1
|
Umar AK, Limpikirati PK, Rivai B, Ardiansah I, Sriwidodo S, Luckanagul JA. Complexed hyaluronic acid-based nanoparticles in cancer therapy and diagnosis: Research trends by natural language processing. Heliyon 2025; 11:e41246. [PMID: 39811313 PMCID: PMC11729671 DOI: 10.1016/j.heliyon.2024.e41246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 12/10/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Hyaluronic acid (HA) is a popular surface modifier in targeted cancer delivery due to its receptor-binding abilities. However, HA alone faces limitations in lipid solubility, biocompatibility, and cell internalization, making it less effective as a standalone delivery system. This comprehensive study aimed to explore a dynamic landscape of complexation in HA-based nanoparticles in cancer therapy, examining diverse aspects from influential modifiers to emerging trends in cancer diagnostics. We discovered that certain active substances, such as 5-aminolevulinic acid, adamantane, and protamine, have been on trend in terms of their usage over the past decade. Dextran, streptavidin, and catechol emerge as intriguing conjugates for HA, coupled with nanostar, quantum dots, and nanoprobe structures for optimal drug delivery and diagnostics. Strategies like hypoxic conditioning, dual responsiveness, and pulse laser activation enhance controlled release, targeted delivery, and real-time diagnostic techniques like ultrasound imaging and X-ray computed tomography (X-ray CT). Based on our findings, conventional bibliometric tools fail to highlight relevant topics in this area, instead producing merely abstract and broad-meaning keywords. Extraction using Named Entity Recognition and topic search with Latent Dirichlet Allocation successfully revealed five representative topics with the ability to exclude irrelevant keywords. A shift in research focuses from optimizing chemical toxicity to particular targeting tactics and precise release mechanisms is evident. These findings reflect the dynamic landscape of HA-based nanoparticle research in cancer therapy, emphasizing advancements in targeted drug delivery, therapeutic efficacy, and multimodal diagnostic approaches to improve overall patient outcomes.
Collapse
Affiliation(s)
- Abd Kakhar Umar
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Medical Informatics Laboratory, ETFLIN, Palu City, 94225, Indonesia
| | - Patanachai K. Limpikirati
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Food and Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Metabolomics for Life Sciences Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Bachtiar Rivai
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Medical Informatics Laboratory, ETFLIN, Palu City, 94225, Indonesia
| | - Ilham Ardiansah
- Department of Animal Husbandry, Faculty Veterinary Science, Chulalongkorn University, Bangkok, 10330, Thailand
- Medical Informatics Laboratory, ETFLIN, Palu City, 94225, Indonesia
| | - Sriwidodo Sriwidodo
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Jittima Amie Luckanagul
- Pharmaceutical Sciences and Technology Program, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
- Center of Excellence in Plant-produced Pharmaceuticals, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
2
|
Zhang X, Gao X, Xu J, Zhang Z, Lin T, Zhang X, Kang X. The role of lncRNA and miRNA on the effects of occurrence and development of osteosarcoma. Int Immunopharmacol 2025; 144:113726. [PMID: 39615111 DOI: 10.1016/j.intimp.2024.113726] [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: 10/10/2024] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 12/15/2024]
Abstract
Osteosarcoma is a common primary malignant bone tumor with a high incidence in children and adolescents, with high invasiveness and lung metastases. Even after traditional surgical excision, chemoradiotherapy, and comprehensive treatment, the survival rate of patients is still low, and the prognosis is not ideal. As an important part of non-coding RNA family, lncRNA and miRNA have significant regulatory effects on the growth, proliferation, metastasis and apoptosis of osteosarcoma cells. Therefore, exploring the roles of lncRNAs and miRNAs in the occurrence and development of osteosarcoma is of great help for the subsequent diagnosis, treatment, and prognosis of osteosarcoma. This paper mainly reviews the current research progress on the effects and mechanisms of lncRNAs and miRNAs on osteosarcoma cells, in order to provide new ideas for future research on the development process, treatment methods, and prognosis of osteosarcoma.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiao Tong University, 710054 Xi'an, Shaanxi Province, China
| | - Xidan Gao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiao Tong University, 710054 Xi'an, Shaanxi Province, China
| | - Jing Xu
- The Second Clinical Medical College of Lanzhou University, 730000 Lanzhou, Gansu, China
| | - Zhuoya Zhang
- The First Clinical Medical College of Lanzhou University, 730000 Lanzhou, Gansu, China
| | - Tingtong Lin
- The Second Clinical Medical College of Lanzhou University, 730000 Lanzhou, Gansu, China
| | - Xueyan Zhang
- Institute of Biochemistry and Molecular Biology and School of Basic Medical Sciences, Lanzhou University, 730000 Lanzhou, Gansu, China
| | - Xin Kang
- Department of Sports Medicine, Honghui Hospital, Xi'an Jiao Tong University, 710054 Xi'an, Shaanxi Province, China.
| |
Collapse
|
3
|
Wang Y, Lin M, Fan T, Zhou M, Yin R, Wang X. Advances of Stimuli-Responsive Amphiphilic Copolymer Micelles in Tumor Therapy. Int J Nanomedicine 2025; 20:1-24. [PMID: 39776491 PMCID: PMC11700880 DOI: 10.2147/ijn.s495387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Amphiphilic copolymers are composed of both hydrophilic and hydrophobic chains, which can self-assemble into polymeric micelles in aqueous solution via the hydrophilic/hydrophobic interactions. Due to their unique properties, polymeric micelles have been widely used as drug carriers. Poorly soluble drugs can be covalently attached to polymer chains or non-covalently incorporated in the micelles, with improved pharmacokinetic profiles and enhanced efficacy. In recent years, stimuli-responsive amphiphilic copolymer micelles have attracted significant attention. These micelles can respond to specific stimuli, including physical triggers (light, temperature, etc). chemical stimuli (pH, redox, etc). and physiological factors (enzymes, ATP, etc). Under these stimuli, the structures or properties of the micelles can change, enabling targeted therapy and controlled drug release in tumors. These stimuli-responsive strategies offer new avenues and approaches to enhance the tumor efficacy and reduce drug side effects. We will review the applications of different types of stimuli-responsive amphiphilic copolymer micelles in tumor therapy, aiming to provide valuable guidance for future research directions and clinical translation.
Collapse
Affiliation(s)
- Yao Wang
- Department of pharmacy, west china hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Meng Lin
- Department of pharmacy, west china hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Tianfei Fan
- Department of pharmacy, west china hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Minglu Zhou
- Department of pharmacy, west china hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Ruxi Yin
- Department of pharmacy, west china hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xueyan Wang
- Department of pharmacy, west china hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
4
|
Raveendran RL, Lekshmi GS, Anirudhan TS. Self-assembled sustainable bionanocomposite hydrogels from chitosan for the combination chemotherapy of hydrophobic and hydrophilic drugs. Int J Biol Macromol 2024; 283:137881. [PMID: 39571842 DOI: 10.1016/j.ijbiomac.2024.137881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 11/17/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024]
Abstract
Self-assembled hydrogels derived from naturally sourced polymers have gained significant interest in drug delivery applications, owing to their potential, exceptional biocompatibility and sustainable properties. This work presents the development and application of self-assembled nanocomposite hydrogels from chitosan and nanosilver as a pH responsive drug delivery system for the controlled release of doxorubicin and paclitaxel in anticancer therapy. Chitosan was functionalized with 4-formyl benzoic acid for incorporating both hydrophobic and hydrophilic anticancer drugs. The self-assembled nanocomposite hydrogels formed from chitosan and 4-formyl benzoic acid by various non-covalent interactions were studied by FT-IR, Dynamic Light Scattering (DLS), and rheology analysis. Rheology studies demonstrated the hydrogel's shear-thinning nature, enabling easy injection. The antibacterial activity can be evidenced by agar-well diffusion assay and MIC values were measured. The antibacterial effect was analyzed by agar-well diffusion assays and H2-DCFDA assay, providing a comprehensive understanding. In-vivo pharmacokinetic studies on Wistar rats demonstrated promising and effective systemic circulation of drug loaded material in blood, thus supporting its potential for therapeutic applications. All these studies and results demonstrates feasibility and a novel synergistic dual drug delivery approach, promising the synergy between hydrophobic paclitaxel (PTX) and hydrophilic Doxorubicin hydrochloride (Dox.HCl), for improved anticancer efficacy.
Collapse
Affiliation(s)
- Reshma L Raveendran
- Department of Chemistry, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India.
| | - G S Lekshmi
- Department of Chemistry, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India
| | - T S Anirudhan
- Department of Chemistry, University of Kerala, Kariavattom, Thiruvananthapuram 695581, Kerala, India.
| |
Collapse
|
5
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
6
|
Xie XK, Zhao XJ, Li RH, Cheng YR, Bing X, Yang J, Wang LY, Zhu HJ, Chen TH, Chen JF. Use of Period Analysis to Timely Assess Five-Year Relative Survival for the Patients With Bone Cancer. World J Oncol 2024; 15:675-681. [PMID: 38993259 PMCID: PMC11236380 DOI: 10.14740/wjon1875] [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: 03/25/2024] [Accepted: 05/13/2024] [Indexed: 07/13/2024] Open
Abstract
Background While timely assessment of long-term survival for patients with bone cancer is essential for evaluation on early detection and prognosis level of treatment of bone cancer, those data are extremely scarce in China. We aimed to timely and accurately assess long-term survival for patients with bone cancer in Eastern China. Methods Patients diagnosed with bone cancer during 2004 - 2018 from four cancer registries with high-quality data from Taizhou, Eastern China were included. Five-year relative survival (RS) of bone cancer patients was calculated by period analysis for overall and the stratification. We further predicted 5-year RS during upcoming 2019 - 2023 using a model-based period analysis and survival data during 2004 - 2018. Results Overall, 5-year RS for patients with bone cancer during 2014 - 2018 reached 46.6%, being 40.8% for male and 51.0% for female. Five-year RS declined along with aging, decreasing from 58.9% for age < 45 years to 41.5% for age > 60 years, while 5-year RS for urban area was higher compared to rural area (59.1% vs. 44.3%). The 5-year RS during upcoming 2019 - 2023 reached 48.3%. We found a clear upward trend in 5-year RS during 2004 - 2023 for overall and the stratification by sex, age at diagnosis, and region. Conclusions We found that, for first time in China using period analysis, most up-to-date 5-year RS for patients with bone cancer reached 46.6% during 2014 - 2018, and is projected to reach 48.3% for the period 2019 - 2023, which has important implications for timely evaluation on early detection and prognosis level of treatment for patients with bone cancer in Eastern China.
Collapse
Affiliation(s)
- Xian Kuan Xie
- Department of Orthopedics, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310053, China
- These authors contribute equally to this work
| | - Xiao Jiao Zhao
- School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China
- These authors contribute equally to this work
| | - Run Hua Li
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China
| | - Yong Ran Cheng
- School of Public Health, Hangzhou Medical College, Hangzhou 311399, China
| | - Xin Bing
- School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Jun Yang
- School of Public Health, Hangzhou Normal University, Hangzhou 311121, China
| | - Liang You Wang
- Department of Non-Communicable Chronic Disease Control and Prevention, Taizhou Municipal Center for Disease Control and Prevention, Taizhou 318000, Zhejiang Province, China
| | - Hui Jun Zhu
- Cadre Health Center of Taizhou City, Taizhou 318000, Zhejiang Province, China
| | - Tian Hui Chen
- Department of Cancer Prevention, Zhejiang Cancer Hospital, Hangzhou 310022, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China
| | - Jin Fei Chen
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| |
Collapse
|
7
|
Sun H, Li X, Liu Q, Sheng H, Zhu L. pH-responsive self-assembled nanoparticles for tumor-targeted drug delivery. J Drug Target 2024; 32:672-706. [PMID: 38682299 DOI: 10.1080/1061186x.2024.2349124] [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: 10/07/2023] [Accepted: 04/23/2024] [Indexed: 05/01/2024]
Abstract
Recent advances in the field of drug delivery have opened new avenues for the development of novel nanodrug delivery systems (NDDS) in cancer therapy. Self-assembled nanoparticles (SANPs) based on tumour microenvironment have great advantages in improving antitumor effect, and pH-responsive SANPs prepared by the combination of pH-responsive nanomaterials and self-assembly technology can effectively improve the efficacy and reduce the systemic toxicity of antitumor drugs. In this review, we describe the characteristics of self-assembly and its driving force, the mechanism of pH-responsive NDDS, and the nanomaterials for pH-responsive SANPs type. A series of pH-responsive SANPs for tumour-targeted drug delivery are discussed, with an emphasis on the relation between structural features and theranostic performance.
