1
|
Gao Q, Wang W, Sun S, Yang Y, Mao K, Yang Y, Wu ZS. Bundling gold nanorods with RCA-produced DNA tape into an intelligently reconfigurable nanocluster bomb for multimodal precision cancer therapy. Mater Today Bio 2025; 32:101718. [PMID: 40236812 PMCID: PMC11999372 DOI: 10.1016/j.mtbio.2025.101718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/10/2025] [Accepted: 03/28/2025] [Indexed: 04/17/2025] Open
Abstract
Via proposing an innovative assembly technique, we bundle cell-targeting aptamer-modified gold nanorods (AuNRs) with RCA product (RCA-p) tape into a reconfigurable nanocluster (ARGN) bomb for multimodal precision cancer therapy. Because each ARGN has 10 individual AuNRs, the short time of laser irradiation can make the temperature increase to 75 °C much higher than the lethal temperature of tumor cells, enabling the efficient photothermal therapy (PTT). Moreover, both siRNA-Plk1 (2820 per ARGN) and chemotherapeutic agents (15860 per ARGN) can be loaded into two specifically-designed containers in the internal cavity. Because the glomeroplasmatic structure enhances the resistance to enzymatic degradation, ARGN bomb can protect siRNAs from the digestion and avoid Dox leakage during in vivo circulation. Moreover, the spontaneous structural reorganization allows aptamers in the interior cavity move outward to the exterior surface, which magically offers the compensation of degraded aptamers and impair persistent in vivo cell targeting ability. The external stimuli (laser irradiation) promotes the release of chemotherapeutic agents and initiates the PTT/chemotherapy outcome, while endogenous stimuli (intracellular biomarkers) causes almost 100 % release of siRNA-Plk1 species and induces RNA interference therapy, completely inhibiting tumor growth without detectable off-target toxicity.
Collapse
Affiliation(s)
- Qian Gao
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 305108, China
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Weijun Wang
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 305108, China
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
- College of Chemistry and Food Science, Nanchang Normal University, Nanchang, 330032, China
| | - Shujuan Sun
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 305108, China
| | - Ya Yang
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 305108, China
| | - Kaili Mao
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuxi Yang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Zai-Sheng Wu
- Cancer Metastasis Alert and Prevention Center, Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 305108, China
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| |
Collapse
|
2
|
Zhao F, Gong L, Wang P, Chen D, Cao S, Yang F, Tang M, Meng Y, Wang Y, Miao L, Li Y, Huang W. Co-encapsulation of norcantharidin prodrugs and lomitapide in nanoparticles to regulate CCL4 expression by inhibiting Wnt/β-catenin pathway for improved anti-tumor immunotherapy. J Nanobiotechnology 2025; 23:369. [PMID: 40394658 PMCID: PMC12093795 DOI: 10.1186/s12951-025-03425-8] [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: 11/13/2024] [Accepted: 05/01/2025] [Indexed: 05/22/2025] Open
Abstract
In the absence of tumor antigen specificity, direct chemokine administration carries the risk of significant "on-target, off-tumor" toxicities, highlighting the need for small-molecule approaches with reduced immunogenicity. This study investigates the synergistic potential of norcantharidin (NCTD) and lomitapide (lomi) in selectively restoring CCL4 expression by deactivating the tumor intrinsic β-catenin pathway. Due to its similar lipophilicity to lomi and potential to suppress β-catenin, NCTD prodrug (C12) was selected to be co-encapsulated with lomi in a nanoparticle-mediated co-delivery system (NP"C12 + lomi"). The NP"C12 + lomi" formulation exhibited a high encapsulation rate, uniform particle size, and suitability for therapeutic use. It effectively inhibited the proliferation of 4T1 cells and restored CCL4 expression. In both primary breast tumor and surgically resected tumor mouse models, NP"C12 + lomi" significantly increased the proportion of CD8+ cells in primary tumors, blood, and lung metastases, approximately doubling their presence. This led to a prolongation of median survival in mice to 59 days. Furthermore, when combined with an immune checkpoint inhibitor, NP"C12 + lomi" substantially inhibited tumor growth and lung metastasis without affecting body weight or causing major tissue or organ damage. This was attributed to the controlled dissociation of the nanoparticle and the subsequent modulation of C12 and lomi, which mitigated CCL4-related toxicity. This study provides valuable insights into the safe production of chemokines using a small-molecule pair through a nanosystem and presents a robust chemo-immunological cascade therapy strategy, demonstrating significant efficacy against malignant metastatic tumors.
Collapse
Affiliation(s)
- Feng Zhao
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Liming Gong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Ping Wang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Dong Chen
- Suzhou Kintor Pharmaceuticals, Inc., Suzhou, 215127, China
| | - Shijie Cao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Feifei Yang
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicines, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, PR China
| | - Manqing Tang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuanyuan Meng
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuming Wang
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Lin Miao
- State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Yunfei Li
- College of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
- Shandong Key Laboratory of Carbohydrate Chemistry and Glycobiology, Shandong University, Jinan, China.
| | - Wei Huang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
3
|
Wu J, Bao Q, Wang X, Chen H, Chen X, Wen Y, Chen J. Research progress of co-delivery nanoparticle drug delivery systems in non-small cell lung cancer: A review. Colloids Surf B Biointerfaces 2025; 254:114795. [PMID: 40403441 DOI: 10.1016/j.colsurfb.2025.114795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 03/25/2025] [Accepted: 05/10/2025] [Indexed: 05/24/2025]
Abstract
Non-small cell lung cancer (NSCLC), as the most commonly diagnosed type of lung cancer, has long been a major focus for cancer drug researchers. Traditional chemotherapy has shown significant efficacy in patients initially diagnosed with NSCLC; however, with the emergence of drug resistance and notable toxic side effects, conventional and single-agent chemotherapy can no longer meet the treatment needs of patients. Nanomedicine systems have gained widespread attention among scholars due to their unique advantages, such as particle size, stable in vivo circulation, and multifunctional carrier materials. However, most single-drug delivery systems fail to meet the treatment expectations for NSCLC patients, prompting the active development of co-delivery nanomedicine systems in preclinical NSCLC research. These systems can utilize surface-modified carriers to co-deliver drugs, genes, photosensitizers, or sonosensitizers with different mechanisms of action. This approach not only achieves the synergistic effects of multiple drugs, multiple pathways, and the combination of chemotherapy with photodynamic/sonodynamic therapy but also, through the encapsulation of inorganic materials, allows for more controllable drug release under external forces such as magnetic fields. This further amplifies the synergistic effects between the drugs, and the results of these studies are significantly superior to those of single-drug treatments. In conclusion, this review summarizes the delivery strategies and the extended use of inorganic materials in the co-delivery of nanoparticles for NSCLC research in recent years, with the hope of providing reference for researchers' drug design strategies.
Collapse
Affiliation(s)
- Jiali Wu
- Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Qiaohong Bao
- Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Xinyu Wang
- Shanghai Wei Er Lab, Shanghai 201707, China.
| | - Hang Chen
- Shanghai Wei Er Lab, Shanghai 201707, China.
| | - Xinmei Chen
- Shanghai Wei Er Lab, Shanghai 201707, China.
| | - Yan Wen
- Department of Pharmacy, Changzheng Hospital, Naval Medical University, No.415, Fengyang Road, Shanghai 200003, China.
| | - Jianming Chen
- Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| |
Collapse
|
4
|
Deng Q, Yue S, You F, Zhai Z, Sun H, Liang L, Li C, Yang L, Zhong Z. Vincristine/Volasertib Polymersome Injection Enables High-Efficiency Synergistic Treatment of Acute Lymphoblastic Leukemia. Acta Biomater 2025:S1742-7061(25)00366-6. [PMID: 40383350 DOI: 10.1016/j.actbio.2025.05.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 05/12/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
Acute lymphoblastic leukemia (ALL), one of the most frequently diagnosed malignancies in children, is associated with a high relapse rate and drug resistance, even with intensive multidrug chemotherapy regimens. The rational combination with molecular targeted agents holds promise for sensitizing patients to chemotherapies and overcoming drug resistance. However, precise codelivery of different drugs in vivo is challenging, often leading to suboptimal therapeutic effects. Herein, we report a vincristine/volasertib polymersome (Ps-VCR/Vol)-based nanocombo for synergistic inhibition of microtubules and polo-like kinase 1, enabling high-efficacy treatment of ALL in vivo. Ps-VCR/Vol, which has a small size (∼26 nm) and tailored VCR/Vol mass ratios from 1:12 to 1:48, exhibited strong synergy in different ALL cells, with 3.3-6.8-fold greater anti-ALL activity than the free VCR/Vol combination. Intriguingly, treatment with Ps-VCR/Vol at a VCR/Vol dosage of 0.25/6 mg/kg markedly inhibited leukemia progression and invasion in orthotopic CCRF-CEM, Nalm-6-Luc and patient-derived xenograft ALL mouse models without inducing toxicity, resulting in a significantly prolonged survival time compared with that of the free drug combination and single-drug polymersome formulations. Ps-VCR/Vol polymersome injection provides a powerful synergistic combination therapy for ALL. STATEMENT OF SIGNIFICANCE: Multidrug combination therapies have increased the remission rates of acute lymphoblastic leukemia (ALL) patients. However, the therapeutic efficacy remains suboptimal due to the dissimilar physicochemical properties of the different drugs involved, and overlapping toxicities pose a critical concern. Herein, we show that intelligent polymersomes mediate the precise codelivery of vincristine sulfate (VCR), a frontline drug for ALL, and volasertib (Vol), a polo-like kinase 1 inhibitor, enabling synergistic treatment of ALL. Compared with free VCR/Vol, VCR/Vol polymersomes with tailored drug ratios substantially inhibited leukemia progression in both cell line- and patient-derived orthotopic ALL models without inducing toxicity, leading to a significant survival benefit. This synergistic polymersome injection may provide a powerful and safe combination therapy for ALL patients.
Collapse
Affiliation(s)
- Qing Deng
- State Key Laboratory of Bioinspired Interfacial Materials Science, and Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Shujing Yue
- State Key Laboratory of Bioinspired Interfacial Materials Science, and Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Fengtao You
- PersonGen BioTherapeutics (Suzhou) Co., Ltd., Suzhou, China
| | - Zhenzhen Zhai
- State Key Laboratory of Bioinspired Interfacial Materials Science, and Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Huanli Sun
- State Key Laboratory of Bioinspired Interfacial Materials Science, and Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China..
| | - Lanlan Liang
- State Key Laboratory of Bioinspired Interfacial Materials Science, and Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Chenming Li
- State Key Laboratory of Bioinspired Interfacial Materials Science, and Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Lin Yang
- PersonGen BioTherapeutics (Suzhou) Co., Ltd., Suzhou, China.; Cyrus Tang Medical Institute, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, China..
| | - Zhiyuan Zhong
- State Key Laboratory of Bioinspired Interfacial Materials Science, and Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China.; College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.; International College of Pharmaceutical Innovation, Soochow University, Suzhou, 215222, China.
| |
Collapse
|
5
|
Benderski K, Lammers T, Sofias AM. Analysis of multi-drug cancer nanomedicine. NATURE NANOTECHNOLOGY 2025:10.1038/s41565-025-01932-1. [PMID: 40374796 DOI: 10.1038/s41565-025-01932-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 04/04/2025] [Indexed: 05/18/2025]
Abstract
Multi-drug nanomedicine is gaining momentum for co-delivering more than one drug to the same site at the same time. Our analysis of 273 pre-clinical tumour growth inhibition studies shows that multi-drug nanotherapy outperforms single-drug therapy, multi-drug combination therapy, and single-drug nanotherapy by 43, 29 and 30%, respectively. Combination nanotherapy also results in the best overall survival rates, with 56% of studies demonstrating complete or partial survival, versus 20-37% for control regimens. Within the multi-drug nanomedicine groups, we analysed the effect of (co-)administration schedule and strategy, passive versus active targeting, nanocarrier material and the type of therapeutic agent. Most importantly, it was found that co-encapsulating two different drugs in the same nanoformulation reduces tumour growth by a further 19% compared with the combination of two individually encapsulated nanomedicines. We finally show that the benefit of multi-drug nanotherapy is consistently observed across different cancer types, in sensitive and resistant tumours, and in xenograft and allograft models. Altogether, this meta-analysis substantiates the value of multi-drug nanomedicine as a potent strategy to improve cancer therapy.
Collapse
Affiliation(s)
- Karina Benderski
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany.
- Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Aachen, Germany.
| | - Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Aachen, Germany.
- Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
6
|
Bhattacharya E, Shaw S, Nayak R, Bose S. Advances in targeted therapy for inflammatory breast cancer: nanomaterials, conventional treatments, and clinical applications. NANOTECHNOLOGY 2025; 36:222002. [PMID: 40294602 DOI: 10.1088/1361-6528/add165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/28/2025] [Indexed: 04/30/2025]
Abstract
Inflammatory breast cancer (IBC) presents a formidable challenge due to its rapid progression and unique clinical characteristics within the various manifestations of breast cancer. Despite being rare, its aggressive nature demands innovative approaches beyond conventional treatments. Nanomedicine offers exciting possibilities for improving all types of breast cancer therapeutics including IBC. In this review, we critically assess the current treatment landscape for IBC, highlighting the limitations of traditional methods and addressing the pressing need for new therapeutic strategies. Although many nanomaterials have been explored for breast cancer therapeutics, either alone or in combination with other therapies, only a limited number of nanotherapeutics have been extensively studied for IBC treatment. This review further explores how advancements in nanotechnology, such as nanoparticle- mediated photothermal therapy, Photodynamic therapy, and nanomedicinal targeted therapies can offer novel avenues for addressing the unique biological, technological, and regulatory challenges posed by IBC. IBC-related various nanomedicines based combinatorial therapies are highlighted in this review. It also provides a forward-looking perspective on key research directions and clinical applications.
