1
|
Chen S, Fan J, Xie Q, Qin Y, Xie H, Xiao C, Wang W, Liu B. Bufotalin loaded biomimetic nanodrug for combined chemo/photodynamic therapy of cancer. Mater Today Bio 2025; 32:101684. [PMID: 40206143 PMCID: PMC11979420 DOI: 10.1016/j.mtbio.2025.101684] [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: 01/13/2025] [Revised: 03/03/2025] [Accepted: 03/17/2025] [Indexed: 04/11/2025] Open
Abstract
The combination of chemotherapy and photodynamic therapy (PDT) for enhancing cancer therapeutic efficiency has attracted tremendous attention recently. However, limitations, such as low local concentration and uncontrollable release of therapeutic agents, reduce combined treatment efficacy. In the present study, we engineered a biomimetic nanodrug employing hollow Prussian blue nanoparticles (HPB NPs) to co-load the chemical agent bufotalin (CS-5) and the photosensitizer chlorin e6 (Ce6) for combined chemo/PDT therapy against cancer. HPB NPs with catalase (CAT)-mimetic activity significantly improved the efficacy of PDT by catalyzing the decomposition of H2O2 into O2, thus alleviating hypoxia, which conversely amplified the efficiency of combination therapy. In vivo assay demonstrated that the encapsulation of a hybrid membrane on the HPB NPs prolonged blood circulation life 3.4-fold compared to free drug. Additionally, this strategy of combinational chemo/PDT therapy exhibits a remarkable cytotoxic effect against gastric cancer (BGC-823) and breast cancer (4T1) through the induction of ferroptosis and pyroptosis while simultaneously activating the immune response, with minimal adverse effects on normal organs. Thus, the co-delivery system based on biomimetic nanocarriers appears to be a promising platform for combined chemo/PDT therapy in tumor treatment.
Collapse
Affiliation(s)
- Simin Chen
- College of Biology, Hunan University, Changsha, 410082, China
| | - Jialong Fan
- College of Biology, Hunan University, Changsha, 410082, China
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, 410219, China
| | - Qian Xie
- Hunan Provincial Maternal and Child Health Care Hospital, Hunan Province, Changsha 410008, China
| | - Yan Qin
- College of Biology, Hunan University, Changsha, 410082, China
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Hailong Xie
- Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Institute of Cancer Research, School of Medicine, University of South China, Hengyang, 421001, China
| | - Chang Xiao
- College of Biology, Hunan University, Changsha, 410082, China
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, 410082, China
| |
Collapse
|
2
|
Sun M, Cao J, Zou Y, Ju H, Lv Y. ZIF-8 composite nanofibrous membranes loaded with bFGF: a new approach for tendon adhesion prevention and repair. Biomater Sci 2025; 13:3058-3073. [PMID: 40260599 DOI: 10.1039/d5bm00062a] [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: 04/23/2025]
Abstract
During tendon injury repair, deficiency of basic fibroblast growth factor (bFGF) is a critical factor leading to unsatisfactory repair results. This study aims to prepare bFGF-loaded zeolite imidazole framework-8 (ZIF-8) nanocrystals using a one-pot synthesis method. Subsequently, a bilayer nanofibrous membrane incorporating these drug-loaded nanocrystals was fabricated through electrospinning technology. The potential of this composite nanofibrous membrane to facilitate the continuous release of bFGF at the site of tendon injury was evaluated, with the aim of enhancing the quality of tendon repair. The efficacy of the nanofibrous membrane in promoting tendon differentiation, preventing tendon adhesion, and facilitating tendon repair was assessed through both in vitro and in vivo experiments. At the site of tendon injury, the degradation of ZIF-8 in an acidic microenvironment resulted in the release of bFGF and Zn2+, which contributed to the enhancement of tendon repair. ZIF-8 nanocrystals achieved an encapsulation efficiency of 50.13% ± 1.42%. Following a continuous release period exceeding 40 days, the cumulative in vitro release rate was determined to be 35.02% ± 4.27%. The incorporation of ZIF-8 nanocrystals into a nanofibrous membrane demonstrated the ability to effectively preserve the bioactivity of bFGF while enabling sustained release at the site of tendon injury, thereby facilitating tendon repair. The findings offer novel insights into the treatment of tendon injuries and provide significant theoretical guidance for the tendon repair process.
Collapse
Affiliation(s)
- Min Sun
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, 430200, P. R. China.
| | - Jinke Cao
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, 430200, P. R. China.
| | - Yang Zou
- School of Resources and Environment, Wuhan Textile University, Wuhan, 430200, P. R. China
| | - Haiyan Ju
- College of Chemistry and Chemical Engineering, Wuhan Textile University, Wuhan 430200, P. R. China
| | - Yonggang Lv
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, 430200, P. R. China.
| |
Collapse
|
3
|
Chen Y, Cai S, Liu FY, Liu M. Advancing oral cancer care: nanomaterial-driven diagnostic and therapeutic innovations. Cell Biol Toxicol 2025; 41:90. [PMID: 40407908 PMCID: PMC12102110 DOI: 10.1007/s10565-025-10027-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 04/13/2025] [Indexed: 05/26/2025]
Abstract
The advent of nanotechnology has significantly advanced the diagnosis and treatment of oral cancer, offering more precise and efficient therapeutic strategies. This review presents a comprehensive overview of recent developments in the application of nanotechnology to oral cancer management. It begins with an overview of the epidemiology of oral cancer and outlines current diagnostic and therapeutic methods. The classification and advantages of various nanomaterials are then introduced. The paper thoroughly explores the use of nanomaterials as drug delivery systems (DDSs), imaging contrast agents, and therapeutic tools, with particular emphasis on multifunctional nanoplatforms that integrate diagnostics and therapy. These platforms enable real-time monitoring and immediate therapeutic response, offering innovative approaches for early detection and intervention. Despite these promising advances, several challenges persist, including issues related to biocompatibility, clearance, targeting specificity, and clinical translation. The review concludes by highlighting current limitations and proposing future directions for the clinical application of nanotechnology in oral cancer treatment.
Collapse
Affiliation(s)
- Yuwen Chen
- Departmentof Orthodontics, School of Stomatology, China Medical University, 117 South Nanjing Street, Heping, Shenyang, Liaoning, 110002, P.R. China
| | - Sijia Cai
- Departmentof Orthodontics, School of Stomatology, China Medical University, 117 South Nanjing Street, Heping, Shenyang, Liaoning, 110002, P.R. China
| | - Fa-Yu Liu
- Department of Oromaxillofacial-Head and Neck, Oral Maxillofacial Surgery, School of Stomatology, China Medical University, Shenyang, Liaoning, 110002, P.R. China
| | - Ming Liu
- Department of Oral Radiology, School of Stomatology, China Medical University, Shenyang, Liaoning, 110002, P.R. China.
| |
Collapse
|
4
|
Sun X, Zhang X, Qin H, Li L. A Needle-Like H 2S-Releasing and H 2O 2 Self-Replenishing Nanoplatform for Enhanced Chemodynamic Tumor Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e06282. [PMID: 40400410 DOI: 10.1002/advs.202506282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/09/2025] [Indexed: 05/23/2025]
Abstract
The tumor microenvironment (TME) significantly restricts chemodynamic therapy (CDT) efficacy through hypoxia and antioxidant defenses. An intelligent cascade nanosystem, PTA-SnS2@GOx, is developed by integrating a tannic acid-modified Prussian blue analogue core, SnS2 shell, and glucose oxidase (GOx) activation module. The needle-like nanostructure enhanced tumor accumulation and cellular uptake. GOx-mediated glucose oxidation generated H2O2 and gluconic acid, triggering pH-responsive H2S release from SnS2. This gas disrupted mitochondrial respiration and catalase activity, alleviating hypoxia while elevating intracellular H2O2 levels. The oxygenated TME subsequently amplified GOx biocatalysis, establishing a self-sustaining cycle of H2O2 production and acidification. Concurrently, Sn4+ ions depleted glutathione, synergistically enhancing Fenton-like reactions in the PTA core for reinforced ROS generation. This multi-tiered strategy achieved effective CDT through the coordinated mechanisms: continuous H2O2 self-supply, pH reduction, and redox homeostasis disruption. Notably, the nanosystem induced immunogenic cell death, promoting dendritic cell maturation and repolarizing tumor-associated macrophages from M2 to M1 phenotype, thereby remodeling immunosuppressive TME and activating systemic antitumor immunity. The synergistic integration of self-amplifying CDT with immune sensitization demonstrates superior tumor suppression in vivo. This study provided an intelligent paradigm for cancer theranostics by combining self-supplying H2S/H2O2-enhanced CDT with sensitized immunotherapy.
Collapse
Affiliation(s)
- Xiaoxiao Sun
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- Department of stomatology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xia Zhang
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Haiyan Qin
- Department of stomatology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Lingling Li
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| |
Collapse
|
5
|
Wang C, Ren K, Yang M, Li X, Li N, Li P, Yang H, Zhang G, Wei X. How Traditional Chinese Medicine Can Play a Role In Nanomedicine? A Comprehensive Review of the Literature. Int J Nanomedicine 2025; 20:6289-6315. [PMID: 40416728 PMCID: PMC12103218 DOI: 10.2147/ijn.s518610] [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: 01/21/2025] [Accepted: 05/13/2025] [Indexed: 05/27/2025] Open
Abstract
Traditional Chinese medicine (TCM), a time-honored practice rooted in natural therapeutics, has served as a cornerstone in safeguarding human health across millennia, aiding in disease mitigation and life vitality preservation. However, many TCM active ingredients suffer from poor solubility, low bioavailability, uncertain toxicity and weak targeting ability. Nanomedicine represents a modern scientific frontier, emerging from the precise engineering of unique nanoscale characteristics, with extensive applications encompassing targeted therapeutic delivery and diverse biomedical fields. Although TCM and nanomedicine diverge fundamentally in historical origins and disciplinary foundations, growing investigations demonstrate their synergistic potential. In this review, nanosized TCM has been revealed as an innovative therapeutic strategy with significant clinical value. Based on the biological activities and structural characteristics of TCM active ingredients, we classify them into two categories: natural nanostructured formulations for TCM and nano-drug delivery systems for TCM. A systematic and comprehensive analysis of preparations specific and functions to two classes of TCM nanomedicines is highlighted. Insights into the advantage of TCM nanomedicines are also introduced. Subsequently, the applications of TCM nanomedicines in the biomedical treatment, including anti-cancer, anti-inflammation and anti-bacterial are summarized. Finally, challenges and future research directions are emphasized, aiming to offer guidance for the modernization of TCM nanomedicines.
Collapse
Affiliation(s)
- Chi Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- Key Laboratory of Enhanced Recovery After Surgery of Intergrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Kaixiang Ren
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- Key Laboratory of Enhanced Recovery After Surgery of Intergrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Mei Yang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- Key Laboratory of Enhanced Recovery After Surgery of Intergrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Xiang Li
- Department of Ophthalmology, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Ningxi Li
- Mianyang Key Laboratory of Anesthesia and Neuroregulation, Department of Anesthesiology, Mianyang Central Hospital, Mianyang, 621000, People’s Republic of China
| | - Peng Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- Key Laboratory of Enhanced Recovery After Surgery of Intergrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Huang Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, People’s Republic of China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310003, People’s Republic of China
| | - Guangjian Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- Key Laboratory of Enhanced Recovery After Surgery of Intergrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| | - Xiaodan Wei
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
- Key Laboratory of Enhanced Recovery After Surgery of Intergrated Chinese and Western Medicine, Administration of Traditional Chinese Medicine of Shaanxi Province, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, People’s Republic of China
| |
Collapse
|
6
|
Aldhubiab B, Almuqbil RM, Nair AB. Harnessing the Power of Nanocarriers to Exploit the Tumor Microenvironment for Enhanced Cancer Therapy. Pharmaceuticals (Basel) 2025; 18:746. [PMID: 40430563 PMCID: PMC12115050 DOI: 10.3390/ph18050746] [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: 04/22/2025] [Revised: 05/14/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
The tumor microenvironment (TME) has a major role in malignancy and its complex nature can mediate tumor survival, metastasis, immune evasion, and drug resistance. Thus, reprogramming or regulating the immunosuppressive TME has a significant contribution to make in cancer therapy. Targeting TME with nanocarriers (NCs) has been widely used to directly deliver anticancer drugs to control TME, which has revealed auspicious outcomes. TME can be reprogrammed by using a range of NCs to regulate immunosuppressive factors and activate immunostimulatory cells. Moreover, TME can be ameliorated via regulating the redox environment, oxygen content, and pH value of the tumor site. NCs have the capacity to provide site-specific delivery of therapeutic agents, controlled release, enhanced solubility and stability, decreased toxicities, and enhanced pharmacokinetics as well as biodistribution. Numerous NCs have demonstrated their potential by inducing distinct anticancer mechanisms by delivering a range of anticancer drugs in various preclinical studies, including metal NCs, liposomal NCs, solid lipid NCs, micelles, nanoemulsions, polymer-based NCs, dendrimers, nanoclays, nanocrystals, and many more. Some of them have already received US Food and Drug Administration approval, and some have entered different clinical phases. However, there are several challenges in NC-mediated TME targeting, including scale-up of NC-based cancer therapy, rapid clearance of NCs by the mononuclear phagocyte system, and TME heterogeneity. In order to harness the full potential of NCs in tumor treatment, there are several factors that need to be carefully studied, including optimization of drug loading into NCs, NC-associated immunogenicity, and biocompatibility for the successful translation of NC-based anticancer therapies into clinical practice. In this review, a range of NCs and their applications in drug delivery to remodel TME for cancer therapy are extensively discussed. Moreover, findings from numerous preclinical and clinical studies with these NCs are also highlighted.
