1
|
Han J, Shen Y, Cao R, Wang W, Duan J, Duan J, Bao C. Active herbal ingredients and drug delivery design for tumor therapy: a review. Chin J Nat Med 2024; 22:1134-1162. [PMID: 39725513 DOI: 10.1016/s1875-5364(24)60741-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Indexed: 12/28/2024]
Abstract
Active herbal ingredients are gaining recognition for their potent anti-tumor efficacy, attributable to various mechanisms including tumor cell inhibition, immune system activation, and tumor angiogenesis inhibition. Recent studies have revealed that numerous anti-tumor herbal ingredients, such as ginsenosides, ursolic acid, oleanolic acid, and Angelica sinensis polysaccharides, can be utilized to develop smart drug carriers like liposomes, micelles, and nanoparticles. These carriers can deliver active herbal ingredients and co-deliver anti-tumor drugs to enhance drug accumulation at tumor sites, thereby improving anti-tumor efficacy. This study provides a comprehensive analysis of the mechanisms by which these active herbal ingredients-derived carriers enhance therapeutic outcomes. Additionally, it highlights the structural properties of these active herbal ingredients, demonstrating how their unique features can be strategically employed to design smart drug carriers with improved anti-tumor efficacy. The insights presented aim to serve as a reference and guide future innovations in the design and application of smart drug carriers for cancer therapy that leverage active herbal ingredients.
Collapse
Affiliation(s)
- Jing Han
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yanxi Shen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ruiying Cao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weiren Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinao Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jialun Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Chunjie Bao
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
2
|
Ma W, Wang X, Zhang D, Mu X. Research Progress of Disulfide Bond Based Tumor Microenvironment Targeted Drug Delivery System. Int J Nanomedicine 2024; 19:7547-7566. [PMID: 39071505 PMCID: PMC11283832 DOI: 10.2147/ijn.s471734] [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: 04/01/2024] [Accepted: 07/17/2024] [Indexed: 07/30/2024] Open
Abstract
Cancer poses a significant threat to human life and health. Chemotherapy is currently one of the effective cancer treatments, but many chemotherapy drugs have cell toxicity, low solubility, poor stability, a narrow therapeutic window, and unfavorable pharmacokinetic properties. To solve the above problems, target drug delivery to tumor cells, and reduce the side effects of drugs, an anti-tumor drug delivery system based on tumor microenvironment has become a focus of research in recent years. The construction of a reduction-sensitive nanomedicine delivery system based on disulfide bonds has attracted much attention. Disulfide bonds have good reductive responsiveness and can effectively target the high glutathione (GSH) levels in the tumor environment, enabling precise drug delivery. To further enhance targeting and accelerate drug release, disulfide bonds are often combined with pH-responsive nanocarriers and highly expressed ligands in tumor cells to construct drug delivery systems. Disulfide bonds can connect drug molecules and polymer molecules in the drug delivery system, as well as between different drug molecules and carrier molecules. This article summarized the drug delivery systems (DDS) that researchers have constructed in recent years based on disulfide bond drug delivery systems targeting the tumor microenvironment, disulfide bond cleavage-triggering conditions, various drug loading strategies, and carrier design. In this review, we also discuss the controlled release mechanisms and effects of these DDS and further discuss the clinical applicability of delivery systems based on disulfide bonds and the challenges faced in clinical translation.
Collapse
Affiliation(s)
- Weiran Ma
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
- Jilin University School of Pharmaceutical Sciences, Changchun, 130021, People’s Republic of China
| | - Xiaoying Wang
- Jilin University School of Pharmaceutical Sciences, Changchun, 130021, People’s Republic of China
| | - Dongqi Zhang
- Department of Urology, The First Hospital of Jilin University, Changchun, 130021, People’s Republic of China
| | - Xupeng Mu
- Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, 130033, People’s Republic of China
| |
Collapse
|
3
|
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
|
4
|
Zhang C, Zhong H, Li X, Xing Z, Liu J, Yu R, Deng X. Design, synthesis and bioactivity investigation of peptide-camptothecin conjugates as anticancer agents with a potential to overcome drug resistance. Int J Pharm 2023; 645:123402. [PMID: 37696345 DOI: 10.1016/j.ijpharm.2023.123402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/22/2023] [Accepted: 09/08/2023] [Indexed: 09/13/2023]
Abstract
Camptothecin (CPT) is a natural plant alkaloid from Camptotheca that exhibits a potent anticancer activity. However, its continued utilization is hindered by drawbacks such as low water solubility and restricted tumor selectivity. Cationic anticancer peptides (CAPs) are generally soluble in water, and exhibit favorable selectivity against malignant cells. In previous study, we have reported a CAP termed KM8-Aib present conspicuous selective anticancer effect. Thus, it is postulated conjugating KM8-Aib with CPT might be a plausible approach to improve the defects of CPT. A series of peptide-CPT conjugates were synthesized and subjected to biological evaluation. Among these compounds, Kb-CC07 displayed the highest selective activity against a set of cancer cell lines including drug-resistant cells, showing the IC50 values in the 0.11-1.01 μM range which is 1.9-22.6 times better than that of CPT, and a wide therapeutic index of 124.5 (vs 5.3 for CPT). The water solubility of Kb-CC07 was also improved by ∼ 100 fold compared with CPT. Further investigation unraveled that Kb-CC07 could effectively penetrate across plasma membranes and delivered more CPT molecules into cancer cells, overcoming the drug-resistance result from efflux drug transporters on tumor surface. In vivo experiments supported that Kb-CC07 has excellent in vivo antiproliferative activity against drug-resistant tumors over CPT (tumor growth inhibition of 98.2% and 37.5% for Kb-CC07 and CPT, respectively, at 5 μmol·kg-1), and prompts CPT accumulation in tumor tissue rather than normal organs, thus producing limited toxicities. To sum up, coupling therapeutic agents to CAPs would be a potential strategy to conquer the shortcomings of anticancer drugs. Additionally, Kb-CC07 is suggested to be a promising anticancer candidate deserving further investigation.