Collapse
Affiliation(s)
- Henglai Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xinyu Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qian Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
8
|
Bellini C, Mancin F, Papini E, Tavano R. Nanotechnological Approaches to Enhance the Potential of α-Lipoic Acid for Application in the Clinic. Antioxidants (Basel) 2024; 13:706. [PMID: 38929145 PMCID: PMC11201002 DOI: 10.3390/antiox13060706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
α-lipoic acid is a naturally occurring compound with potent antioxidant properties that helps protect cells and tissues from oxidative stress. Its incorporation into nanoplatforms can affect factors like bioavailability, stability, reactivity, and targeted delivery. Nanoformulations of α-lipoic acid can significantly enhance its solubility and absorption, making it more bioavailable. While α-lipoic acid can be prone to degradation in its free form, encapsulation within nanoparticles ensures its stability over time, and its release in a controlled and sustained manner to the targeted tissues and cells. In addition, α-lipoic acid can be combined with other compounds, such as other antioxidants, drugs, or nanomaterials, to create synergistic effects that enhance their overall therapeutic benefits or hinder their potential cytotoxicity. This review outlines the advantages and drawbacks associated with the use of α-lipoic acid, as well as various nanotechnological approaches employed to enhance its therapeutic effectiveness, whether alone or in combination with other bioactive agents. Furthermore, it describes the engineering of α-lipoic acid to produce poly(α-lipoic acid) nanoparticles, which hold promise as an effective drug delivery system.
Collapse
Affiliation(s)
- Chiara Bellini
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy; (C.B.); (E.P.)
| | - Fabrizio Mancin
- Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35121 Padova, Italy;
| | - Emanuele Papini
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy; (C.B.); (E.P.)
| | - Regina Tavano
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy; (C.B.); (E.P.)
| |
Collapse
|
9
|
Lee S, Kim HJ, Choi JH, Jang HJ, Cho HB, Kim HR, Park JI, Park KS, Park KH. Light emitting diode (LED) irradiation of liposomes enhances drug encapsulation and delivery for improved cancer eradication. J Control Release 2024; 368:756-767. [PMID: 38499090 DOI: 10.1016/j.jconrel.2024.03.027] [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: 12/19/2023] [Revised: 02/26/2024] [Accepted: 03/15/2024] [Indexed: 03/20/2024]
Abstract
Liposomes are widely used as drug delivery nanoplatforms because of their versatility and biocompatibility; however, their ability to load certain drugs may be suboptimal. In this study, we generated liposomes using a combination of DSPE and DSPE-PEG-2 k lipids and loaded them with doxorubicin (DOX) and paclitaxel (PTX), to investigate the effects of light emitting diode (LED) irradiation on liposome structure and drug loading efficiency. Scanning and transmission electron microscopy revealed that the surface of liposomes irradiated with blue or near-infrared LEDs (LsLipo) was rougher and more irregular than that of non-LED-irradiated liposomes (NsLipo). Nuclear magnetic resonance analysis showed that the hydrogen peak originating from the lipid head groups was lower in LsLipo than in NsLipo preparations, indicating that LED irradiation changed the chemical and physical properties of the liposome. Structural changes, such as reduced rigidity, induced by LED irradiation, increased the loading efficiency of DOX and PTX. In vitro and in vivo experiments showed that LsLipo were more effective at inhibiting the growth of cancer cells than NsLipo. Our findings suggest that LED irradiation enhances the drug delivery efficacy of liposomes and offer new possibilities for improving drug delivery systems.
Collapse
Affiliation(s)
- Sujeong Lee
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Hye Jin Kim
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Jin-Ho Choi
- Department of Biomedical Science, CHA University, Seongnam, Republic of Korea
| | - Hye Jung Jang
- Department of Biomedical Science, CHA University, Seongnam, Republic of Korea
| | - Hui Bang Cho
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Hye-Ryoung Kim
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Ji-In Park
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea
| | - Kyung-Soon Park
- Department of Biomedical Science, CHA University, Seongnam, Republic of Korea.
| | - Keun-Hong Park
- Laboratory of Nano-regenerative Medicine, Department of Biomedical Science, College of Life Science, CHA University, CHA Biocomplex, Sampyeong-Dong, Bundang-gu, Seongnam-si 13488, Republic of Korea.
| |
Collapse
|
10
|
Almeida-Ferreira C, Marto CM, Carmo C, Almeida-Ferreira J, Frutuoso C, Carvalho MJ, Botelho MF, Laranjo M. Efficacy of Cold Atmospheric Plasma vs. Chemotherapy in Triple-Negative Breast Cancer: A Systematic Review. Int J Mol Sci 2024; 25:3254. [PMID: 38542225 PMCID: PMC10970295 DOI: 10.3390/ijms25063254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/05/2024] [Accepted: 03/06/2024] [Indexed: 04/09/2024] Open
Abstract
Breast cancer is a growing disease, with a high worldwide incidence and mortality rate among women. Among the various types, the treatment of triple-negative breast cancer (TNBC) remains a challenge. Considering the recent advances in cold atmospheric plasma (CAP) cancer research, our goal was to evaluate efficacy data from studies based on chemotherapy and CAP in TNBC cell lines and animal models. A search of the literature was carried out in the PubMed, Web of Science, Cochrane Library, and Embase databases. Of the 10,999 studies, there were fifty-four in vitro studies, three in vivo studies, and two in vitro and in vivo studies included. MDA-MB-231 cells were the most used. MTT, MTS, SRB, annexin-V/propidium iodide, trypan blue, and clonogenic assay were performed to assess efficacy in vitro, increasing the reliability and comprehensiveness of the data. There was found to be a decrease in cell proliferation after both chemotherapy and CAP; however, different protocol settings, including an extensive range of drug doses and CAP exposure times, were reported. For both therapies, a considerable reduction in tumor volume was observed in vivo compared with that of the untreated group. The treatment of TNBC cell lines with CAP proved successful, with apoptosis emerging as the predominant type of cellular death. This systematic review presents a comprehensive overview of the treatment landscape in chemotherapy and CAP regarding their efficacy in TNBC cell lines.
Collapse
Affiliation(s)
- Catarina Almeida-Ferreira
- Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (C.A.-F.); (C.M.M.); (C.C.); (C.F.); (M.J.C.); (M.F.B.)
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Carlos Miguel Marto
- Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (C.A.-F.); (C.M.M.); (C.C.); (C.F.); (M.J.C.); (M.F.B.)
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, 3000-354 Coimbra, Portugal
- Institute of Integrated Clinical Practice, Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
- Laboratory for Evidence-Based Sciences and Precision Dentistry, Faculty of Medicine, University of Coimbra, 3000-075 Coimbra, Portugal
- Institute of Experimental Pathology, Faculty of Medicine, University of Coimbra, 3000-354 Coimbra, Portugal
| | - Chrislaura Carmo
- Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (C.A.-F.); (C.M.M.); (C.C.); (C.F.); (M.J.C.); (M.F.B.)
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Coimbra Chemistry Center (CQC), Department of Chemistry, Faculty of Sciences and Technology, University of Coimbra, 3004-535 Coimbra, Portugal
| | | | - Cristina Frutuoso
- Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (C.A.-F.); (C.M.M.); (C.C.); (C.F.); (M.J.C.); (M.F.B.)
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Gynecology Service, Coimbra Hospital and University Centre, Coimbra Health Local Unit, 3004-561 Coimbra, Portugal
| | - Maria João Carvalho
- Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (C.A.-F.); (C.M.M.); (C.C.); (C.F.); (M.J.C.); (M.F.B.)
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal
- Gynecology Service, Coimbra Hospital and University Centre, Coimbra Health Local Unit, 3004-561 Coimbra, Portugal
- Universitary Clinic of Gynecology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Maria Filomena Botelho
- Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (C.A.-F.); (C.M.M.); (C.C.); (C.F.); (M.J.C.); (M.F.B.)
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, 3000-354 Coimbra, Portugal
| | - Mafalda Laranjo
- Institute for Clinical and Biomedical Research (iCBR), Area of Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (C.A.-F.); (C.M.M.); (C.C.); (C.F.); (M.J.C.); (M.F.B.)
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB), 3000-548 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), University of Coimbra, 3000-354 Coimbra, Portugal
| |
Collapse
|
11
|
Barba-Rosado LV, Carrascal-Hernández DC, Insuasty D, Grande-Tovar CD. Graphene Oxide (GO) for the Treatment of Bone Cancer: A Systematic Review and Bibliometric Analysis. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:186. [PMID: 38251150 PMCID: PMC10820493 DOI: 10.3390/nano14020186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024]
Abstract
Cancer is a severe disease that, in 2022, caused more than 9.89 million deaths worldwide. One worrisome type of cancer is bone cancer, such as osteosarcoma and Ewing tumors, which occur more frequently in infants. This study shows an active interest in the use of graphene oxide and its derivatives in therapy against bone cancer. We present a systematic review analyzing the current state of the art related to the use of GO in treating osteosarcoma, through evaluating the existing literature. In this sense, studies focused on GO-based nanomaterials for potential applications against osteosarcoma were reviewed, which has revealed that there is an excellent trend toward the use of GO-based nanomaterials, based on their thermal and anti-cancer activities, for the treatment of osteosarcoma through various therapeutic approaches. However, more research is needed to develop highly efficient localized therapies. It is suggested, therefore, that photodynamic therapy, photothermal therapy, and the use of nanocarriers should be considered as non-invasive, more specific, and efficient alternatives in the treatment of osteosarcoma. These options present promising approaches to enhance the effectiveness of therapy while also seeking to reduce side effects and minimize the damage to surrounding healthy tissues. The bibliometric analysis of photothermal and photochemical treatments of graphene oxide and reduced graphene oxide from January 2004 to December 2022 extracted 948 documents with its search strategy, mainly related to research papers, review papers, and conference papers, demonstrating a high-impact field supported by the need for more selective and efficient bone cancer therapies. The central countries leading the research are the United States, Iran, Italy, Germany, China, South Korea, and Australia, with strong collaborations worldwide. At the same time, the most-cited papers were published in journals with impact factors of more than 6.0 (2021), with more than 290 citations. Additionally, the journals that published the most on the topic are high impact factor journals, according to the analysis performed, demonstrating the high impact of the research field.
Collapse
Affiliation(s)
- Lemy Vanessa Barba-Rosado
- Grupo de Investigación en Fotoquímica y Fotobiología, Programa de Química, Facultad de Ciencias Básicas, Universidad del Atlántico, Puerto Colombia 081008, Colombia; (L.V.B.-R.); (D.C.C.-H.)
| | - Domingo César Carrascal-Hernández
- Grupo de Investigación en Fotoquímica y Fotobiología, Programa de Química, Facultad de Ciencias Básicas, Universidad del Atlántico, Puerto Colombia 081008, Colombia; (L.V.B.-R.); (D.C.C.-H.)