Collapse
Affiliation(s)
- Eshana Bhattacharya
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Siuli Shaw
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Ranu Nayak
- Amity Institute of Nanotechnology, Amity University, Noida, Uttar Pradesh, India
| | - Sudeep Bose
- Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, Uttar Pradesh, India
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh, India
| |
Collapse
|
7
|
Li M, Wu W, Gu M, Su C, Wang X, Pan D, Xu Y, Wang L, Chen C, Yang M, Yan J. Purification-Free Bortezomib-Drug Conjugates Optimize Drug Economy and Cancer Therapeutic Synergy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:23691-23706. [PMID: 40203453 DOI: 10.1021/acsami.5c02160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
As a promising candidate in overcoming resistance, providing synergy, and developing treatments, conjugated combination drugs mostly prevail over drug cocktails in establishing prodrugs and precisely codelivering multiple drugs for combination chemotherapies. However, current drug-drug conjugation methods (e.g., esterification, amidation, etherification, etc.) do not allow quantitative drug conversion and require necessary purification of crude products, resulting in a limited economy of initial drugs. Meanwhile, practical stimulus concentration in vivo usually fails to efficiently activate parent drug release from drug conjugates in target sites, which diminishes their efficacy. Herein, we report a click conjugation strategy based on boronic acid-cis diol complexation, realizing a fast (<30 min), quantitative, and purification-free conjugation of bortezomib (BTZ) and azacytidine (AZA) or capecitabine or doxifluridine. Notably, the BTZ-AZA conjugate spontaneously self-assembles into nanomedicine and exhibits enhanced synergistic efficacy. Furthermore, BTZ and AZA could be conjugated into a polyprodrug with controlled size and composition, and different organelle uptakes augment the synergy of BTZ-AZA conjugate by approximately 1000-fold versus free BTZ toward A549 adenocarcinoma cells (IC50: 0.55 nM versus 536.7 nM). This click strategy would expand the vision for developing smart combination drugs.
Collapse
Affiliation(s)
- Meng Li
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Wei Wu
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Min Gu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Chen Su
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Xinyu Wang
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Donghui Pan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Yuping Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Lizhen Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Chongyang Chen
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Min Yang
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Junjie Yan
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| |
Collapse
|
8
|
Asghar S, Iliescu R, Stiufiuc RI, Dragoi B. Co-Encapsulation of Multiple Antineoplastic Agents in Liposomes by Exploring Microfluidics. Int J Mol Sci 2025; 26:3820. [PMID: 40332493 PMCID: PMC12027889 DOI: 10.3390/ijms26083820] [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: 02/27/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
The inherent complexity of cancer proliferation and malignancy cannot be addressed by the conventional approach of relying on high doses of a single powerful anticancer agent, which is associated with poor efficacy, higher toxicity, and the development of drug resistance. Multiple drug therapy (MDT) rationally designed to target tumor heterogeneity, block alternative survival pathways, modulate the tumor microenvironment, and reduce toxicities would be a viable solution against cancer. Liposomes are the most suitable carrier for anticancer MDT due to their ability to encapsulate both hydrophilic and hydrophobic agents, biocompatibility, and controlled release properties; however, an adequate manufacturing method is important for effective co-encapsulation. Microfluidics involves the manipulation of fluids at the microscale for the controlled synthesis of liposomes with desirable properties. This work critically reviews the use of microfluidics for the synthesis of anticancer MDT liposomes. MDT success not only relies on the identification of synergistic dose combinations of the anticancer modalities but also warrants the loading of multiple therapeutic entities within liposomes in optimal ratios, the protection of the drugs by the nanocarrier during systemic circulation, and the synchronous release at the target site in the same pattern as confirmed in preliminary efficacy studies. Prospects have been identified for the bench-to-bedside translation of anticancer MDT liposomes using microfluidics.
Collapse
Affiliation(s)
- Sajid Asghar
- Nanotechnology Laboratory, TRANSCEND Department, Regional Institute of Oncology, 2-4 General Henri Mathias Berthelot, 700483 Iași, Romania;
- Department of Pharmaceutics, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Radu Iliescu
- Proteomics Laboratory, TRANSCEND Research Center, Regional Institute of Oncology, 2-4 General Henri Mathias Berthelot Street, 700483 Iași, Romania
- Department of Pharmacology, Faculty of Medicine, Grigore T. Popa University of Medicine and Pharmacy, 16 University Street, 700115 Iași, Romania
| | - Rares-Ionut Stiufiuc
- Nanotechnology Laboratory, TRANSCEND Department, Regional Institute of Oncology, 2-4 General Henri Mathias Berthelot, 700483 Iași, Romania;
- Department of NanoSciences, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hatieganu” University of Medicine and Pharmacy, 4-6 Pasteur Street, 400349 Cluj-Napoca, Romania
| | - Brindusa Dragoi
- Nanotechnology Laboratory, TRANSCEND Department, Regional Institute of Oncology, 2-4 General Henri Mathias Berthelot, 700483 Iași, Romania;
- Faculty of Chemistry, Alexandru Ioan Cuza University of Iași, 11 Bd. Carol I, 700506 Iași, Romania
| |
Collapse
|
9
|
Rui R, Li Y, Liu Y, Li X, Zhou G, Zhao C, Han Y. Nanoassemblies of the redox paclitaxel prodrug with the natural active ingredient dihydroartemisinin for therapy of breast cancer. NANOSCALE 2025; 17:8069-8083. [PMID: 40035630 DOI: 10.1039/d4nr05418k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Natural products have attracted attention owing to their multiple antitumor effects, improved chemotherapy sensitivity, and few side effects. The combination of natural active ingredients and chemotherapy drugs could be an effective strategy for synergistic antitumor therapy by preserving their activity to inhibit the growth of tumors, while reducing the side effects of chemotherapy drugs at relatively low doses. Although the feasibility of the delivery of natural products and chemotherapy drugs has been proven, most current carriers cannot be efficiently loaded, thus leading to a discrepancy in the drug release ratio compared to the predefined loading ratio. In this study, simple nanoassemblies with controllable drug release profiles were constructed to co-deliver paclitaxel (PTX) and dihydroartemisinin (DHA) for synergistic treatment of breast cancer. The nanoassemblies demonstrated a notable capacity for loading efficiency, micro-environmental triggering of drug release, and activation of the homodimeric prodrug at the tumor site, thereby facilitating successful combination therapy. The in vitro and in vivo antitumor effects were synergistically improved by combining DHA and PTX through prodrug modifications and nanoassemblies. Our findings provide a simple and efficient strategy for the development of nanoassemblies combining natural active ingredients with chemotherapeutic drugs.
Collapse
Affiliation(s)
- Rucheng Rui
- Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yi Li
- Beijing Institute of Pharmacology and Toxicology, Beijing 100850, PR China
| | - Yiming Liu
- Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xiaocui Li
- Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Gaochao Zhou
- Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Chunai Zhao
- Department of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yang Han
- School of Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
10
|
Gaviria-Soteras L, Sharma AK, Sanmartín C, Plano D. Recent Insights into Bioactive Dichalcogen Derivatives: From Small Molecules to Complex Materials. Int J Mol Sci 2025; 26:2436. [PMID: 40141080 PMCID: PMC11942125 DOI: 10.3390/ijms26062436] [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: 02/06/2025] [Revised: 03/04/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Organodichalcogenides have been explored due to their therapeutic properties. They have been demonstrated to be active against several diseases such as cancer, bacteria, viruses, parasites, or neurological diseases. Among the different classes of dichalcogenides, disulfide derivatives have been widely studied, and many studies cover their therapeutical use. For this reason, this review includes the latest studies of diselenides and ditellurides derivatives with biological applications. With this aim, several bioactive small molecules containing the diselenide or ditelluride bond in their structure have been discussed. Furthermore, it should be highlighted that, in recent years, there has been an increasing interest in the development of nanomaterials for drug delivery due to their therapeutic advantages. In this context, diselenide and ditelluride-containing nanocarriers have emerged as novel approaches. The information compiled in this review includes small molecules and more complex materials containing diselenide or ditelluride bonds in their structure for different therapeutical applications, which could be helpful for the further development of novel drugs for the treatment of different diseases.
Collapse
Affiliation(s)
- Leire Gaviria-Soteras
- Department of Pharmaceutical Sciences, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain; (L.G.-S.); (D.P.)
| | - Arun K. Sharma
- Department of Molecular and Precision Medicine, Penn State Cancer Institute, CH72, 500 University Drive, Hershey, PA 17033, USA;
| | - Carmen Sanmartín
- Department of Pharmaceutical Sciences, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain; (L.G.-S.); (D.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| | - Daniel Plano
- Department of Pharmaceutical Sciences, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain; (L.G.-S.); (D.P.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, E-31008 Pamplona, Spain
| |
Collapse
|
11
|
Zhou H, Yang Z, Jin G, Wang L, Su Y, Liu H, Sun H, Xue L, Mi L, Veselova IA, Li M, Lv S, Chen X. Polymeric Nanoparticles Simultaneously Delivering Paclitaxel Prodrug and Combretastatin A4 with Exceptionally High Drug Loading for Cancer Combination Therapy. NANO LETTERS 2025; 25:3479-3488. [PMID: 39993999 DOI: 10.1021/acs.nanolett.4c05863] [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: 02/26/2025]
Abstract
Nanomedicines capable of delivering multiple drugs have become essential in combination therapy. However, the challenges of low drug loading capacity (DLC) and difficulties in administering dosages between different drugs significantly limit the antitumor efficacy. In this study, a nanomedicine constructed through a rational prodrug and nanocarrier design was reported for cancer combination therapy. Initially, a phenylborate ester (PBE) group-modified paclitaxel (PTX) prodrug (PTX-PBE) was synthesized and could self-assemble in water. Subsequently, combretastatin A4 (CA4) polymer conjugates, mPEG-PCA4 (PCA4), were synthesized as nanocarriers to facilitate the exceptionally high drug loading of PTX-PBE in a precisely controlled manner. Both the in vitro and in vivo experiments demonstrated that the PCA4 loading PTX-PBE nanoparticles (PCA4/PTX-PBE NPs) exhibited potent antitumor efficacy and favorable biocompatibility. Our approach provides a straightforward, efficient, and controllable strategy for the co-delivery of pharmaceuticals in clinical cancer combination therapy.
Collapse
Affiliation(s)
- Huicong Zhou
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Zhaofan Yang
- School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Guanyu Jin
- School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Lanqing Wang
- School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Yuanzhen Su
- School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Hao Liu
- The Central Laboratory, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Hai Sun
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Lingwei Xue
- Yaoshan laboratory, Pingdingshan University, Pingdingshan, Henan 467000, China
| | - Liwei Mi
- Yaoshan laboratory, Pingdingshan University, Pingdingshan, Henan 467000, China
| | - Irina A Veselova
- Department of Chemistry, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510630, China
| | - Shixian Lv
- School of Materials Science and Engineering, Peking University, Beijing 100871, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| |
Collapse
|
12
|
Yuan T, Zhu Y, Dai Y, Bi F, Lin Y, Yang J. Research on an effective, accurate, and universally applicable method for dual-loaded liposomes encapsulation efficiency. J Pharm Sci 2025; 114:1583-1596. [PMID: 39892869 DOI: 10.1016/j.xphs.2025.01.026] [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: 10/28/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/04/2025]
Abstract
OBJECTIVE Dual-loaded liposomes have become increasingly popular in the field of liposomal research. The encapsulation rate of dual-loaded drugs is an important indicator of the quality and efficacy of the dual-loaded liposomes. But it is difficult to determine the encapsulation efficiency of two drugs using a single method when the physicochemical properties of the drugs differ significantly. The aim of this study is to identify a method that is suitable for the different physicochemical properties of drugs and that efficiently and accurately determines the encapsulation efficiency of the two drugs in dual-loaded liposomes. METHODS This study uses three different types of dual-loaded liposomes that simultaneously encapsulate lipophilic and hydrophilic drugs to examine the separation efficiency, encapsulation rate error, and applicability of various currently available methods for determining the encapsulation efficiency of dual-loaded liposomes. These methods include centrifugation, dialysis, ultrafiltration, microcolumn centrifugation, nanoparticle exclusion chromatography (nPEC), and polyethylene glycol-single-chain variable fragment (PEG-scFv) induced sedimentation. RESULTS The results indicate that microcolumn centrifugation, nPEC, and PEG-scFv induced sedimentation methods achieved >90 % separation efficiency for both lipophilic and hydrophilic drugs. Among these, microcolumn centrifugation is cumbersome to operate, and the PEG-scFv induced sedimentation method is only applicable to PEGylated liposomes. In contrast, the nPEC method requires no pre-treatment and is suitable for the separation of all nanoparticles and free drugs. CONCLUSIONS This study concludes through a comparative analysis that the nPEC method is an effective, accurate, and universally applicable method for assessing the encapsulation efficiency of dual-loaded liposomes.
Collapse
Affiliation(s)
- Tong Yuan
- Key Laboratory of Drug Metabolism & Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, PR China
| | - Yuqi Zhu
- Jiangsu Key Laboratory of Carcinogenesis and Intervention,School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, PR China
| | - Yaqi Dai
- Key Laboratory of Drug Metabolism & Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, PR China
| | - Fulin Bi
- Key Laboratory of Drug Metabolism & Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, PR China
| | - Yan Lin
- Department of Clinical Pharmacy, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, PR China.
| | - Jin Yang
- Key Laboratory of Drug Metabolism & Pharmacokinetics, School of Pharmacy, China Pharmaceutical University, Nanjing, PR China.
| |
Collapse
|
13
|
Ma S, Jiang L, Yang W, Liu F, Wang D, Wang F, Huang J. Advances of Nanomaterials in Cancer Photocatalysis Therapy. MATERIALS TODAY SUSTAINABILITY 2025; 29:101023. [DOI: 10.1016/j.mtsust.2024.101023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
14
|
Chen J, Huang Z, Chen Y, Tian H, Chai P, Shen Y, Yao Y, Xu S, Ge S, Jia R. Lactate and lactylation in cancer. Signal Transduct Target Ther 2025; 10:38. [PMID: 39934144 PMCID: PMC11814237 DOI: 10.1038/s41392-024-02082-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 02/13/2025] Open
Abstract
Accumulated evidence has implicated the diverse and substantial influence of lactate on cellular differentiation and fate regulation in physiological and pathological settings, particularly in intricate conditions such as cancer. Specifically, lactate has been demonstrated to be pivotal in molding the tumor microenvironment (TME) through its effects on different cell populations. Within tumor cells, lactate impacts cell signaling pathways, augments the lactate shuttle process, boosts resistance to oxidative stress, and contributes to lactylation. In various cellular populations, the interplay between lactate and immune cells governs processes such as cell differentiation, immune response, immune surveillance, and treatment effectiveness. Furthermore, communication between lactate and stromal/endothelial cells supports basal membrane (BM) remodeling, epithelial-mesenchymal transitions (EMT), metabolic reprogramming, angiogenesis, and drug resistance. Focusing on lactate production and transport, specifically through lactate dehydrogenase (LDH) and monocarboxylate transporters (MCT), has shown promise in the treatment of cancer. Inhibitors targeting LDH and MCT act as both tumor suppressors and enhancers of immunotherapy, leading to a synergistic therapeutic effect when combined with immunotherapy. The review underscores the importance of lactate in tumor progression and provides valuable perspectives on potential therapeutic approaches that target the vulnerability of lactate metabolism, highlighting the Heel of Achilles for cancer treatment.