Collapse
Affiliation(s)
- Chenyu Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinic al Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China; Department of Pharmacy, Guangzhou Chest Hospital, 62 Hengzhigang Road, Guangzhou 510095, China
| | - Honglan Zhong
- Department of Pharmacy, Guangzhou Chest Hospital, 62 Hengzhigang Road, Guangzhou 510095, China
| | - Xiang Li
- Department of Pharmacy, Guangzhou Chest Hospital, 62 Hengzhigang Road, Guangzhou 510095, China
| | - Zhenjian Xing
- Department of Pharmacy, Guangzhou Chest Hospital, 62 Hengzhigang Road, Guangzhou 510095, China
| | - Jiaqi Liu
- Analytical Applications Center, Shimadzu (China) Co., Ltd. Guangzhou Branch, 230 Gaotang Road, Guangzhou 510656, China
| | - Rui Yu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinic al Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| | - Xin Deng
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinic al Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| |
Collapse
|
5
|
Almajidi YQ, Kadhim MM, Alsaikhan F, Turki Jalil A, Hassan Sayyid N, Alexis Ramírez-Coronel A, Hassan Jawhar Z, Gupta J, Nabavi N, Yu W, Ertas YN. Doxorubicin-loaded micelles in tumor cell-specific chemotherapy. ENVIRONMENTAL RESEARCH 2023; 227:115722. [PMID: 36948284 DOI: 10.1016/j.envres.2023.115722] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 03/13/2023] [Accepted: 03/18/2023] [Indexed: 05/08/2023]
Abstract
Nanomedicine is a field that combines biology and engineering to improve disease treatment, particularly in cancer therapy. One of the promising techniques utilized in this area is the use of micelles, which are nanoscale delivery systems that are known for their simple preparation, high biocompatibility, small particle size, and the ability to be functionalized. A commonly employed chemotherapy drug, Doxorubicin (DOX), is an effective inhibitor of topoisomerase II that prevents DNA replication in cancer cells. However, its efficacy is frequently limited by resistance resulting from various factors, including increased activity of drug efflux transporters, heightened oncogenic factors, and lack of targeted delivery. This review aims to highlight the potential of micelles as new nanocarriers for delivering DOX and to examine the challenges involved with employing chemotherapy to treat cancer. Micelles that respond to changes in pH, redox, and light are known as stimuli-responsive micelles, which can improve the targeted delivery of DOX and its cytotoxicity by facilitating its uptake in tumor cells. Additionally, micelles can be utilized to administer a combination of DOX and other drugs and genes to overcome drug resistance mechanisms and improve tumor suppression. Furthermore, micelles can be used in phototherapy, both photodynamic and photothermal, to promote cell death and increase DOX sensitivity in human cancers. Finally, the alteration of micelle surfaces with ligands can further enhance their targeted delivery for cancer suppression.
Collapse
Affiliation(s)
| | - Mustafa M Kadhim
- Department of Dentistry, Kut University College, Kut, Wasit, 52001, Iraq; Medical Laboratory Techniques Department, Al-Farahidi University, Baghdad, 10022, Iraq
| | - Fahad Alsaikhan
- College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj, Saudi Arabia
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, 51001, Iraq.
| | | | - Andrés Alexis Ramírez-Coronel
- Azogues Campus Nursing Career, Health and Behavior Research Group (HBR), Psychometry and Ethology Laboratory, Catholic University of Cuenca, Ecuador; Epidemiology and Biostatistics Research Group, CES University, Colombia; Educational Statistics Research Group(GIEE), National University of Education, Ecuador
| | - Zanko Hassan Jawhar
- Department of Medical Laboratory Science, College of Health Sciences, Lebanese French University, Erbil, Iraq; Clinical Biochemistry Department, College of Health Sciences, Hawler Medical University, Erbil, Iraq
| | - Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, Pin Code 281406, U.P, India
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6, Vancouver, BC, Canada
| | - Wei Yu
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437100, China.
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Türkiye; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Türkiye.
| |
Collapse
|
6
|
Wang M, Zhang M, Hu X, Wang W, Zhang Y, Zhang L, Wang J. Lipid-functionalized gold nanorods with plug-to-direct mitochondria targeting ligand for synergetic photothermal-chemotherapy of tumor therapy. Eur J Pharm Biopharm 2023; 185:71-81. [PMID: 36828240 DOI: 10.1016/j.ejpb.2023.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/19/2022] [Accepted: 02/19/2023] [Indexed: 02/25/2023]
Abstract
Mitochondria targeting therapeutic strategies are promising for more effective and precise cancer therapy. Photothermal therapy are extensively studied as noninvasive cancer treatment. With regards to all-in-one nanocarrier-mediated drug delivery platform, it is still a challenge to enhance one of the features but not compromise other merits. Herein, we present a mitochondrial targeting photothermal-chemotherapy all-in-one nanoplatform involving lipid-functionalized gold nanorods (AuNR) with plug-to-direct mitochondria targeting ligand for synergetic enhanced tumor therapy. Firstly, AuNR were modified by DSPE-PEG-SH owing to the special affinity of sulfhydryl group and gold. And then, DSPE-PEG-DOX with mitochondrial targeting character was directly inserted into DSPE-PEG-SH layer. Meanwhile, paclitaxel (PTX) was loaded in hydrophobic region of the lipid layer. Quite different from introducing additional mitochondrial targeting molecules, we incorporated amphiphilic DSPE-PEG-DOX into a DSPE-PEG-SH layer modified around AuNR to achieve both mitochondrial targeting, photothermal and dual drug loading in a simple AuNR-lipid-DOX/PTX platform, in the case that efficiently enhanced production of reactive oxygen species (ROS) in mitochondria and excellent anti-tumor efficacy were achieved. With good biocompatibility, the constructed nanoplatform based on lipid-functionalized AuNR synergistically combined mitochondrial targeted DSPE-PEG-DOX with mitochondrial-acted PTX and photothermal therapy (PTT), which provided a feasible strategy for organelle-targeted combination PTT-chemotherapy to improve therapeutic effects.