- Departamento de Química y Biología, División de Ciencias Básicas, Universidad del Norte, Km 5 Vía Puerto Colombia, Barranquilla 081007, Colombia;
| | - Daniel Insuasty
- Departamento de Química y Biología, División de Ciencias Básicas, Universidad del Norte, Km 5 Vía Puerto Colombia, Barranquilla 081007, Colombia;
| | - Carlos David Grande-Tovar
- Grupo de Investigación en Fotoquímica y Fotobiología, Programa de Química, Facultad de Ciencias Básicas, Universidad del Atlántico, Puerto Colombia 081008, Colombia; (L.V.B.-R.); (D.C.C.-H.)
| |
Collapse
|
12
|
Wu K, Zhou Z, Liu T, Liu C, Mu X, Jiang J. Co-delivery of curcumin and si-STAT3 with a bioinspired tumor homing for polydopamine nanoparticles for synergistic osteosarcoma therapy. Cancer Nanotechnol 2023; 14:66. [DOI: 10.1186/s12645-023-00215-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 06/02/2023] [Indexed: 01/06/2025] Open
Abstract
Abstract
Purpose
Owing to the complexity of cancer, a synergistic combination of chemotherapy and gene therapy can be a promising therapeutic strategy. This study aimed to use stem cell membrane (SCM)-camouflaged polydopamine nanoparticles for simultaneous delivery of curcumin (CUR) and siRNA-targeting STAT3 (CPDA/siSTAT3@SCM NPs) for osteosarcoma (OS).
Methods
Transmission electron microscopy, UV–Vis absorbance spectra, zeta potential, cell co-localization, and Coomassie bright blue staining were used to characterize CPDA/siSTAT3@SCM NPs constructed by the self-assembly method. Drug release, cellular uptake, cell proliferation, apoptosis, wound healing, and transwell assays were evaluated in vitro. The expression levels of epithelial–mesenchymal transition (EMT)- and apoptosis-related proteins were measured by western blotting. Furthermore, the biodistribution, antitumor efficacy, and biosafety of CPDA/siSTAT3@SCM NPs in an MG63 xenograft mouse model were evaluated.
Results
CPDA/siSTAT3@SCM NPs were successfully synthesized to deliver CUR and siRNA simultaneously, and they showed osteosarcoma-targeting ability. Furthermore, it showed high cellular uptake and excellent synergistic antitumor effects in vitro. CPDA/siSTAT3@SCM NPs suppressed OS cell proliferation, migration, invasion, and EMT progression, and promoted the apoptotic process. In tumor-bearing mice, the treatment with CPDA/siSTAT3@SCM NPs showed an excellent antitumor effect with no side effects in major organs.
Conclusion
This study revealed that CPDA/siSTAT3@SCM NPs can target drug delivery by biomimetic multifunctional nanoparticles to treat OS through chemo-gene combined therapy.
Collapse
|
13
|
Yu T, Cai Z, Chang X, Xing C, White S, Guo X, Jin J. Research Progress of Nanomaterials in Chemotherapy of Osteosarcoma. Orthop Surg 2023; 15:2244-2259. [PMID: 37403654 PMCID: PMC10475694 DOI: 10.1111/os.13806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 05/05/2023] [Accepted: 05/30/2023] [Indexed: 07/06/2023] Open
Abstract
Osteosarcoma (OS) is a common malignant bone tumor that occurs mostly in children and adolescents. At present, surgery after chemotherapy or postoperative adjuvant chemotherapy is the main treatment plan. However, the efficacy of chemotherapeutic drugs is limited by the occurrence of chemotherapeutic resistance, toxicity to normal cells, poor pharmacokinetic performance, and drug delivery failure. The delivery of chemotherapy drugs to the bone to treat OS may fail for a variety of reasons, such as a lack of selectivity for OS cells, initial sudden release, short-term release, and the presence of biological barriers (such as the blood-bone marrow barrier). Nanomaterials are new materials with at least one dimension on the nanometer scale (1-100 nm) in three-dimensional space. These materials have the ability to penetrate biological barriers and can accumulate preferentially in tumor cells. Studies have shown that the effective combination of nanomaterials and traditional chemotherapy can significantly improve the therapeutic effect. Therefore, this article reviews the latest research progress on the use of nanomaterials in OS chemotherapy.
Collapse
Affiliation(s)
- Tianci Yu
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
| | - Zongyan Cai
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
| | - Xingyu Chang
- The First Clinical Medical CollegeLanzhou UniversityLanzhouChina
| | - Chengwei Xing
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
| | - Sylvia White
- Pathology DepartmentYale School of MedicineNew HavenCTUSA
| | - Xiaoxue Guo
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
| | - Jiaxin Jin
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouChina
- Orthopaedics Key Laboratory of Gansu ProvinceLanzhouChina
- Department of OrthopaedicsThe Second Hospital of Lanzhou UniversityLanzhouChina
| |
Collapse
|
14
|
Luo S, Lv Z, Yang Q, Chang R, Wu J. Research Progress on Stimulus-Responsive Polymer Nanocarriers for Cancer Treatment. Pharmaceutics 2023; 15:1928. [PMID: 37514114 PMCID: PMC10386740 DOI: 10.3390/pharmaceutics15071928] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/28/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
As drug carriers for cancer treatment, stimulus-responsive polymer nanomaterials are a major research focus. These nanocarriers respond to specific stimulus signals (e.g., pH, redox, hypoxia, enzymes, temperature, and light) to precisely control drug release, thereby improving drug uptake rates in cancer cells and reducing drug damage to normal cells. Therefore, we reviewed the research progress in the past 6 years and the mechanisms underpinning single and multiple stimulus-responsive polymer nanocarriers in tumour therapy. The advantages and disadvantages of various stimulus-responsive polymeric nanomaterials are summarised, and the future outlook is provided to provide a scientific and theoretical rationale for further research, development, and utilisation of stimulus-responsive nanocarriers.
Collapse
Affiliation(s)
- Shicui Luo
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Zhuo Lv
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Qiuqiong Yang
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Renjie Chang
- Center of Digestive Endoscopy, The First Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming 650021, China
| | - Junzi Wu
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming 650500, China
- Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming 650500, China
| |
Collapse
|
15
|
Cheng F, Huang QF, Li YH, Huang ZJ, Wu QX, Wang W, Liu Y, Wang GH. Combined chemo and photo therapy of programmable prodrug carriers to overcome delivery barriers against nasopharyngeal carcinoma. BIOMATERIALS ADVANCES 2023; 151:213451. [PMID: 37150081 DOI: 10.1016/j.bioadv.2023.213451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/13/2023] [Accepted: 04/25/2023] [Indexed: 05/09/2023]
Abstract
Indocyanine green (ICG) has been employed in medical diagnostics due to its superior photophysical characteristics. However, these advantages are offset by its quick body clearance and inferior photo-stability. In this work, programmable prodrug carriers for chemotherapy/PDT/PTT against nasopharyngeal carcinoma (NPC) were created in order to increase photo-stability and get around biochemical hurdles. The programmable prodrug carriers (PEG-PLA@DIT-PAMAM) that proactively penetrated deeply into NPC tumors and produced the deep phototherapy and selective drug release under laser irradiation was created by dendrimer-DOX/ICG/TPP (DIT-PAMAM) and PEGylated poly (α-lipoic acid) (PLA) copolymer. Long circulation times and minimal toxicity to mammalian cells are two benefits of PEG-coated carriers. The overexpressed GSH on the tumor cell or vascular endothelial cell of the NPC disintegrated the PEG-g-PLA chains and released the DIT-PAMAM nanoparticles after the carriers had reached the NPC tumor periphery. Small, positively charged DIT-PAMAM nanoparticles may penetrate tumors effectively and remain inside tumor for an extended period of time. In addition, the induced ROS cleaved the thioketal linkers for both DOX and nanoparticles and product hyperthermia (PTT) to kill cancer cells under laser irradiation, facilitating faster diffusion of nanoparticles and more effective tumor penetration with a programmable publication of DOX. The programmable prodrug carries showed high photo-stability high photo-stability, which enabled very effective PDT, PTT, and tumor-specific DOX release. With the goal of combining the effects of chemotherapy, PDT, and PTT against NPC, this research showed the great efficacy of programmable prodrug carriers.
Collapse
Affiliation(s)
- Fan Cheng
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Qun-Fa Huang
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Yan-Hong Li
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Zeng-Jin Huang
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Quan-Xin Wu
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China
| | - Wei Wang
- Scientific Research Service Center, Guangdong Medical University, Dongguan 523808, China
| | - Yun Liu
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China.
| | - Guan-Hai Wang
- School of Pharmacy, Guangdong Medical University, Dongguan 523808, China; Key Laboratory of Polymeric Composite and Functional Materials of Ministry of Education, Sun Yet-Sen University, Guangzhou 510275, China.
| |
Collapse
|
16
|
Rezaei N, Kazem Arki M, Miri-Lavasani Z, Solhi R, Khoramipour M, Rashedi H, Asadzadeh Aghdaei H, Hossein-Khannazer N, Mostafavi E, Vosough M. Co-delivery of Doxorubicin and Paclitaxel via Noisome Nanocarriers Attenuates Cancerous Phenotypes in Gastric Cancer Cells. Eur J Pharm Biopharm 2023:S0939-6411(23)00102-9. [PMID: 37105361 DOI: 10.1016/j.ejpb.2023.04.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/13/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023]
Abstract
Gastric cancer (GC) is known as a deadly malignancy all over the world, yet none of the current therapeutic regimens have achieved efficacy. this current study has aimed to optimize and reduce treatment doses and overcome multidrug resistance in GC by developing optimum niosomal formulation for the delivery of doxorubicin (DXR), paclitaxel (PTX), and their co-delivery. The particles' size, polydispersity index (PDI), and entrapment efficacy (EE%) were optimized using statistical techniques, i.e., Box-Behnken and Central Composite Design. In contrast to soluble drug formulations, the release rate of medicines from nanoparticles were higher in physiological and acidic pH. Niosomes were more stable at 4°C, compared to 25°C. The MTT assay revealed that the IC50 of drug-loaded niosomes was the lowest among all developed formulations. The apoptosis-related genes (CASPASE-3, CASPASE-8, and CASPASE-9) and tumor suppressor genes (BAX, BCL2) were evaluated in cancer cells before and after treatment. In comparison to control cells and cells treated with soluble forms of DXR and PTX, while the expression of BCL2 decreased, the expression of BAX, CASPASE-3, CASPASE-8, and CASPASE-9 was enhanced in cells treated with drug-loaded niosomes. Drug-loaded niosomes inhibited colony formation capacity and increased apoptosis in human AGS gastric cancer cells. Our results indicate that co-delivery of DXR and PTX-loaded niosomes may be an effective and innovative therapeutic approach to gastric cancer.
Collapse
Affiliation(s)
- Niloofar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mandana Kazem Arki
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohre Miri-Lavasani
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Solhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Khoramipour
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Rashedi
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Ebrahim Mostafavi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
17
|
Jiang Y, He K. Nanobiotechnological approaches in osteosarcoma therapy: Versatile (nano)platforms for theranostic applications. ENVIRONMENTAL RESEARCH 2023; 229:115939. [PMID: 37088317 DOI: 10.1016/j.envres.2023.115939] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/08/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
Constructive achievements in the field of nanobiotechnology and their translation into clinical course have led to increasing attention towards evaluation of their use for treatment of diseases, especially cancer. Osteosarcoma (OS) is one of the primary bone malignancies that affects both males and females in childhood and adolescence. Like other types of cancers, genetic and epigenetic mutations account for OS progression and several conventional therapies including chemotherapy and surgery are employed. However, survival rate of OS patients remains low and new therapies in this field are limited. The purpose of the current review is to provide a summary of nanostructures used in OS treatment. Drug and gene delivery by nanoplatforms have resulted in an accumulation of therapeutic agents for tumor cell suppression. Furthermore, co-delivery of genes and drugs by nanostructures are utilized in OS suppression to boost immunotherapy. Since tumor cells have distinct features such as acidic pH, stimuli-responsive nanoparticles have been developed to appropriately target OS. Besides, nanoplatforms can be used for biosensing and providing phototherapy to suppress OS. Furthermore, surface modification of nanoparticles with ligands can increase their specificity and selectivity towards OS cells. Clinical translation of current findings suggests that nanoplatforms have been effective in retarding tumor growth and improving survival of OS patients.