Collapse
Affiliation(s)
- Bandar Aldhubiab
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa 31982, Saudi Arabia; (R.M.A.); (A.B.N.)
| | | | | |
Collapse
|
7
|
Al-Shehaby N, Elshoky HA, Zidan M, Salaheldin TA, Gaber MH, Ali MA, El-Sayed NM. In vitro localization of modified zinc oxide nanoparticles showing selective anticancer effects against colorectal carcinoma using biophysical techniques. Sci Rep 2025; 15:16811. [PMID: 40369004 PMCID: PMC12078601 DOI: 10.1038/s41598-025-00434-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/28/2025] [Indexed: 05/16/2025] Open
Abstract
In recent decades, despite advancements in conventional cancer therapies, their serious side effects on both healthy and tumor cells remain a major concern. Aiming to address indiscriminate drug distribution, unwanted toxicity, and high chemotherapy doses, this study explores the targeted delivery of zinc oxide nanoparticles (ZnO NPs). ZnO NPs were synthesized and coated with bovine serum albumin (BSA) and tetraethoxysilane (TEOS) to control cellular uptake and enhance anticancer activity. Characterized by UV-visible spectroscopy, DLS, FTIR, XRD, and TEM, ZnO, ZnOB, and ZnOT particles displayed sizes of 140 ± 13.6 nm, 342 ± 8.4 nm, and 145 ± 23.8 nm, respectively, with ZnOT showing a positive charge of + 19.3 ± 4.16 mV, enhancing stability and cellular interaction. Cytotoxicity assays revealed ZnO's potent anticancer effect in Caco-2 cells with an IC50 of 219 µg/ml, while ZnOB and ZnOT showed moderate toxicity (IC50 values of 308 µg/ml and 235 µg/ml). HepG2 cells maintained viability close to 100%, highlighting ZnO NPs' selectivity for Caco-2 cells. Flow cytometry and confocal microscopy indicated differential uptake, with ZnOB showing the highest uptake in Caco-2 cells after 24 h at 37 °C, increasing fluorescence intensity by over 80% compared to ZnO. ZnOT notably increased late apoptotic cells by 65% in Caco-2 lines and caused a 40% rise in G2/M phase arrest. Mitochondrial function assays showed that ZnO reduced mitochondrial membrane potential by over 30%, indicating stress induction. These results support the potential of ZnO-based nanoparticles in colorectal cancer treatment, offering selective cytotoxicity, enhanced cellular uptake, and clear apoptotic activity, making them a promising alternative to conventional chemotherapy.
Collapse
Affiliation(s)
- Nouran Al-Shehaby
- Tumor Biology Research Program, Basic Research Unit, Research Department, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt
| | - Hisham A Elshoky
- Tumor Biology Research Program, Basic Research Unit, Research Department, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt.
- Nanotechnology and Advanced Materials Central Lab, Agricultural Research Center, P.O. 588 Orman, 9 Elgamaa St., Giza, 12619, Egypt.
- Regional Center for Food and Feed, Agricultural Research Center, Giza, 12619, Egypt.
| | - Mona Zidan
- Immunology research program, Basic Research Unit, Research Department, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt
| | - Taher A Salaheldin
- Tumor Biology Research Program, Basic Research Unit, Research Department, Children's Cancer Hospital Egypt 57357, Cairo, 11441, Egypt
- SUNY Schenectady Community College, New York, USA
| | - Mohamed H Gaber
- Biophysics Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Maha A Ali
- Biophysics Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - Nayera M El-Sayed
- Physics Department, Faculty of Science, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
8
|
Yan X, Dong H, Gao L, Liu M, Wang C. Mechanism of selenium-doped black phosphorus nanosheets wrapped with biomimetic tumor cell membrane for prostate cancer immunotherapy. BIOMATERIALS ADVANCES 2025; 176:214339. [PMID: 40393102 DOI: 10.1016/j.bioadv.2025.214339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 04/28/2025] [Accepted: 05/05/2025] [Indexed: 05/22/2025]
Abstract
Prostate cancer (PCa) is commonly considered a "cold tumor" due to its immunosuppressive microenvironment. Cold tumors are typically identified by the absence of T-cell infiltration within the tumor, while other immune populations and myeloid cells can be observed in these tumors. To achieve light-heat combined immunotherapy checkpoint inhibitor treatment for castration-resistant prostate cancer, we aimed to transforming "cold tumors" into "hot tumors". We designed and synthesized a two-dimensional material, selenium-doped black phosphorus (BP), to enhance the photothermal conversion efficiency, and formed Se@BPNSs by liquid-phase exfoliation. To address the issue of enhanced permeability and retention effect, and to achieve efficient targeting, we coated the Se@BPNSs with RM-1 cell membrane derived from mouse prostate cancer cells. By injecting a certain dose of Se@BPNSs into the tumor and irradiating with a 808 nm laser, the Se@BPNSs converted light energy into heat to kill tumor cells at high temperatures while releasing antigens captured by dendritic cells. In addition, we combined the immunotherapy checkpoint inhibitor anti-PD1 to enhance the immune response and promote immune cell infiltration. The successful preparation of Se@BPNSs was verified through material characterization, cell-level and animal-level experiments, and the antitumor effect was meanwhile verified, which further provided guidance for prostate cancer treatment by photothermal synergistic immunotherapy.
Collapse
Affiliation(s)
- Xingjian Yan
- Department of Urology, The First Hospital of Jilin University, Changchun City, Jilin Province 130021, China.
| | - Han Dong
- Department of Geriatric, The First Hospital of Jilin University, Changchun City, Jilin Province 130021, China
| | - Liyin Gao
- Department of Urology, The First Hospital of Jilin University, Changchun City, Jilin Province 130021, China
| | - Mengqi Liu
- Key Laboratory of Bionic Engineering, Ministry of Education, College of Biological and Agricultural Engineering, Jilin University, Changchun City, Jilin Province 130022, China
| | - Chunxi Wang
- Department of Urology, The First Hospital of Jilin University, Changchun City, Jilin Province 130021, China.
| |
Collapse
|
9
|
Sagayaraj C, Kumar KB, Vimal S, Danya U. Assessment of Phytochemical Profile, Antioxidant, and Anticancer Activity Against Colon Cancer-HT-29: A Potent Therapeutic Medicinal Plant (Tarenna alpestris) in Megamalai Hills, Western Ghats, India. Appl Biochem Biotechnol 2025; 197:3382-3406. [PMID: 39946058 DOI: 10.1007/s12010-025-05193-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2025] [Indexed: 05/11/2025]
Abstract
Medicinal plants have long been recognized as a valuable source of human health due to their therapeutic potential in treating a variety of diseases. Tarenna alpestris, a plant native to the Megamalai Hills in the Western Ghats of India, has traditionally been used for numerous medicinal purposes. However, despite its extensive use in folklore, scientific validation of its therapeutic properties remains limited. This study aims to evaluate the phytochemical composition of Tarenna alpestris, assess its antioxidant properties, and explore its potential anticancer effects against HT-29 colon cancer cells. The phytochemical profile was determined using preliminary screening and gas chromatography-mass spectrometry (GC-MS). Antioxidant activity was measured through DPPH, FRAP, H₂O₂, and N₂O₂ assays. The anticancer effects were investigated using the MTT assay for cell viability, AO/EtBr staining for apoptosis detection, DAPI staining for nuclear fragmentation analysis, and flow cytometry for cell cycle analysis. The phytochemical analysis identified several bioactive compounds, including flavonoids, alkaloids, terpenoids, and phenolic acids. Sixteen phytocomponents were detected from the extract by GCMS analysis, the major compounds are 9-octadecynoic acid (21%), N-hexadecanoic acid (16%), methylene diamine, N,N'-diacetyl (15%), 1-allyl-cyclohexane-1,2-diol (11%) 2-methyl-6-methylene-octa-1,7-dien-3-ol (5%), squalene (4%), and lupeol (3%) respectively. The antioxidant assays demonstrated significant free radical scavenging activity, with IC50 values comparable to known antioxidant standards. The antioxidant enzymatic activity of Tarenna alpestris extract suggests a potent ability to neutralize reactive oxygen species and protect against oxidative damage. In vitro studies revealed that Tarenna alpestris extract significantly inhibited the proliferation of HT-29 colon cancer cells and induced apoptosis, based on concentration dependent manner. The concentration needed to inhibit 50% of cell growth, known as the IC50 value, was found to be 26 ± 0.20 μg/mL. Additionally, cell cycle analysis showed G0/G1 phase arrest in treated cells. Tarenna alpestris exhibits a robust phytochemical profile with substantial antioxidant and anticancer properties. These findings support its potential as a therapeutic agent for cancer prevention and treatment. Further research, including in vivo studies, is warranted to fully elucidate its therapeutic efficacy and mechanisms of action.
Collapse
Affiliation(s)
- Chinnappan Sagayaraj
- Department of Botany, Nirmala College for Women, Tamil Nadu, Redfields, Coimbatore, 641018, India
| | - Kumar Bharath Kumar
- Department of Biochemistry, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Tamil Nadu, Thandalam, Chennai, 602105, India
| | - Sugumar Vimal
- Department of Biochemistry, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Tamil Nadu, Thandalam, Chennai, 602105, India
| | - Uthaman Danya
- Department of Botany, Nirmala College for Women, Tamil Nadu, Redfields, Coimbatore, 641018, India.
| |
Collapse
|
10
|
Song B, Shuang L, Zhang S, Tong C, Chen Q, Li Y, Hao M, Niu W, Jin CH. Research progress of nano drug delivery systems in the anti-tumor treatment of traditional Chinese medicine monomers. PeerJ 2025; 13:e19332. [PMID: 40292112 PMCID: PMC12034246 DOI: 10.7717/peerj.19332] [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: 12/04/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Tumors pose a serious threat to global public health and are usually treated from two aspects: tumor cells and tumor microenvironment. Compared with traditional chemotherapy drugs, traditional Chinese medicine (TCM) monomers have advantages in tumor treatment, such as multiple targets, multiple levels and synergistic intervention. However, most TCM active ingredients have disadvantages such as poor water solubility and stability, which restrict their clinical application. Nano drug delivery systems have the functions of improving the bioavailability of TCM anti-tumor active ingredients, enhancing tissue targeting, achieving controlled drug release, and inhibiting tumor multidrug resistance. Compared with free monomers, they have higher therapeutic effects and fewer side effects. This article summarizes five commonly used anti-tumor TCM monomer nanocarriers, including lipid nanomaterials, exosomes, polymer micelles, carbon nanotubes, and dendrimers, and explains their anti-tumor mechanisms after combining with TCM, such as inhibiting tumor cell proliferation and metastasis, regulating tumor microenvironment, etc. At the same time, the potential of nano drug delivery systems combined with radiotherapy and immunotherapy is discussed, as well as the current problems of potential toxicity, long-term stability, and complex amplification process, as well as future development directions, aiming to provide a reference for promoting the clinical application of nano drug delivery systems for TCM anti-tumor active ingredients.
Collapse
Affiliation(s)
- Bocui Song
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Li Shuang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Shuang Zhang
- Comprehensive Service Center, Yongji Economic Development Zone, Jilin, Jilin, China
| | - Chunyu Tong
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Qian Chen
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Yuqi Li
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Meihan Hao
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Wenqi Niu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
| | - Cheng-Hao Jin
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, China
- College of Life Science and Technology, College of Life Science and Technology, Daqing, Heilongjiang, China
| |
Collapse
|
11
|
Sharma R, Kumar S, Komal K, Ghosh R, Thakur S, Pal RR, Kumar M. Comprehensive insights into pancreatic cancer treatment approaches and cutting-edge nanocarrier solutions: from pathology to nanomedicine. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04094-y. [PMID: 40202672 DOI: 10.1007/s00210-025-04094-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025]
Abstract
Pancreatic cancer is one of the most lethal malignancies worldwide. It is characterized by poor prognosis, high mortality, and recurrence rates. Various modifiable and non-modifiable risk factors are associated with pancreatic cancer incidence. Available treatments for pancreatic cancer include surgery, chemotherapy, radiotherapy, photodynamic therapy, supportive care, targeted therapy, and immunotherapy. However, the survival rates for PC are very low. Regrettably, despite efforts to enhance prognosis, the survival rate of pancreatic cancer remains relatively low. Therefore, it is essential to investigate new approaches to improve pancreatic cancer treatment. By synthesizing current knowledge and identifying existing gaps, this article provides a comprehensive overview of risk factors, pathology, conventional treatments, targeted therapies, and recent advancements in nanocarriers for its treatment, along with various clinical trials and patents that justify the safety and efficacy of innovative carriers for drug delivery systems. Ultimately, this review underscores the potential of these innovative formulations to improve outcomes and contribute significantly to the advancement of Pancreatic Cancer treatment. Together, these insights highlight nano-formulations as a promising frontier for effectively treating Pancreatic Cancer.