Collapse
Affiliation(s)
- Mi Wang
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Mo Zhang
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Xiaoxiao Hu
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Wenli Wang
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Yao Zhang
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Lina Zhang
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, People's Republic of China
| | - Jing Wang
- School of Pharmacy, Key Laboratory of Innovative Drug Development and Evaluation, Hebei Medical University, Shijiazhuang 050017, People's Republic of China.
| |
Collapse
|
7
|
Du K, Xia QS, Zhang LH, Wen J, Huang Z, Zhu ZS. Copolymers induced co-assembly for constructing novel micellar carriers by computer simulations. Chem Phys Lett 2022. [DOI: 10.1016/j.cplett.2022.139874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
8
|
Sun L, Zhao P, Chen M, Leng J, Luan Y, Du B, Yang J, Yang Y, Rong R. Taxanes prodrug-based nanomedicines for cancer therapy. J Control Release 2022; 348:672-691. [PMID: 35691501 DOI: 10.1016/j.jconrel.2022.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/04/2022] [Accepted: 06/04/2022] [Indexed: 11/16/2022]
Abstract
Malignant tumor remains a huge threat to human health and chemotherapy still occupies an important place in clinical tumor treatment. As a kind of potent antimitotic agent, taxanes act as the first-line broad-spectrum cancer drug in clinical use. However, disadvantages such as prominent hydrophobicity, severe off-target toxicity or multidrug resistance lead to unsatisfactory therapeutic effects, which restricts its wider usage. The efficient delivery of taxanes is still quite a challenge despite the rapid developments in biomaterials and nanotechnology. Great progress has been made in prodrug-based nanomedicines (PNS) for cancer therapy due to their outstanding advantages such as high drug loading efficiency, low carrier induced immunogenicity, tumor stimuli-responsive drug release, combinational therapy and so on. Based on the numerous developments in this filed, this review summarized latest updates of taxanes prodrugs-based nanomedicines (TPNS), focusing on polymer-drug conjugate-based nanoformulations, small molecular prodrug-based self-assembled nanoparticles and prodrug-encapsulated nanosystems. In addition, the new trends of tumor stimuli-responsive TPNS were also discussed. Moreover, the future challenges of TPNS for clinical translation were highlighted. We here expect this review will inspire researchers to explore more practical taxanes prodrug-based nano-delivery systems for clinical use.
Collapse
Affiliation(s)
- Linlin Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Pan Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Menghan Chen
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Jiayi Leng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yixin Luan
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Baoxiang Du
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Jia Yang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yong Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| |
Collapse
|
9
|
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
|
10
|
Chen G, Zheng Q, Dai J, Liu J, Yin J, Xu X, Chen A, Ren L. Reduction-sensitive mixed micelles based on mPEG-SS-PzLL /TPGS to enhance anticancer efficiency of doxorubicin. REACT FUNCT POLYM 2022. [DOI: 10.1016/j.reactfunctpolym.2022.105242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
11
|
Chen X, Ji J, Zhou K, Fan X, Li L, Xu W. A novel multifunctional nanoparticles formed by molecular recognition between AS1411 aptamer and redox-responsive paclitaxel-nucleoside analogue prodrug for combination treatment of β-lapachone and paclitaxel. Colloids Surf B Biointerfaces 2022; 212:112345. [PMID: 35074639 DOI: 10.1016/j.colsurfb.2022.112345] [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: 11/24/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 10/19/2022]
Abstract
Despite its high antitumor activity, the clinical application of chemotherapy is greatly impeded by lacking of specific accumulation and poor solubility. To address the above challenges, we designed a AS1411 aptamer modified nanoparticles based on molecular recognition of nucleobases. Firstly, a redox sensitive Paclitaxel-SS-Zidovudine (PZ) prodrug was synthesized. Then PZ/β-lapachone/AS1411/DSPE-PEG nanoparticles were prepared and AS1411 aptamer was connected through molecular recognition between the nucleoside analogue Zidovudine (ZDV) and the thymine on aptamer. DSPE-PEG (DP) was incorporated into nanoparticles to prolong the residence time of nanoparticles in the blood circulation. Furthermore, to realize the combination treatment, β-lapachone (LAP) has been incorporated into nanoparticles with high drug loading efficiency through the interaction of π-π stacking force and H-bonding between LAP and Paclitaxel (PTX). LAP can generate abundant exogenous reactive oxygen species (ROS) via the bioactivation of NAD(P)H: quinone oxidoreductase-1 (NQO1). Moreover, the connection of Zidovudine (ZDV) and AS1411 through molecular recognition of nucleobases further optimized the nanoparticles with high affinity to nucleolin which overexpressed on tumor cell membrane, thereby inducing the specific accumulation of nanoparticles in tumor sites. In vivo and in vitro studies showed that the obtained nanoparticles of PZ/LAP/AS1411/DP exhibited better tumor growth inhibition and lower systemic side effects. Herein, we have rationally conducted a novel self-codelivery system for effectively synergistic antitumor treatment.