Collapse
Affiliation(s)
- Jie Chen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ziyue Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Ya Chen
- Department of Radiology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Hao Tian
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Peiwei Chai
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Yongning Shen
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China
| | - Shiqiong Xu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, PR China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, PR China.
| |
Collapse
|
15
|
Chang CH, Yang SJ, Young TH, Yao WC. Effect of co-loaded vitamin D3 on intravenous injectable raloxifene delivery system. Colloids Surf B Biointerfaces 2025; 246:114379. [PMID: 39566355 DOI: 10.1016/j.colsurfb.2024.114379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 11/22/2024]
Abstract
Owing to its promising advantages, including improved drug bioavailability and therapeutic efficiency at low doses and frequency, increased patient convenience and compliance, and prolonged storage life, nanomedicine has received heightened attention over conventional pharmaceuticals. Human serum albumin (HSA)-based nanoparticles have been used as drug carriers in injectable formulations, with great success and versatility. In this study, raloxifene and vitamin D3 were co-encapsulated in HSA-based nanoparticles (Ral/VitaD/HSA/PSS NPs) as an intravenously injected pharmaceutical formulation in order to enhance their availability in the body. The lyophilization-hydration method was utilized to develop the Ral/VitaD/HSA/PSS NPs. In addition, the characteristics and stability of the NP and the effect of the co-loading of vitamin D3 on raloxifene release in vitro and in vivo were discussed. The raloxifene and vitamin D3 molecules were successfully encapsulated and well dispersed in an amorphous state within Ral/VitaD/HSA/PSS NPs. The prepared Ral/VitaD/HSA/PSS NPs were lyophilized for long-term storage and were both biocompatible and hemocompatible, enhancing alkaline phosphtase activity in osteoblasts. Delivered via intravenous injection, Ral/VitaD/HSA/PSS NPs addressed the low bioavailability of raloxifene and vitamin D3 caused by oral administration, and improved their compatibility and residence time in the body. Overall, the established raloxifene-vitamin D3-co-loaded NPs may be a potential nanomedicine contender for treating postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Chih-Hao Chang
- Department of Orthopedics, National Taiwan University Hospital and National Taiwan University College of Medicine, No. 1, Section 1, Jen-Ai Road, Taipei 100, Taiwan.
| | - Shu-Jyuan Yang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei 100, Taiwan.
| | - Tai-Horng Young
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Wei-Cheng Yao
- Department of Anesthesiology and Pain Medicine, Min-Sheng General Hospital, No. 168, Jingguo Road, Taoyuan Dis., Taoyuan City 330, Taiwan
| |
Collapse
|
16
|
Dai X, Yin Y, Wang C, Xu H. Hyaluronic acid regulated facile synthesis of size-tunable multifunctional nanomedicine for effective cancer therapy. Int J Biol Macromol 2025; 288:138668. [PMID: 39667478 DOI: 10.1016/j.ijbiomac.2024.138668] [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: 10/06/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
The complex and heterogeneous nature of cancer necessitates the development of innovative multifunctional nanomedicines (MN). Hyaluronic acid (HA) is a functional carbohydrate polysaccharide that is widely used in various biomedical fields. In this study, we employed HA as a stabilizer and regulator for the synthesis of a size-tunable nanomedicine comprising ferric ions, doxorubicin, and epigallocatechin gallate (EGCG), referred to as HDE-MN, for cancer therapy. A change in the HA ratio can yield HDE-MNs with sizes varying from ~20 nm to over 100 nm. Modified HA can respond to hyaluronidase (HAase) to provide controllable pH/HAase dual-responsive drug release for improved cancer therapy. Moreover, HA can mediate the targeted delivery of HDE-MNs both in vitro and in vivo to cancer cells. In addition, HDE-MNs reversed multidrug resistance owing to the incorporation of EGCG, inducing ferroptosis due to the involvement of ferric ions. More importantly, HDE-MNs have a photothermal conversion effect, enabling photothermal therapy, photothermally enhanced drug release, and ferroptosis, which collectively contribute to significantly improved cancer therapy. Therefore, the HDE-MNs with laser irradiation achieved full ablation of the in vivo tumors. Together with its good biocompatibility, HDE-MNs may be promising for effective cancer therapy.
Collapse
Affiliation(s)
- Xiuliang Dai
- Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, China
| | - Yina Yin
- Obstetrics and Gynecology Department, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, China
| | - Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu 213164, China
| | - Hongbin Xu
- Obstetrics and Gynecology Department, The Second People's Hospital of Changzhou, the Third Affiliated Hospital of Nanjing Medical University, Changzhou, China.
| |
Collapse
|
17
|
Maghsoudian S, Yektakasmaei MP, Shaabani A, Perseh S, Fatahi Y, Nouri Z, Gholami M, Sayyari N, Hoseinzadeh HA, Motasadizadeh H, Dinarvand R. Synergistic effects of doxorubicin loaded silk fibroin nanoparticles and Cu-TiO 2 nanoparticles for local chemo-sonodynamic therapy against breast cancer. Int J Biol Macromol 2025; 289:138910. [PMID: 39701260 DOI: 10.1016/j.ijbiomac.2024.138910] [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: 08/26/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
A promising new approach to mitigate the adverse effects of chemotherapeutic drugs on healthy tissues involves combining sonodynamic therapy with topical chemotherapy to enhance the therapeutic efficacy of anticancer drugs. In this study, we introduce a multi-functional in situ chitosan hydrogel (CS) containing silk fibroin nanoparticles (SFNPs) loaded with doxorubicin (DOXSFNPs) and CuO/TiO2 nanoparticles (CTNPs) for combination therapy. The developed DOXSFNPs exhibited a size of 257 ± 6 nm, a zeta potential of -14.3 ± 1.8 mV, and a high loading capacity of 12.38 ± 1.73 %. The pH-dependent controlled release of DOX from DOXSF2/CS2 was observed to be more pronounced than that from DOX/CS2. MTT results indicated dose-dependent toxicity of CT/CS2 in response to ultrasaound radiation (US). Our findings revealed a 1.83-fold increase in reactive oxygen species (ROS) production with therapy, with the IC50 of CT3-DOXSF2/CS2-US showing a 58 % reduction compared to CT3/DOXSF2/CS2. In vivo outcomes and histopathological staining demonstrated that the CT3/DOXSF2/CS2-US treatment group exhibited the highest tumor growth inhibition rate, reaching approximately 83.65 %. These findings underscore the potential of this approach in minimizing the adverse effects of chemotherapy while maximizing therapeutic outcomes, offering a valuable contribution to the field of cancer therapy.
Collapse
Affiliation(s)
- Samane Maghsoudian
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Alireza Shaabani
- Department of Polymer and Materials Chemistry, Faculty of Chemistry and Petroleum Sciences, Shahid Beheshti University, Iran
| | - Sahra Perseh
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Yousef Fatahi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zeinab Nouri
- Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdi Gholami
- Faculty of Pharmacy and Pharmaceutical Science Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nastaran Sayyari
- School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Hesam Aldin Hoseinzadeh
- Department of Clinical Science, Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | - Rassoul Dinarvand
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Nanotechnology Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Leicester School of Pharmacy, De Montfort University, Leicester, UK.
| |
Collapse
|
18
|
Izadiyan Z, Misran M, Kalantari K, Webster TJ, Kia P, Basrowi NA, Rasouli E, Shameli K. Advancements in Liposomal Nanomedicines: Innovative Formulations, Therapeutic Applications, and Future Directions in Precision Medicine. Int J Nanomedicine 2025; 20:1213-1262. [PMID: 39911259 PMCID: PMC11794392 DOI: 10.2147/ijn.s488961] [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: 07/27/2024] [Accepted: 01/01/2025] [Indexed: 02/07/2025] Open
Abstract
Liposomal nanomedicines have emerged as a pivotal approach for the treatment of various diseases, notably cancer and infectious diseases. This manuscript provides an in-depth review of recent advancements in liposomal formulations, highlighting their composition, targeted delivery strategies, and mechanisms of action. We explore the evolution of liposomal products currently in clinical trials, emphasizing their potential in addressing diverse medical challenges. The integration of immunotherapeutic agents within liposomes marks a paradigm shift, enabling the design of 'immuno-modulatory hubs' capable of orchestrating precise immune responses while facilitating theranostic applications. The recent COVID-19 pandemic has accelerated research in liposomal-based vaccines and antiviral therapies, underscoring the need for improved delivery mechanisms to overcome challenges like rapid clearance and organ toxicity. Furthermore, we discuss the potential of "smart" liposomes, which can respond to specific disease microenvironments, enhancing treatment efficacy and precision. The integration of artificial intelligence and machine learning in optimizing liposomal designs promises to revolutionize personalized medicine, paving the way for innovative strategies in disease detection and therapeutic interventions. This comprehensive review underscores the significance of ongoing research in liposomal technologies, with implications for future clinical applications and enhanced patient outcomes.
Collapse
Affiliation(s)
- Zahra Izadiyan
- Department of Chemistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Misni Misran
- Department of Chemistry, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Katayoon Kalantari
- Department of Chemical Engineering, Northeastern University, Boston, MA, USA
| | - Thomas J Webster
- Biomedical Engineering, Hebei University of Technology, Tianjin, People’s Republic of China
- School of Engineering, Saveetha University, Chennai, India
| | - Pooneh Kia
- Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | | | - Elisa Rasouli
- Department of Electrical and Electronics Engineering, Nanyang Technological University, Nanyang, Singapore
| | - Kamyar Shameli
- School of Medicine, Institute of Virology, Technical University of Munich, Munich, Germany
| |
Collapse
|
19
|
Xiong H, Wang R, Zhang H, Zhang Q, Qin Y, Du C, Zhang X, Ye J, Shi C, Shen H, Zhu Z, Zhou Z, Chen X, Zhang J. Preclinical and First-in-Human Study of a Compact Radionuclide Labeled Self-Assembly Nanomedicine for Chemo-Radio-Theranostics of Cancer. ACS NANO 2025; 19:3953-3965. [PMID: 39806279 DOI: 10.1021/acsnano.4c18489] [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: 01/16/2025]
Abstract
The emerging combination of chemotherapy and radionuclide therapy has been actively investigated to overcome the limitations of monotherapy and augment therapeutic efficacy. However, it remains a challenge to design a single delivery vehicle that can incorporate chemotherapeutics and radionuclides into a compact structure. Here, a chelator DOTA- or NOTA-modified Evans blue conjugated camptothecin molecule (EB-CPT) nanoprodrug was synthesized, which could self-assemble into nanoparticles due to its inherent amphiphilicity. The nanoparticles could then be effectively labeled with therapeutic radionuclide lutetium-177 (177Lu) or diagnostic radionuclides gallium-68 (68Ga)/copper-64 (64Cu) with high radiolabeling efficiency and radiochemical stability. Impressively, a single-dose chemoradiation therapy of [177Lu]Lu-DOTA-EB-CPT plus EB-CPT effectively inhibited tumor growth in HCT116 tumor-bearing mice compared to the respective individual therapeutic approach. The [64Cu]Cu-NOTA-EB-CPT nanoparticles also exhibited excellent in vivo characteristics including favorable blood circulation properties and prolonged tumor retention in tumor-bearing mice. The safety, feasibility, tolerability, and biodistribution of [68Ga]Ga-NOTA-EB-ss-CPT were also preliminarily characterized in a first-in-human study. This study presents a simple but robust EB-CPT radiopharmaceutical that leverages EB as an albumin binder to strike a delicate balance between enhanced tumor accumulation, safety, and diagnostic efficacy, facilitating an integrated theranostic strategy within a single molecular structure. This radionuclide-labeled EB-CPT nanomedicine presents a step toward clinical translation of the combination of chemotherapy and radiotheranostics.
Collapse
Affiliation(s)
- Hehe Xiong
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Rongxi Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, 119074, Singapore
| | - Heng Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Qianyu Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Yatong Qin
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Chao Du
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xinyi Zhang
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jinmin Ye
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Changrong Shi
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, 119074, Singapore
| | - Huaxiang Shen
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Zhaohui Zhu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Zijian Zhou
- State Key Laboratory of Vaccines for Infectious Diseases, Xiang An Biomedicine Laboratory & Center for Molecular Imaging and Translational Medicine, National Innovation Platform for Industry-Education Integration in Vaccine Research, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, 138667, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Department of Chemical and Biomolecular Engineering, College of Design and Engineering, National University of Singapore, 117575 Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117575 Singapore
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore 117544, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos Singapore 138673, Singapore
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, 119074, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Theranostics Center of Excellence (TCE), Yong Loo Lin School of Medicine, National University of Singapore, 11 Biopolis Way, Helios, 138667, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| |
Collapse
|
20
|
He X, Karlsson PA, Xiong R, Moodie LWK, Wang H, Bergström CAS, Hubert M. Liquid crystal nanoparticles for oral combination antibiotic therapies: A strategy towards protecting commensal gut bacteria during treatment. J Colloid Interface Sci 2025; 678:287-300. [PMID: 39245019 DOI: 10.1016/j.jcis.2024.08.230] [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: 07/10/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024]
Abstract
Antibiotics are essential for treating infections and reducing risks during medical interventions. However, many commonly used antibiotics lack the physiochemical properties for an efficient oral administration when treating systemic infection. Instead, we are reliant on intravenous delivery, which presents complications outside of clinical settings. Developing novel formulations for oral administration is a potential solution to this problem. We engineered hexosome and cubosome liquid crystal nanoparticles (LCNPs) characterized by small-angle X-ray scattering and cryogenic transmission electron microscopy, and could encapsulate the antibiotics vancomycin (VAN) and clarithromycin (CLA) with high loading efficiencies. By rationally choosing stable lipid building blocks, the loaded LCNPs demonstrated excellent resilience against enzymatic degradation in an in vitro gut model LCNP stability is crucial as premature antibiotic leakage can negatively impact the gut microbiota. In screens against the representative gut bacteria Enterococcus faecalis and Escherichia coli, our LCNPs provided a protective effect. Furthermore, we explored co-administration and dual loading strategies of VAN and CLA, and demonstrated effective loading, stability and protection for E. faecalis and E. coli. This work represents a proof of concept for the early-stage development of antibiotic-loaded LCNPs to treat systemic infection via oral administration, opening opportunities for combination antibiotic therapies.