Collapse
Affiliation(s)
- Rohit Sharma
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Ravi Raj Pal
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
12
|
Tu L, Xing B, Ma S, Zou Z, Wang S, Feng J, Cheng M, Jin Y. A review on polysaccharide-based tumor targeted drug nanodelivery systems. Int J Biol Macromol 2025; 304:140820. [PMID: 39933669 DOI: 10.1016/j.ijbiomac.2025.140820] [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: 10/25/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/13/2025]
Abstract
The tumor-targeted drug delivery system (TTDNS) uses nanocarriers to transport chemotherapeutic agents to target tumor cells or tissues precisely. This innovative approach considerably increases the effective concentration of these drugs at the tumor site, thereby enhancing their therapeutic efficacy. Many chemotherapeutic agents face challenges, such as low bioavailability, high cytotoxicity, and inadequate drug resistance. To address these obstacles, TTDNS comprising natural polysaccharides have gained increasing popularity in the field of nanotechnology owing to their ability to improve safety, bioavailability, and biocompatibility while reducing toxicity. In addition, it enhances permeability and allows for controlled drug delivery and release. This review focuses on the sources of natural polysaccharides and their direct and indirect mechanisms of anti-tumor activity. We also explored the preparation of various polysaccharide-based nanocarriers, including nanoparticles, nanoemulsions, nanohydrogels, nanoliposomes, nanocapsules, nanomicelles, nanocrystals, and nanofibers. Furthermore, this review delves into the versatile applications of polysaccharide-based nanocarriers, elucidating their capabilities for in vivo targeting, controlled release, and responsiveness to endogenous and exogenous stimuli, such as pH, reactive oxygen species, glutathione, light, ultrasound, and magnetic fields. This sophisticated design substantially enhances the chemotherapeutic efficacy of the encapsulated drugs at tumor sites and provides a basis for preclinical and clinical research. However, the in vivo stability, drug loading, and permeability of these preparations into tumor tissues still need to be improved. Most of the currently developed biomarker-sensitive polysaccharide nanocarriers are still in the laboratory stage, more innovative delivery mechanisms and clinical studies are needed to develop commercial nanocarriers for medical use.
Collapse
Affiliation(s)
- Liangxing Tu
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Banghuai Xing
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Shufei Ma
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Zijian Zou
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Siying Wang
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China
| | - Jianfang Feng
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China; Guangxi University of Chinese Medicine, Nanning 530200, PR China.
| | - Meng Cheng
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China.
| | - Yi Jin
- Jiangxi University of Chinese Medicine, Nanchang 330006, PR China.
| |
Collapse
|
13
|
Ovechkina VS, Andrianova SK, Shimanskaia IO, Suvorova PS, Ryabinina AY, Blagonravov ML, Belousov VV, Mozhaev AA. Advances in Optogenetics and Thermogenetics for Control of Non-Neuronal Cells and Tissues in Biomedical Research. ACS Chem Biol 2025; 20:553-572. [PMID: 40056098 DOI: 10.1021/acschembio.4c00842] [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: 03/22/2025]
Abstract
Optogenetics and chemogenetics are relatively new biomedical technologies that emerged 20 years ago and have been evolving rapidly since then. This has been made possible by the combined use of genetic engineering, optics, and electrophysiology. With the development of optogenetics and thermogenetics, the molecular tools for cellular control are continuously being optimized, studied, and modified, expanding both their applications and their biomedical uses. The most notable changes have occurred in the basic life sciences, especially in neurobiology and the activation of neurons to control behavior. Currently, these methods of activation have gone far beyond neurobiology and are being used in cardiovascular research, for potential cancer therapy, to control metabolism, etc. In this review, we provide brief information on the types of molecular tools for optogenetic and thermogenetic methods─microbial rhodopsins and proteins of the TRP superfamily─and also consider their applications in the field of activation of non-neuronal tissues and mammalian cells. We also consider the potential of these technologies and the prospects for the use of optogenetics and thermogenetics in biomedical research.
Collapse
Affiliation(s)
- Vera S Ovechkina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
| | - Sofya K Andrianova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Iana O Shimanskaia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Polina S Suvorova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| | - Anna Y Ryabinina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- V.A. Frolov Department of General Pathology and Pathological Physiology, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Mikhail L Blagonravov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- V.A. Frolov Department of General Pathology and Pathological Physiology, Institute of Medicine, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
| | - Vsevolod V Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, 117513, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow, 121205, Russia
| | - Andrey A Mozhaev
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, 117997, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia
- National Research University Higher School of Economics, Moscow, 101000, Russia
| |
Collapse
|
14
|
Ghadami A, Fathi-Karkan S, Siddiqui B, Gondal SA, Rahdar A, Garousi NA, Kharaba Z, Ghotekar S. Nanotechnology in Imatinib delivery: advancing cancer treatment through innovative nanoparticles. Med Oncol 2025; 42:116. [PMID: 40100578 DOI: 10.1007/s12032-025-02660-1] [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: 01/01/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
Nanotechnology-based drug delivery systems have improved target medicines' therapeutic efficacy and specificity in cancer therapy. Imatinib, one of the tyrosine kinase inhibitors widely used for treating chronic myeloid leukemia and gastrointestinal stromal tumors (GIST), faces many drawbacks, such as poor solubility, reduced bioavailability, and the development of resistance. The paper critically reviews advances in nanotechnology-based approaches toward the delivery of Imatinib, relating to polymeric, lipid-based, carbon-based, and stimuli-responsive nanoparticles. These methods enhance solubility, stability, and targeted distribution and are often used to facilitate the co-delivery of other anticancer drugs with considerable problems in cancer treatment. Although much potential for these technologies exists, scalability, safety, and regulatory approval, among other features, need resolution before real cost can meet clinical efficacy. Further directions would go toward bio-inspired system development, enhancing regulatory frameworks, and cost-effective manufacturing processes that bring nanotechnology into the realm of standard treatment for cancer.
Collapse
Affiliation(s)
- Azam Ghadami
- Department of Chemical and Polymer Engineering, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Sonia Fathi-Karkan
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94531-55166, Iran.
- Department of Medical Nanotechnology, School of Medicine, North Khorasan University of Medical Science, Bojnurd, Iran.
| | - Bazla Siddiqui
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-I-Azam University, Islamabad, Pakistan
| | - Sonia Ashfaq Gondal
- School of Pharmacy, University of Management and Technology, 72-A Raiwand Road, Dubai Chowk, Lahore, Pakistan
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol, Iran.
| | | | - Zelal Kharaba
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Suresh Ghotekar
- Centre for Herbal Pharmacology and Environmental Sustainability, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, 603103, India.
| |
Collapse
|
15
|
Wang J, Zhang G, Xing K, Wang B, Liu Y, Xue Y, Liu S, Leong DT. Influencing inter-cellular junctions with nanomaterials. Adv Colloid Interface Sci 2025; 336:103372. [PMID: 39671889 DOI: 10.1016/j.cis.2024.103372] [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: 10/25/2024] [Revised: 11/29/2024] [Accepted: 11/30/2024] [Indexed: 12/15/2024]
Abstract
Cell-cell junctions are essential for maintaining tissue integrity and regulating a wide range of physiological processes. While the disruption of intercellular junctions may lead to pathological conditions, it also presents an opportunity for therapeutic interventions. Nanomaterials have emerged as promising tools for modulating cell-cell junctions, offering new avenues for innovative treatments. In this review, we provide a comprehensive overview of the various nanomaterials interaction with cell-cell junctions. We discussed their underlying mechanisms, heterogenous effects on cellular behavior, and the therapeutic strategies of applying nanomaterial-induced intercellular junction disruption. Additionally, we address the challenges and opportunities involved in translating these strategies into clinical practice and discuss future directions for this rapidly advancing field.
Collapse
Affiliation(s)
- Jinping Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China; Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| | - Guoying Zhang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Kuoran Xing
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| | - Baoteng Wang
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - Yanping Liu
- School of Chemistry and Chemical Engineering, University of Jinan, Jinan 250022, China
| | - Yuling Xue
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore
| | - Shankui Liu
- School of Biological Science and Technology, University of Jinan, Jinan 250022, China
| | - David Tai Leong
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 4 Engineering Drive 4, Singapore 117585, Singapore.
| |
Collapse
|
16
|
Yu Y, Tao Y, Ma J, Li J, Song Z. Targeting the tumor microenvironment with mesenchymal stem cells based delivery approach for efficient delivery of anticancer agents: An updated review. Biochem Pharmacol 2025; 232:116725. [PMID: 39746456 DOI: 10.1016/j.bcp.2024.116725] [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: 11/14/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Drug delivery to cancer cells continues to present a major therapeutic challenge. Mesenchymal stem cells (MSCs) possess an intrinsic ability to migrate specifically to tumor tissues, making them promising candidates for targeted drug delivery. Evidence from preclinical studies indicates that MSCs loaded with therapeutic anti-cancer agents exhibit considerable anti-tumor activity. Moreover, several clinical trials are currently evaluating their effectiveness in cancer patients. The integration of MSCs with synthetic nanoparticles (NPs) enhances their therapeutic potential, particularly through the use of cell membrane-coated NPs, which represent a significant advancement in the field. This review systematically investigates the tumor microenvironment, the sources of MSCs, the tumor homing mechanisms, and the methods of loading and releasing anticancer drugs from MSCs. Furthermore, cutting-edge strategies to improve the efficacy of MSCs based drug delivery systems (DDS) including the innovative use of MSC membrane coated nanoparticles have been discussed. The study concludes with an overview of the therapeutic use of MSCs as drug carriers, including a detailed analysis of the mechanisms by which MSCs deliver therapeutics to cancer cells, enabling targeted drug delivery. It aims to elucidate the current state of this approach, identify key areas for development, and outline potential future directions for advancing MSCs based cancer therapies.
Collapse
Affiliation(s)
- Yang Yu
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun 130000, China
| | - Ying Tao
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun 130000, China
| | - Jingru Ma
- Department of Clinical Laboratory, the Second Hospital of Jilin University, Changchun 130000, China
| | - Jian Li
- Department of Emergency and Critical Care, the Second Hospital of Jilin University, Changchun 130000, China
| | - Zhidu Song
- Department of Ophthalmology, the Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
17
|
Austria E, Bilek M, Varamini P, Akhavan B. Breaking biological barriers: Engineering polymeric nanoparticles for cancer therapy. NANO TODAY 2025; 60:102552. [DOI: 10.1016/j.nantod.2024.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
18
|
Chen Z, Hu Y, Mei H. Harmonizing the symphony of chimeric antigen receptor T cell immunotherapy with the elegance of biomaterials. Trends Biotechnol 2025; 43:333-347. [PMID: 39181760 DOI: 10.1016/j.tibtech.2024.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/27/2024]
Abstract
Chimeric antigen receptor T cell (CAR-T) immunotherapy has become a heated field of cancer research, demonstrating revolutionary efficacy in refractory and relapsed hematologic malignancies. However, CAR-T therapy has still encountered tough challenges, including complicated and lengthy manufacturing procedures, mediocre targeted delivery, limited therapeutic effect against solid tumors and difficulties in real-time in vivo monitoring. To overcome these limitations, various versatile biomaterials have been used in the above aspects and have improved CAR-T therapy impressively. This review mainly summarizes the latest research progress of biomaterials promoting CAR-T therapy in manufacturing, enhancing targeted delivery and tumor infiltration, and dramatic in vivo tracking to provide new insights and inspiration for clinical treatment.
Collapse
Affiliation(s)
- Zhaozhao Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei, China; Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, Wuhan 430022, China.
| |
Collapse
|
19
|
Tang L, Yang X, He L, Zhu C, Chen Q. Preclinical advance in nanoliposome-mediated photothermal therapy in liver cancer. Lipids Health Dis 2025; 24:31. [PMID: 39891269 PMCID: PMC11783920 DOI: 10.1186/s12944-024-02429-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/31/2024] [Indexed: 02/03/2025] Open
Abstract
Liver cancer is a highly lethal malignant tumor with a high incidence worldwide. Therefore, its treatment has long been a focus of medical research. Although traditional treatment methods such as surgery, radiotherapy, and chemotherapy have increased the survival rate of patients, their efficacy remains unsatisfactory owing to the nonspecific distribution of drugs, high toxicity, and drug resistance of tumor tissues. In recent years, the application of nanotechnology in the medical field has opened a new avenue for the treatment of liver cancer. Among these treatment methods, photothermal therapy (PTT) based on nanoliposomes has attracted wide attention owing to its unique targeting and high efficiency. This article reviews the latest preclinical research progress of nanoliposome-based PTT for liver cancer and its metastasis, discusses the preclinical challenges in this field, and proposes directions for improvement, with the aim of improving the effectiveness of liver cancer treatment.