Collapse
Affiliation(s)
- Xuling Chen
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China
| | - Ke Zhou
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China
| | | | - Lingbing Li
- Department of Pharmaceutics, Key Laboratory of Chemical Biology of Ministry of Education, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, Shandong Province 250012, China.
| | - Wei Xu
- Shandong Provincial Qianfoshan Hospital, the First Hospital Affiliated with Shandong First Medical University, China; Shandong Provincial Qianfoshan Hospital, Shandong University, China.
| |
Collapse
|
12
|
Li Y, Zhu C, Wang Y, Wen F, Zhang X. Tumor reduction-sensitive self-delivery molecular prodrug nanomedicine for enhancing the therapeutic efficacy of chemotherapy. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2021.128106] [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]
|
13
|
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
|
14
|
Li Y, Hou H, Zhang P, Zhang Z. Co-delivery of doxorubicin and paclitaxel by reduction/pH dual responsive nanocarriers for osteosarcoma therapy. Drug Deliv 2021; 27:1044-1053. [PMID: 32633576 PMCID: PMC7470123 DOI: 10.1080/10717544.2020.1785049] [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: 12/22/2022] Open
Abstract
Nanoparticle-based drug delivery system offers a promising platform for combination cancer therapy. However, the inefficient drug release in cells reduces the therapeutic efficacy of cancer nanomedicines. Herein, a PEGylated poly(α-lipoic acid) copolymer (mPEG-PαLA) was prepared and used as a reduction/pH dual responsive nanocarrier to simultaneously deliver paclitaxel (PTX) and doxorubicin (DOX) for osteosarcoma therapy. The amphiphilic mPEG-PαLA could efficiently encapsulate both PTX and DOX during its self-assembly into micelles in aqueous solution to generate PTX and DOX co-loaded nanoparticles (NP-PTX-DOX). The as-prepared NP-PTX-DOX showed enhanced PTX and DOX release in response to reductive and acidic stimuli. Moreover, the dual-drug loaded nanoparticles were efficiently internalized by K7 osteosarcoma cells and released drugs intracellularly, as confirmed by flow cytometry analysis and confocal laser scanning microscopy. Consequently, NP-PTX-DOX exhibited synergistic therapeutic effects and induced enhanced cell apoptosis in K7 cells. Furthermore, NP-PTX-DOX presented improved biodistribution and higher tumor growth inhibition efficacy compared to the control groups in a murine osteosarcoma model. Altogether, the results of this work indicate that the proposed strategy is promising for osteosarcoma therapy using mPEG-PαLA copolymer as a dual-responsive nanocarrier to co-deliver anticancer drugs.
Collapse
Affiliation(s)
- Yongshuang Li
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, P. R. China
| | - Hao Hou
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, P. R. China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P. R. China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, P. R. China
| |
Collapse
|
15
|
Wu T, Gong Y, Li Z, Li Y, Xiong X. Preparation and in vitro/vivo evaluation of folate-conjugated pluronic F87-PLGA/TPGS mixed nanoparticles for targeted drug delivery. Curr Drug Deliv 2021; 18:1505-1514. [PMID: 33845742 DOI: 10.2174/1567201818666210412123210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 02/01/2021] [Accepted: 02/22/2021] [Indexed: 11/22/2022]
Abstract
AIM Folate-conjugated Pluronic F87-poly(lactic-co-glycolic acid) block copolymer (FA-F87-PLGA) was synthesized to encapsulate anticancer drug Paclitaxel (PTX) for targeted drug delivery. To further improve the curative effect, D-α-tocopheryl poly(ethylene glycol) 1000 succinate (TPGS or Vitamin E TPGS) was added to form FA-F87-PLGA/TPGS mixed NPs. METHODS FA-F87-PLGA was synthesized by the ring-opening polymerization and the structure was characterized. PTX-loaded nanoparticles were prepared with the nanoprecipitation method. The physicochemical characteristics were studied to determine the appropriate dose ratio of the FA-F87-PLGA to TPGS. The cytotoxicity against Ovarian Cancer Cells (OVCAR-3) was determined by MTT assay. The Area-Under-the Curve (AUC) and half-life were measured in the vivo pharmacokinetic studies. RESULTS Based on the optimization of particle size and embedding rate of PTX-loaded mixed NPs, the appropriate dosage ratio of FA-F87-PLGA to TPGS was finally determined to be 5:3. According to in vitro release studies, the cumulative release rate of PTX-loaded FA-F87-PLGA/TPGS mixed NPs was 92.04%, which was higher than that of nanoparticles without TPGS. The cytotoxicity studies showed that the IC50 value of PTX-loaded FA-F87-PLGA/TPGS decreased by 75.4 times and 19.7 times after 72 h treatment compared with free PTX injections and PTX-loaded FA-F87-PLGA NPs, respectively. In vivo pharmacokinetic studies indicated that FA-F87-PLGA/TPGS mixed NPs had a longer drug metabolism time and a larger Area-Under-the-Curve (AUC) compared with free PTX injections. CONCLUSION FA-F87-PLGA/TPGS mixed NPs are potential candidates for targeted drug delivery systems.