Collapse
Affiliation(s)
- Yao Jiang
- Department of Diagnostic and Interventional Radiology, University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt Am Main, Germany.
| | - Ke He
- Minimally Invasive Tumor Therapies Center, Guangdong Second Provincial General Hospital, Guangzhou, China.
| |
Collapse
|
18
|
Kovshova T, Mantrov S, Boiko S, Malinovskaya J, Merkulova M, Osipova N, Moiseeva N, Akimov M, Dudina P, Senchikhin I, Ermolenko Y, Gelperina S. Co-delivery of paclitaxel and etoposide prodrug by human serum albumin and PLGA nanoparticles: synergistic cytotoxicity in brain tumour cells. J Microencapsul 2023; 40:246-262. [PMID: 36880479 DOI: 10.1080/02652048.2023.2188943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2023]
Abstract
The aims of this study were to develop co-delivery systems of paclitaxel (PTX) and etoposide prodrug (4'-O-benzyloxycarbonyl-etoposide, ETP-cbz) based on non-cross-linked human serum albumin (HSA) and poly(lactide-co-glycolide) nanoparticles and to evaluate the synergistic potential of these drugs in vitro. The nanoformulations were prepared by the high-pressure homogenisation technique and characterised using DLS, TEM, SEM, AFM, HPLC, CZE, in-vitro release, and cytotoxicity in human and murine glioma cells. All nanoparticles had 90-150 nm in size and negative ζ-potentials. The Neuro2A cells were the most sensitive to both HSA- and PLGA-based co-delivery systems (IC50 0.024 µM and 0.053 µM, respectively). The drugs' synergistic effect (combination index < 0.9) was observed in the GL261 cells for both types of co-delivery formulations and in the Neuro2A cells for the HSA-based system. These nanodelivery systems may be useful to improve combination chemotherapy for brain tumour treatment. To our knowledge, this is the first report describing the non-cross-linked HSA-based co-delivery nanosuspension which was prepared using nab™ technology.
Collapse
Affiliation(s)
- Tatyana Kovshova
- D. Mendeleev University of Chemical Technology of Russia, Moscow, Russia
| | - Sergey Mantrov
- D. Mendeleev University of Chemical Technology of Russia, Moscow, Russia
| | - Svetlana Boiko
- D. Mendeleev University of Chemical Technology of Russia, Moscow, Russia
| | - Julia Malinovskaya
- D. Mendeleev University of Chemical Technology of Russia, Moscow, Russia
| | - Maria Merkulova
- D. Mendeleev University of Chemical Technology of Russia, Moscow, Russia
| | - Nadezhda Osipova
- D. Mendeleev University of Chemical Technology of Russia, Moscow, Russia
| | - Natalia Moiseeva
- N.N. Blokhin National Medical Research Center of Oncology, Moscow, Russia
| | - Mikhail Akimov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Polina Dudina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Ivan Senchikhin
- Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, Moscow, Russia
| | - Yulia Ermolenko
- D. Mendeleev University of Chemical Technology of Russia, Moscow, Russia
| | - Svetlana Gelperina
- D. Mendeleev University of Chemical Technology of Russia, Moscow, Russia
| |
Collapse
|
19
|
Tian H, Cao J, Li B, Nice EC, Mao H, Zhang Y, Huang C. Managing the immune microenvironment of osteosarcoma: the outlook for osteosarcoma treatment. Bone Res 2023; 11:11. [PMID: 36849442 PMCID: PMC9971189 DOI: 10.1038/s41413-023-00246-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 12/17/2022] [Accepted: 12/29/2022] [Indexed: 03/01/2023] Open
Abstract
Osteosarcoma, with poor survival after metastasis, is considered the most common primary bone cancer in adolescents. Notwithstanding the efforts of researchers, its five-year survival rate has only shown limited improvement, suggesting that existing therapeutic strategies are insufficient to meet clinical needs. Notably, immunotherapy has shown certain advantages over traditional tumor treatments in inhibiting metastasis. Therefore, managing the immune microenvironment in osteosarcoma can provide novel and valuable insight into the multifaceted mechanisms underlying the heterogeneity and progression of the disease. Additionally, given the advances in nanomedicine, there exist many advanced nanoplatforms for enhanced osteosarcoma immunotherapy with satisfactory physiochemical characteristics. Here, we review the classification, characteristics, and functions of the key components of the immune microenvironment in osteosarcoma. This review also emphasizes the application, progress, and prospects of osteosarcoma immunotherapy and discusses several nanomedicine-based options to enhance the efficiency of osteosarcoma treatment. Furthermore, we examine the disadvantages of standard treatments and present future perspectives for osteosarcoma immunotherapy.
Collapse
Affiliation(s)
- Hailong Tian
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Jiangjun Cao
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Bowen Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041 China
| | - Edouard C. Nice
- grid.1002.30000 0004 1936 7857Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800 Australia
| | - Haijiao Mao
- Department of Orthopaedic Surgery, The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, 315020, People's Republic of China.
| | - Yi Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Canhua Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
20
|
Liu C, Wu K, Li J, Mu X, Gao H, Xu X. Nanoparticle-mediated therapeutic management in cholangiocarcinoma drug targeting: Current progress and future prospects. Biomed Pharmacother 2023; 158:114135. [PMID: 36535198 DOI: 10.1016/j.biopha.2022.114135] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Patients with cholangiocarcinoma (CCA) often have an unfavorable prognosis because of its insidious nature, low resectability rate, and poor response to anticancer drugs and radiotherapy, which makes early detection and treatment difficult. At present, CCA has a five-year overall survival rate (OS) of only 5%, despite advances in therapies. New an increasing number of evidence suggests that nanoplatforms may play a crucial role in enhancing the pharmacological effects and in reducing both short- and long-term side effects of cancer treatment. This document reviews the advantages and shortcomings of nanoparticles such as liposomes, polymeric nanoparticle,inorganic nanoparticle, nano-metals and nano-alloys, carbon dots, nano-micelles, dendrimer, nano-capsule, bio-Nanomaterials in the diagnosis and treatment of CCA and discuss the current challenges in of nanoplatforms for CCA.
Collapse
Affiliation(s)
- Chunkang Liu
- Department of Gastrointestinal Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Kunzhe Wu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jianyang Li
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xupeng Mu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Huan Gao
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaohua Xu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
21
|
Liu X, Liu Y, Qiang L, Ren Y, Lin Y, Li H, Chen Q, Gao S, Yang X, Zhang C, Fan M, Zheng P, Li S, Wang J. Multifunctional 3D-printed bioceramic scaffolds: Recent strategies for osteosarcoma treatment. J Tissue Eng 2023; 14:20417314231170371. [PMID: 37205149 PMCID: PMC10186582 DOI: 10.1177/20417314231170371] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/31/2023] [Indexed: 05/21/2023] Open
Abstract
Osteosarcoma is the most prevalent bone malignant tumor in children and teenagers. The bone defect, recurrence, and metastasis after surgery severely affect the life quality of patients. Clinically, bone grafts are implanted. Primary bioceramic scaffolds show a monomodal osteogenesis function. With the advances in three-dimensional printing technology and materials science, while maintaining the osteogenesis ability, scaffolds become more patient-specific and obtain additional anti-tumor ability with functional agents being loaded. Anti-tumor therapies include photothermal, magnetothermal, old and novel chemo-, gas, and photodynamic therapy. These strategies kill tumors through novel mechanisms to treat refractory osteosarcoma due to drug resistance, and some have shown the potential to reverse drug resistance and inhibit metastasis. Therefore, multifunctional three-dimensional printed bioceramic scaffolds hold excellent promise for osteosarcoma treatments. To better understand, we review the background of osteosarcoma, primary 3D-printed bioceramic scaffolds, and different therapies and have a prospect for the future.
Collapse
Affiliation(s)
- Xingran Liu
- Shanghai Key Laboratory of Orthopedic
Implant, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of
Medicine, Shanghai, China
| | - Yihao Liu
- Shanghai Key Laboratory of Orthopedic
Implant, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of
Medicine, Shanghai, China
| | - Lei Qiang
- Southwest Jiaotong University, Chengdu,
China
| | - Ya Ren
- Southwest Jiaotong University, Chengdu,
China
| | - Yixuan Lin
- Shanghai Key Laboratory of Orthopedic
Implant, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han Li
- Shanghai Jiao Tong University School of
Medicine, Shanghai, China
| | - Qiuhan Chen
- Shanghai Jiao Tong University School of
Medicine, Shanghai, China
| | - Shuxin Gao
- Shanghai Jiao Tong University School of
Medicine, Shanghai, China
| | - Xue Yang
- Southwest Jiaotong University, Chengdu,
China
| | - Changru Zhang
- Shanghai Key Laboratory of Orthopedic
Implant, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of
Medicine, Shanghai, China
| | - Minjie Fan
- Department of Orthopaedic Surgery,
Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Pengfei Zheng
- Department of Orthopaedic Surgery,
Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Shuai Li
- Department of Orthopedics, The First
Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinwu Wang
- Shanghai Key Laboratory of Orthopedic
Implant, Department of Orthopedic Surgery, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of
Medicine, Shanghai, China
- Southwest Jiaotong University, Chengdu,
China
- Shanghai Jiao Tong University,
Shanghai, China
- Weifang Medical University School of
Rehabilitation Medicine, Weifang, Shandong Province, China
| |
Collapse
|
22
|
Li J, Li J, Wang H, Chen Y, Qin J, Zeng H, Wang K, Wang S. Microscopic Raman illustrating antitumor enhancement effects by the combination drugs of γ-secretase inhibitor and cisplatin on osteosarcoma cells. JOURNAL OF BIOPHOTONICS 2022; 15:e202200189. [PMID: 36057844 DOI: 10.1002/jbio.202200189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/02/2022] [Accepted: 08/31/2022] [Indexed: 06/15/2023]
Abstract
By using Raman microspectroscopy, it aims to elucidate the cellular variations caused by the combination drug of γ-secretase inhibitor (DAPT) and cisplatin in osteosarcoma (OS) cells. Illustrated by the obtained results of spectral analysis, the intracellular composition significantly changed after combined drug actions compared to the solo DAPT treatment, indicating the synergistic effect of DAPT combined with cisplatin on OS cells. Meanwhile, multivariate curve resolution-alternating least squares (MCR-ALS) algorithm was utilized to address the biochemical constitution changes in all investigated groups including the untreated (UT), DAPT (40D) and combined drug (40D + 20C) treated cells. K-means cluster and univariate imaging were both utilized to visualize the changes in subcellular morphology and biochemical distribution. The presented study provides a unique understanding on the cellular responses to DAPT combined with cisplatin from the natural biochemical perspectives, and laids an experimental foundation for exploring the therapeutic strategies of other combined anticancer drugs in cancer cell model.
Collapse
Affiliation(s)
- Jie Li
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
- School of Physics and Electronic Engineering, Xianyang Normal University, Xianyang, Shaanxi, China
| | - Jing Li
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haifeng Wang
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Yishen Chen
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Jie Qin
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Haishan Zeng
- Imaging Unit-Integrative Oncology Department, BC Cancer Research Center, Vancouver, Canada
| | - Kaige Wang
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| | - Shuang Wang
- Institute of Photonics and Photon-Technology, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
23
|
Lv SY, He S, Ling XL, Wang YQ, Huang C, Long JR, Wang JQ, Qin Y, Wei H, Yu CY. Review of lipoic acid: From a clinical therapeutic agent to various emerging biomaterials. Int J Pharm 2022; 627:122201. [PMID: 36115465 DOI: 10.1016/j.ijpharm.2022.122201] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/20/2022] [Accepted: 09/09/2022] [Indexed: 01/25/2023]
Abstract
Lipoic acid (LA), an endogenous small molecule in organisms, has been extensively used for the highly efficient clinical treatment of malignant diseases, which include diabetes, Alzheimer's disease, and cancer over the past seven decades. Tremendous progresses have been made on the use of LA in nanomedicine for the development of various biomaterials because of its unique biological properties and highly adaptable structure since the first discovery. However, there are few reviews thus far, to our knowledge, summarizing this hot subject of research of LA and its derived biomaterials. For this purpose, we present herein the first comprehensive summary on the design and development of LA and its derived materials for biomedical applications. This review first discusses the therapeutic use of LA followed by the description of synthesis and preclinical study of LA-derived-small molecules. The applications of various LA and poly (lipoic acid) (PLA)-derived-biomaterials are next summarized in detail with an emphasis on the use of LA for the design of biomaterials and the diverse properties. This review describes the development of LA from a clinical therapeutic agent to a building unit of various biomaterials field, which will promote the further discovery of new therapeutic uses of LA as therapeutic agents and facile development of LA-based derivates with greater performance for biomedical applications.