Collapse
Affiliation(s)
- Xiguo He
- Department of Pharmacy, Uppsala University, Uppsala, Sweden; The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Uppsala, Sweden; Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden
| | - Philip A Karlsson
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Ruisheng Xiong
- Department of Chemistry, Uppsala University, Uppsala, Sweden
| | - Lindon W K Moodie
- Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden; Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Helen Wang
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Christel A S Bergström
- Department of Pharmacy, Uppsala University, Uppsala, Sweden; The Swedish Drug Delivery Center, Department of Pharmacy, Uppsala University, Uppsala, Sweden; Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden
| | - Madlen Hubert
- Department of Pharmacy, Uppsala University, Uppsala, Sweden; Uppsala Antibiotic Center, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
21
|
Schiavoni V, Emanuelli M, Sartini D, Salvolini E, Pozzi V, Campagna R. Curcumin and its Analogues in Oral Squamous Cell Carcinoma: State-of-the-art and Therapeutic Potential. Anticancer Agents Med Chem 2025; 25:313-329. [PMID: 38757321 DOI: 10.2174/0118715206297840240510063330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 05/18/2024]
Abstract
Oral Squamous Cell Carcinoma (OSCC) is the most common cancer arising from squamous epithelium in the oral cavity and is characterized by high aggressiveness and metastatic potential, which together with a late diagnosis results in a 5-year survival rate of only 50% of patients. The therapeutic options for OSCC management are limited and largely influenced by the cancer stage. While radical surgery can be curative in early stage of disease, most cases require adjuvant therapies, including chemotherapy and radiotherapy which, however, often achieve poor curative rates and are associated with important negative effects. Therefore, there is an urgent need to discover new alternative treatment strategies to improve patients' outcomes. Several medicinal herbs are being studied for their preventive or therapeutic effect in several diseases, including cancer. In particular, the Indian spice curcumin, largely used in oriental countries, has been studied as a chemopreventive or adjuvant agent for different malignancies. Indeed, curcumin is characterized by important biological properties, including antioxidant, anti-inflammatory, and anticancer effects, which could also be exploited in OSCC. However, due to its limited bioavailability and poor aqueous solubility, this review is focused on studies designing new synthetic analogues and developing novel types of curcumin delivery systems to improve its pharmacokinetic and biological properties. Thus, this review analyses the potential therapeutic role of curcumin in OSCC by providing an overview of current in vitro and in vivo studies demonstrating the beneficial effects of curcumin and its analogues in OSCC.
Collapse
Affiliation(s)
- Valentina Schiavoni
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, 60020, Italy
| | - Monica Emanuelli
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, 60020, Italy
- New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, Ancona, 60131, Italy
| | - Davide Sartini
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, 60020, Italy
| | - Eleonora Salvolini
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, 60020, Italy
| | - Valentina Pozzi
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, 60020, Italy
| | - Roberto Campagna
- Department of Clinical Sciences, Polytechnic University of Marche, Ancona, 60020, Italy
| |
Collapse
|
22
|
Czuczi T, Murányi J, Móra I, Gurbi B, Varga A, Papp D, Schlosser G, Csala M, Csámpai A. Development of Novel Imipridones with Alkyne- and Triazole-Linked Warheads on the Tricyclic Skeleton, Showing Superior Ability to Eradicate PANC-1 and Fadu Cells Compared to ONC201. Int J Mol Sci 2024; 25:13176. [PMID: 39684886 DOI: 10.3390/ijms252313176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/18/2024] Open
Abstract
Our ongoing research focuses on the development of new imipridone derivatives. We aim to design compounds that can completely and selectively eradicate cancer cells after relatively short treatment. We have synthetized systematically designed novel hybrids and evaluated their antiproliferative activity against PANC-1 and Fadu cell lines. We have also conducted preliminary studies on the mechanism, including colony formation as well as dose-response tests in HEK293T wild-type (WT) and HEK293T CLPP-/- cells. Following gradual structural fine-tuning based on high throughput screening, we identified two imipridone hybrids as the most potent derivatives. Their unique substitution pattern includes N-methylated propargylamine and ferrocenyl/phenyltriazole moieties on the benzyl groups attached to opposite sides of the imipridone core. We found that the compounds with IC50 values similar to those of ONC201 completely eradicated cancer cells at about 4 μM, while ONC201 treatment at even higher concentrations left 30-50% of viable cells behind. Both compounds exerted equal activity in WT and CLPP-/- HEK293T cells, indicating a ClpP-independent mechanism. Further development is needed to improve the tumor selectivity of the two potent imipridone derivatives. By preserving tumor cytotoxicity, we aim to generate new drug candidates that evade resistance and can be applied in a sufficiently broad therapeutic window.
Collapse
Affiliation(s)
- Tamás Czuczi
- Department of Organic Chemistry, Eötvös Loránd University (ELTE), Pázmány P. Sétány 1/A, H-1117 Budapest, Hungary
- Hevesy György PhD School of Chemistry, Pázmány P. Sétány 1/A, H-1117 Budapest, Hungary
| | - József Murányi
- Department of Organic Chemistry, Eötvös Loránd University (ELTE), Pázmány P. Sétány 1/A, H-1117 Budapest, Hungary
| | - István Móra
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
| | - Bianka Gurbi
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
| | - Attila Varga
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
| | - Dávid Papp
- Hevesy György PhD School of Chemistry, Pázmány P. Sétány 1/A, H-1117 Budapest, Hungary
- MTA-ELTE Lendület (Momentum) Ion Mobility Mass Spectrometry Research Group, Institute of Chemistry, ELTE Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary
| | - Gitta Schlosser
- MTA-ELTE Lendület (Momentum) Ion Mobility Mass Spectrometry Research Group, Institute of Chemistry, ELTE Eötvös Loránd University, Pázmány Péter Sétány 1/A, H-1117 Budapest, Hungary
| | - Miklós Csala
- Department of Molecular Biology, Semmelweis University, Tűzoltó u. 37-47, H-1094 Budapest, Hungary
| | - Antal Csámpai
- Department of Organic Chemistry, Eötvös Loránd University (ELTE), Pázmány P. Sétány 1/A, H-1117 Budapest, Hungary
| |
Collapse
|
23
|
Nambiar NR, Gaur S, Ramachandran G, Pandey RS, M S, Nath LR, Dutta T, Sudheesh MS. Remote loading in liposome: a review of current strategies and recent developments. J Liposome Res 2024; 34:658-670. [PMID: 38343137 DOI: 10.1080/08982104.2024.2315449] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 11/28/2024]
Abstract
Liposomes have gained prominence as nanocarriers in drug delivery, and the number of products in the market is increasing steadily, particularly in cancer therapeutics. Remote loading of drugs in liposomes is a significant step in the translation and commercialization of the first liposomal product. Low drug loading and drug leakage from liposomes is a translational hurdle that was effectively circumvented by the remote loading process. Remote loading or active loading could load nearly 100% of the drug, which was not possible with the passive loading procedure. A major drawback of conventional remote loading is that only a very small percentage of the drugs are amenable to this method. Therefore, methods for drug loading are still a problem for several drugs. The loading of multiple drugs in liposomes to improve the efficacy and safety of nanomedicine has gained prominence recently with the introduction of a marketed formulation (Vyxeos) that improves overall survival in acute myeloid leukemia. Different strategies for modifying the remote loading process to overcome the drawbacks of the conventional method are discussed here. The review aims to discuss the latest developments in remote loading technology and its implications in liposomal drug delivery.
Collapse
Affiliation(s)
- Navami Rajan Nambiar
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi, India
| | - Shreya Gaur
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi, India
| | - Gayathri Ramachandran
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi, India
| | - Ravi Shankar Pandey
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | - Sabitha M
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi, India
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi, India
| | | | - M S Sudheesh
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Health Science Campus, Amrita Vishwa Vidyapeetham, Ponekkara, Kochi, India
| |
Collapse
|
24
|
Yuan Y, Li J, Chen M, Zhao Y, Zhang B, Chen X, Zhao J, Liang H, Chen Q. Nano-encapsulation of drugs to target hepatic stellate cells: Toward precision treatments of liver fibrosis. J Control Release 2024; 376:318-336. [PMID: 39413846 DOI: 10.1016/j.jconrel.2024.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Liver fibrosis is characterized by excessive extracellular matrix (ECM) deposition triggered by hepatic stellate cells (HSCs). As central players in fibrosis progression, HSCs are the most important therapeutic targets for antifibrotic therapy. However, owing to the limitations of systemic drug administration, there is still no suitable and effective clinical treatment. In recent years, nanosystems have demonstrated expansive therapeutic potential and evolved into a clinical modality. In liver fibrosis, nanosystems have undergone a paradigm shift from targeting the whole liver to locally targeted modifying processes. Nanomedicine delivered to HSCs has significant potential in managing liver fibrosis, where optimal management would benefit from targeted delivery, personalized therapy based on the specific site of interest, and minor side effects. In this review, we present a brief overview of the role of HSCs in the pathogenesis of liver fibrosis, summarize the different types of nanocarriers and their specific delivery applications in liver fibrosis, and highlight the biological barriers associated with the use of nanosystems to target HSCs and approaches available to solve this issue. We further discuss in-depth all the molecular target receptors overexpressed during HSC activation in liver fibrosis and their corresponding ligands that have been used for drug or gene delivery targeting HSCs.
Collapse
Affiliation(s)
- Yue Yuan
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Jiaxuan Li
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Min Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Ying Zhao
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China
| | - Jianping Zhao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China.
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China; Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan, China.
| | - Qian Chen
- Division of Gastroenterology, Department of Internal Medicine at Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China.
| |
Collapse
|
25
|
Inam A, Zhang S, Zhang S, Wu D. AQ4N nanocomposites for hypoxia-associated tumor combination therapy. Biomater Sci 2024; 12:5883-5911. [PMID: 39431892 DOI: 10.1039/d4bm00883a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Hypoxia in solid tumors increases their invasiveness and resistance to therapy, presenting a formidable obstacle in tumor therapy. The hypoxia prodrug banoxantrone (AQ4N) undergoes conversion into its topoisomerase II inhibitor form AQ4 under hypoxic conditions, which inhibits tumor cells while leaving normal cells unharmed. Numerous studies have found that AQ4N significantly enhances the tumor effect while minimizing toxicity to normal tissues when combined with other drugs or therapeutic approaches. Thus, to maximize AQ4N's effectiveness, co-delivery of AQ4N with other therapeutic agents to the tumor site is paramount, leading to the development of multifunctional multicomponent AQ4N nanocomposites thereby emerging as promising candidates for combination therapy in tumor treatment. However, currently there is a lack of systematic analysis and reviews focusing on AQ4N. Herein, this review provides a comprehensive retrospect and analysis of the recent advancements in AQ4N nanocomposites. Specifically, we discuss the synergistic effects observed when AQ4N is combined with chemotherapeutic drugs, radiotherapy, phototherapy, starvation, sonodynamic therapy and immunotherapy in preclinical models. Moreover, the advantages, limitations, and future perspectives of different AQ4N nanocomposites are highlighted, providing researchers from diverse fields with novel insights into tumor treatment.
Collapse
Affiliation(s)
- Amrah Inam
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.
| | - Shuo Zhang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.
| | - Shuai Zhang
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.
| | - Daocheng Wu
- Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, P.R. China.
| |
Collapse
|
26
|
Gogikar SK, Sen S, Daravath S, Aalhate M, Mahajan S, Ateeq MAM, Singh PK, Dikundwar AG. Bioanalytical method development and validation of docetaxel and carvacrol in mice plasma using LC-QqQ-MS/MS. EUROPEAN JOURNAL OF MASS SPECTROMETRY (CHICHESTER, ENGLAND) 2024; 30:235-245. [PMID: 39428991 DOI: 10.1177/14690667241289001] [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: 10/22/2024]
Abstract
Present work describes the development of a liquid chromatography tandem mass spectrometry-based bioanalytical method for the reliable simultaneous quantification of docetaxel (DXL) and carvacrol (CVL) in the mice plasma. A rapid and sensitive bioanalytical method was developed and optimized in mice plasma using Paclitaxel as an internal standard. Validation of the bioanalytical method was performed according to the ICH M10 guideline covering the range of 9.62-1923.08 ng/mL in the mice plasma milleu at the low, mid, and high-quality control concentrations of 28.86 ng/mL, 961.54 ng/mL, and 1346.15 ng/mL, respectively for both the analytes. Validation parameters such as accuracy, precision, carryover-test, matrix effect, and reinjection reproducibility were carried out and were found in limits. Stability studies (Benchtop, autosampler, freeze-thaw, and long-term) were performed and found to be within limits. The developed bioanalytical method was found to be suitable for the simultaneous quantification of DXL and CVL in the mice plasma.
