities. An alternative approach that has not been studied as extensively is biomedical ultrasound, including techniques such as multiparametric contrast-enhanced ultrasound (mpCEUS), doppler, elastography, and super-resolution processing. Ultrasound is safe, inexpensive, noninvasive, and capable of imaging the entire tumor with high temporal and spatial resolution. In this work, we summarize the in vivo imaging tools that have been used to predict nanoparticle distribution and treatment efficacy in oncology. We emphasize ultrasound imaging and the recent developments in the field concerning CEUS. The successful implementation of an imaging strategy for prediction of nanoparticle accumulation in tumors could lead to increased clinical translation of nanomedicines, and subsequently, improved patient outcomes. This article is categorized under: Diagnostic Tools In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery Nanomedicine for Oncologic Disease Therapeutic Approaches and Drug Discovery Emerging Technologies.
Collapse
Affiliation(s)
- Michaela B Cooley
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Dana Wegierak
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Agata A Exner
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Radiology, Case Western Reserve University and University Hospitals of Cleveland, Cleveland, Ohio, USA
| |
Collapse
|
47
|
Wang S, Liu T, Huang Y, Du C, Wang D, Wang X, Lv Q, He Z, Zhai Y, Sun B, Sun J. The effect of lengths of branched-chain fatty alcohols on the efficacy and safety of docetaxel-prodrug nanoassemblies. Acta Pharm Sin B 2024; 14:1400-1411. [PMID: 38486988 PMCID: PMC10934334 DOI: 10.1016/j.apsb.2023.09.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/18/2023] [Accepted: 09/14/2023] [Indexed: 03/17/2024] Open
Abstract
The self-assembly prodrugs are usually consisted of drug modules, activation modules, and assembly modules. Keeping the balance between efficacy and safety by selecting suitable modules remains a challenge for developing prodrug nanoassemblies. This study designed four docetaxel (DTX) prodrugs using disulfide bonds as activation modules and different lengths of branched-chain fatty alcohols as assembly modules (C16, C18, C20, and C24). The lengths of the assembly modules determined the self-assembly ability of prodrugs and affected the activation modules' sensitivity. The extension of the carbon chains improved the prodrugs' self-assembly ability and pharmacokinetic behavior while reducing the cytotoxicity and increased cumulative toxicity. The use of C20 can balance efficacy and safety. These results provide a great reference for the rational design of prodrug nanoassemblies.
Collapse
Affiliation(s)
- Shuo Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Tian Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yuetong Huang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chaoying Du
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Danping Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiyan Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qingzhi Lv
- School of Pharmacy, Binzhou Medical University, Binzhou 256600, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yinglei Zhai
- School of Medical Devices, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bingjun Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
48
|
Cheng Y, Zhang H, Wei H, Yu CY. Injectable hydrogels as emerging drug-delivery platforms for tumor therapy. Biomater Sci 2024; 12:1151-1170. [PMID: 38319379 DOI: 10.1039/d3bm01840g] [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: 02/07/2024]
Abstract
Tumor therapy continues to be a prominent field within biomedical research. The development of various drug carriers has been propelled by concerns surrounding the side effects and targeting efficacy of various chemotherapeutic drugs and other therapeutic agents. These carriers strive to enhance drug concentration at tumor sites, minimize systemic side effects, and improve therapeutic outcomes. Among the reported delivery systems, injectable hydrogels have emerged as an emerging candidate for the in vivo delivery of chemotherapeutic drugs due to their minimal invasive drug delivery properties. This review systematically summarizes the composition and preparation methodologies of injectable hydrogels and further highlights the delivery mechanisms of diverse drugs using these hydrogels for tumor therapy, along with an in-depth discussion on the optimized therapeutic efficiency of drugs encapsulated within the hydrogels. The work concludes by providing a dynamic forward-looking perspective on the potential challenges and possible solutions of the in situ injectable hydrogels for non-surgical and real-time diagnostic applications.
Collapse
Affiliation(s)
- Yao Cheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China.
| | - Haitao Zhang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China.
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China.