Collapse
Affiliation(s)
- Shao-Yang Lv
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Suisui He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Xiao-Li Ling
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yue-Qin Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Cong Huang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Jin-Rong Long
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Jia-Qi Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yang Qin
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pharmacy & Pharmacology, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China.
| |
Collapse
|
24
|
Chen Y, Wu X, Li J, Jiang Y, Xu K, Su J. Bone-Targeted Nanoparticle Drug Delivery System: An Emerging Strategy for Bone-Related Disease. Front Pharmacol 2022; 13:909408. [PMID: 35712701 PMCID: PMC9195145 DOI: 10.3389/fphar.2022.909408] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/27/2022] [Indexed: 12/28/2022] Open
Abstract
Targeted delivery by either systemic or local targeting of therapeutics to the bone is an attractive treatment for various bone metabolism diseases such as osteoporosis, osteoarthritis, osteosarcoma, osteomyelitis, etc. To overcome the limitations of direct drug delivery, the combination of bone-targeted agents with nanotechnology has the opportunity to provide a more effective therapeutic approach, where engineered nanoparticles cause the drug to accumulate in the bone, thereby improving efficacy and minimizing side effects. Here, we summarize the current advances in systemic or local bone-targeting approaches and nanosystem applications in bone diseases, which may provide new insights into nanocarrier-delivered drugs for the targeted treatment of bone diseases. We envision that novel drug delivery carriers developed based on nanotechnology will be a potential vehicle for the treatment of currently incurable bone diseases and are expected to be translated into clinical applications.
Collapse
Affiliation(s)
- Yulin Chen
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,School of Medicine, Shanghai University, Shanghai, China.,School of Life Sciences, Shanghai University, Shanghai, China
| | - Xianmin Wu
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, China
| | - Jiadong Li
- Institute of Translational Medicine, Shanghai University, Shanghai, China.,School of Medicine, Shanghai University, Shanghai, China.,School of Life Sciences, Shanghai University, Shanghai, China
| | - Yingying Jiang
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Ke Xu
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, China
| |
Collapse
|
25
|
Nechaeva AM, Artyukhov AA, Luss AL, Shtilman MI, Svistunova AY, Motyakin MV, Levina II, Krivoborodov EG, Toropygin I, Chistyakov EM, Tsatsakis AM, Gurevich L, Mezhuev YO. The Synthesis and Properties of a New Carrier for Paclitaxel and Doxorubicin Based on the Amphiphilic Copolymer of N‐vinyl‐2‐pyrrolidone and Acrylic Acid. MACROMOL CHEM PHYS 2022. [DOI: 10.1002/macp.202200081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2022]
Affiliation(s)
- Anna M. Nechaeva
- Mendeleev University of Chemical Technology of Russia Moscow 125047 Russia
| | | | - Anna L. Luss
- Mendeleev University of Chemical Technology of Russia Moscow 125047 Russia
| | | | | | - Mikhail V. Motyakin
- Emanuel Institute of Biochemical Physics Russian Academy of Sciences Moscow 119334 Russia
- Semenov Federal Research Center for Chemical Physics Russian Academy of Sciences Moscow 119991 Russia
| | - Irina I. Levina
- Emanuel Institute of Biochemical Physics Russian Academy of Sciences Moscow 119334 Russia
| | | | - IlyaYu Toropygin
- V.N. Orekhovich Research Institute of Biomedical Chemistry Russian Academy of Medical Sciences Moscow 119832 Russia
| | | | - Aristides M. Tsatsakis
- Center of Toxicology Science & Research Division of Morphology Medical School University of Crete Voutes Campus Heraklion Crete 71003 Greece
| | - Leonid Gurevich
- Department of Materials and Production Aalborg University Skjernvej 4A Aalborg 9220 Denmark
| | | |
Collapse
|
26
|
Diao W, Yang B, Sun S, Wang A, Kou R, Ge Q, Shi M, Lian B, Sun T, Wu J, Bai J, Qu M, Wang Y, Yu W, Gao Z. PNA-Modified Liposomes Improve the Delivery Efficacy of CAPIRI for the Synergistic Treatment of Colorectal Cancer. Front Pharmacol 2022; 13:893151. [PMID: 35784721 PMCID: PMC9240350 DOI: 10.3389/fphar.2022.893151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/09/2022] [Indexed: 01/10/2023] Open
Abstract
Tumor-associated antigen mucin 1 (MUC1) is highly expressed in colorectal cancer and is positively correlated with advanced stage at diagnosis and poor patient outcomes. The combination of irinotecan and capecitabine is standard chemotherapy for metastatic colorectal cancer and is known as XELIRI or CAPIRI, which significantly prolongs the progression-free survival and overall survival of colorectal cancer patients compared to a single drug alone. We previously reported that peanut agglutinin (PNA)-conjugated liposomes showed enhanced drug delivery efficiency to MUC1-positive liver cancer cells. In this study, we prepared irinotecan hydrochloride (IRI) and capecitabine (CAP)-coloaded liposomes modified by peanut agglutinin (IRI/CAP-PNA-Lips) to target MUC1-positive colorectal cancer. The results showed that IRI/CAP-PNA-Lips showed an enhanced ability to target MUC1-positive colorectal cancer cells compared to unmodified liposomes. Treatment with IRI/CAP-PNA-Lips also increased the proportion of apoptotic cells and inhibited the proliferation of colorectal cancer cells. The targeting specificity for tumor cells and the antitumor effects of PNA-modified liposomes were significantly increased in tumor-bearing mice with no severe cytotoxicity to normal tissues. These results suggest that PNA-modified liposomes could provide a new delivery strategy for the synergistic treatment of colorectal cancer with clinical chemotherapeutic agents.
Collapse
Affiliation(s)
- Wenbin Diao
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
| | - Ben Yang
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
| | - Sipeng Sun
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
| | - Anping Wang
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
| | - Rongguan Kou
- School of Life Science and Technology, Weifang Medical University, Weifang, China
| | - Qianyun Ge
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
| | - Mengqi Shi
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
| | - Bo Lian
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
| | - Tongyi Sun
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
| | - Jingliang Wu
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
| | - Jingkun Bai
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
| | - Meihua Qu
- Translational Medical Center, Second People’s Hospital of Weifang, Weifang, China
| | - Yubing Wang
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
- *Correspondence: Yubing Wang, ; Wenjing Yu, ; Zhiqin Gao,
| | - Wenjing Yu
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
- *Correspondence: Yubing Wang, ; Wenjing Yu, ; Zhiqin Gao,
| | - Zhiqin Gao
- School of Life Science and Technology, Weifang Medical University, Weifang, China
- Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang, China
- *Correspondence: Yubing Wang, ; Wenjing Yu, ; Zhiqin Gao,
| |
Collapse
|
27
|
Ren G, Duan D, Wang G, Wang R, Li Y, Zuo H, Zhang Q, Zhang G, Zhao Y, Wang R, Zhang S. Construction of reduction-sensitive heterodimer prodrugs of doxorubicin and dihydroartemisinin self-assembled nanoparticles with antitumor activity. Colloids Surf B Biointerfaces 2022; 217:112614. [PMID: 35700564 DOI: 10.1016/j.colsurfb.2022.112614] [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: 03/02/2022] [Revised: 05/30/2022] [Accepted: 06/04/2022] [Indexed: 11/20/2022]
Abstract
Doxorubicin (DOX) is used as a first-line chemotherapeutic drug, whereas dihydroartemisinin (DHA) also shows a certain degree of antitumor activity. Disulfide bonds (-SS-) in prodrug molecules can be degraded in highly reducing environments. Thus, heterodimer prodrugs of DOX and DHA linked by a disulfide bond was designed and subsequently prepared as reduction-responsive self-assembled nanoparticles (DOX-SS-DHA NPs). In an in vitro release study, DOX-SS-DHA NPs exhibited reduction-responsive activity. Upon cellular evaluation, DOX-SS-DHA NPs were found to have better selectivity toward tumor cells and less cytotoxicity to normal cells. Compared to free DiR, DOX-SS-DHA NPs showed improved accumulation at the tumor site and even had a longer clearance half-life. More importantly, DOX-SS-DHA NPs possessed a much higher tumor inhibition efficacy than DOX-sol and MIX-sol in 4T1 tumor-bearing mice. Our results suggested the superior antitumor efficacy of DOX-SS-DHA NPs with less cytotoxicity.
Collapse
Affiliation(s)
- Guolian Ren
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Danyu Duan
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Geng Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rongrong Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yujie Li
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Hengtong Zuo
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Qichao Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guoshun Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongdan Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Ruili Wang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Shuqiu Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
28
|
Bhattacharjee S. Craft of Co-encapsulation in Nanomedicine: A Struggle To Achieve Synergy through Reciprocity. ACS Pharmacol Transl Sci 2022; 5:278-298. [PMID: 35592431 PMCID: PMC9112416 DOI: 10.1021/acsptsci.2c00033] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Indexed: 12/19/2022]
Abstract
Achieving synergism, often by combination therapy via codelivery of chemotherapeutic agents, remains the mainstay of treating multidrug-resistance cases in cancer and microbial strains. With a typical core-shell architecture and surface functionalization to ensure facilitated targeting of tissues, nanocarriers are emerging as a promising platform toward gaining such synergism. Co-encapsulation of disparate theranostic agents in nanocarriers-from chemotherapeutic molecules to imaging or photothermal modalities-can not only address the issue of protecting the labile drug payload from a hostile biochemical environment but may also ensure optimized drug release as a mainstay of synergistic effect. However, the fate of co-encapsulated molecules, influenced by temporospatial proximity, remains unpredictable and marred with events with deleterious impact on therapeutic efficacy, including molecular rearrangement, aggregation, and denaturation. Thus, more than just an art of confining multiple therapeutics into a 3D nanoscale space, a co-encapsulated nanocarrier, while aiming for synergism, should strive toward achieving a harmonious cohabitation of the encapsulated molecules that, despite proximity and opportunities for interaction, remain innocuous toward each other and ensure molecular integrity. This account will inspect the current progress in co-encapsulation in nanocarriers and distill out the key points toward accomplishing such synergism through reciprocity.
Collapse
Affiliation(s)
- Sourav Bhattacharjee
- School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
29
|
Design of Smart Nanomedicines for Effective Cancer Treatment. Int J Pharm 2022; 621:121791. [PMID: 35525473 DOI: 10.1016/j.ijpharm.2022.121791] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/25/2022] [Accepted: 04/28/2022] [Indexed: 12/22/2022]
Abstract
Nanomedicine is a novel field of study that involves the use of nanomaterials to address challenges and issues that are associated with conventional therapeutics for cancer treatment including, but not limited to, low bioavailability, low water-solubility, narrow therapeutic window, nonspecific distribution, and multiple side effects of the drugs. Multiple strategies have been exploited to reduce the nonspecific distribution, and thus the side effect of the active pharmaceutical ingredients (API), including active and passive targeting strategies and externally controllable release of the therapeutic cargo. Site-specific release of the drug prevents it from impacting healthy cells, thereby significantly reducing side effects. API release triggers can be either externally applied, as in ultrasound-mediated activation, or induced by the tumor. To rationally design such nanomedicines, a thorough understanding of the differences between the tumor microenvironment versus that of healthy tissues must be pared with extensive knowledge of stimuli-responsive biomaterials. Herein, we describe the characteristics that differentiate tumor tissues from normal tissues. Then, we introduce smart materials that are commonly used for the development of smart nanomedicines to be triggered by stimuli such as changes in pH, temperature, and enzymatic activity. The most recent advances and their impact on the field of cancer therapy are further discussed.
