1
|
Cui J, Hu B, Fu Y, Xu Z, Li Y. pH-Sensitive nanodiamond co-delivery of retinal and doxorubicin boosts breast cancer chemotherapy. RSC Adv 2023; 13:27403-27414. [PMID: 37711368 PMCID: PMC10498152 DOI: 10.1039/d3ra03907b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/28/2023] [Indexed: 09/16/2023] Open
Abstract
Herein for the first time we take the advantage of nanodiamonds (NDs) to covalently immobilize all-trans retinal (NPA) by an imine bond, allowing pH-mediated drug release. DOX is then physically adsorbed onto NPA to form an NPA@D co-loaded double drug in the sodium citrate medium, which is also susceptible to pH-triggered DOX dissociation. The cytotoxicity results showed that NPA@D could markedly inhibit the growth of DOX-sensitive MCF-7 cells in a synergetic way compared to the NP@D system of single-loaded DOX, while NPA basically showed no cytotoxicity and weak inhibition of migration. In addition, NPA@D can overcome the drug resistance of MCF-7/ADR cells, indicating that this nanodrug could evade the pumping of DOX by drug-resistant cells, but free DOX is nearly ineffective against these cells. More importantly, the fluorescence imaging of tumor-bearing mice in vivo and ex vivo demonstrated that the NPA@D was mainly accumulated in the tumor site rather than any other organ by intraperitoneal injection after 24 h, in which the fluorescence intensity of NPA@D was 19 times that of the free DOX, suggesting that a far reduced off-target effect and side effects would be expected. Therefore, this work presents a new paradigm for improving chemotherapy and reversing drug resistance using the ND platform for co-delivery of DOX and ATR.
Collapse
Affiliation(s)
- Jicheng Cui
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University Taiyuan 030006 P. R. China
| | - Bo Hu
- China Institute for Radiation Protection Taiyuan 030006 P. R. China
| | - Yuejun Fu
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University Taiyuan 030006 China
| | - Zhengkun Xu
- Faculty of Science, McMaster University Hamilton L8S 4K1 ON Canada
| | - Yingqi Li
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Molecular Science, Shanxi University Taiyuan 030006 P. R. China
- School of Chemistry and Chemical Engineering, Shanxi University Taiyuan 030006 PR China
| |
Collapse
|
2
|
Fu CP, Cai XY, Chen SL, Yu HW, Fang Y, Feng XC, Zhang LM, Li CY. Hyaluronic Acid-Based Nanocarriers for Anticancer Drug Delivery. Polymers (Basel) 2023; 15:polym15102317. [PMID: 37242892 DOI: 10.3390/polym15102317] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Hyaluronic acid (HA), a main component of the extracellular matrix, is widely utilized to deliver anticancer drugs due to its biocompatibility, biodegradability, non-toxicity, non-immunogenicity and numerous modification sites, such as carboxyl and hydroxyl groups. Moreover, HA serves as a natural ligand for tumor-targeted drug delivery systems, as it contains the endocytic HA receptor, CD44, which is overexpressed in many cancer cells. Therefore, HA-based nanocarriers have been developed to improve drug delivery efficiency and distinguish between healthy and cancerous tissues, resulting in reduced residual toxicity and off-target accumulation. This article comprehensively reviews the fabrication of anticancer drug nanocarriers based on HA in the context of prodrugs, organic carrier materials (micelles, liposomes, nanoparticles, microbubbles and hydrogels) and inorganic composite nanocarriers (gold nanoparticles, quantum dots, carbon nanotubes and silicon dioxide). Additionally, the progress achieved in the design and optimization of these nanocarriers and their effects on cancer therapy are discussed. Finally, the review provides a summary of the perspectives, the lessons learned so far and the outlook towards further developments in this field.
