1
|
Li L, Chen M, Reis RL, Kundu SC, Xiao B, Shi X. Advancements of nanoscale drug formulations for combination treatment of colorectal cancer. Int J Pharm 2025; 674:125508. [PMID: 40132771 DOI: 10.1016/j.ijpharm.2025.125508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 03/10/2025] [Accepted: 03/21/2025] [Indexed: 03/27/2025]
Abstract
Combination chemotherapy is widely utilized in treating colorectal cancer (CRC), particularly for patients who are ineligible for surgery or those with metastatic CRC (mCRC). While this therapeutic method has demonstrated efficacy in managing CRC and mCRC, its broader clinical application is limited due to the unique physical properties, mechanisms of action, and pharmacokinetics of different chemotherapeutic drugs. Consequently, achieving satisfactory treatment outcomes proves to be challenging. Nanotechnology has given rise to innovative drug systems that are precise, controllable, and highly efficient in drug delivery. These nanoscale drug delivery systems can integrate the advantageous aspects of various therapeutic modalities, including chemotherapy, gene therapy, and immunotherapy. This review aims to explain the application of nano-drug delivery system in the treatment of colorectal cancer. Through its unique physical/chemical properties and biological functions, it can solve the limitations of traditional therapy and achieve more accurate, efficient and safe treatment. The advantages/disadvantages, physical and chemical characteristics of various drug delivery systems are described in detail, and suggestions on selecting reasonable NDDSs according to different drug combination methods are given to achieve the best therapeutic effect. This review paper presents an exhaustive summary of the diverse range of drugs utilized in chemotherapy, in addition to outlining strategies for effectively integrating chemotherapy with other treatment modalities. Furthermore, it delves into the principle of selecting carriers for various drug combinations.
Collapse
Affiliation(s)
- Liqi Li
- Department of General Surgery, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Maohua Chen
- Department of Pharmacy, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Rui L Reis
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetic, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4800-058, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetic, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Barco, Guimarães 4805-017, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães 4800-058, Portugal
| | - Bo Xiao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Xiaoxiao Shi
- State Key Laboratory of Resource Insects, College of Sericulture, Textile, and Biomass Sciences, Southwest University, Beibei, Chongqing 400715, China.
| |
Collapse
|
2
|
Hou D, Zhao W, Yang Q, Wang F, Wu W, Xu L, Yao W, Sun D, Zhu Y, Wu X. Curcumol promotes immune cell invasion and inhibits angiogenesis in colon cancer by decreasing IGF2BP3 expression. Biochem Biophys Res Commun 2025; 750:151394. [PMID: 39899937 DOI: 10.1016/j.bbrc.2025.151394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 01/05/2025] [Accepted: 01/23/2025] [Indexed: 02/05/2025]
Abstract
The study investigates the potential mechanisms by which curcumol exerts colon anti-cancer effects through angiogenesis and immune infiltration. Using The Cancer Genome Atlas (TCGA) database, we found abnormal levels of IGF2BP3 in both paraneoplastic and colon cancer tissues. Firstly, in vivo experiments were conducted to detect the colon anti-cancer and pro-apoptotic effects of curcumol. Immunofluorescence and immunoblotting experiments elucidated that curcumol regulates immune infiltration and angiogenesis in colon cancer through IGF2BP3, and the interaction between curcumol and IGF2BP3 was predicted. Additionally, IGF2BP3 overexpression (IGF2BP3-OE) was further used to verify the colon anti-cancer mechanism of curcumol. The results of the experiment revealed that IGF2BP3 expression is upregulated in colon cancer tissues and correlates with poor patient survival. Our study demonstrated that curcumol significantly inhibits colon cancer tumor growth by promoting apoptosis and inhibiting proliferation through the induction of apoptosis-related proteins Bax and Cleaved-Caspase 3, and the depletion of Bcl-2. Curcumol also downregulated IGF2BP3, promoting the infiltration of immune-activated helper T cells (CD4+ T), cytotoxic T cells (CD8+ T), and natural killer (NK) cells in tumor tissues, while reducing the number of immune-suppressing regulatory T cells (Treg) and inhibiting the angiogenesis-related protein CD31. Molecular docking experiments identified IGF2BP3 as a direct target of curcumol. When IGF2BP3 was overexpressed, the inhibitory effect of curcumol on angiogenesis in colon cancer tissues was reversed. In summary, curcumol promotes immune cell infiltration in tumor tissues by downregulating IGF2BP3, thereby inhibiting the proliferation and angiogenesis of colon cancer cells.