Collapse
Affiliation(s)
- Lixuan Tang
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xiao Yang
- The department of oncology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Liwen He
- School of Medicine, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Chaogeng Zhu
- The department of hepatobiliary pancreatic hernia surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China.
| | - Qingshan Chen
- The department of hepatobiliary pancreatic hernia surgery, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410208, China.
| |
Collapse
|
20
|
Muniandy MT, Chee CF, Rahman NA, Wong TW. Enhancing Aqueous Solubility and Anticancer Efficacy of Oligochitosan-Folate-Cisplatin Conjugates through Oleic Acid Grafting for Targeted Nanomedicine Development. ACS OMEGA 2025; 10:2428-2441. [PMID: 39895753 PMCID: PMC11780459 DOI: 10.1021/acsomega.4c03529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 10/23/2024] [Accepted: 10/29/2024] [Indexed: 02/04/2025]
Abstract
Oligochitosan is an anticancer water-soluble biomaterial. Conjugating cisplatin (anticancer drug) and folic acid (targeting ligand) with oligochitosan reduces its aqueous solubility, thus requiring excessive drug dose to be biologically active and organic instead of aqueous processing into nanomedicine. Covalent grafting of oleic acid onto oligochitosan-folate-cisplatin conjugate is envisaged to promote aqueous solubility via reducing interchain interaction, but it is challenging where multiple functional moieties are covalently attached onto a short oligomer (<5 kDa). This study produced oligochitosan-oleate-folate-cisplatin conjugate dissolvable in aqueous media pH 3-7, which represents common processing pH in drug vehicle development and tumor microenvironmental pHs. Oligochitosan-oleate conjugation was effected through O-acylation to provide amino groups of oligochitosan for folate and cisplatin grafting. Oligochitosan-folate-cisplatin conjugate was poorly soluble in aqueous and organic media. A degree of oleic acid substitution (DS) < 10% conferred aqueous solubility beyond which became less soluble due to hydrophobicity rise. Oligochitosan-oleate-folate-cisplatin conjugate with 4.51 ± 0.32% DS, 8.50 ± 0.57% folate content, and 0.94 ± 0.80% cisplatin content was dissolvable in aqueous media pH 3.3-7, conferring processing safety with improved cancer cytotoxicity in the nanoparticulate form at the acidic tumor microenvironment.
Collapse
Affiliation(s)
- M. Tamilarasi Muniandy
- Department
of Chemistry, Faculty of Science, Universiti
Malaya, 50603 Kuala Lumpur, Malaysia
- Non-Destructive
Biomedical and Pharmaceutical Research Centre, Smart Manufacturing
Research Institute, Universiti Teknologi
MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia
| | - Chin Fei Chee
- Nanotechnology
and Catalysis Research Centre, Universiti
Malaya, 50603 Kuala Lumpur, Malaysia
| | - Noorsaadah Abdul Rahman
- Department
of Chemistry, Faculty of Science, Universiti
Malaya, 50603 Kuala Lumpur, Malaysia
- Institute
for Advanced Studies, Universiti Malaya, 50603 Kuala Lumpur, Malaysia
| | - Tin Wui Wong
- Non-Destructive
Biomedical and Pharmaceutical Research Centre, Smart Manufacturing
Research Institute, Universiti Teknologi
MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia
- Particle
Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, 42300 Puncak Alam, Selangor, Malaysia
- Department
of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| |
Collapse
|
21
|
He M, Chen S, Yu H, Fan X, Wu H, Wang Y, Wang H, Yin X. Advances in nanoparticle-based radiotherapy for cancer treatment. iScience 2025; 28:111602. [PMID: 39834854 PMCID: PMC11743923 DOI: 10.1016/j.isci.2024.111602] [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] [Indexed: 01/22/2025] Open
Abstract
Radiotherapy has long been recognized as an effective conventional approach in both clinical and scientific research, primarily through mechanisms involving DNA destruction or the generation of reactive oxygen species to target tumors. However, significant challenges persist, including the unavoidable damage to normal tissues and the development of radiation resistance. As a result, nanotechnology-based radiotherapy has garnered considerable attention for its potential to enhance precision in irradiation, improve radiosensitization, and achieve therapeutic advancements. Importantly, radiotherapy alone frequently falls short of fully eradicating tumors. Consequently, to augment the efficacy of radiotherapy, it is often integrated with other therapeutic strategies. This review elucidates the mechanisms of radiotherapy sensitization based on diverse nanoparticles. Typically, radiotherapy is sensitized through augmenting reactive oxygen species production, targeted radiotherapy, hypoxia relief, enhancement of antitumor immune microenvironment, and G2/M cell cycle arrest. Moreover, the incorporation of nanoparticle-based anti-tumor strategies with radiotherapy markedly enhances the current state of radiotherapy. Additionally, a compilation of clinical trials utilizing nano-radioenhancers is presented. Finally, future prospects for clinical translation in this field are thoroughly examined.
Collapse
Affiliation(s)
- Meijuan He
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Shixiong Chen
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai General Hospital Branch of National Center for Translational Medicine (Shanghai), Shanghai 201620, China
| | - Hongwei Yu
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Xuhui Fan
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Hong Wu
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Yihui Wang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai General Hospital Branch of National Center for Translational Medicine (Shanghai), Shanghai 201620, China
| | - Han Wang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
- Shanghai General Hospital Branch of National Center for Translational Medicine (Shanghai), Shanghai 201620, China
- Jiading Branch of Shanghai General Hospital, Shanghai 201803, China
| | - Xiaorui Yin
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| |
Collapse
|
22
|
Luo M, Wang YM, Zhao FK, Luo Y. Recent Advances in Nanomaterial-Mediated Cell Death for Cancer Therapy. Adv Healthc Mater 2025; 14:e2402697. [PMID: 39498722 DOI: 10.1002/adhm.202402697] [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: 07/22/2024] [Revised: 10/01/2024] [Indexed: 11/07/2024]
Abstract
Nanomedicine has shown great anticancer potential by disrupting redox homeostasis and increasing the levels of oxidative stress, but the therapeutic effect is limited by factors including the intrinsic self-protection mechanism of tumors. Cancer cell death can be induced by the exploration of different cell death mechanisms, such as apoptosis, pyroptosis, necroptosis, cuproptosis, and ferroptosis. The merging of nanotechnology with biomedicine has provided tremendous opportunities to construct cell death-based nanomedicine for innovative cancer therapy. Nanocarriers are not only used for the targeted delivery of cell death inducers, but also as therapeutic components to induce cell death to achieve efficient tumor treatment. This review focuses on seven cell death modalities mediated by nanomaterials, such as apoptosis, pyroptosis, necroptosis, ferroptosis, cuprotosis, immunogenic cell death, and autophagy. The mechanisms of these seven cell death modalities are described in detail, as well as the preparation of nanomaterials that induce them and the mechanisms, they used to exert their effects. Finally, this work describes the potential future development based on the current knowledge related to cell death induced by nanomaterials.
Collapse
Affiliation(s)
- Min Luo
- Department of Clinical Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| | - Yuan-Min Wang
- Department of Clinical Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| | - Fu-Kun Zhao
- Department of Clinical Medicine, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| | - Yong Luo
- Department of Neurology, The Third Affiliated Hospital of Zunyi Medical University, The First People's Hospital of Zunyi, Zunyi, Guizhou, 563000, China
| |
Collapse
|
23
|
Gedda G, Park YJ, Pang MG. Recent development of nanotechnology-based approaches for gynecologic cancer therapy. Obstet Gynecol Sci 2025; 68:18-29. [PMID: 39591955 PMCID: PMC11788694 DOI: 10.5468/ogs.24180] [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: 07/05/2024] [Revised: 09/29/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Gynecological cancer is a life-threatening malignancy among women. Traditional therapies, including chemotherapy, often face challenges in terms of chemotherapeutic drug solubility and resistance, specificity, tumor site targeting, and toxicity to healthy tissues, leading to shortened efficacy and unfavorable patient outcomes and survival rates in patients with gynecologic malignancies. Recently, nanotechnology-based therapeutic methods such as targeted drug delivery and phototherapies have emerged as an appropriate alternative to overcome issues associated with traditional therapeutic methods. Specifically, nanomaterials and nanomaterial-based methods enhance the delivery of therapeutic/targeting agents to tumor sites and cellular uptakes and improve the tumor-suppressing effect. This review aims to provide an overview and future perspective on the potential impact of nanotechnology-based therapeutic methods for effective therapies for gynecologic cancer.
Collapse
Affiliation(s)
- Gangaraju Gedda
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Korea
- Central Research Laboratory, KS Hegde Medical Academy, NITTE (Deemed to be University), Mangaluru, India
| | - Yoo-Jin Park
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Korea
| | - Myung-Geol Pang
- Department of Animal Science & Technology and BET Research Institute, Chung-Ang University, Anseong, Korea
| |
Collapse
|
24
|
Vedarethinam V, Jeevanandam J. Role of nanotechnology in microbiome drug development. HUMAN MICROBIOME DRUG TARGETS 2025:245-263. [DOI: 10.1016/b978-0-443-15435-5.00018-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
25
|
Shofolawe-Bakare O, Toragall VB, Hulugalla K, Mayatt R, Iammarino P, Bentley JP, Smith AE, Werfel T. Glycopolymeric Nanoparticles Block Breast Cancer Growth by Inhibiting Efferocytosis in the Tumor Microenvironment. ACS APPLIED NANO MATERIALS 2024; 7:28851-28863. [PMID: 40443825 PMCID: PMC12121950 DOI: 10.1021/acsanm.4c06534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 06/02/2025]
Abstract
Conventional inhibitors of immune checkpoints such as anti-programmed death-1 and its ligand (anti-PD-1/PD-L1) and anti-cytotoxic T lymphocyte-associated protein 4 (anti-CTLA4) have revolutionized therapeutic approaches to cancer, establishing immunotherapy as the standard of care for many cancers. A significant number of cancers, however, remain refractory to the inhibition of these immune checkpoints, leading to the search for alternative immune checkpoints that are more relevant to those diseases. Tumor-associated macrophage (TAM)-mediated efferocytosis is an increasingly appreciated immune checkpoint with a profound impact on the phenotype of the tumor microenvironment (TME). TAMs perform their efferocytic function through the receptor MerTK, and MerTK activity correlates with tumor progression. To combat efferocytosis in the TME, we developed poly[[2-(diisopropylamino)ethyl methacrylate]-b-poly(methacrylamidomannose)] nanoparticles (PMAM NPs) capable of encapsulating and preferentially delivering UNC2025 (a MerTK inhibitor) to TAMs. The NPs had suitable physicochemical properties, such as a size of 130 nm and a neutral surface charge. The PMAM NPs encapsulated hydrophobic cargo and released them in a pH-dependent manner, showing suitability for cytosolic delivery. Moreover, the PMAM NPs showed 12-fold greater macrophage internalization than traditional PEGMA NPs. Macrophage internalization was shown to be dependent on the mannose receptor CD206, as the blockade of CD206 led to a significant decrease in PMAM NP internalization. Furthermore, PMAM NPs had a lower internalization than PEGMA NPs in 4T1 cancer cells that do not express CD206, further confirming macrophage selectivity. In vivo biodistribution studies showed the PMAM NPs were capable of internalization by TAMs in the TME. Lastly, UNC2025-PMAM NPs significantly reduced tumor volume compared to free UNC2025, showing greater therapeutic efficacy in a model of triple-negative breast cancer. These glycopolymer-based, efferocytosis-blocking NPs have promise both as a class of standalone cancer immunotherapy and as an adjuvant to improve response rates to checkpoint immunotherapy.
Collapse
Affiliation(s)
| | - Veeresh B. Toragall
- Department of Biomedical Engineering, University of Mississippi, University, MS, USA, 38677
| | - Kenneth Hulugalla
- Department of BioMolecular Sciences, University of Mississippi, University, MS, USA, 38677
| | - Railey Mayatt
- Department of Chemistry, Mississippi State University, Starkville, MS, USA. 39762
| | - Paige Iammarino
- Institute for Imaging and Analytical Technologies, Mississippi State University, Starkville, MS, USA, 39762
| | - John P. Bentley
- Department of Pharmacy Administration, University of Mississippi, University, MS, USA, 38677
| | - Adam E. Smith
- Department of Chemical Engineering, University of Mississippi, University, MS, USA, 38677
- Department of Biomedical Engineering, University of Mississippi, University, MS, USA, 38677
| | - Thomas Werfel
- Department of Chemical Engineering, University of Mississippi, University, MS, USA, 38677
- Department of Biomedical Engineering, University of Mississippi, University, MS, USA, 38677
- Department of BioMolecular Sciences, University of Mississippi, University, MS, USA, 38677
- Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS, USA, 39216
| |
Collapse
|
26
|
Mani Giri P, Banerjee A, Ghosal A, Salu P, Reindl K, Layek B. Mesenchymal stem cell-delivered paclitaxel nanoparticles exhibit enhanced efficacy against a syngeneic orthotopic mouse model of pancreatic cancer. Int J Pharm 2024; 666:124753. [PMID: 39321899 PMCID: PMC11760187 DOI: 10.1016/j.ijpharm.2024.124753] [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: 08/12/2024] [Revised: 09/15/2024] [Accepted: 09/22/2024] [Indexed: 09/27/2024]
Abstract
Pancreatic cancer is considered the deadliest among various solid tumors, with a five-year survival rate of 13 %. One of the major challenges in the management of advanced pancreatic cancer is the inefficient delivery of chemotherapeutics to the tumor site. Even though nanocarriers have been developed to improve tumoral delivery of chemotherapeutics, less than 1 % of the drugs reach tumors, rendering inadequate concentration for effective inhibition of tumors. As a potential alternative, mesenchymal stem cells (MSCs) can effectively deliver their cargo to tumor sites because of their resistance to chemotherapeutics and inherent tumor tropism. In this study, we used MSCs for the delivery of dibenzocyclooctyne (DBCO)-functionalized paclitaxel (PTX)-loaded poly(lactide-co-glycolide)-b-poly (ethylene glycol) (PLGA) nanoparticles. MSCs were modified to generate artificial azide groups on their surface, allowing nanoparticle loading via endocytosis and surface conjugation via click chemistry. This dual drug loading strategy significantly improves the PTX-loading capacity of azide-expressed MSCs (MSC-Az, 55.4 pg/cell) compared to unmodified MSCs (28.1 pg/cell). The in vitro studies revealed that PTX-loaded MSC-Az, nano-MSCs, exhibited cytotoxic effects against pancreatic cancer without altering their inherent phenotype, differentiation abilities, and tumor tropism. In an orthotopic pancreatic tumor model, nano-MSCs demonstrated significant inhibition of tumor growth (p < 0.05) and improved survival (p < 0.0001) compared to PTX solution, PTX nanocarriers, and Abraxane. Thus, nano-MSCs could be an effective delivery system for targeted pancreatic cancer chemotherapy and other solid tumors.