Collapse
Affiliation(s)
- Chao-Ping Fu
- Institute of Biomaterials and Tissue Engineering & Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
- College of Materials Science & Engineering, Huaqiao University, Xiamen 361021, China
- State Key Laboratory of Molecular Engineering of Polymers (Fudan University), Shanghai 200438, China
| | - Xing-Yu Cai
- Institute of Biomaterials and Tissue Engineering & Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
- College of Materials Science & Engineering, Huaqiao University, Xiamen 361021, China
| | - Si-Lin Chen
- Institute of Biomaterials and Tissue Engineering & Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
- College of Materials Science & Engineering, Huaqiao University, Xiamen 361021, China
| | - Hong-Wei Yu
- Institute of Biomaterials and Tissue Engineering & Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
- College of Materials Science & Engineering, Huaqiao University, Xiamen 361021, China
| | - Ying Fang
- College of Materials Science & Engineering, Huaqiao University, Xiamen 361021, China
| | - Xiao-Chen Feng
- College of Materials Science & Engineering, Huaqiao University, Xiamen 361021, China
| | - Li-Ming Zhang
- School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China
| | - Chang-Yong Li
- Institute of Biomaterials and Tissue Engineering & Fujian Provincial Key Laboratory of Biochemical Technology, Huaqiao University, Xiamen 361021, China
| |
Collapse
|
3
|
Junnuthula V, Kolimi P, Nyavanandi D, Sampathi S, Vora LK, Dyawanapelly S. Polymeric Micelles for Breast Cancer Therapy: Recent Updates, Clinical Translation and Regulatory Considerations. Pharmaceutics 2022; 14:1860. [PMID: 36145608 PMCID: PMC9501124 DOI: 10.3390/pharmaceutics14091860] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/15/2022] [Accepted: 09/01/2022] [Indexed: 12/13/2022] Open
Abstract
With the growing burden of cancer, parallel advancements in anticancer nanotechnological solutions have been witnessed. Among the different types of cancers, breast cancer accounts for approximately 25% and leads to 15% of deaths. Nanomedicine and its allied fields of material science have revolutionized the science of medicine in the 21st century. Novel treatments have paved the way for improved drug delivery systems that have better efficacy and reduced adverse effects. A variety of nanoformulations using lipids, polymers, inorganic, and peptide-based nanomedicines with various functionalities are being synthesized. Thus, elaborate knowledge of these intelligent nanomedicines for highly promising drug delivery systems is of prime importance. Polymeric micelles (PMs) are generally easy to prepare with good solubilization properties; hence, they appear to be an attractive alternative over the other nanosystems. Although an overall perspective of PM systems has been presented in recent reviews, a brief discussion has been provided on PMs for breast cancer. This review provides a discussion of the state-of-the-art PMs together with the most recent advances in this field. Furthermore, special emphasis is placed on regulatory guidelines, clinical translation potential, and future aspects of the use of PMs in breast cancer treatment. The recent developments in micelle formulations look promising, with regulatory guidelines that are now more clearly defined; hence, we anticipate early clinical translation in the near future.
Collapse
Affiliation(s)
| | - Praveen Kolimi
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA
| | - Dinesh Nyavanandi
- Pharmaceutical Development Services, Thermo Fisher Scientific, Cincinnati, OH 45237, USA
| | - Sunitha Sampathi
- GITAM School of Pharmacy, GITAM Deemed to be University, Hyderabad 502329, India
| | | | - Sathish Dyawanapelly
- Department of Pharmaceutical Science and Technology, Institute of Chemical Technology, Mumbai 400019, India
| |
Collapse
|
4
|
Curcio M, Vittorio O, Bell JL, Iemma F, Nicoletta FP, Cirillo G. Hyaluronic Acid within Self-Assembling Nanoparticles: Endless Possibilities for Targeted Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12162851. [PMID: 36014715 PMCID: PMC9413373 DOI: 10.3390/nano12162851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/03/2022] [Accepted: 08/16/2022] [Indexed: 05/27/2023]
Abstract
Self-assembling nanoparticles (SANPs) based on hyaluronic acid (HA) represent unique tools in cancer therapy because they combine the HA targeting activity towards cancer cells with the advantageous features of the self-assembling nanosystems, i.e., chemical versatility and ease of preparation and scalability. This review describes the key outcomes arising from the combination of HA and SANPs, focusing on nanomaterials where HA and/or HA-derivatives are inserted within the self-assembling nanostructure. We elucidate the different HA derivatization strategies proposed for this scope, as well as the preparation methods used for the fabrication of the delivery device. After showing the biological results in the employed in vivo and in vitro models, we discussed the pros and cons of each nanosystem, opening a discussion on which approach represents the most promising strategy for further investigation and effective therapeutic protocol development.