Collapse
Affiliation(s)
- Dahai Hou
- Guizhou University of Traditional Chinese Medicine College of Pharmacy, Guiyang, Guizhou, 550025, China
| | - Wuxia Zhao
- Guizhou University of Traditional Chinese Medicine College of Pharmacy, Guiyang, Guizhou, 550025, China
| | - Qi Yang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Fang Wang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Wenya Wu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Linyu Xu
- Department of General Surgery, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, 215600, China
| | - Wenchao Yao
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China
| | - Dongdong Sun
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yimiao Zhu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| | - Xiaoyu Wu
- Department of Surgical Oncology, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
3
|
Elhamipour M, Soleimanjahi H, Abdoli A, Sharifi N, Karimi H, Soleyman Jahi S, Kvistad R. Combination Therapy with Secretome of Reovirus-Infected Mesenchymal Stem Cells and Metformin Improves Anticancer Effects of Irinotecan on Colorectal Cancer Cells in vitro. Intervirology 2024; 68:1-16. [PMID: 39561737 DOI: 10.1159/000542356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/29/2024] [Indexed: 11/21/2024] Open
Abstract
INTRODUCTION Irinotecan, a topoismorase 1 inhibitor, has been used for the treatment of colorectal cancer. It was shown that monotherapy alone is largely ineffective. The combination therapy was used for antitumor activity. The synergistic anticancer effects of oncolytic reovirus-infected secretome in combination with irinotecan and metformin are evaluated in vitro. The aim of research was to assess anticancer impacts of ReoT3D, irinotecan, metformin in combination, against murine colorectal cancer cells (CT26). METHODS The L929 and the CT26 colorectal cancerous cell lines were treated in vitro with irinotecan, metformin, the Dearing strain of reovirus serotype 3 (ReoT3D) (V), and the secretome of intact (S) or reovirus-infected murine adipose-derived mesenchymal stem cells (SV). The cell viability was measured by MTT, and the apoptosis rate was analyzed by annexin V-FITC staining and flow cytometry 48 and 72 h after treatment. RESULTS We found that cells exposed to a combination of SV+Met+I had significantly lower cell viability and higher apoptosis rates as compared to cells exposed to Met+I, 48 and 72 h. These results suggest that metformin in combination with irinotecan and reovirus produces a synergistic effect on cell death, and adding reovirus-infected secretome (SV) to a Met+I regimen induces a higher apoptosis rate compared to Met+I alone. Based on the results, the combination of SV+Met+I has induced more apoptosis than S, SV, SV+I, and SV+Met. Also, all of the combined treatments induced apoptosis significantly versus secretome alone. DISCUSSION In this in vitro study, we found that the combination of T3D reovirus (oncolytic virus) and metformin with the anticancer drug irinotecan resulted in higher rates of growth inhibition and apoptosis induction in the colorectal cancer cell line. This synergistic effect was even more pronounced when using the combination of secretome derived from reovirus-infected AD-MSCs, metformin, and irinotecan. CONCLUSION We highlight that the combination of ReoT3D-derived secretome with irinotecan and metformin showed a synergistic anticancer effect on the CT26 cell line, and this strategy may be considered as a new approach against colorectal cancer in the in vitro and in vivo in future studies.