Collapse
Affiliation(s)
- Paras Mani Giri
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, United States
| | - Anurag Banerjee
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, United States
| | - Arpita Ghosal
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, United States
| | - Philip Salu
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58105, United States
| | - Katie Reindl
- Department of Biological Sciences, North Dakota State University, Fargo, ND 58105, United States
| | - Buddhadev Layek
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, United States.
| |
Collapse
|
27
|
Shabnum SS, Siranjeevi R, Raj CK, Saravanan A, Vickram AS, Chopra H, Malik T. Advancements in nanotechnology-driven photodynamic and photothermal therapies: mechanistic insights and synergistic approaches for cancer treatment. RSC Adv 2024; 14:38952-38995. [PMID: 39659608 PMCID: PMC11629304 DOI: 10.1039/d4ra07114j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 12/01/2024] [Indexed: 12/12/2024] Open
Abstract
Cancer is a disease that involves uncontrolled cell division triggered by genetic damage to the genes that control cell growth and division. Cancer starts as a localized illness, but subsequently spreads to other areas in the human body (metastasis), making it incurable. Cancer is the second most prevalent cause of mortality worldwide. Every year, almost ten million individuals get diagnosed with cancer. Although different cancer treatment options exist, such as chemotherapy, radiation, surgery and immunotherapy, their clinical efficacy is limited due to their significant side effects. New cancer treatment options, such as phototherapy, which employs light for the treatment of cancer, have sparked a growing fascination in the cancer research community. Phototherapies are classified into two types: photodynamic treatment (PDT) and photothermal therapy (PTT). PDT necessitates the use of a photosensitizing chemical and exposure to light at a certain wavelength. Photodynamic treatment (PDT) is primarily based on the creation of singlet oxygen by the stimulation of a photosensitizer, which is then used to kill tumor cells. PDT can be used to treat a variety of malignancies. On the other hand, PTT employs a photothermal molecule that activates and destroys cancer cells at the longer wavelengths of light, making it less energetic and hence less hazardous to other cells and tissues. While PTT is a better alternative to standard cancer therapy, in some irradiation circumstances, it can cause cellular necrosis, which results in pro-inflammatory reactions that can be harmful to therapeutic effectiveness. Latest research has revealed that PTT may be adjusted to produce apoptosis instead of necrosis, which is attractive since apoptosis reduces the inflammatory response.
Collapse
Affiliation(s)
- S Sameera Shabnum
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University Chennai-602105 Tamil Nadu India
| | - R Siranjeevi
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University Chennai-602105 Tamil Nadu India
| | - C Krishna Raj
- Department of Chemistry, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University Chennai-602105 Tamil Nadu India
| | - A Saravanan
- Department of Sustainable Engineering, Institute of Biotechnology, Saveetha School of Engineering, SIMATS Chennai-602105 Tamil Nadu India
| | - A S Vickram
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Saveetha University Chennai-602105 Tamil Nadu India
| | - Hitesh Chopra
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University Rajpura 140401 Punjab India
| | - Tabarak Malik
- Department of Biomedical Sciences, Institute of Health, Jimma University 378 Jimma Ethiopia
- Division of Research & Development, Lovely Professional University Phagwara 144411 India
| |
Collapse
|
28
|
Meng W, Huang L, Guo J, Xin Q, Liu J, Hu Y. Innovative Nanomedicine Delivery: Targeting Tumor Microenvironment to Defeat Drug Resistance. Pharmaceutics 2024; 16:1549. [PMID: 39771528 PMCID: PMC11728492 DOI: 10.3390/pharmaceutics16121549] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/24/2024] [Accepted: 11/30/2024] [Indexed: 01/16/2025] Open
Abstract
Nanodrug delivery systems have revolutionized tumor therapy like never before. By overcoming the complexity of the tumor microenvironment (TME) and bypassing drug resistance mechanisms, nanotechnology has shown great potential to improve drug efficacy and reduce toxic side effects. This review examines the impact of the TME on drug resistance and recent advances in nanomedicine delivery systems to overcome this challenge. Characteristics of the TME such as hypoxia, acidity, and high interstitial pressure significantly reduce the effectiveness of chemotherapy and radiotherapy, leading to increased drug resistance in tumor cells. Then, this review summarizes innovative nanocarrier designs for these microenvironmental features, including hypoxia-sensitive nanoparticles, pH-responsive carriers, and multifunctional nanosystems that enable targeted drug release and improved drug penetration and accumulation in tumors. By combining nanotechnology with therapeutic strategies, this review offers a novel perspective by focusing on the innovative design of nanocarriers that interact with the TME, a dimension often overlooked in similar reviews. We highlight the dual role of these nanocarriers in therapeutic delivery and TME modulation, emphasize their potential to overcome drug resistance, and look at future research directions.
Collapse
Affiliation(s)
- Wenjun Meng
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China (J.L.)
| | - Li Huang
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China (J.L.)
| | - Jiamin Guo
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qing Xin
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiyan Liu
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China (J.L.)
| | - Yuzhu Hu
- Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
29
|
Dutta B, Barick KC, Hassan PA, Tyagi AK. Recent progress and current status of surface engineered magnetic nanostructures in cancer theranostics. Adv Colloid Interface Sci 2024; 334:103320. [PMID: 39515063 DOI: 10.1016/j.cis.2024.103320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/25/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Cancer theranostic is the combination of diagnosis and therapeutic modalities for cancer treatment. It realizes a more flexible, precise and non-invasive treatment of patients. In this aspect, magnetic nanostructures (MNSs) have gained paramount importance and revolutionized the cancer management due to their unique physicochemical properties and inherent magnetic characteristics. MNSs have amazing theranostic ability starting from drug delivery to magnetic hyperthermia and magnetic resonance imaging to multimodal imaging in association with radioisotopes or fluorescent probes. Precise regulation over the synthetic process and their consequent surface functionalization makes them even more fascinating. The ultimate goal is to develop a platform that combines multiple diagnostic and therapeutic functionalities based on MNSs. This perspective has provided an overview of the state-of-art of theranostic applications of MNSs. Special emphasis has been dedicated towards the importance of synthetic approaches of MNSs as well as their subsequent surface engineering and integration with biological/therapeutic molecules that decide the final outcomes of the efficacy of MNSs in theranostic applications. Moreover, the recent advancements, opportunities and allied challenges towards clinical applications of MNSs in cancer management have been demonstrated.
Collapse
Affiliation(s)
- Bijaideep Dutta
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - K C Barick
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India.
| | - P A Hassan
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - A K Tyagi
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India; Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| |
Collapse
|
30
|
Qiao JX, Guo DY, Tian H, Wang ZP, Fan QQ, Tian Y, Sun J, Zhang XF, Zou JB, Cheng JX, Luan F, Zhai BT. Research progress of paclitaxel nanodrug delivery system in the treatment of triple-negative breast cancer. Mater Today Bio 2024; 29:101358. [PMID: 39677523 PMCID: PMC11638641 DOI: 10.1016/j.mtbio.2024.101358] [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: 07/16/2024] [Revised: 10/27/2024] [Accepted: 11/21/2024] [Indexed: 12/17/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer, characterized by the loss or low expression of estrogen receptor (ER), human epidermal growth factor receptor 2 (HER2) and progesterone receptor (PR). Due to the lack of clear therapeutic targets, paclitaxel (PTX) is often used as a first-line standard chemotherapy drug for the treatment of high-risk and locally advanced TNBC. PTX is a diterpenoid alkaloid extracted and purified from Taxus plants, functioning as an anticancer agent by inducing and promoting tubulin polymerization, inhibiting spindle formation in cancer cells, and preventing mitosis. However, its clinical application is limited by low solubility and high toxicity. Nanodrug delivery system (NDDS) is one of the feasible methods to improve the water solubility of PTX and reduce side effects. In this review, we summarize the latest advancements in PTX-targeted NDDS, as well as its combination with other codelivery therapies for TNBC treatment. NDDS includes passive targeting, active targeting, stimuli-responsive, codelivery, and multimode strategies. These systems have good prospects in improving the bioavailability of PTX, enhancing tumor targeting, reducing toxicity, controlling drug release, and reverse tumor multidrug resistance (MDR). This review provides valuable insights into the clinical development and application of PTX-targeted NDDS in the treatment of TNBC.
Collapse
Affiliation(s)
- Jia-xin Qiao
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Dong-yan Guo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Huan Tian
- Department of Pharmacy, National Old Pharmacist Inheritance Studio, Xi'an Hospital of Traditional Chinese Medicine, Xi'an, 710021, China
| | - Zhan-peng Wang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Qiang-qiang Fan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Yuan Tian
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jing Sun
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Xiao-fei Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jun-bo Zou
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Jiang-xue Cheng
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Fei Luan
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| | - Bing-tao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Province Key Laboratory of New Drugs and Chinese Medicine Foundation Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, China
| |
Collapse
|
31
|
Xie B, Liu Y, Li X, Yang P, He W. Solubilization techniques used for poorly water-soluble drugs. Acta Pharm Sin B 2024; 14:4683-4716. [PMID: 39664427 PMCID: PMC11628819 DOI: 10.1016/j.apsb.2024.08.027] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/28/2024] [Accepted: 08/14/2024] [Indexed: 12/13/2024] Open
Abstract
About 40% of approved drugs and nearly 90% of drug candidates are poorly water-soluble drugs. Low solubility reduces the drugability. Effectively improving the solubility and bioavailability of poorly water-soluble drugs is a critical issue that needs to be urgently addressed in drug development and application. This review briefly introduces the conventional solubilization techniques such as solubilizers, hydrotropes, cosolvents, prodrugs, salt modification, micronization, cyclodextrin inclusion, solid dispersions, and details the crystallization strategies, ionic liquids, and polymer-based, lipid-based, and inorganic-based carriers in improving solubility and bioavailability. Some of the most commonly used approved carrier materials for solubilization techniques are presented. Several approved poorly water-soluble drugs using solubilization techniques are summarized. Furthermore, this review summarizes the solubilization mechanism of each solubilization technique, reviews the latest research advances and challenges, and evaluates the potential for clinical translation. This review could guide the selection of a solubilization approach, dosage form, and administration route for poorly water-soluble drugs. Moreover, we discuss several promising solubilization techniques attracting increasing attention worldwide.
Collapse
Affiliation(s)
- Bing Xie
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Yaping Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| |
Collapse
|
32
|
Shah DD, Chorawala MR, Mansuri MKA, Parekh PS, Singh S, Prajapati BG. Biogenic metallic nanoparticles: from green synthesis to clinical translation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:8603-8631. [PMID: 38935128 DOI: 10.1007/s00210-024-03236-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024]
Abstract
Biogenic metallic nanoparticles (NPs) have garnered significant attention in recent years due to their unique properties and various applications in different fields. NPs, including gold, silver, zinc oxide, copper, titanium, and magnesium oxide NPs, have attracted considerable interest. Green synthesis approaches, utilizing natural products, offer advantages such as sustainability and environmental friendliness. The theranostics applications of these NPs hold immense significance in the fields of medicine and diagnostics. The review explores intricate cellular uptake pathways, internalization dynamics, reactive oxygen species generation, and ensuing inflammatory responses, shedding light on the intricate mechanisms governing their behaviour at a molecular level. Intriguingly, biogenic metallic NPs exhibit a wide array of applications in medicine, including but not limited to anti-inflammatory, anticancer, anti-diabetic, anti-plasmodial, antiviral properties and radical scavenging efficacy. Their potential in personalized medicine stands out, with a focus on tailoring treatments to individual patients based on these NPs' unique attributes and targeted delivery capabilities. The article culminates in emphasizing the role of biogenic metallic NPs in shaping the landscape of personalized medicine. Harnessing their unique properties for tailored therapeutics, diagnostics and targeted interventions, these NPs pave the way for a paradigm shift in healthcare, promising enhanced efficacy and reduced adverse effects.
Collapse
Affiliation(s)
- Disha D Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
| | - Mohammad Kaif A Mansuri
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad, Gujarat, 380009, India
| | - Priyajeet S Parekh
- AV Pharma LLC, 1545 University Blvd N Ste A, Jacksonville, FL, 32211, USA
| | - Sudarshan Singh
- Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Bhupendra G Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Mehsana, Gujarat, 384012, India.
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, 73000, Thailand.
| |
Collapse
|
33
|
Ding L, Chang C, Liang M, Dong K, Li F. Plant‐Derived Extracellular Vesicles as Potential Emerging Tools for Cancer Therapeutics. ADVANCED THERAPEUTICS 2024; 7. [DOI: 10.1002/adtp.202400256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Indexed: 01/03/2025]
Abstract
AbstractExtracellular vesicles (EVs) are membranous structures secreted by cells that play important roles in intercellular communication and material transport. Due to its excellent biocompatibility, lipophilicity, and homing properties, EVs have been used as a new generation of drug delivery systems for the diagnosis and treatment of tumors. Despite the potential clinical benefits of animal‐derived extracellular vesicles (AEVs), their large‐scale production remains sluggish due to the exorbitant cost of cell culture, challenging quality control measures, and limited production capabilities. This constraint significantly hinders their widespread clinical application. Plant‐derived extracellular vesicles (PEVs) share similar functionalities with AEVs, yet they hold several advantages including a wide variety of source materials, cost‐effectiveness, ease of preparation, enhanced safety, more stable physicochemical properties, and notable efficacy. These merits position PEVs as promising contenders with broad potential in the biomedical sector. This review will elucidate the advantages of PEVs, delineating their therapeutic mechanisms in cancer treatment, and explore the prospective applications of engineered PEVs as targeted delivery nano‐system for drugs, microRNAs, small interfering RNAs, and beyond. The aim is to heighten researchers’ focus on PEVs and expedite the progression from fundamental research to the transformation of groundbreaking discoveries.