Collapse
Affiliation(s)
- Tianyi Wu
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi. China
| | - Yanchun Gong
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi. China
| | - Ziling Li
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi. China
| | - Yuping Li
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi. China
| | - Xiangyuan Xiong
- School of Life Science, Jiangxi Science and Technology Normal University, Nanchang, Jiangxi. China
| |
Collapse
|
16
|
Li X, Hou S, Chen J, He CE, Gao YE, Lu Y, Jia D, Ma X, Xue P, Kang Y, Xu Z. Engineering silk sericin decorated zeolitic imidazolate framework-8 nanoplatform to enhance chemotherapy. Colloids Surf B Biointerfaces 2021; 200:111594. [PMID: 33561693 DOI: 10.1016/j.colsurfb.2021.111594] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/14/2021] [Accepted: 01/25/2021] [Indexed: 12/26/2022]
Abstract
The low therapeutic effect and strong side-effect are the major barriers for clinical chemotherapy. Herein, a pH-responsive nanoplatform based-silk sericin-zeolitic imidazolate framework-8 was designed for the delivery of chemotherapeutic doxorubicin (denoted as ZIF-8@DOX@SS, ZDS), which can overcome the limitation of poor circulation stability and unexpected drug leakage in blood circulation, producing a satisfactory chemotherapy. Concretely, ZIF-8 structure shows better stability and biocompatibility owing to the protection of a nature and non-toxic sericin protein. When it comes to low pH environment (e.g. in tumor cell microenvironment), the coordination effect in ZIF-8 will be broken and release DOX drugs. The nano-sized morphology endow ZDS an efficient drug uptake and significant tumor permeability efficiency. Furthermore, the tumor-specific biodegradability makes ZDS possible to realize targeted and enhanced chemotherapy. Considering all the advantages in the study, this silk sericin-based nanosystem represent a promising strategy for the design of stimuli-responsive by using natural polymer to improve the treatment effect of chemotherapy.
Collapse
Affiliation(s)
- Xinyi Li
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China
| | - Shengxin Hou
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China
| | - Jiucun Chen
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China.
| | - Cai-E He
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China
| | - Yong-E Gao
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China
| | - Yi Lu
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China
| | - Die Jia
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China
| | - Xianbin Ma
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China
| | - Peng Xue
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China
| | - Yuejun Kang
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China
| | - Zhigang Xu
- State Key Laboratory of Silkworm Genome Biology, School of Materials and Energy, Southwest University, Chongqing, 400715, PR China; Chongqing Engineering Research Center for Micro-Nano Biomedical Materials and Devices, Southwest University, Chongqing, 400715, PR China.
| |
Collapse
|
17
|
Dahri M, Abolmaali SS, Abedanzadeh M, Salmanpour M, Maleki R. Composition and surface chemistry engineering of graphene grafting chitosan for stimuli-responsive cancer therapy: An in-silico study. INFORMATICS IN MEDICINE UNLOCKED 2021. [DOI: 10.1016/j.imu.2021.100627] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
18
|
Chen D, Ge S, Zuo L, Wang S, Liu M, Li S. Adjudin-loaded redox-sensitive paclitaxel-prodrug micelles for overcoming multidrug resistance with efficient targeted Colon cancer therapy. Drug Deliv 2020; 27:1094-1105. [PMID: 32706289 PMCID: PMC7470106 DOI: 10.1080/10717544.2020.1797245] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023] Open
Abstract
Multidrug resistance (MDR) is the primary cause for the failure of chemotherapy in the treatment of colon cancer. Recent research has indicated that the combination of a chemotherapeutic agent and a mitochondrial inhibitor might represent a promising strategy to help overcome MDR. However, for this approach to be clinically effective, it is important that the two drugs can be actively and simultaneously delivered into tumor cells at an optimal ratio and completely released drug within cells. To address these challenges, we designed and prepared a folate receptor-targeted and redox-responsive drug delivery system (FA- ss -P/A) that was able to co-deliver paclitaxel (PTX) and adjudin (ADD) to reverse colon cancer MDR. The PTX prodrug was obtained by conjugating PTX to dextrin via a disulfide-linkage. Then, folic acid (FA) was modified on the PTX prodrug. Finally, ADD, a mitochondrial inhibitor, was encapsulated in the PTX prodrug-formed micelles. A series of in vitro and in vivo experiments subsequently demonstrated that FA- ss -P/A can effectively reverse MDR by increasing cell uptake, inhibiting PTX efflux, and improving drug release.
Collapse
Affiliation(s)
- Deli Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Sitang Ge
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Lugen Zuo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Shuanhu Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Mulin Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Shiqing Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| |
Collapse
|
19
|
Van Gheluwe L, Buchy E, Chourpa I, Munnier E. Three-Step Synthesis of a Redox-Responsive Blend of PEG- block-PLA and PLA and Application to the Nanoencapsulation of Retinol. Polymers (Basel) 2020; 12:E2350. [PMID: 33066418 PMCID: PMC7602167 DOI: 10.3390/polym12102350] [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: 09/14/2020] [Revised: 10/01/2020] [Accepted: 10/12/2020] [Indexed: 01/06/2023] Open
Abstract
Smart polymeric nanocarriers have been developed to deliver therapeutic agents directly to the intended site of action, with superior efficacy. Herein, a mixture of poly(lactide) (PLA) and redox-responsive poly(ethylene glycol)-block-poly(lactide) (PEG-block-PLA) containing a disulfide bond was synthesized in three steps. The nanoprecipitation method was used to prepare an aqueous suspension of polymeric nanocarriers with a hydrodynamic diameter close to 100 nm. Retinol, an anti-aging agent very common in cosmetics, was loaded into these smart nanocarriers as a model to measure their capacity to encapsulate and to protect a lipophilic active molecule. Retinol was encapsulated with a high efficiency with final loading close to 10% w/w. The stimuli-responsive behavior of these nanocarriers was demonstrated in vitro, in the presence of l-Glutathione, susceptible to break of disulfide bond. The toxicity was low on human keratinocytes in vitro and was mainly related to the active molecule. Those results show that it is not necessary to use 100% of smart copolymer in a nanosystem to obtain a triggered release of their content.