Collapse
Affiliation(s)
- Shiva Kumar Gogikar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Sibu Sen
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Sandeep Daravath
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Mohd Aman Mohd Ateeq
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Amol G Dikundwar
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
27
|
Ulusal H, Ulusal F, Özdemir N. Development of mesoporous magnetic nanoparticles supported idarubicin and investigation of apoptotic and cytotoxic effects on cancer cell lines. Mol Biol Rep 2024; 51:975. [PMID: 39259442 DOI: 10.1007/s11033-024-09914-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/04/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Many methods are used for cancer treatment, especially chemotherapy. In addition to the their therapeutic effects, chemotherapeutic drugs also have serious disadvantages, such as not being cell and tissue-specific, causing toxicity in many tissues, and developing drug resistance. Many methods, especially nanocarriers, have been designed to overcome these disadvantages. METHODS AND RESULTS In this study, we synthesized mesoporous silica iron oxide nanoparticles with different pore diameters and loaded idarubicin (6MFe3O4-NH2-IDA and 35MFe3O4-NH2-IDA). The synthesized molecules were characterized using FT-IR, XRD, and SEM methods. The cytotoxic effects of unbound idarubicin and idarubicin-loaded nanoparticles on MCF7 and HL-60 cell lines were examined by MTT test. Additionally, the expression of anti-apoptotic (Survivin and BCL-2) and apoptotic (BAX, PUMA, and NOXA) genes of the nanoparticles were measured by PCR method. As a result of the analyses, it was seen that nanoparticles with the desired properties and sizes were synthesized. In MTT analysis, it was observed that both nanoparticles dramatically decreased the IC50 value in cell lines. However, the 35MFe3O4-NH2-IDA molecule was found to have lower IC50 values. IC50 values for pristine IDA, 6MFe3O4-NH2, and 35MFe3O4-NH2 at 24 h were found to be 3.56, 1.24 and 0.25 µM in the MCF7 cell line and 4.15, 1.16 and 0.34 µM in the HL-60 cell line, respectively. Additionally, apoptotic gene expression increased, and anti-apoptotic gene expression decreased. CONCLUSIONS Our study demonstrates that the effectiveness of idarubicin can be significantly enhanced by its application with mesoporous nanocarriers. This enhancement is attributed to the controlled release of idarubicin from the nanocarrier, which circumvents drug resistance mechanisms, improves drug solubility, and increases the drug-carrying capacity per unit volume due to the porous structure of the carrier. These findings underscore the potential of the synthesized nanocarrier in cancer treatment and provide a clear direction for future research in this field.
Collapse
Affiliation(s)
- Hasan Ulusal
- Department of Medical Biochemistry, Gaziantep University, Gaziantep, 27310, Türkiye.
| | - Fatma Ulusal
- Department of Chemistry and Chemical Process Technologies, Mersin Tarsus Organized Industrial Zone Technical Sciences Vocational School, Tarsus University, Mersin, 33100, Türkiye
| | - Nalan Özdemir
- Department of Chemistry, Erciyes University, Kayseri, 38039, Türkiye
| |
Collapse
|
28
|
Ahmad W, Sajjad W, Zhou Q, Ge Z. Nanomedicine for combination of chemodynamic therapy and immunotherapy of cancers. Biomater Sci 2024; 12:4607-4629. [PMID: 39115141 DOI: 10.1039/d3bm02133e] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Chemodynamic therapy (CDT), as a new type of therapy, has received more and more attention in the field of tumor therapy in recent years. By virtue of the characteristics of weak acidity and excess H2O2 in the tumor microenvironment, CDT uses the Fenton or Fenton-like reactions to catalyze the transformation of H2O2 into strongly oxidizing ˙OH, resulting in increased intracellular oxidative stress for lipid oxidation, protein inactivation, or DNA damage, and finally inducing apoptosis of cancer cells. In particular, CDT has the advantage of tumor specificity. However, the therapeutic efficacy of CDT frequently depends on the catalytic efficiency of the Fenton reaction, which needs the presence of sufficient H2O2 and catalytic metal ions. Relatively low concentrations of H2O2 and the lack of catalytic metal ions usually limit the final therapeutic effect. The combination of CDT with immunotherapy will be an effective means to improve the therapeutic effect. In this review paper, the recent progress related to nanomedicine for the combination of CDT and immunotherapy is summarized. Immunogenic death of tumor cells, immune checkpoint inhibitors, and stimulator of interferon gene (STING) activation as the main immunotherapy strategies to combine with CDT are discussed. Finally, the challenges and prospects for the clinical translation and future development direction are discussed.
Collapse
Affiliation(s)
- Waqas Ahmad
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Wasim Sajjad
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, China
| | - Qinghao Zhou
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| | - Zhishen Ge
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, Shaanxi, China.
| |
Collapse
|
29
|
Yoo H, Kim Y, Kim J, Cho H, Kim K. Overcoming Cancer Drug Resistance with Nanoparticle Strategies for Key Protein Inhibition. Molecules 2024; 29:3994. [PMID: 39274842 PMCID: PMC11396748 DOI: 10.3390/molecules29173994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/06/2024] [Accepted: 08/21/2024] [Indexed: 09/16/2024] Open
Abstract
Drug resistance remains a critical barrier in cancer therapy, diminishing the effectiveness of chemotherapeutic, targeted, and immunotherapeutic agents. Overexpression of proteins such as B-cell lymphoma 2 (Bcl-2), inhibitor of apoptosis proteins (IAPs), protein kinase B (Akt), and P-glycoprotein (P-gp) in various cancers leads to resistance by inhibiting apoptosis, enhancing cell survival, and expelling drugs. Although several inhibitors targeting these proteins have been developed, their clinical use is often hampered by systemic toxicity, poor bioavailability, and resistance development. Nanoparticle-based drug delivery systems present a promising solution by improving drug solubility, stability, and targeted delivery. These systems leverage the Enhanced Permeation and Retention (EPR) effect to accumulate in tumor tissues, reducing off-target toxicity and increasing therapeutic efficacy. Co-encapsulation strategies involving anticancer drugs and resistance inhibitors within nanoparticles have shown potential in achieving coordinated pharmacokinetic and pharmacodynamic profiles. This review discusses the mechanisms of drug resistance, the limitations of current inhibitors, and the advantages of nanoparticle delivery systems in overcoming these challenges. By advancing these technologies, we can enhance treatment outcomes and move towards more effective cancer therapies.
Collapse
Affiliation(s)
- Hyeonji Yoo
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Yeonjin Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Jinseong Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Hanhee Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea
| |
Collapse
|
30
|
Hadkar VM, Mohanty C, Selvaraj CI. Biopolymeric nanocarriers in cancer therapy: unleashing the potency of bioactive anticancer compounds for enhancing drug delivery. RSC Adv 2024; 14:25149-25173. [PMID: 39139249 PMCID: PMC11317881 DOI: 10.1039/d4ra03911d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
Effective cancer treatment is becoming a global concern, and recent developments in nanomedicine are essential for its treatment. Cancer is a severe metabolic syndrome that affects the human population and is a significant contributing factor to deaths globally. In science, nanotechnology offers rapidly developing delivery methods for natural bioactive compounds that are becoming increasingly prominent and can be used to treat diseases in a site-specific way. Chemotherapy and radiotherapy are conventional approaches for preventing cancer progression and have adverse effects on the human body. Many chemically synthesized drugs are used as anticancer agents, but they have several side effects; hence, they are less preferred. Medicinal plants and marine microorganisms represent a vast, mostly untapped reservoir of bioactive compounds for cancer treatment. However, they have several limitations, including nonspecific targeting, weak water solubility and limited therapeutic potential. An alternative option is the use of biopolymeric nanocarriers, which can generate effective targeted treatment therapies when conjugated with natural anticancer compounds. The present review focuses on biopolymeric nanocarriers utilizing natural sources as anticancer drugs with improved tumor-targeting efficiency. This review also covers various natural anticancer compounds, the advantages and disadvantages of natural and synthetic anticancer compounds, the problems associated with natural anticancer drugs and the advantages of biopolymeric nanocarriers over synthetic nanocarriers as drug delivery agents. This review also discusses various biopolymeric nanocarriers for enhancing the controlled delivery of anticancer compounds and the future development of nanomedicines for treating cancer.
Collapse
Affiliation(s)
- Vrushali Manoj Hadkar
- School of Biosciences and Technology, Vellore Institute of Technology (VIT) Vellore 632014 Tamil Nadu India
| | - Chirasmita Mohanty
- School of Biosciences and Technology, Vellore Institute of Technology (VIT) Vellore 632014 Tamil Nadu India
| | - Chinnadurai Immanuel Selvaraj
- Department of Genetics and Plant Breeding, VIT School of Agricultural Sciences and Advanced Learning (VAIAL), VIT Vellore 632014 Tamil Nadu India
| |
Collapse
|
31
|
Shin YB, Choi JY, Yoon MS, Yoo MK, Shin DH, Lee JW. Evaluation of Anticancer Efficacy of D-α-Tocopheryl Polyethylene-Glycol Succinate and Soluplus ® Mixed Micelles Loaded with Olaparib and Rapamycin Against Ovarian Cancer. Int J Nanomedicine 2024; 19:7871-7893. [PMID: 39114180 PMCID: PMC11304412 DOI: 10.2147/ijn.s468935] [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: 03/14/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
Purpose Ovarian cancer has the highest mortality rate and lowest survival rate among female reproductive system malignancies. There are treatment options of surgery and chemotherapy, but both are limited. In this study, we developed and evaluated micelles composed of D-α-tocopheryl polyethylene-glycol (PEG) 1000 succinate (TPGS) and Soluplus® (SOL) loaded with olaparib (OLA), a poly(ADP-ribose)polymerase (PARP) inhibitor, and rapamycin (RAPA), a mammalian target of rapamycin (mTOR) inhibitor in ovarian cancer. Methods We prepared micelles containing different molar ratios of OLA and RAPA embedded in different weight ratios of TPGS and SOL (OLA/RAPA-TPGS/SOL) were prepared and physicochemical characterized. Furthermore, we performed in vitro cytotoxicity experiments of OLA, RAPA, and OLA/RAPA-TPGS/SOL. In vivo toxicity and antitumor efficacy assays were also performed to assess the efficacy of the mixed micellar system. Results OLA/RAPA-TPGS/SOL containing a 4:1 TPGS:SOL weight ratio and a 2:3 OLA:RAPA molar ratio showed synergistic effects and were optimized. The drug encapsulation efficiency of this formulation was >65%, and the physicochemical properties were sustained for 180 days. Moreover, the formulation had a high cell uptake rate and significantly inhibited cell migration (**p < 0.01). In the in vivo toxicity test, no toxicity was observed, with the exception of the high dose group. Furthermore, OLA/RAPA-TPGS/SOL markedly inhibited tumor spheroid and tumor growth in vivo. Conclusion Compared to the control, OLA/RAPA-TPGS/SOL showed significant tumor inhibition. These findings lay a foundation for the use of TPGS/SOL mixed micelles loaded with OLA and RAPA in the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Yu Been Shin
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Ju-Yeon Choi
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Moon Sup Yoon
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Myeong Kyun Yoo
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Dae Hwan Shin
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
- Chungbuk National University Hospital, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Jeong-Won Lee
- Department of Obstetrics and Gynecology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
- Samsung Advanced Institute for Health Sciences & Technology, Sungkyunkwan University School of Medicine, Seoul, 06351, Republic of Korea
| |
Collapse
|
32
|
Chaudhuri A, Naveen Kumar D, Kumar D, Kumar Agrawal A. Functionalized solid lipid nanoparticles combining docetaxel and erlotinib synergize the anticancer efficacy against triple-negative breast cancer. Eur J Pharm Biopharm 2024; 201:114386. [PMID: 38950717 DOI: 10.1016/j.ejpb.2024.114386] [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: 03/18/2024] [Revised: 06/08/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
The goal of the study was to fabricate folic acid functionalized docetaxel (DOC)/erlotinib (ERL)-loaded solid lipid nanoparticles (SLNs) to synergistically increase the anticancer activity against triple-negative breast cancer. DOC/ERL-SLNs were prepared by the high shear homogenization - ultrasound dispersion method (0.1 % w/v for DOC, and 0.3 %w/v for ERL) and optimized using Plackett Burman Design (PBD) followed by Box Behnken Design (BBD). The optimized SLNs demonstrated particle size < 200 nm, PDI < 0.35, and negative zeta potential with entrapment and loading efficiency of ∼80 and ∼4 %, respectively. The SLNs and folic acid functionalized SLNs (FA-SLNs) showed sustained release for both drugs, followed by Higuchi and Korsemeyer-Peppas drug release models, respectively. Further, the in vitro pH-stat lipolysis model demonstrated an approximately 3-fold increase in the bioaccessibility of drugs from SLNs compared to suspension. The TEM images revealed the spherical morphology of the SLNs. DOC/ERL loaded SLNs showed dose- and time-dependent cytotoxicity and exhibited a synergism at a molar ratio of 1:3 in TNBC with a combination index of 0.35 and 0.37, respectively. FA-DOC/ERL-SLNs showed enhanced anticancer activity as evidenced by MMP and ROS assay and further inhibited the colony-forming ability and the migration capacity of TNBC cells. Conclusively, the study has shown that SLNs are encouraging systems to improve the pharmaceutical attributes of poorly bioavailable drugs.
Collapse
Affiliation(s)
- Aiswarya Chaudhuri
- Department of Pharmaceutical Engineering & Technology, IIT (BHU), Varanasi 221005, India
| | - Dulla Naveen Kumar
- Department of Pharmaceutical Engineering & Technology, IIT (BHU), Varanasi 221005, India
| | - Dinesh Kumar
- Department of Pharmaceutical Engineering & Technology, IIT (BHU), Varanasi 221005, India
| | - Ashish Kumar Agrawal
- Department of Pharmaceutical Engineering & Technology, IIT (BHU), Varanasi 221005, India.
| |
Collapse
|
33
|
Martins CF, García-Astrain C, Conde J, Liz-Marzán LM. Nanocomposite hydrogel microneedles: a theranostic toolbox for personalized medicine. Drug Deliv Transl Res 2024; 14:2262-2275. [PMID: 38376619 PMCID: PMC11208216 DOI: 10.1007/s13346-024-01533-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2024] [Indexed: 02/21/2024]
Abstract
Due to the severity and high prevalence of cancer, as well as its complex pathological condition, new strategies for cancer treatment and diagnostics are required. As such, it is important to design a toolbox that integrates multiple functions on a single smart platform. Theranostic hydrogels offer an innovative and personalized method to tackle cancer while also considering patient comfort, thereby facilitating future implementation and translation to the clinic. In terms of theranostic systems used in cancer therapy, nanoparticles are widely used as diagnostic and therapeutic tools. Nanoparticles can achieve systemic circulation, evade host defenses, and deliver drugs and signaling agents at the targeted site, to diagnose and treat the disease at a cellular and molecular level. In this context, hydrogel microneedles have a high potential for multifunctional operation in medical devices, while avoiding the complications associated with the systemic delivery of therapeutics. Compared with oral administration and subcutaneous injection, microneedles offer advantages such as better patient compliance, faster onset of action, and improved permeability and efficacy. In addition, they comprise highly biocompatible polymers with excellent degradability and tunable properties. Nanoparticles and microneedles thus offer the possibility to expand the theranostic potential through combined synergistic use of their respective features. We review herein recent advances concerning processing methods and material requirements within the realm of hydrogel microneedles as theranostic platforms, various approaches toward cancer therapy, and the incorporation of nanoparticles for added functionality.