Collapse
Affiliation(s)
- Maliheh Elhamipour
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran,
| | - Hoorieh Soleimanjahi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Asghar Abdoli
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Negar Sharifi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hesam Karimi
- Department of Virology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | - Saeed Soleyman Jahi
- Division of Gastroenterology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA
| | - Ruth Kvistad
- Department of Biology, University of Missouri - St. Louis, St. Louis, Missouri, USA
- Department of Chemistry and Biochemistry, University of Missouri - St. Louis, St. Louis, Missouri, USA
| |
Collapse
|
4
|
Shewale RS, Gomte SS, Jain A. A sustainable RP-HPLC method for concurrent estimation of capecitabine and celecoxib in liposomal formulation: Greenness and whiteness appraisal. Arch Pharm (Weinheim) 2024:e2400632. [PMID: 39344208 DOI: 10.1002/ardp.202400632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/03/2024] [Accepted: 09/06/2024] [Indexed: 10/01/2024]
Abstract
Liposomes have been reported for combination therapy due to their ability to carry both hydrophilic and lipophilic drugs together. The current investigation aims to develop a novel, eco-friendly, and sustainable reverse-phase high-performance liquid chromatography (RP-HPLC) method for the simultaneous quantification of capecitabine and celecoxib co-encapsulated in liposomes. The method reported herein uses a C18 column (4.6 × 250 mm2, 5 μm) and a mobile phase consisting of water, and acetonitrile/methanol in a ratio of 60:40, containing 0.1% formic acid in both the phases. The flow rate is maintained at 1 mL/min, with an injection volume of 10 μL in the gradient mode. Detection is set at λmax = 240 nm for capecitabine and 252 nm for celecoxib. The developed liposomes are mono-disperse with a surface potential of -6.93 mV. The average size of the liposomes is 142 nm. The percentage entrapment efficiency for capecitabine is 52.39 ± 0.94%, and for celecoxib, it is 77.13 ± 0.74%. The Analytical Greenness Metric of 0.61 and Analytical Eco-Scale Score of 75 signify the greenness of the developed method. Also, the Red-Green-Blue model shows excellent whiteness, with a score of 83.2. Thus, the developed method offers a reliable, accurate, precise, buffer-free, and environment-friendly RP-HPLC approach for the simultaneous analysis of capecitabine and celecoxib co-encapsulated in liposomes.
Collapse
Affiliation(s)
- Rushikesh Sanjay Shewale
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Gandhinagar, Gujarat, India
| | - Shyam Sudhakar Gomte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Gandhinagar, Gujarat, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Ahmedabad (NIPER-A), An Institute of National Importance, Government of India, Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, Gandhinagar, Gujarat, India
| |
Collapse
|
5
|
Jiang D, Nie H, Wang Z, Xiong Y, Shen H, Gao Y, Zhu X, Mao Z. Developing oxaliplatin and IL-15 Co-carried gels as drug depots to enable triple-interlocked combination therapy for colorectal cancer. Colloids Surf B Biointerfaces 2024; 241:113996. [PMID: 38850745 DOI: 10.1016/j.colsurfb.2024.113996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/16/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024]
Abstract
Chemo-immunotherapy, which involves the simultaneous use of chemotherapy drug and immunotherapeutic agent to achieve synergistic effects, plays a crucial role in cancer treatment. However, the immunosuppressive microenvironment, insufficient tumor specificity, and serious systemic side effects hinder their synergistic therapeutic effects and clinical applications. Herein, T cell and natural killer (NK) cell, which are the most important immune effector cells, were both activated to reverse the immunosuppressive microenvironment. To simplify drug carriers, oxaliplatin was selected as the chemotherapy drug which can both induce the ICD effect and activate T cells. IL-15 was selected to activate NK cells. To enhance the productivity of the carrier and reduce side effects, the easy-prepared thermosensitive hydrogel (OXL/IL-15 TG) was developed to co-load oxaliplatin-loaded liposomes (OXL) and IL-15. Colorectal cancer, suitable for in situ administration, was selected as model cancer. The resulting novel triple-interlocked combination therapy could directly kill the tumor cells, induces ICD effect and activate NK cells. After administration, OXL/IL-15 TG was formed serving as a drug depot, slowing releasing OXL and IL-15 non-interferencely. OXL around 165.47±7.04 nm was passively delivered to tumor tissue, killing tumor cells and inducing ICD effect. The results demonstrated that IL-15 stimulated the activation of NK cells. In tumor-bearing mice models, OXL/IL-15 TG exhibited a remarkable and noteworthy anti-tumor efficacy, and expanded survival rate. Notably, OXL/IL-15 TG led to an enhanced infiltration of CD3+CD8+ T cells and CD3-CD49+ NK cells within the tumor tissue. Overall, the triple-interlocked combination therapy provided a new idea for colorectal cancer therapy.