Collapse
|
30
|
Konhäuser M, Kannaujiya VK, Steiert E, Schwickert K, Schirmeister T, Wich PR. Co-Encapsulation of l-Asparaginase and Etoposide in Dextran Nanoparticles for Synergistic Effect in Chronic Myeloid Leukemia Cells. Int J Pharm 2022; 622:121796. [PMID: 35525474 DOI: 10.1016/j.ijpharm.2022.121796] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 04/02/2022] [Accepted: 04/29/2022] [Indexed: 11/17/2022]
Abstract
Co-encapsulation of multiple therapeutic drugs in a single nanocarrier has the potential to enable synergistic interactions, increase drug efficacy, and reduce side effects. The enzyme l-asparaginase and the small molecule drug etoposide have a known synergistic effect against selected cancer types. However, both drugs differ significantly in size, molecular weight, and solubility, which often results in challenges when a simultaneous delivery is required. In this study, we present the co-encapsulation of a large hydrophilic enzyme l-asparaginase and the small hydrophobic drug etoposide into a biodegradable, biocompatible, and acid-responsive dextran-based nanoparticle system. These dual drug-loaded nanoparticles show an excellent cellular uptake in chronic myeloid leukemia (CML) K562 cells and a stepwise release of the cytotoxic payloads in a pH-dependent manner. In activity tests, the dual drug-loaded formulation has shown a significant effect on cell viability (down to 31%) compared to those incubated only with l-asparaginase (92%) or etoposide (82%) at a particle concentration of 125 μg∙mL-1. These results show that the simultaneous co-delivery of these two drugs in K562 cells leads to synergistic cytotoxicity, indicating a great potential for the treatment of CML.
Collapse
Affiliation(s)
- M Konhäuser
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - V K Kannaujiya
- School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - E Steiert
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - K Schwickert
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - T Schirmeister
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany
| | - P R Wich
- Institute of Pharmaceutical and Biomedicinal Sciences, Johannes Gutenberg-University Mainz, Staudingerweg 5, Mainz 55128, Germany; School of Chemical Engineering, University of New South Wales, Sydney, NSW 2052, Australia; Australian Centre for NanoMedicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
31
|
Synthesis of Amphiphilic Copolymers of N-Vinyl-2-pyrrolidone and Allyl Glycidyl Ether for Co-Delivery of Doxorubicin and Paclitaxel. Polymers (Basel) 2022; 14:polym14091727. [PMID: 35566896 PMCID: PMC9103997 DOI: 10.3390/polym14091727] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/16/2022] [Accepted: 04/20/2022] [Indexed: 12/22/2022] Open
Abstract
Co-delivery of chemotherapeutics in cancer treatment has been proven essential for overcoming multidrug resistance and improving the outcome of therapy. We report the synthesis of amphiphilic copolymers of N-vinyl-2-pyrrolidone and allyl glycidyl ether of various compositions and demonstrate that they can form nanoaggregates capable of simultaneous covalent immobilization of doxorubicin by the epoxy groups in the shell and hydrophobic-driven incorporation of paclitaxel into the core of nanoparticles. The structure of the obtained copolymers was characterized by 13C NMR, IR, and MALDI spectroscopy, as well as adsorption at the water/toluene interface. A linear increase in the number-average molecular weight of amphiphilic copolymers and a decrease in the number-average diameter of macromolecular aggregates with an increase in the ratio N-vinyl-2-pyrrolidone/allyl glycidyl ether were observed. The assembled nanocarriers were characterized by DLS. The reported novel nanocarriers can be of interest for delivery and co-delivery of a wide range of pharmacological preparations and combined therapy for cancer and other deceases.
Collapse
|
32
|
Wu K, Yu B, Li D, Tian Y, Liu Y, Jiang J. Recent Advances in Nanoplatforms for the Treatment of Osteosarcoma. Front Oncol 2022; 12:805978. [PMID: 35242707 PMCID: PMC8885548 DOI: 10.3389/fonc.2022.805978] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Osteosarcoma (OS) is the most common primary bone tumor in children and young people. Traditional surgical excision combined with chemotherapy presents many limitations, such as resistance and systemic side effects of chemotherapy drugs, postoperative recurrence, and bone defects. Given these limitations, novel therapeutic modalities for OS treatment using nanometer-sized platform-based chemotherapeutic delivery have emerged as a promising alternative therapy. This form of therapy offers multiple advantages, such as accurate delivery of the drug to the tumor site and repair of limited bone defects after tumor resection. In this review, we briefly summarize nanoplatforms, including liposomes, polymeric nanoparticles, inorganic nanoparticles, nanomicelles, dendrimers, nanocapsules, and exosomes. The essential shortcomings involved in these nanoplatforms, such as poor stability, immunogenicity, insufficient circulation, and drug leakage are also discussed, and related solutions are briefly proposed. Finally, the application prospects of nanoplatforms in the treatment of OS are discussed.
Collapse
Affiliation(s)
- Kunzhe Wu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Beibei Yu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Di Li
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yangyang Tian
- Department of Dermatology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yan Liu
- Department of Anesthesiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinlan Jiang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
33
|
Ashrafizadeh M, Saebfar H, Gholami MH, Hushmandi K, Zabolian A, Bikarannejad P, Hashemi M, Daneshi S, Mirzaei S, Sharifi E, Kumar AP, Khan H, Heydari Sheikh Hossein H, Vosough M, Rabiee N, Thakur Kumar V, Makvandi P, Mishra YK, Tay FR, Wang Y, Zarrabi A, Orive G, Mostafavi E. Doxorubicin-loaded graphene oxide nanocomposites in cancer medicine: Stimuli-responsive carriers, co-delivery and suppressing resistance. Expert Opin Drug Deliv 2022; 19:355-382. [PMID: 35152815 DOI: 10.1080/17425247.2022.2041598] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The application of doxorubicin (DOX) in cancer therapy has been limited due to its drug resistance and poor internalization. Graphene oxide (GO) nanostructures have the capacity for DOX delivery while promoting its cytotoxicity in cancer. AREAS COVERED The favorable characteristics of GO nanocomposites, preparation method, and application in cancer therapy are described. Then, DOX resistance in cancer is discussed. The GO-mediated photothermal therapy and DOX delivery for cancer suppression are described. Preparation of stimuli-responsive GO nanocomposites, surface functionalization, hybrid nanoparticles, and theranostic applications are emphasized in DOX chemotherapy. EXPERT OPINION Graphene oxide nanoparticle-based photothermal therapy maximizes the anti-cancer activity of DOX against cancer cells. Apart from DOX delivery, GO nanomaterials are capable of loading anti-cancer agents and genetic tools to minimize drug resistance and enhance the cytolytic impact of DOX in cancer eradication. To enhance DOX accumulation in cancer cells, stimuli-responsive (redox-, light-, enzyme- and pH-sensitive) GO nanoparticles have been developed for DOX delivery. Further development of targeted delivery of DOX-loaded GO nanomaterials against cancer cells may be achieved by surface modification of polymers such as polyethylene glycol, hyaluronic acid, and chitosan. Doxorubicin-loaded GO nanoparticles have demonstrated theranostic potential for simultaneous diagnosis and therapy. Hybridization of GO with other nanocarriers such as silica and gold nanoparticles further broadens their potential anti-cancer therapy applications.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, 34956 Istanbul, Turkey
| | - Hamidreza Saebfar
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Hossein Gholami
- DVM. Graduated, Faculty of Veterinary Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Amirhossein Zabolian
- Department of Orthopedics, School of Medicine, 5th Azar Hospital, Golestan University of Medical Sciences, Golestan, Iran
| | - Pooria Bikarannejad
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Esmaeel Sharifi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, 6517838736 Hamadan, Iran
| | - Alan Prem Kumar
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.,Cancer Science Institute of Singapore and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | | | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Navid Rabiee
- Department of Chemistry, Sharif University of Technology, Tehran, Iran.,School of Engineering, Macquarie University, Sydney, New South Wales, 2109, Australia
| | - Vijay Thakur Kumar
- Biorefining and Advanced Materials Research Center, Scotland's Rural College (SRUC), Kings Buildings, West Mains Road, Edinburgh EH9 3JG, U.K.,School of Engineering, University of Petroleum & Energy Studies (UPES), Dehradun 248007, Uttarakhand, India
| | - Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Materials Interface, viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy
| | - Yogendra Kumar Mishra
- Mads Clausen Institute, NanoSYD, University of Southern Denmark, 6400 Sønderborg, Denmark
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA, USA
| | - Yuzhuo Wang
- Department of Urological Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Sariyer 34396, Istanbul, Turkey
| | - Gorka Orive
- NanoBioCel Research Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN). Vitoria-Gasteiz, Spain.,University Institute for Regenerative Medicine and Oral Implantology - UIRMI (UPV/EHUFundación Eduardo Anitua). Vitoria-Gasteiz, Spain.,Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.,Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.,Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| |
Collapse
|
34
|
Yu XQ, Zhan YR, Tan J, Hei MW, Zhang S, Zhang J. Construction of GSH-triggered cationic fluoropolymer as two-in-one nanoplatform for combined chemo/gene therapy. J Mater Chem B 2022; 10:1308-1318. [DOI: 10.1039/d1tb02602j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The combined chemo-gene therapy has become a promising approach for enhanced anti-cancer treatment. However, effective co-delivery of therapeutic gene and drug into target cells and tissues remains a major obstacle....
Collapse
|
35
|
OUP accepted manuscript. J Pharm Pharmacol 2022; 74:1027-1039. [DOI: 10.1093/jpp/rgac013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 02/19/2022] [Indexed: 11/13/2022]
|
36
|
Liang W, Dong Y, Shao R, Zhang S, Wu X, Huang X, Sun B, Zeng B, Zhao J. Application of Nanoparticles in Drug Delivery for the Treatment of Osteosarcoma: Focusing on the Liposomes. J Drug Target 2021; 30:463-475. [PMID: 34962448 DOI: 10.1080/1061186x.2021.2023160] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Osteosarcoma (OS) is one of the most common primary bone malignancies in children and adolescents. The toxicity to healthy tissues from conventional therapeutic strategies, including chemotherapy and radiotherapy, and drug resistance, severely affect OS patients' quality of life and cancer-specific outcomes. Many efforts have been made to develop various nanomaterial-based drug delivery systems with specific properties to overcome these limitations. Among the developed nanocarriers, liposomes are the most successful and promising candidates for providing targeted tumor therapy and enhancing the safety and therapeutic effect of encapsulated agents. Liposomes have low immunogenicity, high biocompatibility, prolonged half-life, active group protection, cell-like membrane structure, safety, and effectiveness. This review will discuss various nanomaterial-based carriers in cancer therapy and then the characteristics and design of liposomes with a particular focus on the targeting feature. We will also summarize the recent advances in the liposomal drug delivery system for OS treatment in preclinical and clinical studies.