Collapse
Affiliation(s)
- Louise Van Gheluwe
- EA 6295 Nanomédicaments et Nanosondes, Faculté de Pharmacie, Université de Tours, 31 Avenue Monge, 37 200 Tours, France; (L.V.G.); (I.C.)
| | - Eric Buchy
- Laboratoires Eriger, 39 Rue des Granges Galand, 37550 Saint-Avertin, France;
| | - Igor Chourpa
- EA 6295 Nanomédicaments et Nanosondes, Faculté de Pharmacie, Université de Tours, 31 Avenue Monge, 37 200 Tours, France; (L.V.G.); (I.C.)
| | - Emilie Munnier
- EA 6295 Nanomédicaments et Nanosondes, Faculté de Pharmacie, Université de Tours, 31 Avenue Monge, 37 200 Tours, France; (L.V.G.); (I.C.)
| |
Collapse
|
20
|
Wang Q, Gao F, Zhou X. Redox-responsive AIE micelles for intracellular paclitaxel delivery. Colloid Polym Sci 2020. [DOI: 10.1007/s00396-020-04679-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
21
|
Hyaluronic acid targeted and pH-responsive nanocarriers based on hollow mesoporous silica nanoparticles for chemo-photodynamic combination therapy. Colloids Surf B Biointerfaces 2020; 194:111166. [PMID: 32521461 DOI: 10.1016/j.colsurfb.2020.111166] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 05/24/2020] [Accepted: 06/01/2020] [Indexed: 12/18/2022]
Abstract
In this work, a pH-responsive and tumor targeted multifunctional drug delivery system (RB-DOX@HMSNs-N = C-HA) was designed to realize chemo-photodynamic combination therapy. Hollow mesoporous silica nanoparticles (HMSNs) was served as the host material to encapsulate doxorubicin (DOX) and photosensitizer rose bengal (RB). Hyaluronic acid (HA) was modified on the surface of HMSNs via pH-sensitive Schiff base bonds as gatekeeper as well as targeted agent. Characterization results indicated the successful preparation of HMSNs-N = C-HA with appropriate diameter of 170 nm around and the nanocarriers displayed superior drug loading capacity (15.30 % for DOX and 12.78 % for RB). Notably, the results of in vitro drug release experiments confirmed that the system possessed good pH-sensitivity, which made it possible to release cargoes in slight acid tumor micro-environments. Significantly, the in vitro cell uptake and cytotoxicity assay results fully proved that RB-DOX@HMSNs-N = C-HA could precisely target murine mammary carcinoma (4T1) cells and effectively inhibit tumor cells viability with chemo-photodynamic synergistic therapy. Overall, our work (RB-DOX@HMSNs-N = C-HA) provides an efficient approach for the development of chemo-photodynamic combination therapy.
Collapse
|
22
|
Tumor-targeted and self-assembled mixed micelles as carriers for enhanced anticancer efficacy of gemcitabine. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
23
|
Liu S, Li R, Qian J, Sun J, Li G, Shen J, Xie Y. Combination Therapy of Doxorubicin and Quercetin on Multidrug-Resistant Breast Cancer and Their Sequential Delivery by Reduction-Sensitive Hyaluronic Acid-Based Conjugate/d-α-Tocopheryl Poly(ethylene glycol) 1000 Succinate Mixed Micelles. Mol Pharm 2020; 17:1415-1427. [PMID: 32159961 DOI: 10.1021/acs.molpharmaceut.0c00138] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The therapeutic efficacy of chemotherapy in many types of hematological malignancies and solid tumors is dramatically hindered by multidrug resistance (MDR). This work presents a combination strategy of pretreatment of MDA-MB-231/MDR1 cells with quercetin (QU) followed by doxorubicin (DOX) to overcome MDR, which can be delivered by mixed micelles composed of the reduction-sensitive hyaluronic acid-based conjugate and d-α-tocopheryl poly(ethylene glycol) 1000 succinate. The combination strategy can enhance the cytotoxicity of DOX on MDA-MB-231/MDR1 cells by increasing intracellular DOX accumulation and facilitating DOX-induced apoptosis. The probable MDR reversal mechanisms are that the pretreatment cells with QU-loaded mixed micelles downregulate P-glycoprotein expression to decrease DOX efflux as well as initiate mitochondria-dependent apoptotic pathways to accelerate DOX-induced apoptosis. In addition, this combination strategy can not only potentiate in vivo tumor-targeting efficiency but also enhance the antitumor effect in MDA-MB-231/MDR1-bearing nude mice without toxicity or side effects. This research suggests that the co-administration of natural compounds and chemotherapeutic drugs could be an effective strategy to overcome tumor MDR, which deserves further exploration.