Collapse
Affiliation(s)
- Lin Ding
- The First Affiliated Hospital (Shenzhen People's Hospital),Southern University of Science and Technology,The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital) Shenzhen 518055 China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell Therapy Shenzhen 518020 China
- Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation Shenzhen 518020 China
- Shenzhen Immune Cell Therapy Public Service Platform Shenzhen 518020 China
| | - Chih‐Jung Chang
- School of Medicine and Medical Research Center Xiamen Chang Gung Hospital Hua Qiao University Xiamen Fujian 362017 China
- Department of Dermatology Drug Hypersensitivity Clinical and Research Center Chang Gung Memorial Hospital Linkou Taoyuan 244330 Taiwan
| | - Min‐Li Liang
- The First Affiliated Hospital (Shenzhen People's Hospital),Southern University of Science and Technology,The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital) Shenzhen 518055 China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell Therapy Shenzhen 518020 China
- Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation Shenzhen 518020 China
- Shenzhen Immune Cell Therapy Public Service Platform Shenzhen 518020 China
| | - Kang‐Mei Dong
- Xiamen Lifeint Technology Co., Ltd. Fujian 361000 China
| | - Fu‐Rong Li
- The First Affiliated Hospital (Shenzhen People's Hospital),Southern University of Science and Technology,The Second Clinical Medical College, Jinan University (Shenzhen People's Hospital) Shenzhen 518055 China
- Guangdong Engineering Technology Research Center of Stem Cell and Cell Therapy Shenzhen 518020 China
- Shenzhen Key Laboratory of Stem Cell Research and Clinical Transformation Shenzhen 518020 China
- Shenzhen Immune Cell Therapy Public Service Platform Shenzhen 518020 China
| |
Collapse
|
34
|
Gupta S, Dutta B, Shelar SB, Gangwar A, Bhattacharyya K, Bairwa KK, Hassan PA, Barick KC. Polyphosphate-Mediated Crystallographic and Colloidal Stabilization of CuS Nanoparticles: Enhanced NIR-Responsive Chemo-Photothermal Efficacy. ACS APPLIED BIO MATERIALS 2024; 7:6641-6655. [PMID: 39257063 DOI: 10.1021/acsabm.4c00838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Photothermal therapy (PTT) is an emerging treatment modality for cancer management. However, the photothermal agents (PTAs) used in PTT should have sufficient biocompatibility, water dispersibility, and good photoresponsive. In this aspect, water-dispersible and biocompatible linear polyphosphate (LP)-functionalized CuS nanoparticles (LP-CuS NPs) were developed using sodium tripolyphosphate (LP molecule) as a surface passivating agent. The successful formation of the green covellite CuS phase was confirmed by X-ray diffraction and TEM analyses, and its surface functionalization with the LP ligand was evident from X-ray photoelectron spectroscopy, Fourier transform infrared, thermogravimetric analysis, and light scattering measurements. It has been found that the use of LP not only stabilizes the crystallographic covellite CuS phase by overcoming the requirement of a soft ligand but also provides long-term aqueous colloidal stability, which is essential for PTT applications. The aqueous suspension of LP-CuS NPs showed excellent heating efficacy under near infrared (NIR) light irradiation (980 nm) and has a strong binding affinity towards anticancer drug, doxorubicin hydrochloride (DOX). The drug-loaded systems (DOX@LP-CuS NPs) revealed a pH-dependent drug release behavior with higher concentrations in a mild acidic environment. The in vitro studies showed substantial cellular uptake of DOX-loaded systems in cancer cell lines and enhanced toxicity towards them upon irradiation of NIR light through apoptotic induction, suggesting their potential application in chemo-photothermal therapy.
Collapse
Affiliation(s)
- Sonali Gupta
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - Bijaideep Dutta
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - S B Shelar
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - Asnit Gangwar
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - K Bhattacharyya
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - K K Bairwa
- Radiation and Photochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
| | - P A Hassan
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| | - K C Barick
- Chemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai 400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai 400094, India
| |
Collapse
|
35
|
Nan J, Cao HX, Park JO, Choi E, Kang B. Tunable Acoustic Tweezer System for Precise Three-Dimensional Particle Manipulation. MICROMACHINES 2024; 15:1240. [PMID: 39459114 PMCID: PMC11509511 DOI: 10.3390/mi15101240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024]
Abstract
This study introduces a tunable acoustic tweezer system designed for precise three-dimensional particle trapping and manipulation. The system utilizes a dual-liquid-layer acoustic lens, which enables the dynamic control of the focal length through the adjustable curvature of a latex membrane. This tunability is essential for generating the acoustic forces necessary for effective manipulation of particles, particularly along the direction of acoustic wave propagation (z-axis). Experiments conducted with spherical particles as small as 1.5 mm in diameter demonstrated the system's capability for stable trapping and manipulation. Performance was rigorously evaluated through both z-axis and 3D manipulation tests. In the z-axis experiments, the system achieved a manipulation range of 33.4-53.4 mm, with a root-mean-square error and standard deviation of 0.044 ± 0.045 mm, which highlights its precision. Further, the 3D manipulation experiments showed that particles could be accurately guided along complex paths, including multilayer rectangular and helical trajectories, with minimal deviation. A visual feedback-based particle navigation system significantly enhanced positional accuracy, reducing errors relative to open-loop control. These results confirm that the tunable acoustic tweezer system is a robust tool for applications requiring precise control of particles with diameter of 1.5 mm in three-dimensional environments. Considering its ability to dynamically adjust the focal point and maintain stable trapping, this system is well suited for tasks demanding high precision, such as targeted particle delivery and other applications involving advanced material manipulation.
Collapse
Affiliation(s)
- Jiyun Nan
- School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea;
- Korea Institute of Medical Microrobotics, Gwangju 61000, Republic of Korea; (H.X.C.); (J.-O.P.)
| | - Hiep Xuan Cao
- Korea Institute of Medical Microrobotics, Gwangju 61000, Republic of Korea; (H.X.C.); (J.-O.P.)
| | - Jong-Oh Park
- Korea Institute of Medical Microrobotics, Gwangju 61000, Republic of Korea; (H.X.C.); (J.-O.P.)
| | - Eunpyo Choi
- School of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea;
- Korea Institute of Medical Microrobotics, Gwangju 61000, Republic of Korea; (H.X.C.); (J.-O.P.)
| | - Byungjeon Kang
- Korea Institute of Medical Microrobotics, Gwangju 61000, Republic of Korea; (H.X.C.); (J.-O.P.)
- College of AI Convergence, Chonnam National University, Gwangju 61186, Republic of Korea
- Graduate School of Data Science, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
36
|
Li Y, Gao X, Li Y, Yan S, Zhang Y, Zheng X, Gu Q. Endocytosis: the match point of nanoparticle-based cancer therapy. J Mater Chem B 2024; 12:9435-9458. [PMID: 39192831 DOI: 10.1039/d4tb01227e] [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: 08/29/2024]
Abstract
Nanomedicine has inspired a ground-breaking strategy for cancer therapy. By intelligently assembling diverse moieties to form nanoparticles, numerous functionalities such as controlled release, synergistic efficiency, and in situ killing can be achieved. The emerging nanoparticles have been designed with elevated targeting efficiency as targeting cancer cells is the primary requirement for nanoparticles. However, effective targeting does not guarantee therapeutic effects as endocytosis is a prerequisite for nanoparticles to exert effects. The recent decade has witnessed the rapid development of endocytosis-oriented nanoparticles, and this review subtly analyzes, categorizes, and exemplifies these nanoparticles according to their biological internalization patterns, and the correlation between the endocytosis mechanism and the property of nanoparticles is bridged. Based on the interdisciplinary vision, the present challenges and future perspectives of nanoparticle design for successful endocytosis are discussed, highlighting the potential strategies for the future development of endocytosis-oriented nanoparticles, thus facilitating the endocytosis-oriented strategy from bench to bedside. The undeniable fact is that endocytosis-oriented nanoparticles will definitely bring new blood to the next generation of advanced cancer therapies.
Collapse
Affiliation(s)
- Yonglu Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China.
| | - Xin Gao
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China.
| | - Yapeng Li
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China.
| | - Shihai Yan
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China.
| | - Yiru Zhang
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-food Processing, Fuli Institute of Food Science, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Xiaodong Zheng
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-food Processing, Fuli Institute of Food Science, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang University, Hangzhou 310058, People's Republic of China
| | - Qing Gu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, Zhejiang, People's Republic of China.
| |
Collapse
|
37
|
Soroushmanesh M, Dinari M, Farrokhpour H. Comprehensive Computational Investigation of the Porphyrin-Based COF as a Nanocarrier for Delivering Anti-Cancer Drugs: A Combined MD Simulation and DFT Calculation. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:19073-19085. [PMID: 39189806 DOI: 10.1021/acs.langmuir.4c02154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
As nanomaterials have gained prominence in drug delivery technology, exploring their feasibility through computational methods is beneficial before practical tests. In this study, we aim to evaluate the capability of the porphyrin-based covalent organic framework COF-366 as a nanocarrier for two anticancer drugs, irinotecan (IRI) and doxorubicin (DOX). The optimal binding conformation of the drug molecules on the COF surface was predicted by using molecular docking. Subsequently, molecular dynamic simulation (MD) was performed to assess the adsorption mechanism of drug molecules on the COF in the aqueous environment. The free energy of adsorption for DOX and IRI was estimated to be -20.07 and -23.89 kcal/mol, respectively. The adsorption of both drugs on the COF surface is mainly influenced by the π-π interaction. Furthermore, density functional theory (DFT) calculation, natural bond orbital (NBO), and quantum theory of atoms in molecules (QTAIM) analyses were employed to investigate the structural stability of Drug@COF complexes and gain a detailed understanding of the interaction between them at the molecular level. Based on DFT results, it was found that in addition to π-π interaction, the bis-piperidine-phenylene interaction affects the adsorption of IRI on the COF surface. Moreover, the diffusion behavior of the drug molecule inside the COF pore was simulated using a ten-layer COF. Based on the mean square displacement analysis, the diffusion coefficients of DOX and IRI within the COF pore were calculated to be 108 and 97 um2/s, respectively. This computational study sheds light on how different types of interactions between the drug molecule and COF affect the adsorption and diffusion process. Our findings validated that the porphyrin-based COF-366 can serve as a nanobased platform for delivering DOX and IRI.
Collapse
Affiliation(s)
- Mohsen Soroushmanesh
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Islamic Republic of Iran
| | - Mohammad Dinari
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Islamic Republic of Iran
| | - Hossein Farrokhpour
- Department of Chemistry, Isfahan University of Technology, Isfahan 84156-83111, Islamic Republic of Iran
| |
Collapse
|
38
|
Zhang Y, Li Y, Gu J, Wu J, Ma Y, Lu G, Barboiu M, Chen J. Glycopolymeric Micellar Nanoparticles for Platelet-Mediated Tumor-Targeted Delivery of Docetaxel for Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:44528-44537. [PMID: 39155662 DOI: 10.1021/acsami.4c09548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
The high level of accumulation of therapeutic agents in tumors is crucial for cancer treatment. Compared to the passive tumor-targeting effect, active tumor-targeting delivery systems, primarily mediated by peptides with high production costs and reduced circulation time, are highly desired. Platelet-driven technologies have opened new avenues for targeted drug delivery prevalently through a membrane coating strategy that involves intricate manufacturing procedures or the fucoidan-mediated hitchhiking method with limited platelet affinity. Here, a novel type of amphiphilic glycopolymer self-assembled micellar nanoparticle has been developed to adhere to naturally activated platelets in the blood. The simultaneous integration of fucose and sialic acid segments into glycopolymers enables closer mimicry of the structure of P-selectin glycoprotein ligand-1 (PSGL-1), thereby increasing the affinity for activated platelets. It results in the formation of glycopolymeric micelle-platelet hybrids, facilitating targeted drug delivery to tumors. The selective platelet-assisted cellular uptake of docetaxel (DTX)-loaded glycopolymeric micelles leads to lower IC50 values against 4T1 cells than that of free DTX. The directed tumor-targeting effect of activated platelets has significantly improved the tumor accumulation capacity of the glycopolymeric nanoparticles, with up to 21.0% found in tumors within the initial 0.2 h. Additionally, with acid-responsive drug release and inherent antimetastasis properties, the glycopolymeric nanoparticles ensured potent therapeutic efficacy, prolonged survival time, and reduced cardiotoxicity, presenting a new and unexplored strategy for platelet-directed drug delivery to tumors, showing promising prospects in treating localized tumors and preventing tumor metastasis.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Yi Li
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Jieyu Gu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Jun Wu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Yongxin Ma
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Guodong Lu
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| | - Mihail Barboiu
- Institut Europeen des Membranes, Adaptive Supramolecular Nanosystems Group, University of Montpellier, ENSCM-CNRS, UMR5635, Place E. Bataillon CC047, 34095 Montpellier, France
| | - Jinghua Chen
- Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, P. R. China
| |
Collapse
|
39
|
Harisa GI, Faris TM, Sherif AY, Alzhrani RF, Alanazi SA, Kohaf NA, Alanazi FK. Coding Therapeutic Nucleic Acids from Recombinant Proteins to Next-Generation Vaccines: Current Uses, Limitations, and Future Horizons. Mol Biotechnol 2024; 66:1853-1871. [PMID: 37578574 DOI: 10.1007/s12033-023-00821-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/04/2023] [Indexed: 08/15/2023]
Abstract
This study aims to highlight the potential use of cTNAs in therapeutic applications. The COVID-19 pandemic has led to significant use of coding therapeutic nucleic acids (cTNAs) in terms of DNA and mRNA in the development of vaccines. The use of cTNAs resulted in a paradigm shift in the therapeutic field. However, the injection of DNA or mRNA into the human body transforms cells into biological factories to produce the necessary proteins. Despite the success of cTNAs in the production of corona vaccines, they have several limitations such as instability, inability to cross biomembranes, immunogenicity, and the possibility of integration into the human genome. The chemical modification and utilization of smart drug delivery cargoes resolve cTNAs therapeutic problems. The success of cTNAs in corona vaccine production provides perspective for the eradication of influenza viruses, Zika virus, HIV, respiratory syncytial virus, Ebola virus, malaria, and future pandemics by quick vaccine design. Moreover, the progress cTNAs technology is promising for the development of therapy for genetic disease, cancer therapy, and currently incurable diseases.