Collapse
Affiliation(s)
- Catarina F Martins
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas, NMSFCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Clara García-Astrain
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014, Donostia-San Sebastián, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y, Nanomedicina (CIBER-BBN), 20014, Donostia-San Sebastián, Spain
| | - João Conde
- ToxOmics, NOVA Medical School, Faculdade de Ciências Médicas, NMSFCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Luis M Liz-Marzán
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014, Donostia-San Sebastián, Spain.
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y, Nanomedicina (CIBER-BBN), 20014, Donostia-San Sebastián, Spain.
- Ikerbasque, Basque Foundation for Science, 48009, Bilbao, Spain.
| |
Collapse
|
34
|
Ren S, Xu Y, Dong X, Mu Q, Chen X, Yu Y, Su G. Nanotechnology-empowered combination therapy for rheumatoid arthritis: principles, strategies, and challenges. J Nanobiotechnology 2024; 22:431. [PMID: 39034407 PMCID: PMC11265020 DOI: 10.1186/s12951-024-02670-7] [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: 03/25/2024] [Accepted: 06/25/2024] [Indexed: 07/23/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease with multifactorial etiology and intricate pathogenesis. In RA, repeated monotherapy is frequently associated with inadequate efficacy, drug resistance, and severe side effects. Therefore, a shift has occurred in clinical practice toward combination therapy. However, conventional combination therapy encounters several hindrances, including low selectivity to arthritic joints, short half-lives, and varying pharmacokinetics among coupled drugs. Emerging nanotechnology offers an incomparable opportunity for developing advanced combination therapy against RA. First, it allows for co-delivering multiple drugs with augmented physicochemical properties, targeted delivery capabilities, and controlled release profiles. Second, it enables therapeutic nanomaterials development, thereby expanding combination regimens to include multifunctional nanomedicines. Lastly, it facilitates the construction of all-in-one nanoplatforms assembled with multiple modalities, such as phototherapy, sonodynamic therapy, and imaging. Thus, nanotechnology offers a promising solution to the current bottleneck in both RA treatment and diagnosis. This review summarizes the rationale, advantages, and recent advances in nano-empowered combination therapy for RA. It also discusses safety considerations, drug-drug interactions, and the potential for clinical translation. Additionally, it provides design tips and an outlook on future developments in nano-empowered combination therapy. The objective of this review is to achieve a comprehensive understanding of the mechanisms underlying combination therapy for RA and unlock the maximum potential of nanotechnology, thereby facilitating the smooth transition of research findings from the laboratory to clinical practice.
Collapse
Affiliation(s)
- Shujing Ren
- Department of Pharmacy, Affiliated Hospital 2 of Nantong University, Nantong, 226000, PR China
| | - Yuhang Xu
- School of Pharmacy, Nantong University, Nantong, 226000, PR China
| | - Xingpeng Dong
- School of Pharmacy, Nantong University, Nantong, 226000, PR China
| | - Qingxin Mu
- Department of Pharmaceutics, University of Washington, Seattle, WA, 98195, USA
| | - Xia Chen
- Department of Pharmacy, Affiliated Hospital 2 of Nantong University, Nantong, 226000, PR China.
| | - Yanyan Yu
- School of Pharmacy, Nantong University, Nantong, 226000, PR China.
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong, 226000, PR China.
| |
Collapse
|
35
|
Jasim SA, Farber IM, Noraldeen SAM, Bansal P, Alsaab HO, Abdullaev B, Alkhafaji AT, Alawadi AH, Hamzah HF, Mohammed BA. Incorporation of immunotherapies and nanomedicine to better normalize angiogenesis-based cancer treatment. Microvasc Res 2024; 154:104691. [PMID: 38703993 DOI: 10.1016/j.mvr.2024.104691] [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: 02/19/2024] [Revised: 04/11/2024] [Accepted: 04/27/2024] [Indexed: 05/06/2024]
Abstract
Neoadjuvant targeting of tumor angiogenesis has been developed and approved for the treatment of malignant tumors. However, vascular disruption leads to tumor hypoxia, which exacerbates the treatment process and causes drug resistance. In addition, successful delivery of therapeutic agents and efficacy of radiotherapy require normal vascular networks and sufficient oxygen, which complete tumor vasculopathy hinders their efficacy. In view of this controversy, an optimal dose of FDA-approved anti-angiogenic agents and combination with other therapies, such as immunotherapy and the use of nanocarrier-mediated targeted therapy, could improve therapeutic regimens, reduce the need for administration of high doses of chemotherapeutic agents and subsequently reduce side effects. Here, we review the mechanism of anti-angiogenic agents, highlight the challenges of existing therapies, and present how the combination of immunotherapies and nanomedicine could improve angiogenesis-based tumor treatment.
Collapse
Affiliation(s)
| | - Irina M Farber
- Department of children's diseases of the F. Filatov clinical institute of children's health, I. M. Sechenov First Moscow State Medical University of Health of Russian Federation (Sechenov University), Moscow, Russia
| | | | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif 21944, Saudi Arabia
| | - Bekhzod Abdullaev
- Research Department of Biotechnology, New Uzbekistan University, Mustaqillik Avenue 54, Tashkent 100007, Uzbekistan; Department of Oncology, School of Medicine, Central Asian University, Milliy Bog Street 264, Tashkent 111221, Uzbekistan..
| | | | - Ahmed Hussien Alawadi
- College of Technical Engineering, the Islamic University, Najaf, Iraq; College of Technical Engineering, the Islamic University of Al Diwaniyah, Qadisiyyah, Iraq; College of Technical Engineering, the Islamic University of Babylon, Babylon, Iraq
| | - Hamza Fadhel Hamzah
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | |
Collapse
|
36
|
Lakshmi Anvitha N, A G, S V, S B, I G K I. Facile Fabrication of Titanium Carbide (Ti3C2)-Bismuth Vanadate (BiVO4) Nano-Coupled Oxides for Anti-cancer Activity. Cureus 2024; 16:e61492. [PMID: 38952587 PMCID: PMC11216123 DOI: 10.7759/cureus.61492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 06/01/2024] [Indexed: 07/03/2024] Open
Abstract
Background MXene is a newly discovered substance consisting of 2D transition metal carbides or nitrides, produced through the disintegration and etching of aluminum layers. It possesses numerous properties, including a high surface area, conductivity, strength, stiffness, negative zeta potential, and excellent volumetric capacitance. MXene is utilized in detecting anti-cancer medicine, while bismuth vanadate (BiVO4) is synthesized to form an optimized material for anti-cancer activity applications. BiVO4 exhibits visible light absorption, strong chemical stability, and non-toxic properties. However, when loaded onto target stem cells, it can cause skin and respiratory irritation. Aim This study aimed to evaluate the facile fabrication of titanium carbide (Ti3C2)-BiVO4 nanomaterials coupled with oxides for anti-cancer activity. Moreover, it aimed to create Ti3C2-BiVO4 nanomaterials in combination with oxides using X-ray diffraction (XRD) and scanning electron microscopy (SEM) to assess their potential as efficient and targeted anti-cancer agents. Methods and materials To prepare the 2D Ti3C2 MXene, 2.5 g of titanium aluminum carbide (Ti3AlC2) powder was dissolved in 60 mL of a 40% hydrofluoric acid (HF) solution in a polytetrafluoroethylene(PTFE) container. The etching process was made more efficient and completed in 24 hours by using a magnetic stirring system to keep the mixture stirred and heated continuously. The centrifugation was performed at 4000 rpm for five minutes. Subsequently, deionized water was used to wash the solution many times until its pH reached around 7. The appropriate Ti3C2 powder was made by vacuum drying the acquired sediment at 80°C for 24 hours. Monoclinic BiVO4 samples were synthesized via a hydrothermal method. Typically, 10 mmol of Bi(NO3)3.5H2O was dissolved in 100 mL of a 2 mol/L HNO3 solution and stirred uniformly. Subsequently, 10 mmol of ammonium metavanadate (NH4VO3) was added to the mixed solution. After being stirred for one hour, the mixture was transferred into a 100 mL sealed Teflon-lined stainless steel autoclave at 180°C for 16 hours. After cooling to room temperature, the sediment was washed three times with deionized water, ethanol, and acetone, respectively. Finally, the suspension was dried at 80°C, followed by calcination at 450°C for three hours to obtain BiVO4. Ti3C2-BiVO4 heterostructures were prepared by surface modification Ti3C2 using BiVO4 suspensions by a simple, cost-effective approach. Results Ti3C2 nanosheets were observed with BiVO4 particles, and the high crystalline nature of the compound was confirmed after XRD analysis and energy-dispersive spectroscopy (EDS) analysis. The compound was found to be pure without any impurities and exhibited anti-cancer activity. Conclusion The XRD, field emission scanning electron microscopy(FESEM), and EDS investigations provide an in-depth analysis of the structural, morphological, and compositional characteristics of Ti3C2-BiVO4 sheets. The XRD analysis proves the successful combination of different materials and the presence of crystalline phases. The FESEM imaging technique exposes the shape and arrangement of particles in sheets, while the EDS analysis verifies the elemental composition and uniform distribution. These investigations show that Ti3C2-BiVO4 composites have been successfully synthesized, indicating their potential for use in anti-cancer applications.
Collapse
Affiliation(s)
- Nagubandi Lakshmi Anvitha
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS) Saveetha University, Chennai, IND
| | - Geetha A
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS) Saveetha University, Chennai, IND
| | - Vasugi S
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS) Saveetha University, Chennai, IND
| | - Balachandran S
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS) Saveetha University, Chennai, IND
| | - Ilangovar I G K
- Department of Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS) Saveetha University, Chennai, IND
| |
Collapse
|
37
|
Geng WC, Jiang ZT, Chen SL, Guo DS. Supramolecular interaction in the action of drug delivery systems. Chem Sci 2024; 15:7811-7823. [PMID: 38817563 PMCID: PMC11134347 DOI: 10.1039/d3sc04585d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 04/27/2024] [Indexed: 06/01/2024] Open
Abstract
Complex diseases and diverse clinical needs necessitate drug delivery systems (DDSs), yet the current performance of DDSs is far from ideal. Supramolecular interactions play a pivotal role in various aspects of drug delivery, encompassing biocompatibility, drug loading, stability, crossing biological barriers, targeting, and controlled release. Nevertheless, despite having some understanding of the role of supramolecular interactions in drug delivery, their incorporation is frequently overlooked in the design and development of DDSs. This perspective provides a brief analysis of the involved supramolecular interactions in the action of drug delivery, with a primary emphasis on the DDSs employed in the clinic, mainly liposomes and polymers, and recognized phenomena in research, such as the protein corona. The supramolecular interactions implicated in various aspects of drug delivery systems, including biocompatibility, drug loading, stability, spatiotemporal distribution, and controlled release, were individually analyzed and discussed. This perspective aims to trigger a comprehensive and systematic consideration of supramolecular interactions in the further development of DDSs. Supramolecular interactions embody the true essence of the interplay between the majority of DDSs and biological systems.
Collapse
Affiliation(s)
- Wen-Chao Geng
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Ze-Tao Jiang
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Shi-Lin Chen
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| | - Dong-Sheng Guo
- College of Chemistry, Key Laboratory of Functional Polymer Materials (Ministry of Education), State Key Laboratory of Elemento-Organic Chemistry, Frontiers Science Center for New Organic Matter, Collaborative Innovation Center of Chemical Science and Engineering, Nankai University Tianjin 300071 China
| |
Collapse
|
38
|
Wang C, Pan J, Chen S, Qiu L, Hu H, Ji L, Wang J, Liu W, Ni X. Polyvinylpyrrolidone Assisted One-Pot Synthesis of Size-Tunable Cocktail Nanodrug for Multifunctional Combat of Cancer. Int J Nanomedicine 2024; 19:4339-4356. [PMID: 38774026 PMCID: PMC11107942 DOI: 10.2147/ijn.s459428] [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: 01/26/2024] [Accepted: 04/30/2024] [Indexed: 05/24/2024] Open
Abstract
Background The in vivo barriers and multidrug resistance (MDR) are well recognized as great challenges for the fulfillment of antitumor effects of current drugs, which calls for the development of novel therapeutic agents and innovative drug delivery strategies. Nanodrug (ND) combining multiple drugs with distinct modes of action holes the potential to circumvent these challenges, while the introduction of photothermal therapy (PTT) can give further significantly enhanced efficacy in cancer therapy. However, facile preparation of ND which contains dual drugs and photothermal capability with effective cancer treatment ability has rarely been reported. Methods In this study, we selected curcumin (Cur) and doxorubicin (Dox) as two model drugs for the creation of a cocktail ND (Cur-Dox ND). We utilized polyvinylpyrrolidone (PVP) as a stabilizer and regulator to prepare Cur-Dox ND in a straightforward one-pot method. Results The size of the resulting Cur-Dox ND can be easily adjusted by tuning the charged ratios. It was noted that both loaded drugs in Cur-Dox ND can realize their functions in the same target cell. Especially, the P-glycoprotein inhibition effect of Cur can synergistically cooperate with Dox, leading to enhanced inhibition of 4T1 cancer cells. Furthermore, Cur-Dox ND exhibited pH-responsive dissociation of loaded drugs and a robust photothermal translation capacity to realize multifunctional combat of cancer for photothermal enhanced anticancer performance. We further demonstrated that this effect can also be realized in 3D multicellular model, which possibly attributed to its superior drug penetration as well as photothermal-enhanced cellular uptake and drug release. Conclusion In summary, Cur-Dox ND might be a promising ND for better cancer therapy.