Collapse
Affiliation(s)
- Manuela Curcio
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Orazio Vittorio
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sidney, NSW 2052, Australia
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Jessica Lilian Bell
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sidney, NSW 2052, Australia
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Francesca Iemma
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Fiore Pasquale Nicoletta
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Giuseppe Cirillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
5
|
Advances in understanding the role of P-gp in doxorubicin resistance: Molecular pathways, therapeutic strategies, and prospects. Drug Discov Today 2021; 27:436-455. [PMID: 34624510 DOI: 10.1016/j.drudis.2021.09.020] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 07/22/2021] [Accepted: 09/29/2021] [Indexed: 12/11/2022]
Abstract
P-glycoprotein (P-gp) is a drug efflux transporter that triggers doxorubicin (DOX) resistance. In this review, we highlight the molecular avenues regulating P-gp, such as Nrf2, HIF-1α, miRNAs, and long noncoding (lnc)RNAs, to reveal their participation in DOX resistance. These antitumor compounds and genetic tools synergistically reduce P-gp expression. Furthermore, ATP depletion impairs P-gp activity to enhance the antitumor activity of DOX. Nanoarchitectures, including liposomes, micelles, polymeric nanoparticles (NPs), and solid lipid nanocarriers, have been developed for the co-delivery of DOX with anticancer compounds and genes enhancing DOX cytotoxicity. Surface modification of nanocarriers, for instance with hyaluronic acid (HA), can promote selectivity toward cancer cells. We discuss these aspects with a focus on P-gp expression and activity.
Collapse
|
6
|
Li L, Zhang Q, Li J, Tian Y, Kang Y, Ren G, Liu W, Wang H, Wang B, Yan L, Guo L, Diao H. Targeted Delivery of Doxorubicin Using Transferrin-Conjugated Carbon Dots for Cancer Therapy. ACS APPLIED BIO MATERIALS 2021; 4:7280-7289. [PMID: 35006957 DOI: 10.1021/acsabm.1c00811] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A transferrin receptor (TfR)-targeted nanodrug [green fluorescence emission carbon dot (GCD)-polyethylene glycol (PEG)-transferrin (Tf)@doxorubicin (Dox)] for cancer therapy was developed by functionalizing GCDs with PEG, Tf, and Dox. GCDs were synthesized by the one-step hydrothermal method, followed by conjugating PEG and Tf by covalent bonds and loading Dox by electrostatic interactions. The nanodrug exhibits high stability under neutral conditions and effectively releases Dox at pH of 5.5. GCD-PEG-Tf@Dox can be selectively internalized by TfR-overexpressed tumor cells (MCF-7 and K150) via receptor-mediated endocytosis and further release Dox to the nuclei. As a result, GCD-PEG-Tf@Dox exhibits significant lethality to tumor cells (MCF-7 and K150) but greatly reduced toxicity to normal cells [Chinese hamster ovary cell line (CHO)] compared with free Dox. In vivo studies have confirmed that GCD-PEG-Tf@Dox can effectively inhibit tumor proliferation with negligible side effects.