Collapse
Affiliation(s)
- Wenqing Liang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China
| | - Yongqiang Dong
- Department of Orthopedics, Xinchang People's Hospital, Shaoxing 312500, China
| | - Ruyi Shao
- Department of Orthopedics, Zhuji People's Hospital, Shaoxing 312500, China
| | - Songou Zhang
- College of Medicine, Shaoxing University, Shaoxing 312000, China
| | - Xudong Wu
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China
| | - Xiaogang Huang
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China
| | - Bin Sun
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China
| | - Bin Zeng
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China
| | - Jiayi Zhao
- Department of Orthopedics, Zhoushan Hospital of Traditional Chinese Medicine Affiliated to Zhejiang Chinese Medical University, Zhoushan 316000, China
| |
Collapse
|
37
|
Ma X, Gao Y, Zhao D, Zhang W, Zhao W, Wu M, Cui Y, Li Q, Zhang Z, Ma C. Titanium Implants and Local Drug Delivery Systems Become Mutual Promoters in Orthopedic Clinics. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 12:47. [PMID: 35009997 PMCID: PMC8746425 DOI: 10.3390/nano12010047] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 06/14/2023]
Abstract
Titanium implants have always been regarded as one of the gold standard treatments for orthopedic applications, but they still face challenges such as pain, bacterial infections, insufficient osseointegration, immune rejection, and difficulty in personalizing treatment in the clinic. These challenges may lead to the patients having to undergo a painful second operation, along with increased economic burden, but the use of drugs is actively solving these problems. The use of systemic drug delivery systems through oral, intravenous, and intramuscular injection of various drugs with different pharmacological properties has effectively reduced the levels of inflammation, lowered the risk of endophytic bacterial infection, and regulated the progress of bone tumor cells, processing and regulating the balance of bone metabolism around the titanium implants. However, due to the limitations of systemic drug delivery systems-such as pharmacokinetics, and the characteristics of bone tissue in the event of different forms of trauma or disease-sometimes the expected effect cannot be achieved. Meanwhile, titanium implants loaded with drugs for local administration have gradually attracted the attention of many researchers. This article reviews the latest developments in local drug delivery systems in recent years, detailing how various types of drugs cooperate with titanium implants to enhance antibacterial, antitumor, and osseointegration effects. Additionally, we summarize the improved technology of titanium implants for drug loading and the control of drug release, along with molecular mechanisms of bone regeneration and vascularization. Finally, we lay out some future prospects in this field.
Collapse
|
38
|
Zhang P, Li M, Xiao C, Chen X. Stimuli-responsive polypeptides for controlled drug delivery. Chem Commun (Camb) 2021; 57:9489-9503. [PMID: 34546261 DOI: 10.1039/d1cc04053g] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Controlled drug delivery systems, which could release loaded therapeutics upon physicochemical changes imposed by physiological triggers in the desired zone and during the required period of time, offer numerous advantages over traditional drug carriers including enhanced therapeutic effects and reduced toxicity. A polypeptide is a biocompatible and biodegradable polymer, which can be conveniently endowed with stimuli-responsiveness by introducing natural amino acid residues with innate stimuli-responsive characteristics or introducing responsive moieties to its side chains using simple conjugating methods, rendering it an ideal biomedical material for controlled drug delivery. This feature article summarizes our recent work and other relevant studies on the development of polypeptide-based drug delivery systems that respond to single or multiple physiological stimuli (e.g., pH, redox potential, glucose, and hypoxia) for controlled drug delivery applications. The material designs, synthetic strategies, loading and controlled-release mechanisms of drugs, and biomedical applications of these stimuli-responsive polypeptides-based drug delivery systems are elaborated. Finally, the challenges and opportunities in this field are briefly discussed.
Collapse
Affiliation(s)
- Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China. .,Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Mingqian Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China. .,Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China. .,Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, P. R. China
| |
Collapse
|
39
|
Dai G, Zheng D, Liu G, Song Q. Synergistic Anticancer Effects of Cisplatin Combined with Combretastatin A4 Phosphate on Human Osteosarcoma-Xenografted Mice. Cells Tissues Organs 2021; 210:293-300. [PMID: 34433168 DOI: 10.1159/000517446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/12/2021] [Indexed: 11/19/2022] Open
Abstract
This study aimed to investigate the effectiveness of anticancer therapy combining a cytotoxic chemotherapeutic agent, cisplatin (DDP), and a vascular disruptive drug, combretastatin A4 phosphate (CA4P), in osteosarcoma. First, a human osteosarcoma-xenografted mice model was established. Second, the transplanted tumor models were treated with DDP and CA4P in combination or as monotherapy. Third, the therapeutic effects and the mechanism of the drug combination in the inhibition of transplanted tumors was studied. Finally, the toxic effects of the drugs were observed and recorded. The results showed that DDP combined with CA4P significantly inhibited the growth and lung metastasis of transplanted tumors compared with the monotherapy drug group and vehicle control group. Histopathological analysis revealed that apoptotic and necrotic cell death significantly increased in the combination group, and combined treatment significantly inhibited the proliferation of osteosarcoma cells compared with either agent alone or the vehicle control. Additionally, no obvious toxic effects were observed in the combination group. These results indicate that the combined effects of DDP and CA4P on the progression of human osteosarcoma in vivo were superior to that of either agent alone. DDP combined with CA4P exerted synergistic effects at lower concentrations and promoted apoptosis and necrosis, as well as inhibited proliferation of osteosarcoma cells, but it did not increase the systemic toxic effects of chemotherapy. Our findings highlight CA4P as an effective anticancer agent candidate for combination with DDP in clinical applications to treat osteosarcoma.
Collapse
Affiliation(s)
- Guo Dai
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Di Zheng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Gaiwei Liu
- Department of Orthopedics, Jingzhou Central Hospital, Jingzhou, China
| | - Qi Song
- Department of Trauma Surgery, Wuhan No. 1 Hospital, Wuhan, China
| |
Collapse
|
40
|
Hao D, Zhang Z, Ji Y. Responsive polymeric drug delivery systems for combination anticancer therapy: experimental design and computational insights. INT J POLYM MATER PO 2021. [DOI: 10.1080/00914037.2021.1960340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Dule Hao
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Zheng Zhang
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Yuanhui Ji
- Jiangsu Province Hi-Tech Key Laboratory for Bio-medical Research, School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| |
Collapse
|
41
|
Zhang S, Guo W. β-Elemene Enhances the Sensitivity of Osteosarcoma Cells to Doxorubicin via Downregulation of Peroxiredoxin-1. Onco Targets Ther 2021; 14:3599-3609. [PMID: 34113126 PMCID: PMC8184248 DOI: 10.2147/ott.s303152] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 05/06/2021] [Indexed: 12/31/2022] Open
Abstract
Background Doxorubicin (Dox) resistance is a primary obstacle for the treatment of osteosarcoma. Meanwhile, β-Elemene was shown to exhibit an anti-proliferative effect on osteosarcoma cells. However, the role of a combination of Dox with β-Elemene on osteosarcoma cells remains unclear. Thus, this study aimed to investigate the role of the combination of Dox with β-Elemene on the proliferation, apoptosis and oxidative stress of Dox-resistance osteosarcoma cells. Methods CKC-8, EdU staining and flow cytometry assays were used to determine the viability, proliferation and apoptosis of Dox-resistance osteosarcoma cells, respectively. Meanwhile, the expression of antioxidant protein peroxiredoxin-1 (Prx-1) in Dox-resistance osteosarcoma cells was detected with Western blot assay. Results In this study, the inhibitory effects of Dox on the viability and proliferation of Dox-resistance osteosarcoma cells were significantly enhanced by β-Elemene. In addition, the combination of Dox and β-Elemene markedly induced the apoptosis and oxidative stress in Dox-resistance osteosarcoma cells. Moreover, combination treatment notably downregulated the expression of Prx-1 in Dox-resistance osteosarcoma cells, indicating that combination treatment inhibited the antioxidant capacity of Dox-resistance osteosarcoma cells. In vivo experiments confirmed that β-Elemene could enhance the anti-tumor effect of Dox in Saos-2/Dox xenograft model. Conclusion We found that β-Elemene could reverse Dox resistance in Dox-resistance osteosarcoma cells via inhibition of Prx-1. Therefore, combining Dox with β-Elemene might be considered as a therapeutic approach for the treatment of Dox-resistant osteosarcoma.
Collapse
Affiliation(s)
- Shaochun Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China.,Department of Orthopedics, Ezhou Central Hospital, Ezhou, Hubei, 436000, People's Republic of China
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, People's Republic of China
| |
Collapse
|
42
|
Song M, Xia W, Tao Z, Zhu B, Zhang W, Liu C, Chen S. Self-assembled polymeric nanocarrier-mediated co-delivery of metformin and doxorubicin for melanoma therapy. Drug Deliv 2021; 28:594-606. [PMID: 33729072 PMCID: PMC7996084 DOI: 10.1080/10717544.2021.1898703] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Malignant melanoma is a life-threatening form of skin cancer with a low response rate to single-agent chemotherapy. Although combined therapies of metformin (MET) and doxorubicin (DOX) are effective in treating a variety of cancers, including breast cancer, their different physicochemical properties and administration routines reduce the effective co-accumulation of both drugs in tumors. Nanoparticles (NPs) have been demonstrated to potentially improve drug delivery efficiency in cancer therapy of, for example, liver and lung cancers. Hence, in this study, we prepared pH-sensitive, biocompatible, tumor-targeting NPs based on the conjugation of biomaterials, including sodium alginate, cholesterol, and folic acid (FCA). As expected, since cholesterol and folic acid are two essentials, but insufficient, substrates for melanoma growth, we observed that the FCA NPs specifically and highly effectively accumulated in xenograft melanoma tumors. Taking advantage of the FCA NP system, we successfully co-delivered a combination of MET and DOX into melanoma tumors to trigger pyroptosis, apoptosis, and necroptosis (PANoptosis) of the melanoma cells, thus blocking melanoma progression. Combined, the establishment of such an FCA NP system provides a promising vector for effective drug delivery into melanoma and increases the possibility and efficiency of drug combinations for cancer treatment.
Collapse
Affiliation(s)
- Mingming Song
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wentao Xia
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zixuan Tao
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Bin Zhu
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Wenxiang Zhang
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chang Liu
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Siyu Chen
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
43
|
Jia R, Teng L, Gao L, Su T, Fu L, Qiu Z, Bi Y. Advances in Multiple Stimuli-Responsive Drug-Delivery Systems for Cancer Therapy. Int J Nanomedicine 2021; 16:1525-1551. [PMID: 33658782 PMCID: PMC7920594 DOI: 10.2147/ijn.s293427] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 01/27/2021] [Indexed: 12/15/2022] Open
Abstract
Nanomedicines afford unique advantages in therapeutic intervention against tumors. However, conventional nanomedicines have failed to achieve the desired effect against cancers because of the presence of complicated physiological fluids and the tumor microenvironment. Stimuli-responsive drug-delivery systems have emerged as potential tools for advanced treatment of cancers. Versatile nano-carriers co-triggered by multiple stimuli in different levels of organisms (eg, extracorporeal, tumor tissue, cell, subcellular organelles) have aroused widespread interest because they can overcome sequential physiological and pathological barriers to deliver diverse therapeutic “payloads” to the desired targets. Furthermore, multiple stimuli-responsive drug-delivery systems (MSR-DDSs) offer a good platform for co-delivery of agents and reversing multidrug resistance. This review affords a comprehensive overview on the “landscape” of MSR-DDSs against tumors, highlights the design strategies of MSR-DDSs in recent years, discusses the putative advantage of oncotherapy or the obstacles that so far have hindered the clinical translation of MSR-DDSs.
Collapse
Affiliation(s)
- Ruixin Jia
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, People's Republic of China
| | - Lesheng Teng
- School of Life Science, Jilin University, Changchun, Jilin, People's Republic of China
| | - Lingyu Gao
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, People's Republic of China
| | - Ting Su
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, People's Republic of China
| | - Lu Fu
- College of Life Science, Jilin Agricultural University, Changchun, Jilin, People's Republic of China
| | - Zhidong Qiu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, People's Republic of China
| | - Ye Bi
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, Jilin, People's Republic of China.,Practice Training Center, Changchun University of Chinese Medicine, Changchun, Jilin, People's Republic of China
| |
Collapse
|
44
|
Barani M, Mukhtar M, Rahdar A, Sargazi S, Pandey S, Kang M. Recent Advances in Nanotechnology-Based Diagnosis and Treatments of Human Osteosarcoma. BIOSENSORS 2021; 11:55. [PMID: 33672770 PMCID: PMC7924594 DOI: 10.3390/bios11020055] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/13/2021] [Accepted: 02/15/2021] [Indexed: 12/24/2022]
Abstract
Osteosarcoma (OSA) is a type of bone cancer that begins in the cells that form bones.OSA is a rare mesenchymal bone neoplasm derived from mesenchymal stem cells. Genome disorganization, chromosomal modifications, deregulation of tumor suppressor genes, and DNA repair defects are the factors most responsible for OSA development. Despite significant advances in the diagnosing and treatment of OSA, patients' overall survival has not improved within the last twenty years. Lately, advances in modern nanotechnology have spurred development in OSA management and offered several advantages to overcome the drawbacks of conventional therapies. This technology has allowed the practical design of nanoscale devices combined with numerous functional molecules, including tumor-specific ligands, antibodies, anti-cancer drugs, and imaging probes. Thanks to their small sizes, desirable drug encapsulation efficiency, and good bioavailability, functionalized nanomaterials have found wide-spread applications for combating OSA progression. This review invokes the possible utility of engineered nanomaterials in OSA diagnosis and treatment, motivating the researchers to seek new strategies for tackling the challenges associated with it.