Collapse
Affiliation(s)
- Dandan Jiang
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450003, China
| | - Haiqian Nie
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ziang Wang
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yuhan Xiong
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Huimin Shen
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ya Gao
- State Key Laboratory of Esophageal Cancer Prevention & Treatment; Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China; Key Laboratory of Henan Province for Drug Quality and Evaluation, Henan Province; Institute of Drug Discovery and Development; School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Xiali Zhu
- School of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| | - Zhenkun Mao
- Department of Pharmacy, Henan Provincial People's Hospital; People's Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450003, China.
| |
Collapse
|
6
|
Wang L, Liu J, Wang X, Li X, Zhang X, Yuan L, Wu Y, Liu M. Effect of the combined binding of topotecan and catechin/protocatechuic acid to a pH-sensitive DNA tetrahedron on release and cytotoxicity: Spectroscopic and calorimetric studies. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 314:124179. [PMID: 38522375 DOI: 10.1016/j.saa.2024.124179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024]
Abstract
The therapeutic efficacy of chemotherapy drugs can be effectively improved through the dual effects of their combination with natural polyphenols and the delivery of targeted DNA nanostructures. In this work, the interactions of topotecan (TPT), (+)-catechin (CAT), or protocatechuic acid (PCA) with a pH-sensitive DNA tetrahedron (MUC1-TD) in the binary and ternary systems at pHs 5.0 and 7.4 were investigated by fluorescence spectroscopy and calorimetry. The intercalative binding mode of TPT/CAT/PC to MUC1-TD was confirmed, and their affinity was ranked in the order of PCA > CAT > TPT. The effects of the pH-sensitivity of MUC1-TD and different molecular structures of CAT and PCA on the loading, release, and cytotoxicity of TPT were discussed. The weakened interaction under acidic conditions and the co-loading of CAT/PCA, especially PCA, improved the release of TPT loaded by MUC1-TD. The targeting of MUC1-TD and the synergistic effect with CAT/PCA, especially CAT, enhanced the cytotoxicity of TPT on A549 cells. For L02 cells, the protective effect of CAT/PCA reduced the damage caused by TPT. The single or combined TPT loaded by MUC1-TD was mainly concentrated in the nucleus of A549 cells. This work will provide key information for the combined application of TPT and CAT/PCA loaded by DNA nanostructures to improve chemotherapy efficacy and reduce side effects.
Collapse
Affiliation(s)
- Lu Wang
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng 252059, China
| | - Jie Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, China
| | - Xiangtai Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, China
| | - Xinyu Li
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng 252059, China
| | - Xinpeng Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, China
| | - Lixia Yuan
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, China
| | - Yushu Wu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, China
| | - Min Liu
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng 252059, China; Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, China.
| |
Collapse
|
7
|
Yadav R, Bhawale R, Srivastava V, Pardhi E, Bhalerao HA, Sonti R, Mehra NK. Innovative Nanoparticulate Strategies in Colon Cancer Treatment: A Paradigm Shift. AAPS PharmSciTech 2024; 25:52. [PMID: 38429601 DOI: 10.1208/s12249-024-02759-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/06/2024] [Indexed: 03/03/2024] Open
Abstract
As a major public health issue, colorectal cancer causes 9.4% of total cancer-related deaths and comprises 10% of new cancer diagnoses worldwide. In the year 2023, an estimated 153,020 people are expected to receive an identification of colorectal cancer (CRC), resulting in roughly 52,550 fatalities anticipated as a result of this illness. Among those impacted, approximately 19,550 cases and 3750 deaths are projected to occur in individuals under the age of 50. Irinotecan (IRN) is a compound derived from the chemical structure of camptothecin, a compound known for its action in inhibiting DNA topoisomerase I. It is employed in the treatment strategy for CRC therapies. Comprehensive in vivo and in vitro studies have robustly substantiated the anticancer efficacy of these compounds against colon cancer cell lines. Blending irinotecan in conjunction with other therapeutic cancer agents such as oxaliplatin, imiquimod, and 5 fluorouracil enhanced cytotoxicity and improved chemotherapeutic efficacy. Nevertheless, it is linked to certain serious complications and side effects. Utilizing nano-formulated prodrugs within "all-in-one" carrier-free self-assemblies presents an effective method to modify the pharmacokinetics and safety portfolio of cytotoxic chemotherapeutics. This review focuses on elucidating the mechanism of action, exploring synergistic effects, and innovating novel delivery approaches to enhance the therapeutic efficacy of irinotecan.