Collapse
Affiliation(s)
- Lihong Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China.,College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China.,Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, PR China
| | - Qi Zhang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Jinyao Li
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Yafei Tian
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Yu Kang
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Guodong Ren
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China
| | - Wen Liu
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China.,Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, PR China
| | - Haojiang Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Bin Wang
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Lili Yan
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Lixia Guo
- College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China
| | - Haipeng Diao
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan 030001, PR China.,College of Basic Medical Sciences, Shanxi Medical University, Taiyuan 030001, PR China.,Key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan 030001, PR China
| |
Collapse
|
7
|
Liu S, Khan AR, Yang X, Dong B, Ji J, Zhai G. The reversal of chemotherapy-induced multidrug resistance by nanomedicine for cancer therapy. J Control Release 2021; 335:1-20. [PMID: 33991600 DOI: 10.1016/j.jconrel.2021.05.012] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022]
Abstract
Multidrug resistance (MDR) of cancer is a persistent problem in chemotherapy. Scientists have considered the overexpressed efflux transporters responsible for MDR and chemotherapy failure. MDR extremely limits the therapeutic effect of chemotherapy in cancer treatment. Many strategies have been applied to solve this problem. Multifunctional nanoparticles may be one of the most promising approaches to reverse MDR of tumor. These nanoparticles can keep stability in the blood circulation and selectively accumulated in the tumor microenvironment (TME) either by passive or active targeting. The stimuli-sensitive or organelle-targeting nanoparticles can release the drug at the targeted-site without exposure to normal tissues. In order to better understand reversal of MDR, three main strategies are concluded in this review. First strategy is the synergistic effect of chemotherapeutic drugs and ABC transporter inhibitors. Through directly inhibiting overexpressed ABC transporters, chemotherapeutic drugs can enter into resistant cells without being efflux. Second strategy is based on nanoparticles circumventing over-expressed efflux transporters and directly targeting resistance-related organelles. Third approach is the combination of multiple therapy modes overcoming cancer resistance. At last, numerous researches demonstrated cancer stem-like cells (CSCs) had a deep relation with drug resistance. Here, we discuss two different drug delivery approaches of nanomedicine based on CSC therapy.
Collapse
Affiliation(s)
- Shangui Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - Abdur Rauf Khan
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - Bo Dong
- Department of cardiovascular medicine, Shandong Provincial Hospital, Jinan 250021, PR China
| | - Jianbo Ji
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
8
|
Wang T, Dong J, Yuan X, Wen H, Wu L, Liu J, Sui H, Deng W. A New Chalcone Derivative C49 Reverses Doxorubicin Resistance in MCF-7/DOX Cells by Inhibiting P-Glycoprotein Expression. Front Pharmacol 2021; 12:653306. [PMID: 33927626 PMCID: PMC8076869 DOI: 10.3389/fphar.2021.653306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/01/2021] [Indexed: 12/30/2022] Open
Abstract
Objective: C49 is a chalcone derivative. The aim of the current study is to illuminate the efficacy of C49 in reversing multidrug resistance (MDR) in MCF-7/DOX cells and its underlying molecular mechanism. Methods: The cytotoxic effects of C49 on MCF-7/DOX cells were evaluated by MTT assay using different concentration (0-250 μmol/L) of C49. Cell proliferation was evaluated by colony formation assay. Cell death was examined by morphological analysis using Hoechst 33,258 staining. Flow cytometry and immunofluorescence were utilized to evaluate the intracellular accumulation of doxorubicin (DOX) and cell apoptosis. The differentially expressed genns between MCF-7 and MCF-7/DOX cells were analyzed by GEO database. The expression of PI3K/Akt pathway proteins were assessed by Western blot The activities of C49 combined with DOX was evaluated via xenograft tumor model in female BALB/c nude mice. Results: C49 inhibited the growth of MCF-7 cells (IC50 = 59.82 ± 2.10 μmol/L) and MCF-7/DOX cells (IC50 = 65.69 ± 8.11 μmol/L) with dosage-dependent and enhanced the cellular accumulation of DOX in MCF-7/DOX cells. The combination of C49 and DOX inhibited cell proliferation and promoted cell apoptosis. MCF-7/DOX cells regained drug sensibility with the combination treatment through inhibiting the expression of P-gp, p-PI3K and p-Akt proteins. Meanwhile, C49 significantly increased the anticancer efficacy of DOX in vivo. Conclusion: C49 combined with DOX restored DOX sensitivity in MCF-7/DOX cells through inhibiting P-gp protein.