Collapse
Affiliation(s)
- Shuo Liu
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Rui Li
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Jin Qian
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Jiabin Sun
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Guowen Li
- Pharmacy Department, Shanghai TCM-integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Jianliang Shen
- School of Ophthalmology & Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, Zhejiang 325035, China.,Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Yan Xie
- Research Center for Health and Nutrition, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| |
Collapse
|
24
|
Roque MC, Franco MS, Vilela JMC, Andrade MS, de Barros ALB, Leite EA, Oliveira MC. Development of Long-Circulating and Fusogenic Liposomes Co-encapsulating Paclitaxel and Doxorubicin in Synergistic Ratio for the Treatment of Breast Cancer. Curr Drug Deliv 2019; 16:829-838. [DOI: 10.2174/1567201816666191016112717] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/26/2019] [Accepted: 09/19/2019] [Indexed: 11/22/2022]
Abstract
Background:
The co-encapsulation of paclitaxel (PTX) and doxorubicin (DXR) in liposomes
has the potential to offer pharmacokinetic and pharmacodynamic advantages, providing delivery of both
drugs to the tumor at the ratio required for synergism.
Objective:
To prepare and characterize long-circulating and fusogenic liposomes co-encapsulating PTX
and DXR in the 1:10 molar ratio (LCFL-PTX/DXR).
Methods:
LCFL-PTX/DXR was prepared by the lipid film formation method. The release of PTX and
DXR from liposomes was performed using a dialysis method. Studies of cytotoxicity, synergism, and
cellular uptake were also carried out.
Results:
The encapsulation percentage of PTX and DXR was 74.1 ± 1.8 % and 89.6 ± 12.3%, respectively,
and the mean diameter of the liposomes was 244.4 ± 28.1 nm. The vesicles remained stable for
30 days after their preparation. The drugs were simultaneously released from vesicles during 36 hours,
maintaining the drugs combination in the previously established ratio. Cytotoxicity studies using 4T1
breast cancer cells showed lower inhibitory concentration 50% (IC50) value for LCFL-PTX/DXR treatment
(0.27 ± 0.11 µm) compared to the values of free drugs treatment. In addition, the combination
index (CI) assessed for treatment with LCFL-PTX/DXR was equal to 0.11 ± 0.04, showing strong synergism
between the drugs. Cell uptake studies have confirmed that the molar ratio between PTX and
DXR is maintained when the drugs are administered in liposomes.
Conclusion:
It was possible to obtain LCFL-PTX/DXR suitable for intravenous administration, capable
of releasing the drugs in a fixed synergistic molar ratio in the tumor region.
Collapse
Affiliation(s)
- Marjorie Coimbra Roque
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marina Santiago Franco
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | | | - André Luís Branco de Barros
- Department of Clinical and Toxicological Analyzes, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Elaine Amaral Leite
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mônica Cristina Oliveira
- Department of Pharmaceutical Products, Faculty of Pharmacy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
25
|
Khan DH, Bashir S, Correia A, Khan MI, Figueiredo P, Santos HA, Peltonen L. Utilization of green formulation technique and efficacy estimation on cell line studies for dual anticancer drug therapy with niosomes. Int J Pharm 2019; 572:118764. [PMID: 31628977 DOI: 10.1016/j.ijpharm.2019.118764] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 09/11/2019] [Accepted: 10/02/2019] [Indexed: 01/22/2023]
Abstract
The aim of the present study was to prepare niosome formulations for the simultaneous encapsulation, dual drug therapy, of two anticancer drugs by the ecological probe sonication method. Poloxamer and sorbitan monostearate were used as surface active agents in niosomes, and the water soluble doxorubicin and poorly-water soluble paclitaxel were used as anticancer drugs. Thorough physicochemical analysis were performed for the niosomes, and their cytotoxicity and activity were evaluated on MCF-7 and PC3-MM2 cancer cell lines. Prepared niosomes were small in size with sizes ranging from 137 nm to 893 nm, and entrapment efficiencies were high, ranging from 91.24% to 99.99%. During the four weeks stability testing, the particle size remained stable. The niosomal formulations showed in vitro sustained drug release profiles for doxorubicin and clearly increased the dissolution rate of poorly water soluble paclitaxel. The incorporation of both the drugs into niosomes improved cell penetration and antiproliferative activity of the drugs PC3-MM2 cell lines. As a conclusion, doxorubicin and paclitaxel loaded niosome formulations resulted in relatively stable, small sized niosomes with improved drug release profiles, low toxicity, better cell penetration and antiproliferative activity. The niosomes showed synergistic effect due to the presence of both drugs, which can overcome multidrug resistance.
Collapse
Affiliation(s)
- Daulat Haleem Khan
- College of Pharmacy, University of Sargodha, Sargodha, Pakistan; Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, FI-00014, University of Helsinki, Finland; Lahore College of Pharmaceutical Sciences, 54000 Lahore, Pakistan
| | - Sajid Bashir
- College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Alexandra Correia
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, FI-00014, University of Helsinki, Finland
| | - Muhammad Imran Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, 54000 Lahore, Pakistan
| | - Patrícia Figueiredo
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, FI-00014, University of Helsinki, Finland
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, FI-00014, University of Helsinki, Finland; Helsinki Institute of Life Science (HiLIFE), FI-00014, University of Helsinki, Finland
| | - Leena Peltonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, FI-00014, University of Helsinki, Finland.