Collapse
Affiliation(s)
- Gamaleldin I Harisa
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia.
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 11451, Saudi Arabia.
- Department of Biochemistry and Molecular Biology, College of Pharmacy, Al-Azhar University, Nasr City, Cairo, Egypt.
| | - Tarek M Faris
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Abdelrahman Y Sherif
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 11451, Saudi Arabia
| | - Riyad F Alzhrani
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 11451, Saudi Arabia
- Nanobiotechnology Research Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh A Alanazi
- Pharmaceutical Care Services, King Abdulaziz Medical City, Riyadh, Saudi Arabia
- King Saud Bin Abdulaziz University for Health Science Collage of Pharmacy, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Neveen A Kohaf
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Azhar University, Cairo, 11651, Egypt
| | - Fars K Alanazi
- Kayyali Chair for Pharmaceutical Industry, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box: 2457, Riyadh, 11451, Saudi Arabia
| |
Collapse
|
40
|
Wu X, Xin Y, Zhang H, Quan L, Ao Q. Biopolymer-Based Nanomedicine for Cancer Therapy: Opportunities and Challenges. Int J Nanomedicine 2024; 19:7415-7471. [PMID: 39071502 PMCID: PMC11278852 DOI: 10.2147/ijn.s460047] [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: 02/15/2024] [Accepted: 05/18/2024] [Indexed: 07/30/2024] Open
Abstract
Cancer, as the foremost challenge among human diseases, has plagued medical professionals for many years. While there have been numerous treatment approaches in clinical practice, they often cause additional harm to patients. The emergence of nanotechnology has brought new directions for cancer treatment, which can deliver anticancer drugs specifically to tumor areas. This article first introduces the application scenarios of nanotherapies and treatment strategies of nanomedicine. Then, the noteworthy characteristics exhibited by biopolymer materials were described, which make biopolymers stand out in polymeric nanomedicine delivery. Next, we focus on summarizing the state-of-art studies of five categories of proteins (Albumin, Gelatin, Silk fibroin, Zein, Ferritin), nine varieties of polysaccharides (Chitosan, Starch, Hyaluronic acid, Dextran, cellulose, Fucoidan, Carrageenan, Lignin, Pectin) and liposomes in the field of anticancer drug delivery. Finally, we also provide a summary of the advantages and limitations of these biopolymers, discuss the prevailing impediments to their application, and discuss in detail the prospective research directions. This review not only helps readers understand the current development status of nano anticancer drug delivery systems based on biopolymers, but also is helpful for readers to understand the properties of various biopolymers and find suitable solutions in this field through comparative reading.
Collapse
Affiliation(s)
- Xixi Wu
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Yuan Xin
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Hengtong Zhang
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Liang Quan
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| | - Qiang Ao
- NMPA Key Laboratory for Quality Research and Control of Tissue Regenerative Biomaterial, & Institute of Regulatory Science for Medical Device, & National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064, People’s Republic of China
| |
Collapse
|
41
|
Zhang D, Zhai B, Sun J, Cheng J, Zhang X, Guo D. Advances on Delivery System of Active Ingredients of Dried Toad Skin and Toad Venom. Int J Nanomedicine 2024; 19:7273-7305. [PMID: 39050871 PMCID: PMC11268768 DOI: 10.2147/ijn.s469742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/29/2024] [Indexed: 07/27/2024] Open
Abstract
Dried toad skin (TS) and toad venom (TV) are the dried skin of the Bufo bufo gargarizans Cantor and the Bufo melanostictus Schneider, which remove the internal organs and the white secretions of the skin and retroauricular glands. Since 2005, cinobufacini preparations have been approved by the State Food and Drug Administration for use as adjuvant therapies in the treatment of various advanced cancers. Meanwhile, bufalenolides has been identified as the main component of TS/TV, exhibiting antitumor activity, inducing apoptosis of cancer cells and inhibiting cancer cell proliferation or metastasis through a variety of signaling pathways. However, clinical agents frequently face limitations such as inherent toxicity at high concentrations and insufficient tumor targeting. Additionally, the development and utilization of these active ingredients are hindered by poor water solubility, low bioavailability, and rapid clearance from the bloodstream. To address these challenges, the design of a targeted drug delivery system (TDDS) aims to enhance drug bioavailability, improve targeting within the body, increase drug efficacy, and reduce adverse reactions. This article reviews the TDDS for TS/TV, and their active components, including passive, active, and stimuli-responsive TDDS, to provide a reference for advancing their clinical development and use.
Collapse
Affiliation(s)
- Dan Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Bingtao Zhai
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Jing Sun
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Jiangxue Cheng
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Xiaofei Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| | - Dongyan Guo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi University of Chinese Medicine, Xi’an, 712046, People’s Republic of China
| |
Collapse
|
42
|
Yanikoglu R, Karakas CY, Ciftci F, Insel MA, Karavelioglu Z, Varol R, Yilmaz A, Cakir R, Uvet H, Ustundag CB. Development of Graphene Oxide-Based Anticancer Drug Combination Functionalized with Folic Acid as Nanocarrier for Targeted Delivery of Methotrexate. Pharmaceutics 2024; 16:837. [PMID: 38931957 PMCID: PMC11207743 DOI: 10.3390/pharmaceutics16060837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Graphene has become a prominent material in cancer research in recent years. Graphene and its derivatives also attract attention as carriers in drug delivery systems. In this study, we designed a graphene oxide (GO)-based methotrexate (MTX)-loaded and folic acid (FA)-linked drug delivery system. MTX and FA were bound to GO synthesized from graphite. MTX/FA/GO drug delivery system and system components were characterized using Fourier transform infrared spectroscopy (FTIR), differential calorimetric analysis (DSC), scanning electron microscopy (SEM), transmission electron microscopy (TEM), zeta potential analysis, and dimension measurement (DLS) studies. SEM and TEM images confirmed the nanosheet structure of GO synthesized from graphite, and it was shown that MTX/FA binding to GO transformed the two-dimensional GO into a three-dimensional structure. FTIR and DSC graphs confirmed that oxygen atoms were bound to GO with the formation of carboxylic, hydroxyl, epoxide, and carbonyl groups as a result of the oxidation of graphite, and GO was successfully synthesized. Additionally, these analyses showed that MTX and FA bind physicochemically to the structure of GO. The in vitro Franz diffusion test was performed as a release kinetic test. The release kinetics mathematical model and correlation coefficient (R2) of MTX-loaded GO/FA nanomaterials were found to be the Higuchi model and 0.9785, respectively. Stiffness analyses showed that adding FA to this release system facilitated the entry of the drug into the cell by directing the system to target cells. As a result of the stiffness analyses, the stiffness values of the control cell group, free MTX, and MTX/FA/GO applied cells were measured as 2.34 kPa, 1.87 kPa, and 1.56 kPa, respectively. According to these results, it was seen that MTX/FA/GO weakened the cancer cells. Combined use of the MTX/FA/GO drug delivery system had a higher cytotoxic effect than free MTX on the MDA-MB-231 breast cancer cell line. The results showed that the synthesized MTX/FA/GO material has promising potential in cancer cell-specific targeted therapy for MTX as a drug delivery system.
Collapse
Affiliation(s)
- Reyhan Yanikoglu
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, İstanbul 34210, Türkiye
| | - Canan Yagmur Karakas
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, İstanbul 34210, Türkiye
| | - Fatih Ciftci
- Department of Biomedical Engineering, Fatih Sultan Mehmet Vakif University, Istanbul 34445, Türkiye
| | - Mert Akın Insel
- Department of Chemical Engineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, İstanbul 34210, Türkiye
| | - Zeynep Karavelioglu
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, İstanbul 34210, Türkiye
- Department of Health Sciences and Technology, ETH Zurich, 8092 Zurich, Switzerland
| | - Rahmetullah Varol
- Department of Mechatronics Engineering, Yildiz Technical University, Istanbul 34349, Türkiye
- Business Administration Department, Bundeswehr University Munich, 85579 Munich, Germany
| | - Abdurrahim Yilmaz
- Department of Mechatronics Engineering, Yildiz Technical University, Istanbul 34349, Türkiye
| | - Rabia Cakir
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, İstanbul 34210, Türkiye
- Türkiye Biotechnology Institute, Health Institutes of Türkiye, Istanbul 34718, Türkiye
| | - Hüseyin Uvet
- Department of Mechatronics Engineering, Yildiz Technical University, Istanbul 34349, Türkiye
| | - Cem Bulent Ustundag
- Department of Bioengineering, Faculty of Chemical and Metallurgical Engineering, Yıldız Technical University, İstanbul 34210, Türkiye
- Health Biotechnology Joint Research and Application Center of Excellence, Istanbul 34220, Türkiye
| |
Collapse
|
43
|
Amin H, Ibrahim IM, Hassanein EHM. Weaponizing chitosan and its derivatives in the battle against lung cancer. Int J Biol Macromol 2024; 272:132888. [PMID: 38844273 DOI: 10.1016/j.ijbiomac.2024.132888] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 05/28/2024] [Accepted: 06/02/2024] [Indexed: 06/11/2024]
Abstract
Lung cancer (LC) is a crisis of catastrophic proportions. It is a global problem and urgently requires a solution. The classic chemo drugs are lagging behind as they lack selectivity, where their side effects are spilled all over the body, and these adverse effects would be terribly tragic for LC patients. Therefore, they could make a bad situation worse, inflict damage on normal cells, and inflict pain on patients. Since our confidence in classic drugs is eroding, chitosan can offer a major leap forward in LC therapy. It can provide the backbone and the vehicle that enable chemo drugs to penetrate the hard shell of LC. It could be functionalized in a variety of ways to deliver a deadly payload of toxins to kill the bad guys. It is implemented in formulation of polymeric NPs, lipidic NPs, nanocomposites, multiwalled carbon nanotubes, and phototherapeutic agents. This review is a pretty clear proof of chitosan's utility as a weapon in battling LC. Chitosan-based formulations could work effectively to kill LC cells. If a researcher is looking for a vehicle for medication for LC therapy, chitosan can be an appropriate choice.
Collapse
Affiliation(s)
- Haitham Amin
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt.
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt.
| |
Collapse
|
44
|
Darguzyte M, Rama E, Rix A, Baier J, Hermann J, Rezvantalab S, Khedri M, Jankowski J, Kiessling F. Riboflavin-targeted polymers improve tolerance of paclitaxel while maintaining therapeutic efficacy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2024; 58:102751. [PMID: 38705222 DOI: 10.1016/j.nano.2024.102751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 03/15/2024] [Accepted: 04/26/2024] [Indexed: 05/07/2024]
Abstract
Active targeting can enhance precision and efficacy of drug delivery systems (DDS) against cancers. Riboflavin (RF) is a promising ligand for active targeting due to its biocompatibility and high riboflavin-receptor expression in cancers. In this study, RF-targeted 4-arm polyethylene glycol (PEG) stars conjugated with Paclitaxel (PTX), named PEG PTX RF, were evaluated as a targeted DDS. In vitro, PEG PTX RF exhibited higher toxicity against tumor cells compared to the non-targeted counterpart (PEG PTX), while free PTX displayed the highest acute toxicity. In vivo, all treatments were similarly effective, but PEG PTX RF-treated tumors showed fewer proliferating cells, pointing to sustained therapy effects. Moreover, PTX-treated animals' body and liver weights were significantly reduced, whereas both remained stable in PEG PTX and PEG PTX RF-treated animals. Overall, our targeted and non-targeted DDS reduced PTX's adverse effects, with RF targeting promoted drug uptake in cancer cells for sustained therapeutic effect.