Collapse
Affiliation(s)
- Cheng Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Jiaoyang Pan
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Shaoqing Chen
- Department of Radiology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Lin Qiu
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Huaanzi Hu
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Li Ji
- Department of Otorhinolaryngology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Nanjing, Jiangsu Province, People’s Republic of China
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Wenjia Liu
- Department of Gastroenterology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, People’s Republic of China
| | - Xinye Ni
- Department of Radiology, The Affiliated Changzhou No. 2 People’s Hospital of Nanjing Medical University, Changzhou, Jiangsu Province, People’s Republic of China
| |
Collapse
|
39
|
Chen J, Liu Z, Fang H, Su Q, Fan Y, Song L, He S. Therapeutic efficacy of a novel self-assembled immunostimulatory siRNA combining apoptosis promotion with RIG-I activation in gliomas. J Transl Med 2024; 22:395. [PMID: 38685028 PMCID: PMC11057130 DOI: 10.1186/s12967-024-05151-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/30/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Current cancer therapies often fall short in addressing the complexities of malignancies, underscoring the urgent need for innovative treatment strategies. RNA interference technology, which specifically suppresses gene expression, offers a promising new approach in the fight against tumors. Recent studies have identified a novel immunostimulatory small-interfering RNA (siRNA) with a unique sequence (sense strand, 5'-C; antisense strand, 3'-GGG) capable of activating the RIG-I/IRF3 signaling pathway. This activation induces the release of type I and III interferons, leading to an effective antiviral immune response. However, this class of immunostimulatory siRNA has not yet been explored in cancer therapy. METHODS IsiBCL-2, an innovative immunostimulatory siRNA designed to suppress the levels of B-cell lymphoma 2 (BCL-2), contains a distinctive motif (sense strand, 5'-C; antisense strand, 3'-GGG). Glioblastoma cells were subjected to 100 nM isiBCL-2 treatment in vitro for 48 h. Morphological changes, cell viability (CCK-8 assay), proliferation (colony formation assay), migration/invasion (scratch test and Transwell assay), apoptosis rate, reactive oxygen species (ROS), and mitochondrial membrane potential (MMP) were evaluated. Western blotting and immunofluorescence analyses were performed to assess RIG-I and MHC-I molecule levels, and ELISA was utilized to measure the levels of cytokines (IFN-β and CXCL10). In vivo heterogeneous tumor models were established, and the anti-tumor effect of isiBCL-2 was confirmed through intratumoral injection. RESULTS IsiBCL-2 exhibited significant inhibitory effects on glioblastoma cell growth and induced apoptosis. BCL-2 mRNA levels were significantly decreased by 67.52%. IsiBCL-2 treatment resulted in an apoptotic rate of approximately 51.96%, accompanied by a 71.76% reduction in MMP and a 41.87% increase in ROS accumulation. Western blotting and immunofluorescence analyses demonstrated increased levels of RIG-I, MAVS, and MHC-I following isiBCL-2 treatment. ELISA tests indicated a significant increase in IFN-β and CXCL10 levels. In vivo studies using nude mice confirmed that isiBCL-2 effectively impeded the growth and progression of glioblastoma tumors. CONCLUSIONS This study introduces an innovative method to induce innate signaling by incorporating an immunostimulatory sequence (sense strand, 5'-C; antisense strand, 3'-GGG) into siRNA, resulting in the formation of RNA dimers through Hoogsteen base-pairing. This activation triggers the RIG-I signaling pathway in tumor cells, causing further damage and inducing a potent immune response. This inventive design and application of immunostimulatory siRNA offer a novel perspective on tumor immunotherapy, holding significant implications for the field.
Collapse
Affiliation(s)
- Junxiao Chen
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Ziyuan Liu
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Haiting Fang
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Qing Su
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Yiqi Fan
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Luyao Song
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Shuai He
- Department of Pharmacy, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, Guangdong, China.
| |
Collapse
|
40
|
Mugundhan SL, Mohan M. Nanoscale strides: exploring innovative therapies for breast cancer treatment. RSC Adv 2024; 14:14017-14040. [PMID: 38686289 PMCID: PMC11056947 DOI: 10.1039/d4ra02639j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024] Open
Abstract
Breast cancer (BC) is a predominant malignancy in women that constitutes approximately 30% of all cancer cases and has a mortality rate of 14% in recent years. The prevailing therapies include surgery, chemotherapy, and radiotherapy, each with its own limitations and challenges. Despite oral or intravenous administration, there are numerous barriers to accessing anti-BC agents before they reach the tumor site, including physical, physiological, and biophysical barriers. The complexity of BC pathogenesis, attributed to a combination of endogenous, chronic, intrinsic, extrinsic and genetic factors, further complicates its management. Due to the limitations of existing cancer treatment approaches, there is a need to explore novel, efficacious solutions. Nanodrug delivery has emerged as a promising avenue in cancer chemotherapy, aiming to enhance drug bioavailability while mitigating adverse effects. In contrast to conventional chemotherapy, cancer nanotechnology leverages improved permeability to achieve comprehensive disruption of cancer cells. This approach also presented superior pharmacokinetic profiles. The application of nanotechnology in cancer therapeutics includes nanotechnological tools, but a comprehensive review cannot cover all facets. Thus, this review concentrates specifically on BC treatment. The focus lies in the successful implementation of systematic nanotherapeutic strategies, demonstrating their superiority over conventional methods in delivering anti-BC agents. Nanotechnology-driven drug delivery holds immense potential in treating BC. By surmounting multiple barriers and capitalizing on improved permeability, nanodrug delivery has demonstrated enhanced efficacy and reduced adverse effects compared to conventional therapies. This review highlights the significance of systematic nanotherapy approaches, emphasizing the evolving landscape of BC management.
Collapse
Affiliation(s)
- Sruthi Laakshmi Mugundhan
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology SRM Nagar Kattankulathur 603203 Tamil Nadu India
| | - Mothilal Mohan
- Department of Pharmaceutics, SRM College of Pharmacy, SRM Institute of Science and Technology SRM Nagar Kattankulathur 603203 Tamil Nadu India
| |
Collapse
|
41
|
Ahmad N, Bukhari SNA, Hussain MA, Ejaz H, Munir MU, Amjad MW. Nanoparticles incorporated hydrogels for delivery of antimicrobial agents: developments and trends. RSC Adv 2024; 14:13535-13564. [PMID: 38665493 PMCID: PMC11043667 DOI: 10.1039/d4ra00631c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/01/2024] [Indexed: 04/28/2024] Open
Abstract
The prevention and treatment of microbial infections is an imminent global public health concern due to the poor antimicrobial performance of the existing antimicrobial regime and rapidly emerging antibiotic resistance in pathogenic microbes. In order to overcome these problems and effectively control bacterial infections, various new treatment modalities have been identified. To attempt this, various micro- and macro-molecular antimicrobial agents that function by microbial membrane disruption have been developed with improved antimicrobial activity and lesser resistance. Antimicrobial nanoparticle-hydrogels systems comprising antimicrobial agents (antibiotics, biological extracts, and antimicrobial peptides) loaded nanoparticles or antimicrobial nanoparticles (metal or metal oxide) constitute an important class of biomaterials for the prevention and treatment of infections. Hydrogels that incorporate nanoparticles can offer an effective strategy for delivering antimicrobial agents (or nanoparticles) in a controlled, sustained, and targeted manner. In this review, we have described an overview of recent advancements in nanoparticle-hydrogel hybrid systems for antimicrobial agent delivery. Firstly, we have provided an overview of the nanoparticle hydrogel system and discussed various advantages of these systems in biomedical and pharmaceutical applications. Thereafter, different hybrid hydrogel systems encapsulating antibacterial metal/metal oxide nanoparticles, polymeric nanoparticles, antibiotics, biological extracts, and antimicrobial peptides for controlling infections have been reviewed in detail. Finally, the challenges and future prospects of nanoparticle-hydrogel systems have been discussed.
Collapse
Affiliation(s)
- Naveed Ahmad
- Department of Pharmaceutics, College of Pharmacy, Jouf University Sakaka 72388 Aljouf Saudi Arabia
| | - Syed Nasir Abbas Bukhari
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University Sakaka 72388 Aljouf Saudi Arabia
| | - Muhammad Ajaz Hussain
- Centre for Organic Chemistry, School of Chemistry, University of the Punjab Lahore 54590 Pakistan
| | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University Sakaka 72388 Aljouf Saudi Arabia
| | - Muhammad Usman Munir
- Australian Institute for Bioengineering & Nanotechnology, The University of Queensland Brisbane Queens-land 4072 Australia
| | - Muhammad Wahab Amjad
- 6 Center for Ultrasound Molecular Imaging and Therapeutics, School of Medicine, University of Pittsburgh 15213 Pittsburgh Pennsylvania USA
| |
Collapse
|
42
|
Ranasinghe R, Mathai M, Abdullah Alshawsh M, Zulli A. Nanocarrier-mediated cancer therapy with cisplatin: A meta-analysis with a promising new paradigm. Heliyon 2024; 10:e28171. [PMID: 39839154 PMCID: PMC11747978 DOI: 10.1016/j.heliyon.2024.e28171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 01/23/2025] Open
Abstract
Aims Cisplatin is a frontline chemotherapeutic utilized to attenuate multiple cancers in the clinic. Given its side-effects, a new cisplatin formulation which could prevent cytotoxicity, metabolic deficiencies and metastasis is much needed. This study investigates whether nanocarriers can provide a better mode of drug delivery in preclinical cancer models seeking a potent anticancer therapeutic agent. Materials and methods The PubMed database was searched, and 242 research articles were screened from which 94 articles qualified for selection from those published by December 31, 2023 and the data was synthesized using the Review Manager software. Key findings Cisplatin encapsulated as a nanomedicine confirmed the versatility of nanocarriers in significantly diminishing cancer cell viability, half maximal inhibitory concentration, tumour volume, biodistribution of platinum in tumours and kidney; at p < 0.00001 and a 95% confidence interval. Significance An estimated 19.3 million global cancer incidence is reported with 50% mortality worldwide for which nanocarrier-mediated cisplatin therapy is most promising. Our findings offer new vistas for future cancer treatment when combined with chemo-immunotherapy that utilizes the recently advanced nanozymes.
Collapse
Affiliation(s)
- Ranmali Ranasinghe
- Institute for Health and Sport, College of Health and Medicine, Victoria University, Melbourne, Victoria, Australia
| | - Michael Mathai
- Institute for Health and Sport, College of Health and Medicine, Victoria University, Melbourne, Victoria, Australia
| | - Mohammed Abdullah Alshawsh
- Department of Paediatrics, School of Clinical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
- Department of Pharmacology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, 50603, Malaysia
| | - Anthony Zulli
- Institute for Health and Sport, College of Health and Medicine, Victoria University, Melbourne, Victoria, Australia
| |
Collapse
|
43
|
Doostmohammadi A, Jooya H, Ghorbanian K, Gohari S, Dadashpour M. Potentials and future perspectives of multi-target drugs in cancer treatment: the next generation anti-cancer agents. Cell Commun Signal 2024; 22:228. [PMID: 38622735 PMCID: PMC11020265 DOI: 10.1186/s12964-024-01607-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/05/2024] [Indexed: 04/17/2024] Open
Abstract
Cancer is a major public health problem worldwide with more than an estimated 19.3 million new cases in 2020. The occurrence rises dramatically with age, and the overall risk accumulation is combined with the tendency for cellular repair mechanisms to be less effective in older individuals. Conventional cancer treatments, such as radiotherapy, surgery, and chemotherapy, have been used for decades to combat cancer. However, the emergence of novel fields of cancer research has led to the exploration of innovative treatment approaches focused on immunotherapy, epigenetic therapy, targeted therapy, multi-omics, and also multi-target therapy. The hypothesis was based on that drugs designed to act against individual targets cannot usually battle multigenic diseases like cancer. Multi-target therapies, either in combination or sequential order, have been recommended to combat acquired and intrinsic resistance to anti-cancer treatments. Several studies focused on multi-targeting treatments due to their advantages include; overcoming clonal heterogeneity, lower risk of multi-drug resistance (MDR), decreased drug toxicity, and thereby lower side effects. In this study, we'll discuss about multi-target drugs, their benefits in improving cancer treatments, and recent advances in the field of multi-targeted drugs. Also, we will study the research that performed clinical trials using multi-target therapeutic agents for cancer treatment.
Collapse
Affiliation(s)
- Ali Doostmohammadi
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Hossein Jooya
- Biochemistry Group, Department of Chemistry, Faculty of Science, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Kimia Ghorbanian
- Student Research Committee, Semnan University of Medical Sciences, Semnan, Iran
| | - Sargol Gohari
- Department of Biology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Mehdi Dadashpour
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran.
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran.
| |
Collapse
|
44
|
Huang Y, Fan H, Ti H. Tumor microenvironment reprogramming by nanomedicine to enhance the effect of tumor immunotherapy. Asian J Pharm Sci 2024; 19:100902. [PMID: 38595331 PMCID: PMC11002556 DOI: 10.1016/j.ajps.2024.100902] [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: 08/28/2023] [Revised: 12/22/2023] [Accepted: 01/16/2024] [Indexed: 04/11/2024] Open
Abstract
With the rapid development of the fields of tumor biology and immunology, tumor immunotherapy has been used in clinical practice and has demonstrated significant therapeutic potential, particularly for treating tumors that do not respond to standard treatment options. Despite its advances, immunotherapy still has limitations, such as poor clinical response rates and differences in individual patient responses, largely because tumor tissues have strong immunosuppressive microenvironments. Many tumors have a tumor microenvironment (TME) that is characterized by hypoxia, low pH, and substantial numbers of immunosuppressive cells, and these are the main factors limiting the efficacy of antitumor immunotherapy. The TME is crucial to the occurrence, growth, and metastasis of tumors. Therefore, numerous studies have been devoted to improving the effects of immunotherapy by remodeling the TME. Effective regulation of the TME and reversal of immunosuppressive conditions are effective strategies for improving tumor immunotherapy. The use of multidrug combinations to improve the TME is an efficient way to enhance antitumor immune efficacy. However, the inability to effectively target drugs decreases therapeutic effects and causes toxic side effects. Nanodrug delivery carriers have the advantageous ability to enhance drug bioavailability and improve drug targeting. Importantly, they can also regulate the TME and deliver large or small therapeutic molecules to decrease the inhibitory effect of the TME on immune cells. Therefore, nanomedicine has great potential for reprogramming immunosuppressive microenvironments and represents a new immunotherapeutic strategy. Therefore, this article reviews strategies for improving the TME and summarizes research on synergistic nanomedicine approaches that enhance the efficacy of tumor immunotherapy.