| |
Collapse
|
26
|
Wang S, Liu M, Wang W, Li T, Cui M, Sun W, Yang X, Song S. Preparation and Evaluation of mPEG-PLGA Block Copolymer Micelles Loaded with a Sarsasapogenin Derivative. AAPS PharmSciTech 2019; 20:280. [PMID: 31399832 DOI: 10.1208/s12249-019-1491-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/23/2019] [Indexed: 02/07/2023] Open
Abstract
Sarsasapogenin derivative 5n (SGD 5n) is a new compound with potent antitumor efficacy, but the low solubility severely affects its absorption and bioavailability. Therefore, the SGD 5n-loaded mPEG-PLGA block copolymer micelles were developed to improve the value of SGD 5n in clinical application. The polymeric micelles were prepared by an organic solvent evaporation method, and the encapsulation efficiency (EE), drug loading (DL), critical micelle concentrations (CMC), morphology, particle size, and zeta potential were determined. The cytotoxicity was examined by the MTT assay, and the cellular uptake study was performed by confocal laser scanning microscopy. A model of tumor-bearing mouse was established to study the antitumor activity in vivo. The results demonstrated that the particle size of the prepared micelle was 124.6 ± 9.6 nm, the encapsulation efficiency was 82.0 ± 2.9%, and the drug loading was 8.3 ± 0.4%. The results of cytotoxicity and cellular uptake demonstrated that the SGD 5n-loaded micelles could efficiently enter tumor cells, and the cellular uptake of SGD 5n presented concentration and time dependence. This study demonstrated that the prepared SGD 5n-loaded polymeric micelles had significant antitumor activity and provided a basis for clinical development of new compound SGD 5n.
Collapse
|
27
|
Zeng YY, Zeng YJ, Zhang NN, Li CX, Xie T, Zeng ZW. The Preparation, Determination of a Flexible Complex Liposome Co-Loaded with Cabazitaxel and β-Elemene, and Animal Pharmacodynamics on Paclitaxel-Resistant Lung Adenocarcinoma. Molecules 2019; 24:E1697. [PMID: 31052317 PMCID: PMC6539285 DOI: 10.3390/molecules24091697] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 04/25/2019] [Accepted: 04/30/2019] [Indexed: 01/31/2023] Open
Abstract
Paclitaxel is highly effective at killing many malignant tumors; however, the development of drug resistance is common in clinical applications. The issue of overcoming paclitaxel resistance is a difficult challenge at present. In this study, we developed nano drugs to treat paclitaxel-resistant lung adenocarcinoma. We selected cabazitaxel and β-elemene, which have fewer issues with drug resistance, and successfully prepared cabazitaxel liposome, β-elemene liposome and cabazitaxel-β-elemene complex liposome with good flexibility. The encapsulation efficiencies of cabazitaxel and β-elemene in these liposomes were detected by precipitation microfiltration and microfiltration centrifugation methods, respectively. Their encapsulation efficiencies were all above 95%. The release rates were detected by a dialysis method. The release profiles of cabazitaxel and β-elemene in these liposomes conformed to the Weibull equation. The release of cabazitaxel and β-elemene in the complex liposome were almost synchronous. The pharmacodynamics study showed that cabazitaxel flexible liposome and β-elemene flexible liposome were relatively good at overcoming paclitaxel resistance on paclitaxel-resistant lung adenocarcinoma. As the flexible complex liposome, the dosage of cabazitaxel could be reduced to 25% that of the cabazitaxel injection while retaining a similar therapeutic effect. It showed that β-elemene can replace some of the cabazitaxel, allowing the dosage of cabazitaxel to be reduced, thereby reducing the drug toxicity.
Collapse
Affiliation(s)
- Yi-Ying Zeng
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China.
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, 1378 Wenyi Road, Hangzhou 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, China.
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, China.
| | - Yi-Jun Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, 1378 Wenyi Road, Hangzhou 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, China.
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, China.
| | - Na-Na Zhang
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, 1378 Wenyi Road, Hangzhou 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, China.
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, China.
| | - Chen-Xi Li
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, 1378 Wenyi Road, Hangzhou 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, China.
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, China.
| | - Tian Xie
- Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China.
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, 1378 Wenyi Road, Hangzhou 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, China.
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, China.
| | - Zhao-Wu Zeng
- Holistic Integrative Pharmacy Institutes, Hangzhou Normal University, 1378 Wenyi Road, Hangzhou 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicine of Zhejiang Province, Hangzhou 311121, China.
- Engineering Laboratory of Development and Application of Traditional Chinese Medicine from Zhejiang Province, Hangzhou 311121, China.
| |
Collapse
|
28
|
Eftekhari RB, Maghsoudnia N, Samimi S, Zamzami A, Dorkoosh FA. Co-Delivery Nanosystems for Cancer Treatment: A Review. Pharm Nanotechnol 2019; 7:90-112. [PMID: 30907329 DOI: 10.2174/2211738507666190321112237] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/08/2019] [Accepted: 03/18/2019] [Indexed: 06/09/2023]
Abstract
Massive data available on cancer therapy more than ever lead our mind to the general concept that there is no perfect treatment for cancer. Indeed, the biological complexity of this disease is too excessive to be treated by a single therapeutic approach. Current delivery systems containing a specific drug or gene have their particular opportunities and restrictions. It is worth noting that a considerable number of studies suggest that single- drug delivery systems result in insufficient suppression of cancer growth. Therefore, one of the main ideas of co-delivery system designing is to enhance the intended response or to achieve the synergistic/combined effect compared to the single drug strategy. This review focuses on various strategies for co-delivery of therapeutic agents in the treatment of cancer. The primary approaches within the script are categorized into co-delivery of conventional chemotherapeutics, gene-based molecules, and plant-derived materials. Each one is explained in examples with the recent researches. In the end, a brief summary is provided to conclude the gist of the review.
Collapse
Affiliation(s)
- Reza Baradaran Eftekhari
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloufar Maghsoudnia
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Shabnam Samimi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Zamzami
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Farid Abedin Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Medical Biomaterial Research Center (MBRC), Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|