Collapse
Affiliation(s)
- Ting Wang
- Department of Medical Oncology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingjing Dong
- Shanghai Bailijia Health Pharmaceutical Technology, Shanghai, China
| | - Xu Yuan
- Department of Medical Oncology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Haotian Wen
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Linguangjin Wu
- Department of Medical Oncology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianwen Liu
- State Key Laboratory of Bioreactor Engineering and Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Hua Sui
- Medical Experiment Center, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanli Deng
- Department of Medical Oncology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
9
|
Xie X, Yuan Z, Yuan Q, Huang Y, Yu Q, Ren J, Liang L, Jin H, Yu J. Preparation and characterization of amphiphilic nanoparticles based on chondroitin sulfate A conjugated with hydrophobic drug for enhanced doxorubicin delivery. Colloid Polym Sci 2020. [DOI: 10.1007/s00396-020-04778-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
10
|
Lan Y, Liang Q, Sun Y, Cao A, Liu L, Yu S, Zhou L, Liu J, Zhu R, Liu Y. Codelivered Chemotherapeutic Doxorubicin via a Dual-Functional Immunostimulatory Polymeric Prodrug for Breast Cancer Immunochemotherapy. ACS APPLIED MATERIALS & INTERFACES 2020; 12:31904-31921. [PMID: 32551517 DOI: 10.1021/acsami.0c06120] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Immunochemotherapy is viewed as a promising approach for cancer therapy via combination treatment with immune-modulating drugs and chemotherapeutic drugs. A novel dual-functional immunostimulatory polymeric prodrug carrier PEG2k-Fmoc-1-MT was developed for simultaneously delivering 1-methyl tryptophan (1-MT) of an indoleamine 2,3-dioxygenase (IDO) inhibitor and chemotherapeutic doxorubicin (DOX) for breast cancer immunochemotherapy. DOX/PEG2k-Fmoc-1-MT micelles were more effective in cell proliferation inhibition and apoptosis induction in 4T1 cells. PEG2k-Fmoc-1-MT prodrug micelles presented enhanced inhibition ability of IDO with decreased kynurenine production and increased the proliferation in dose-dependent manners of effector CD4+ and CD8+ T cells. DOX/PEG2k-Fmoc-1-MT micelles exhibited prolonged blood circulation time and superior accumulation of DOX and 1-MT in tumors compared to that of DOX and 1-MT solutions. A significantly enhanced immune response of the DOX/PEG2k-Fmoc-1-MT micelles was observed with the decreasing tryptophan/kynurenine ratio in blood and tumor tissue, promoting effector CD4+ and CD8+ T cells while reducing regulatory T cell (Tregs) expression. Meanwhile, the coreleased DOX-triggered immunogenic cell death action combined with the cleaved 1-MT promoted the related cytokine secretion of tumor necrosis factor-α, interleukin-2, and interferon-γ, further facilitating the T cell-mediated immune responses. More importantly, the DOX-loaded micelles led to a significantly improved inhibition on tumor growth and prolonged animal survival rate in a 4T1 murine breast cancer model. In conclusion, DOX codelivered by a PEG2k-Fmoc-1-MT immunostimulatory polymeric prodrug showed a maximum immunochemotherapy efficacy against breast cancer.