Collapse
Affiliation(s)
- Yu Huang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Hui Fan
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Huihui Ti
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, China
- Guangdong Province Precise Medicine Big Date of Traditional Chinese Medicine Engineering Technology Research Center, Guangdong Pharmaceutical University, Guangzhou 510006, China
| |
Collapse
|
45
|
Wang J, Zhao W, Zhang Z, Liu X, Xie T, Wang L, Xue Y, Zhang Y. A Journey of Challenges and Victories: A Bibliometric Worldview of Nanomedicine since the 21st Century. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2308915. [PMID: 38229552 DOI: 10.1002/adma.202308915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/18/2023] [Indexed: 01/18/2024]
Abstract
Nanotechnology profoundly affects the advancement of medicine. Limitations in diagnosing and treating cancer and chronic diseases promote the growth of nanomedicine. However, there are very few analytical and descriptive studies regarding the trajectory of nanomedicine, key research powers, present research landscape, focal investigative points, and future outlooks. Herein, articles and reviews published in the Science Citation Index Expanded of Web of Science Core Collection from first January 2000 to 18th July 2023 are analyzed. Herein, a bibliometric visualization of publication trends, countries/regions, institutions, journals, research categories, themes, references, and keywords is produced and elaborated. Nanomedicine-related academic output is increasing since the COVID-19 pandemic, solidifying the uneven global distribution of research performance. While China leads in terms of publication quantity and has numerous highly productive institutions, the USA has advantages in academic impact, commercialization, and industrial value. Nanomedicine integrates with other disciplines, establishing interdisciplinary platforms, in which drug delivery and nanoparticles remain focal points. Current research focuses on integrating nanomedicine and cell ferroptosis induction in cancer immunotherapy. The keyword "burst testing" identifies promising research directions, including immunogenic cell death, chemodynamic therapy, tumor microenvironment, immunotherapy, and extracellular vesicles. The prospects, major challenges, and barriers to addressing these directions are discussed.
Collapse
Affiliation(s)
- Jingyu Wang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| | - Wenling Zhao
- Beijing National Laboratory for Molecular Sciences, CAS Laboratory of Colloid and Interface and Thermodynamics CAS Research/Education Center for Excellence in Molecular Sciences, Center for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Zhao Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| | - Xingzi Liu
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| | - Tong Xie
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| | - Lan Wang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| | - Yuzhou Xue
- Department of Cardiology, Institute of Vascular Medicine, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, State Key Laboratory of Vascular Homeostasis and Remodeling Peking University, Beijing Key Laboratory of Cardiovascular Receptors Research, Peking University Third Hospital, Beijing, 100191, China
| | - Yuemiao Zhang
- Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Key Laboratory of Renal Disease, Ministry of Health of China, Key Laboratory of Chronic Kidney Disease Prevention and Treatment (Peking University), Ministry of Education, Beijing, 100034, China
| |
Collapse
|
46
|
Jamali S, Jamali B, Abedi F, Firoozrai M, Davaran S, Vaghefi Moghaddam S. Folate receptor-mediated delivery system based on chitosan coated polymeric nanoparticles for combination therapy of breast cancer. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:605-627. [PMID: 38271010 DOI: 10.1080/09205063.2024.2303196] [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: 10/18/2023] [Accepted: 01/04/2024] [Indexed: 01/27/2024]
Abstract
Combination therapy using two or more drugs with different mechanisms of action is an effective strategy for treating cancer. This is because of the synergistic effect of complementary drugs that enhances their effectiveness. However, this approach has some limitations, such as non-specific distribution of the drugs in the tumor and the occurrence of dose-dependent toxicity to healthy tissues. To overcome these issues, we have developed a folate receptor-mediated co-delivery system that improves the access of chemotherapy drugs to the tumor site. We prepared a nanoplatform by encapsulating paclitaxel (PTX) and curcumin (CUR) in poly(caprolactone)-poly(ethylene glycol)-poly(caprolactone) (PCL-PEG-PCL) co-polymer using a double emulsion method and coating nanoparticles with pH-responsive chitosan-folic acid (CS-FA) conjugate. The nanocarrier's physicochemical properties were studied, confirming successful preparation with appropriate size and morphology. PTX and CUR could be released synchronously in a controlled and acid-facilitated manner. The dual drug-loaded nanocarrier exhibited excellent anti-tumor efficiency in MDA-MB-231 cells in vitro. The active targeting effect of FA concluded from the high inhibitory effect of dual drug-loaded nanocarrier on MDA-MB-231 cells, which have overexpressed folate receptors on their surface, compared to Human umbilical vein endothelial cells (HUVEC). Overall, the nanoengineered folate receptor-mediated co-delivery system provides great potential for safe and effective cancer therapy.
Collapse
Affiliation(s)
- Sajjad Jamali
- Department of Clinical Biochemistry, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | - Behzad Jamali
- Department of Clinical Biochemistry, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | - Fatemeh Abedi
- Clinical Research Development, Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohsen Firoozrai
- Department of Clinical Biochemistry, Shahrood Branch, Islamic Azad University, Shahrood, Iran
| | - Soodabeh Davaran
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Medicinal Chemistry, Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| | - Sevil Vaghefi Moghaddam
- Clinical Research Development, Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
47
|
Liu M, Srivastava G, Ramanujam J, Brylinski M. SynerGNet: A Graph Neural Network Model to Predict Anticancer Drug Synergy. Biomolecules 2024; 14:253. [PMID: 38540674 PMCID: PMC10967862 DOI: 10.3390/biom14030253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 02/16/2024] [Accepted: 02/19/2024] [Indexed: 01/03/2025] Open
Abstract
Drug combination therapy shows promise in cancer treatment by addressing drug resistance, reducing toxicity, and enhancing therapeutic efficacy. However, the intricate and dynamic nature of biological systems makes identifying potential synergistic drugs a costly and time-consuming endeavor. To facilitate the development of combination therapy, techniques employing artificial intelligence have emerged as a transformative solution, providing a sophisticated avenue for advancing existing therapeutic approaches. In this study, we developed SynerGNet, a graph neural network model designed to accurately predict the synergistic effect of drug pairs against cancer cell lines. SynerGNet utilizes cancer-specific featured graphs created by integrating heterogeneous biological features into the human protein-protein interaction network, followed by a reduction process to enhance topological diversity. Leveraging synergy data provided by AZ-DREAM Challenges, the model yields a balanced accuracy of 0.68, significantly outperforming traditional machine learning. Encouragingly, augmenting the training data with carefully constructed synthetic instances improved the balanced accuracy of SynerGNet to 0.73. Finally, the results of an independent validation conducted against DrugCombDB demonstrated that it exhibits a strong performance when applied to unseen data. SynerGNet shows a great potential in detecting drug synergy, positioning itself as a valuable tool that could contribute to the advancement of combination therapy for cancer treatment.
Collapse
Affiliation(s)
- Mengmeng Liu
- Division of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA; (M.L.)
| | - Gopal Srivastava
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - J. Ramanujam
- Division of Electrical and Computer Engineering, Louisiana State University, Baton Rouge, LA 70803, USA; (M.L.)
- Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Michal Brylinski
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
- Center for Computation and Technology, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
48
|
Bertão AR, Teixeira F, Ivasiv V, Parpot P, Almeida-Aguiar C, Fonseca AM, Bañobre-López M, Baltazar F, Neves IC. Machine Learning-Assisted Optimization of Drug Combinations in Zeolite-Based Delivery Systems for Melanoma Therapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:5696-5707. [PMID: 38271191 PMCID: PMC10859889 DOI: 10.1021/acsami.3c18224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/27/2024]
Abstract
Two independent artificial neural network (ANN) models were used to determine the optimal drug combination of zeolite-based delivery systems (ZDS) for cancer therapy. The systems were based on the NaY zeolite using silver (Ag+) and 5-fluorouracil (5-FU) as antimicrobial and antineoplastic agents. Different ZDS samples were prepared, and their characterization indicates the successful incorporation of both pharmacologically active species without any relevant changes to the zeolite structure. Silver acts as a counterion of the negative framework, and 5-FU retains its molecular integrity. The data from the A375 cell viability assays, involving ZDS samples (solid phase), 5-FU, and Ag+ aqueous solutions (liquid phase), were used to train two independent machine learning (ML) models. Both models exhibited a high level of accuracy in predicting the experimental cell viability results, allowing the development of a novel protocol for virtual cell viability assays. The findings suggest that the incorporation of both Ag and 5-FU into the zeolite structure significantly potentiates their anticancer activity when compared to that of the liquid phase. Additionally, two optimal AgY/5-FU@Y ratios were proposed to achieve the best cell viability outcomes. The ZDS also exhibited significant efficacy against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus); the predicted combination ratio is also effective against S. aureus, underscoring the potential of this approach as a therapeutic option for cancer-associated bacterial infections.
Collapse
Affiliation(s)
- Ana Raquel Bertão
- CQUM,
Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- Life
and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s
- PT Government Associate Laboratory, University
of Minho, 4710-057 Braga/Guimarães, Portugal
- Advanced
(Magnetic) Theranostic Nanostructures Lab, Nanomedicine Group, International
Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| | - Filipe Teixeira
- CQUM,
Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Viktoriya Ivasiv
- CQUM,
Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Pier Parpot
- CQUM,
Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- CEB
- Centre of Biological Engineering, University
of Minho, 4710-057 Braga, Portugal
| | - Cristina Almeida-Aguiar
- CBMA - Centre
of Molecular and Environmental Biology, University of Minho, 4710-057 Braga, Portugal
| | - António M. Fonseca
- CQUM,
Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- CEB
- Centre of Biological Engineering, University
of Minho, 4710-057 Braga, Portugal
| | - Manuel Bañobre-López
- Advanced
(Magnetic) Theranostic Nanostructures Lab, Nanomedicine Group, International
Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga, 4715-330 Braga, Portugal
| | - Fátima Baltazar
- Life
and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s
- PT Government Associate Laboratory, University
of Minho, 4710-057 Braga/Guimarães, Portugal
| | - Isabel C. Neves
- CQUM,
Centre of Chemistry, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- CEB
- Centre of Biological Engineering, University
of Minho, 4710-057 Braga, Portugal
| |
Collapse
|
49
|
Ahmed T, Liu FCF, Wu XY. An update on strategies for optimizing polymer-lipid hybrid nanoparticle-mediated drug delivery: exploiting transformability and bioactivity of PLN and harnessing intracellular lipid transport mechanism. Expert Opin Drug Deliv 2024; 21:245-278. [PMID: 38344771 DOI: 10.1080/17425247.2024.2318459] [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: 01/09/2023] [Accepted: 02/09/2024] [Indexed: 02/20/2024]
Abstract
INTRODUCTION Polymer-lipid hybrid nanoparticle (PLN) is an emerging nanoplatform with distinct properties and functionalities from other nanocarrier systems. PLN can be optimized to overcome various levels of drug delivery barriers to achieve desired therapeutic outcomes via rational selection of polymer and lipid combinations based on a thorough understanding of their properties and interactions with therapeutic agents and biological systems. AREAS COVERED This review provides an overview of PLN including the motive and history of PLN development, types of PLN, preparation methods, attestations of their versatility, and design strategies to circumvent various barriers for increasing drug delivery accuracy and efficiency. It also highlights recent advances in PLN design including: rationale selection of polymer and lipid components to achieve spatiotemporal drug targeting and multi-targeted cascade drug delivery; utilizing the intracellular lipid transport mechanism for active targeting to desired organelles; and harnessing bioreactive lipids and polymers to magnify therapeutic effects. EXPERT OPINION A thorough understanding of properties of PLN components and their biofate is important for enhancing disease site targeting, deep tumor tissue penetration, cellular uptake, and intracellular trafficking of PLN. For futuristic PLN development, active lipid transport and dual functions of lipids and polymers as both nanocarrier material and pharmacological agents can be further explored.
Collapse
Affiliation(s)
- Taksim Ahmed
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - Fuh-Ching Franky Liu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | | |
Collapse
|
50
|
Kim K, Park MH. Advancing Cancer Treatment: Enhanced Combination Therapy through Functionalized Porous Nanoparticles. Biomedicines 2024; 12:326. [PMID: 38397928 PMCID: PMC10887220 DOI: 10.3390/biomedicines12020326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 01/25/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Cancer remains a major global health challenge, necessitating the development of innovative treatment strategies. This review focuses on the functionalization of porous nanoparticles for combination therapy, a promising approach to enhance cancer treatment efficacy while mitigating the limitations associated with conventional methods. Combination therapy, integrating multiple treatment modalities such as chemotherapy, phototherapy, immunotherapy, and others, has emerged as an effective strategy to address the shortcomings of individual treatments. The unique properties of mesoporous silica nanoparticles (MSN) and other porous materials, like nanoparticles coated with mesoporous silica (NP@MS), metal-organic frameworks (MOF), mesoporous platinum nanoparticles (mesoPt), and carbon dots (CDs), are being explored for drug solubility, bioavailability, targeted delivery, and controlled drug release. Recent advancements in the functionalization of mesoporous nanoparticles with ligands, biomaterials, and polymers are reviewed here, highlighting their role in enhancing the efficacy of combination therapy. Various research has demonstrated the effectiveness of these nanoparticles in co-delivering drugs and photosensitizers, achieving targeted delivery, and responding to multiple stimuli for controlled drug release. This review introduces the synthesis and functionalization methods of these porous nanoparticles, along with their applications in combination therapy.
Collapse
Affiliation(s)
- Kibeom Kim
- Convergence Research Center, Nanobiomaterials Institute, Sahmyook University, Seoul 01795, Republic of Korea;
| | - Myoung-Hwan Park
- Convergence Research Center, Nanobiomaterials Institute, Sahmyook University, Seoul 01795, Republic of Korea;
- Department of Chemistry and Life Science, Sahmyook University, Seoul 01795, Republic of Korea
- Department of Convergence Science, Sahmyook University, Seoul 01795, Republic of Korea
| |
Collapse
|