Collapse
Affiliation(s)
- Milita Darguzyte
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf, Center for Molecular Medicine Cologne, 50931 Cologne, Germany; Institute for Translational Immune-Oncology, Cancer Research Center Cologne-Essen (CCCE), University of Cologne, 50931 Cologne, Germany
| | - Elena Rama
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Anne Rix
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Jasmin Baier
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany
| | - Juliane Hermann
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Sima Rezvantalab
- Department of Chemical Engineering, Urmia University of Technology, 57166-419, Urmia, Iran
| | - Mohammad Khedri
- Computational Biology and Chemistry Group (CBCG) Universal Scientific Education and Research Network (USERN), 19839-63113 Tehran, Iran
| | - Joachim Jankowski
- Institute of Molecular Cardiovascular Research, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, University Hospital Aachen, Forckenbeckstrasse 55, 52074 Aachen, Germany; Fraunhofer MEVIS, Institute for Medical Image Computing, Forckenbeckstrasse 55, 52074 Aachen, Germany.
| |
Collapse
|
45
|
Long Q, Zhao X, Gao L, Liu M, Pan F, Gao X, Zhan C, Chen Y, Wang J, Qian J. Effects of Surface IR783 Density on the In Vivo Behavior and Imaging Performance of Liposomes. Pharmaceutics 2024; 16:744. [PMID: 38931866 PMCID: PMC11206891 DOI: 10.3390/pharmaceutics16060744] [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: 04/20/2024] [Revised: 05/16/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Background: Nanoparticles conjugated with fluorescent probes have versatile applications, serving not only for targeted fluorescent imaging but also for evaluating the in vivo profiles of designed nanoparticles. However, the relationship between fluorophore density and nanoparticle behavior remains unexplored. Methods: The IR783-modified liposomes (IR783-sLip) were prepared through a modified ethanol injection and extrusion method. The cellular uptake efficiency of IR783-sLip was characterized by flow cytometry and fluorescence microscope imaging. The effects of IR783 density on liposomal in vivo behavior were investigated by pharmacokinetic studies, biodistribution studies, and in vivo imaging. The constitution of protein corona was analyzed by the Western blot assay. Results: Dense IR783 modification improved cellular uptake of liposomes in vitro but hindered their blood retention and tumor imaging performance in vivo. We found a correlation between IR783 density and protein corona absorption, particularly IgM, which significantly impacted the liposome performance. Meanwhile, we observed that increasing IR783 density did not consistently improve the effectiveness of tumor imaging. Conclusions: Increasing the density of modified IR783 on liposomes is not always beneficial for tumor near-infrared (NIR) imaging yield. It is not advisable to prematurely evaluate novel nanomaterials through fluorescence dye conjugation without carefully optimizing the density of the modifications.
Collapse
Affiliation(s)
- Qianqian Long
- School of Pharmacy, Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; (Q.L.); (X.Z.); (M.L.); (F.P.)
| | - Xinmin Zhao
- School of Pharmacy, Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; (Q.L.); (X.Z.); (M.L.); (F.P.)
| | - Lili Gao
- Department of Pathology, Pudong New Area People’s Hospital, Shanghai 201299, China;
| | - Mengyuan Liu
- School of Pharmacy, Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; (Q.L.); (X.Z.); (M.L.); (F.P.)
| | - Feng Pan
- School of Pharmacy, Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; (Q.L.); (X.Z.); (M.L.); (F.P.)
| | - Xihui Gao
- School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; (X.G.); (C.Z.)
| | - Changyou Zhan
- School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; (X.G.); (C.Z.)
| | - Yang Chen
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China;
| | - Jialei Wang
- School of Pharmacy, Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; (Q.L.); (X.Z.); (M.L.); (F.P.)
| | - Jun Qian
- School of Pharmacy, Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai 200032, China; (Q.L.); (X.Z.); (M.L.); (F.P.)
| |
Collapse
|
46
|
Bokhari SS, Ali T, Naeem M, Hussain F, Nasir A. Recent advances in nanoformulation-based delivery for cancer immunotherapy. Nanomedicine (Lond) 2024; 19:1253-1269. [PMID: 38717427 PMCID: PMC11285355 DOI: 10.1080/17435889.2024.2343273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 07/25/2024] Open
Abstract
Cancer is one of the leading causes of mortality worldwide, and its treatment faces several challenges. Phytoconstituents derived from recently discovered medicinal plants through nanotechnology potentially target cancer cells via PI3K/Akt/mTOR pathways and exert their effects selectively through the generation of reactive oxygen species through β-catenin inhibition, DNA damage, and increasing caspase 3/9 and p53 expression. These nanocarriers act specifically against different cancer cell lines such as HT-29, MOLT-4 human leukemia cancer and MCF-7 cell lines SKOV-3, Caov-3, SW-626, HepG2, A-549, HeLa, and MCF-7. This review comprehensively elaborates on the cellular and molecular mechanisms, and therapeutic prospects of various plant-mediated nanoformulations to attain a revolutionary shift in cancer immunotherapy.
Collapse
Affiliation(s)
- Seyedeh Saimeh Bokhari
- Clinico-Molecular Biochemistry Laboratory, Department of Biochemistry, University of Agriculture, 38000, Faisalabad, Pakistan
| | - Tayyab Ali
- Clinico-Molecular Biochemistry Laboratory, Department of Biochemistry, University of Agriculture, 38000, Faisalabad, Pakistan
| | - Muhammad Naeem
- College of Life Science, Hebei Normal University, Shijiazhuang, 050024, China
| | - Fatma Hussain
- Clinico-Molecular Biochemistry Laboratory, Department of Biochemistry, University of Agriculture, 38000, Faisalabad, Pakistan
| | - Abdul Nasir
- Medical Research Center, Second Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, Henan, China
| |
Collapse
|
47
|
Nguyen KT, Kee H, Go G, Kim S, Choi E, Park J, Park S, Kim J. Field‐Free Region Scanning‐Based Magnetic Microcarrier Targeting in Multibifurcation Vessels. ADVANCED INTELLIGENT SYSTEMS 2024; 6. [DOI: 10.1002/aisy.202300700] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Indexed: 01/05/2025]
Abstract
Navigation of microcarriers in complex environments as a vascular network remains an open challenge due to limited solutions for effective targeting strategy. Simultaneous real‐time visualization and manipulation of microcarriers at any depth in the human body is far to be achieved even though one of each task has been successfully proven. Herein, a novel targeting strategy is proposed that employs field‐free region (FFR) scanning to guide microcarriers through multiple bifurcations within a predefined vessel network. The main challenge of this method lies on how, where, and when to activate FFR to steer a particle to a desired direction, regardless of its spatial feedback. To achieve it, first, a mathematical model of particle motion in a vessel network is developed to predict particle behaviors and positions. Subsequently, an optimization algorithm is formulated to place FFR well‐coordinated around each bifurcation at a designated moment. The established solution for targeting a magnetic microcarrier is preemptively evaluated through finite element simulations and then successfully implemented in in vitro multibranched vessels.
Collapse
Affiliation(s)
- Kim Tien Nguyen
- Korea Institute of Medical Microrobotics Gwangju 61011 Korea
| | - Hyeonwoo Kee
- Department of Robotics and Mechatronics Engineering Daegu Gyeongbuk Institute of Science & Technology (DGIST) Daegu 42988 Korea
| | - Gwangjun Go
- Department of Mechanical Engineering Chosun University Gwangju 61452 Korea
| | - Seok‐Jae Kim
- Korea Institute of Medical Microrobotics Gwangju 61011 Korea
| | - Eunpyo Choi
- School of Mechanical Engineering Chonnam National University Gwangju 61186 Korea
| | - Jong‐Oh Park
- Korea Institute of Medical Microrobotics Gwangju 61011 Korea
| | - Sukho Park
- Department of Robotics and Mechatronics Engineering Daegu Gyeongbuk Institute of Science & Technology (DGIST) Daegu 42988 Korea
| | - Jayoung Kim
- Korea Institute of Medical Microrobotics Gwangju 61011 Korea
- Department of Biosystems Engineering Chungbuk National University Cheongju 28644 Korea
| |
Collapse
|
48
|
Jaber SA. The antioxidant and anticancer activity of Quercus coccifera plant leaves extracts. Saudi J Biol Sci 2024; 31:103979. [PMID: 38510526 PMCID: PMC10951596 DOI: 10.1016/j.sjbs.2024.103979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
Quercus species are one of the medicinal plants that commonly used in the treatment of different diseases. Quercus coccifera (Q. coccifera) is part of the Quercus species which grow in Jordan and used in traditional folklore medicine. The aim of this study is to confirm the ability of (Q. coccifera) leaves extracts to exert anticancer activity. In this study, an extraction method of the dried-leaves using different polarity solvents was used. Extracts were pre-evaluated for antioxidant and anticancer activities while active extracts were used to measure half maximal effective concentration (EC50) against 2,2-Diphenyl-1-picrylhydrazyl (DPPH), and Half-maximal inhibitory concentration (IC50) against cancer cells. Methanol, boiled and microwaved water extracts had greater than 80 % antioxidant activity, and the strongest activity, of more than 99 %, was boiled water extract. Similarly, the pre-evaluation treatments of cancer cell lines indicated a strong biological activity of more than 70 % from the previously mentioned extracts, and the highest activity, of greater than 90 %, was from boiled water extracts against all cancer cell lines. The highest EC50 against DPPH was obtained by using 0.009 mg/ml boiled water extracts, which was lower than positive control quercetin. In the same manner, lung, breast, and prostate cancer cell lines were highly affected by boiled water extracts with IC50 of 14.1, 7.2, and 25.1 µg/ml, respectively, and a selectivity index (SI) of greater than 4.71. Q. coccifera leaves extracts show promising ability to be a source of a new anticancer therapeutics.
Collapse
Affiliation(s)
- Saif Aldeen Jaber
- Faculty of Pharmacy, Middle East University, Amman, Jordan
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
| |
Collapse
|
49
|
Akbarzadeh F, Khoshgard K. Enhancement of the effect of novel targeted 5-aminolevulinic acid conjugated bismuth oxide nanoparticles-based photodynamic therapy by simultaneous radiotherapy on KB cells. Photodiagnosis Photodyn Ther 2024; 46:104025. [PMID: 38403143 DOI: 10.1016/j.pdpdt.2024.104025] [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: 08/25/2023] [Revised: 01/30/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND Selective accumulation of photosensitizers into cancerous cells is one of the most important factors affecting photodynamic therapy (PDT) efficacy. 5-aminolevulinic acid (5-ALA) is the precursor of a strong photosensitizer, protoporphyrin-IX; but it has poor permeability into the cells. Folate receptors are overexpressed on the surface of many tumor cells. In the present study, folic acid (FA) and 5-ALA conjugated bismuth oxide nanoparticles were synthesized; and used in PDT, radiotherapy (RT), and concurrent PDT & RT against nasopharyngeal carcinoma (KB cell line). METHODS The KB cells were incubated with the synthesized nanoparticles (NPs) for 2 h; then illuminated using a custom-made LED lamp at the light dose of 26 J/cm2. Irradiation of the cells was carried out using X-ray 6 MV (2 Gy); and synergistic effect of the simultaneous RT and PDT treatments was evaluated using fractional product values. Efficacy of the treatments was determined using MTT and Caspase-3 enzyme activity assays. RESULTS Targeting of folic acid receptors enables the selective endocytosis of the conjugated NPs. RT results in the presence of Bi2O3 NPs showed a significant radiosensitizer potential of these NPs. Fractional product values of 1.49±0.05, 1.36±0.06, and 1.05±0.06 obtained in the presence of FA-5-ALA conjugated NPs, 5-ALA conjugated NPs, and in the absence of the NPs, respectively. Therefore, simultaneous RT and PDT in the presence of these conjugated NPs is superior to RT in the presence of the NPs. CONCLUSION Simultaneous PDT and RT in the presence of FA-5-ALA conjugated bismuth oxide NPs can be introduced as a promising therapeutic approach in controlling KB cancer cells.
Collapse
Affiliation(s)
- Fatemeh Akbarzadeh
- Students Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Karim Khoshgard
- Department of Medical Physics, School of Medicine, Kermanshah University of Medical Sciences, Sorkheh-Lizhe Blvd, Kermanshah, P.O.Box:1568, Iran.
| |
Collapse
|
50
|
López-Estévez AM, Lapuhs P, Pineiro-Alonso L, Alonso MJ. Personalized Cancer Nanomedicine: Overcoming Biological Barriers for Intracellular Delivery of Biopharmaceuticals. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309355. [PMID: 38104275 DOI: 10.1002/adma.202309355] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/09/2023] [Indexed: 12/19/2023]
Abstract
The success of personalized medicine in oncology relies on using highly effective and precise therapeutic modalities such as small interfering RNA (siRNA) and monoclonal antibodies (mAbs). Unfortunately, the clinical exploitation of these biological drugs has encountered obstacles in overcoming intricate biological barriers. Drug delivery technologies represent a plausible strategy to overcome such barriers, ultimately facilitating the access to intracellular domains. Here, an overview of the current landscape on how nanotechnology has dealt with protein corona phenomena as a first and determinant biological barrier is presented. This continues with the analysis of strategies facilitating access to the tumor, along with conceivable methods for enhanced tumor penetration. As a final step, the cellular barriers that nanocarriers must confront in order for their biological cargo to reach their target are deeply analyzed. This review concludes with a critical analysis and future perspectives of the translational advances in personalized oncological nanomedicine.
Collapse
Affiliation(s)
- Ana María López-Estévez
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacology, Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Philipp Lapuhs
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacology, Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Laura Pineiro-Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacology, Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - María José Alonso
- Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), Department of Pharmacology, Pharmaceutics and Pharmaceutical Technology, School of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| |
Collapse
|