Collapse
Affiliation(s)
- Yang Lan
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Qiangwei Liang
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Yue Sun
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Aichen Cao
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Lu Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Shuangyu Yu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Liyue Zhou
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Jinxia Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
| | - Rongyue Zhu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
- Key Laboratory of Hui Ethnic Medicine Modernization, Ningxia Medical University, Yinchuan 750004, China
| | - Yanhua Liu
- Department of Pharmaceutics, School of Pharmacy, Ningxia Medical University, No. 1160, Shengli Street, Yinchuan 750004, China
- Key Laboratory of Hui Ethnic Medicine Modernization, Ningxia Medical University, Yinchuan 750004, China
| |
Collapse
|
11
|
The potential impact of trigonelline loaded micelles on Nrf2 suppression to overcome oxaliplatin resistance in colon cancer cells. Mol Biol Rep 2020; 47:5817-5829. [PMID: 32661875 DOI: 10.1007/s11033-020-05650-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) has a pivotal role in promoting chemoresistance by regulation of antioxidants and detoxification enzymes. Trigonelline is one of the major alkaloids in raw coffee which has been recently introduced as potent inhibitor of Nrf2. This study investigated the role of trigonelline and trigonelline loaded micelles in Nrf2 inhibition to break down oxaliplatin resistance in colon cancer cells. The PCL-PEG-PCL and PLA-PCL-PEG-PCL-PLA copolymers and trigonelline loaded micelles were prepared and characterized for fourier transforms infrared (FTIR), hydrogen nuclear magnetic resonance (1H-NMR), carbon nuclear magnetic resonance (13C-NMR) spectroscopy, particle size, zeta potential, scanning electron microscopy (SEM) and entrapment efficiency. Cell viability and apoptosis were evaluated by using MTT and flow cytometry assays, respectively. Nrf2, MRP1, NQO1, HO-1, Bax, and Bcl2 gene expressions were examined by qRT-PCR. Our results revealed that micelles had spherical shapes with narrow sizes and zeta potential indexes of - 9.06 ± 6.94 mV for trigonelline loaded 3Block and - 7.47 ± 6.08 mV for trigonelline loaded 5Block micelles. After Nrf2 inhibition by trigonelline, antioxidant response element (ARE) related gene expressions were decreased (p < 0.05) with a significantly higher impact by trigonelline loaded micelles (p < 0.05). Trigonelline loaded micelles also strongly decreased IC50 value of oxaliplatin in resistant colon cancer cells (p < 0.05). Furthermore, trigonelline loaded 5Block micelle increased oxaliplatin-induced apoptosis in a Nrf2/ARE dependent manner. Altogether, the current study suggests that delivery of trigonelline loaded micelles as potent Nrf2 inhibitors can be considered as a promising strategy to overcome oxaliplatin resistance in colon cancer patients.
Collapse
|
12
|
Acid-responsive dextran-based therapeutic nanoplatforms for photodynamic-chemotherapy against multidrug resistance. Int J Biol Macromol 2020; 155:233-240. [DOI: 10.1016/j.ijbiomac.2020.03.197] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 11/21/2022]
|
13
|
Advances in anti-breast cancer drugs and the application of nano-drug delivery systems in breast cancer therapy. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101662] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
14
|
Ma H, Yang X, Ke J, Wang C, Peng L, Hu F, Yuan H. Smart Assembled Human Serum Albumin Nanocarrier Enhanced Breast Cancer Treatment and Antitumor Immunity by Chemo- photothermal Therapy. ACS Biomater Sci Eng 2020; 6:3217-3229. [DOI: 10.1021/acsbiomaterials.0c00286] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Affiliation(s)
- Huisong Ma
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Xiqin Yang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Jia Ke
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Cheng Wang
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Lijun Peng
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Fuqiang Hu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| | - Hong Yuan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China