| |
Collapse
|
49
|
Wang L, Wu Y, Yang N, Yin W, Yang H, Li C, Zhuang Y, Song Z, Cheng X, Shi S, Wu Y. Self-assembly of maltose-albumin nanoparticles for efficient targeting delivery and therapy in liver cancer. Int J Biol Macromol 2024; 258:128691. [PMID: 38072344 DOI: 10.1016/j.ijbiomac.2023.128691] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/03/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024]
Abstract
The effective delivery and targeted release of drugs within tumor cells are critical factors in determining the therapeutic efficacy of nanomedicine. To achieve this objective, a conjugate of maltose (Mal) and bovine serum albumin (BSA) was synthesized by the Maillard reaction and self-assembled into nanoparticles with active-targeting capabilities upon pH/heating induction. This nanoparticle could be effectively loaded with doxorubicin (DOX) to form stable nanodrugs (Mal-BSA/DOX) that were sensitive to low pH or high glutathione (GSH), thereby achieving a rapid drug release (96.82 % within 24 h). In vitro cell experiments indicated that maltose-modified BSA particles efficiently enhance cellular internalization via glucose transporters (GLUT)-mediated endocytosis, resulting in increased intracellular DOX levels and heightened expression of γ-H2AX. Consequently, these results ultimately lead to selective tumor cells death, as evidenced by an IC50 value of 3.83 μg/mL in HepG2 cells compared to 5.87 μg/mL in 293t cells. The efficacy of Mal-BSA/DOX in tumor targeting therapy has been further confirmed by in vivo studies, as it effectively delivered a higher concentration of DOX to tumor tissue. This targeted delivery approach not only reduces the systemic toxicity of DOX but also effectively inhibits tumor growth (TGI, 75.95 %). These findings contribute valuable insights into the advancement of targeting-albumin nanomedicine and further support its potential in tumor treatment.
Collapse
Affiliation(s)
- Lu Wang
- Collaborative Innovation Center of targeted Development of Medicinal Resources, Anqing Normal University, Anqing 246133, PR China
| | - Yirui Wu
- Collaborative Innovation Center of targeted Development of Medicinal Resources, Anqing Normal University, Anqing 246133, PR China
| | - Niuniu Yang
- Collaborative Innovation Center of targeted Development of Medicinal Resources, Anqing Normal University, Anqing 246133, PR China
| | - Wenting Yin
- Collaborative Innovation Center of targeted Development of Medicinal Resources, Anqing Normal University, Anqing 246133, PR China
| | - Huang Yang
- Collaborative Innovation Center of targeted Development of Medicinal Resources, Anqing Normal University, Anqing 246133, PR China
| | - Conghu Li
- Collaborative Innovation Center of targeted Development of Medicinal Resources, Anqing Normal University, Anqing 246133, PR China; Belt and Road Model International Science and Technology Cooperation Base for Biodiversity Conservation and Utilization in Basins of Anhui Province, Anqing 246133, PR China
| | - Yan Zhuang
- Collaborative Innovation Center of targeted Development of Medicinal Resources, Anqing Normal University, Anqing 246133, PR China
| | - Ziyi Song
- Collaborative Innovation Center of targeted Development of Medicinal Resources, Anqing Normal University, Anqing 246133, PR China
| | - Xu Cheng
- Collaborative Innovation Center of targeted Development of Medicinal Resources, Anqing Normal University, Anqing 246133, PR China; Belt and Road Model International Science and Technology Cooperation Base for Biodiversity Conservation and Utilization in Basins of Anhui Province, Anqing 246133, PR China.
| | - Shuiqing Shi
- Collaborative Innovation Center of targeted Development of Medicinal Resources, Anqing Normal University, Anqing 246133, PR China; Belt and Road Model International Science and Technology Cooperation Base for Biodiversity Conservation and Utilization in Basins of Anhui Province, Anqing 246133, PR China.
| | - Yan Wu
- Collaborative Innovation Center of targeted Development of Medicinal Resources, Anqing Normal University, Anqing 246133, PR China; Belt and Road Model International Science and Technology Cooperation Base for Biodiversity Conservation and Utilization in Basins of Anhui Province, Anqing 246133, PR China
| |
Collapse
|
50
|
Merlin JPJ, Crous A, Abrahamse H. Nano-phototherapy: Favorable prospects for cancer treatment. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1930. [PMID: 37752098 DOI: 10.1002/wnan.1930] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/28/2023]
Abstract
Nanotechnology-based phototherapies have drawn interest in the fight against cancer because of its noninvasiveness, high flexibility, and precision in terms of cancer targeting and drug delivery based on its surface properties and size. Phototherapy has made remarkable development in recent decades. Approaches to phototherapy, which utilize nanomaterials or nanotechnology have emerged to contribute to advances around nanotechnologies in medicine, particularly for cancers. A brief overviews of the development of photodynamic therapy as well as its mechanism in cancer treatment is provided. We emphasize the design of novel nanoparticles utilized in photodynamic therapy while summarizing the representative progress during the recent years. Finally, to forecast important future research in this area, we examine the viability and promise of photodynamic therapy systems based on nanoparticles in clinical anticancer treatment applications and briefly make mention of the elimination of all reactive metabolites pertaining to nano formulations inside living organisms providing insight into clinical mechanistic processes. Future developments and therapeutic prospects for photodynamic treatments are anticipated. Our viewpoints might encourage scientists to create more potent phototherapy-based cancer therapeutic modalities. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- J P Jose Merlin
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Anine Crous
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|