Collapse
Affiliation(s)
- Mahmood Barani
- Department of Chemistry, Shahid Bahonar University of Kerman, Kerman 76169-14111, Iran;
| | - Mahwash Mukhtar
- Faculty of Pharmacy, Institute of Pharmaceutical Technology and Regulatory Affairs, University of Szeged, 6720 Szeged, Hungary;
| | - Abbas Rahdar
- Department of Physics, Faculty of Science, University of Zabol, Zabol 538-98615, Iran
| | - Saman Sargazi
- Cellular and Molecule Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan 98167-43463, Iran;
| | - Sadanand Pandey
- Particulate Matter Research Center, Research Institute of Industrial Science & Technology (RIST), 187-12, Geumho-ro, Gwangyang-si 57801, Korea
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Korea;
| | - Misook Kang
- Department of Chemistry, College of Natural Science, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Korea;
| |
Collapse
|
45
|
Desai SA, Manjappa A, Khulbe P. Drug delivery nanocarriers and recent advances ventured to improve therapeutic efficacy against osteosarcoma: an overview. J Egypt Natl Canc Inst 2021; 33:4. [PMID: 33555490 DOI: 10.1186/s43046-021-00059-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 01/18/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Osteosarcoma (OS) is one of the key cancers affecting the bone tissues, primarily occurred in children and adolescence. Recently, chemotherapy followed by surgery and then post-operative adjuvant chemotherapy is widely used for the treatment of OS. However, the lack of selectivity and sensitivity to tumor cells, the development of multi-drug resistance (MDR), and dangerous side effects have restricted the use of chemotherapeutics. MAIN BODY There is an unmet need for novel drug delivery strategies for effective treatment and management of OS. Advances in nanotechnology have led to momentous progress in the design of tumor-targeted drug delivery nanocarriers (NCs) as well as functionalized smart NCs to achieve targeting and to treat OS effectively. The present review summarizes the drug delivery challenges in OS, and how organic nanoparticulate approaches are useful in overcoming barriers will be explained. The present review describes the various organic nanoparticulate approaches such as conventional nanocarriers, stimuli-responsive NCs, and ligand-based active targeting strategies tested against OS. The drug conjugates prepared with copolymer and ligand having bone affinity, and advanced promising approaches such as gene therapy, gene-directed enzyme prodrug therapy, and T cell therapy tested against OS along with their reported limitations are also briefed in this review. CONCLUSION The nanoparticulate drugs, drug conjugates, and advanced therapies such as gene therapy, and T cell therapy have promising and potential application in the effective treatment of OS. However, many of the above approaches are still at the preclinical stage, and there is a long transitional period before their clinical application.
Collapse
Affiliation(s)
- Sujit Arun Desai
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Rd, Mahal, Jagatpura, Jaipur, Rajasthan, 302017, India. .,Annasaheb Dange College of D Pharmacy, Ashta, Tal: Walwa, Dist., Sangli, Maharashtra, 416301, India.
| | - Arehalli Manjappa
- Tatyasaheb Kore College of Pharmacy, Warananagar, Tal: Panhala, Dist., Kolhapur, Maharashtra, 416113, India
| | - Preeti Khulbe
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Rd, Mahal, Jagatpura, Jaipur, Rajasthan, 302017, India
| |
Collapse
|
46
|
Wang S, Sun L, Cao H, Zhong Y, Shao Z. Development of a Dual-drug-loaded Silk Fibroin Hydrogel and Study on Its Drugs Release Behaviors. ACTA CHIMICA SINICA 2021. [DOI: 10.6023/a21050203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
47
|
Zhang H, Wu Y, Xu X, Chen C, Xue X, Xu B, Li T, Chen Z. Synthesis Characterization of Platinum (IV) Complex Curcumin Backboned Polyprodrugs: In Vitro Drug Release Anticancer Activity. Polymers (Basel) 2020; 13:E67. [PMID: 33375302 PMCID: PMC7795977 DOI: 10.3390/polym13010067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/20/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
The conventional mono-chemotherapy still suffers from unsatisfied potency for cancer therapy due to tumor heterogeneity and the occurrence of drug resistance. Combination chemotherapy based on the nanosized drug delivery systems (nDDSs) has been developed as a promising platform to circumvent the limitations of mono-chemotherapy. In this work, starting from cisplatin and curcumin (Cur), we prepared a dual drug backboned shattering polymeric nDDS for synergistic chemotherapy. By in situ polymerization of the Cur, platinum (IV) complex-based prodrug monomer (DHP), L-lysine diisocyanate (LDI), and then conjugation with a hydrophilic poly (ethylene glycol) monomethyl ether (mPEG) derivative, a backbone-type platinum (IV) and Cur linkage containing mPEG-poly(platinum-co-Cur)-mPEG (PCPt) copolymer was synthesized. Notably, the platinum (IV) (Pt (IV)) and Cur were incorporated into the hydrophobic segment of PCPt with the fixed drugs loading ratio and high drugs loading content. The batch-to-batch variability could be decreased. The resulting prodrug copolymer then self-assembled into nanoparticles (PCPt NPs) with an average diameter around 100 nm, to formulate a synergetic nDDS. Importantly, PCPt NPs could greatly improve the solubility and stability of Cur. In vitro drug release profiles have demonstrated that PCPt NPs were stable in PBS 7.4, rapid burst release was greatly decreased, and the Pt and Cur release could be largely enhanced under reductive conditions due to the complete dissociation of the hydrophobic main chain of PCPt. In vitro cell viability test indicated that PCPt NPs were efficient synergistic chemotherapy units. Moreover, PCPt NPs were synergistic for cisplatin-resistant cell lines A549/DDP cells, and they exhibited excellent reversal ability of tumor resistance to cisplatin. This work provides a promising strategy for the design and synthesis of nDDS for combination chemotherapy.
Collapse
Affiliation(s)
- Honglei Zhang
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (H.Z.); (X.X.); (B.X.); (T.L.)
| | - Yanjuan Wu
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (H.Z.); (X.X.); (B.X.); (T.L.)
| | - Xiao Xu
- Institute of Food Safety and Environment Monitoring, College of Chemistry, Fuzhou University, Fuzhou 350108, China; (X.X.); (C.C.)
| | - Chen Chen
- Institute of Food Safety and Environment Monitoring, College of Chemistry, Fuzhou University, Fuzhou 350108, China; (X.X.); (C.C.)
| | - Xiukun Xue
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (H.Z.); (X.X.); (B.X.); (T.L.)
| | - Ben Xu
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (H.Z.); (X.X.); (B.X.); (T.L.)
| | - Tianduo Li
- Shandong Provincial Key Laboratory of Molecular Engineering, School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China; (H.Z.); (X.X.); (B.X.); (T.L.)
| | - Zhaowei Chen
- Institute of Food Safety and Environment Monitoring, College of Chemistry, Fuzhou University, Fuzhou 350108, China; (X.X.); (C.C.)
| |
Collapse
|
48
|
Bikiaris ND, Ainali NM, Christodoulou E, Kostoglou M, Kehagias T, Papasouli E, Koukaras EN, Nanaki SG. Dissolution Enhancement and Controlled Release of Paclitaxel Drug via a Hybrid Nanocarrier Based on mPEG-PCL Amphiphilic Copolymer and Fe-BTC Porous Metal-Organic Framework. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E2490. [PMID: 33322372 PMCID: PMC7763675 DOI: 10.3390/nano10122490] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/06/2020] [Accepted: 12/09/2020] [Indexed: 01/12/2023]
Abstract
In the present work, the porous metal-organic framework (MOF) Basolite®F300 (Fe-BTC) was tested as a potential drug-releasing depot to enhance the solubility of the anticancer drug paclitaxel (PTX) and to prepare controlled release formulations after its encapsulation in amphiphilic methoxy poly(ethylene glycol)-poly(ε-caprolactone) (mPEG-PCL) nanoparticles. Investigation revealed that drug adsorption in Fe-BTC reached approximately 40%, a relatively high level, and also led to an overall drug amorphization as confirmed by differential scanning calorimetry (DSC) and X-ray diffraction (XRD). The dissolution rate of PTX-loaded MOF was substantially enhanced achieving a complete (100%) release within four days, while the neat drug only reached a 13% maximum rate (3-4 days). This PTX-Fe-BTC nanocomposite was further encapsulated into a mPEG-PCL matrix, a typical aliphatic amphiphilic copolyester synthesized in our lab, whose biocompatibility was validated by in vitro cytotoxicity tests toward human umbilical vein endothelial cells (HUVEC). Encapsulation was performed according to the solid-in-oil-in-water emulsion/solvent evaporation technique, resulting in nanoparticles of about 143 nm, slightly larger of those prepared without the pre-adsorption of PTX on Fe-BTC (138 nm, respectively). Transmission electron microscopy (TEM) imaging revealed that spherical nanoparticles with embedded PTX-loaded Fe-BTC nanoparticles were indeed fabricated, with sizes ranging from 80 to 150 nm. Regions of the composite Fe-BTC-PTX system in the infrared (IR) spectrum are identified as signatures of the drug-MOF interaction. The dissolution profiles of all nanoparticles showed an initial burst release, attributed to the drug amount located at the nanoparticles surface or close to it, followed by a steadily and controlled release. This is corroborated by computational analysis that reveals that PTX attaches effectively to Fe-BTC building blocks, but its relatively large size limits diffusion through crystalline regions of Fe-BTC. The dissolution behaviour can be described through a bimodal diffusivity model. The nanoparticles studied could serve as potential chemotherapeutic candidates for PTX delivery.
Collapse
Affiliation(s)
- Nikolaos D. Bikiaris
- Laboratory of Chemistry and Technology of Polymers and Dyes, Department of Chemistry, Aristotle University of Thessaloniki, GR-541 24 Thessaloniki, Greece; (N.D.B.); (N.M.A.); (E.C.)
| | - Nina Maria Ainali
- Laboratory of Chemistry and Technology of Polymers and Dyes, Department of Chemistry, Aristotle University of Thessaloniki, GR-541 24 Thessaloniki, Greece; (N.D.B.); (N.M.A.); (E.C.)
| | - Evi Christodoulou
- Laboratory of Chemistry and Technology of Polymers and Dyes, Department of Chemistry, Aristotle University of Thessaloniki, GR-541 24 Thessaloniki, Greece; (N.D.B.); (N.M.A.); (E.C.)
| | - Margaritis Kostoglou
- Laboratory of General and Inorganic Chemical Technology, Department of Chemistry, Aristotle University of Thessaloniki, GR-541 24 Thessaloniki, Greece;
| | - Thomas Kehagias
- Laboratory of Electron Microscopy, Department of Physics, Aristotle University of Thessaloniki, GR-541 24 Thessaloniki, Greece;
| | - Emilia Papasouli
- Laboratory of Quantum and Computational Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, GR-541 24 Thessaloniki, Greece; (E.P.); (E.N.K.)
| | - Emmanuel N. Koukaras
- Laboratory of Quantum and Computational Chemistry, Department of Chemistry, Aristotle University of Thessaloniki, GR-541 24 Thessaloniki, Greece; (E.P.); (E.N.K.)
| | - Stavroula G. Nanaki
- Laboratory of Chemistry and Technology of Polymers and Dyes, Department of Chemistry, Aristotle University of Thessaloniki, GR-541 24 Thessaloniki, Greece; (N.D.B.); (N.M.A.); (E.C.)
| |
Collapse
|