| |
Collapse
|
15
|
Kozlovskaya V, Liu F, Yang Y, Ingle K, Qian S, Halade GV, Urban VS, Kharlampieva E. Temperature-Responsive Polymersomes of Poly(3-methyl- N-vinylcaprolactam)- block-poly( N-vinylpyrrolidone) To Decrease Doxorubicin-Induced Cardiotoxicity. Biomacromolecules 2019; 20:3989-4000. [PMID: 31503464 DOI: 10.1021/acs.biomac.9b01026] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Despite being one of the most potent chemotherapeutics, doxorubicin (DOX) facilitates cardiac toxicity by irreversibly damaging the cardiac muscle as well as severely dysregulating the immune system and impairing the resolution of cardiac inflammation. Herein, we report synthesis and aqueous self-assembly of nanosized polymersomes from temperature-responsive poly(3-methyl-N-vinylcaprolactam)-block-poly(N-vinylpyrrolidone) (PMVC-PVPON) diblock copolymers and demonstrate their potential to minimize DOX cardiotoxicity compared to liposomal DOX. RAFT polymerization of vinylpyrrolidone and 3-methyl-N-vinylcaprolactam, which are structurally similar monomers but have drastically different hydrophobicity, allows decreasing the cloud point of PMVCm-PVPONn copolymers below 20 °C. The lower critical solution temperature (LCST) of the PMVC58-PVPONn copolymer varied from 19.2 to 18.6 and to 15.2 °C by decreasing the length of the hydrophilic PVPONn block from n = 98 to n = 65 and to n = 20, respectively. The copolymers assembled into stable vesicles at room temperature when PVPON polymerization degrees were 65 and 98. Anticancer drug DOX was entrapped with high efficiency into the aqueous PMVC58-PVPON65 polymersomal core surrounded by the hydrophobic temperature-sensitive PMVC shell and the hydrophilic PVPON corona. Unlike many liposomal, micellar, or synthetic drug delivery systems, these polymersomes exhibit an exceptionally high loading capacity of DOX (49%) and encapsulation efficiency (95%) due to spontaneous loading of the drug at room temperature from aqueous DOX solution. We also show that C57BL/6J mice injected with the lethal dose of DOX at 15 mg kg-1 did not survive the 14 day treatment, resulting in 100% mortality. The DOX-loaded PMVC58-PVPON65 polymersomes did not cause any mortality in mice indicating that they can be used for successful DOX encapsulation. The gravimetric analyses of the animal organs from mice treated with liposome-encapsulated DOX (Lipo-DOX) and PMVC58-PVPON65 polymersomes (Poly-DOX) revealed that the Lipo-DOX injection caused some toxicity manifesting as decreased body weight compared to Poly-DOX and saline control. Masses of the left ventricle of the heart, lung, and spleen reduced in the Lipo-DOX-treated mice compared to the nontoxic saline control, while no significant decrease of those masses was observed for the Poly-DOX-treated mice. Our results provide evidence for superior stability of synthetic polymersomes in vivo and show promise for the development of next-generation drug carriers with minimal side effects.
Collapse
Affiliation(s)
| | | | | | | | - Shuo Qian
- Neutron Scattering Division, Neutron Sciences Directorate , Oak Ridge National Laboratory , Oak Ridge , Tennessee 37831 , United States
| | | | - Volker S Urban
- Neutron Scattering Division, Neutron Sciences Directorate , Oak Ridge National Laboratory , Oak Ridge , Tennessee 37831 , United States
| | | |
Collapse
|
16
|
Diao L, Shen A, Yang Y, Tao J, Hu Y. CD44-targeted hyaluronic acid–curcumin reverses chemotherapeutics resistance by inhibiting P-gp and anti-apoptotic pathways. RSC Adv 2019; 9:40873-40882. [PMID: 35540044 PMCID: PMC9076387 DOI: 10.1039/c9ra08202f] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 12/02/2019] [Indexed: 01/20/2023] Open
Abstract
Chemotherapeutic drug resistance poses a great challenge in cancer therapy.
Collapse
Affiliation(s)
- Lu Diao
- School of Pharmaceutical Sciences
- Wenzhou Medical University
- Wenzhou
- China
- Zhejiang Pharmaceutical College
| | - Ao Shen
- The University of Queensland
- Brisbane 4072
- Australia
| | - Yunxu Yang
- Zhejiang Pharmaceutical College
- Ningbo
- China
| | - Jin Tao
- Zhejiang Pharmaceutical College
- Ningbo
- China
| | - Ying Hu
- School of Pharmaceutical Sciences
- Wenzhou Medical University
- Wenzhou
- China
- Zhejiang Pharmaceutical College
| |
Collapse
|