Collapse
Affiliation(s)
- Rati Yadav
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, 500 037, India
| | - Rohit Bhawale
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, 500 037, India
| | - Vaibhavi Srivastava
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, 500 037, India
| | - Ekta Pardhi
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, 500 037, India
| | - Harshada Anil Bhalerao
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Rajesh Sonti
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, 500 037, India.
| |
Collapse
|
8
|
Zhang Z, Xu X, Du J, Chen X, Xue Y, Zhang J, Yang X, Chen X, Xie J, Ju S. Redox-responsive polymer micelles co-encapsulating immune checkpoint inhibitors and chemotherapeutic agents for glioblastoma therapy. Nat Commun 2024; 15:1118. [PMID: 38320994 PMCID: PMC10847518 DOI: 10.1038/s41467-024-44963-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 01/11/2024] [Indexed: 02/08/2024] Open
Abstract
Immunotherapy with immune checkpoint blockade (ICB) for glioblastoma (GBM) is promising but its clinical efficacy is seriously challenged by the blood-tumor barrier (BTB) and immunosuppressive tumor microenvironment. Here, anti-programmed death-ligand 1 antibodies (aPD-L1) are loaded into a redox-responsive micelle and the ICB efficacy is further amplified by paclitaxel (PTX)-induced immunogenic cell death (ICD) via a co-encapsulation approach for the reinvigoration of local anti-GBM immune responses. Consequently, the micelles cross the BTB and are retained in the reductive tumor microenvironment without altering the bioactivity of aPD-L1. The ICB efficacy is enhanced by the aPD-L1 and PTX combination with suppression of primary and recurrent GBM, accumulation of cytotoxic T lymphocytes, and induction of long-lasting immunological memory in the orthotopic GBM-bearing mice. The co-encapsulation approach facilitating efficient antibody delivery and combining with chemotherapeutic agent-induced ICD demonstrate that the chemo-immunotherapy might reprogram local immunity to empower immunotherapy against GBM.
Collapse
Affiliation(s)
- Zhiqi Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Xiaoxuan Xu
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Jiawei Du
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Xin Chen
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, 210009, China
| | - Yonger Xue
- Center for BioDelivery Sciences, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jianqiong Zhang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
- Department of Microbiology and Immunology, Medical School, Southeast University, Nanjing, 210009, China
| | - Xue Yang
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore.
| | - Jinbing Xie
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China.
| | - Shenghong Ju
- Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
9
|
Bagheri M, Zandieh MA, Daryab M, Samaei SS, Gholami S, Rahmanian P, Dezfulian S, Eary M, Rezaee A, Rajabi R, Khorrami R, Salimimoghadam S, Hu P, Rashidi M, Ardakan AK, Ertas YN, Hushmandi K. Nanostructures for site-specific delivery of oxaliplatin cancer therapy: Versatile nanoplatforms in synergistic cancer therapy. Transl Oncol 2024; 39:101838. [PMID: 38016356 DOI: 10.1016/j.tranon.2023.101838] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/24/2023] [Accepted: 11/17/2023] [Indexed: 11/30/2023] Open
Abstract
As a clinically approved treatment strategy, chemotherapy-mediated tumor suppression has been compromised, and in spite of introducing various kinds of anticancer drugs, cancer eradication with chemotherapy is still impossible. Chemotherapy drugs have been beneficial in improving the prognosis of cancer patients, but after resistance emerged, their potential disappeared. Oxaliplatin (OXA) efficacy in tumor suppression has been compromised by resistance. Due to the dysregulation of pathways and mechanisms in OXA resistance, it is suggested to develop novel strategies for overcoming drug resistance. The targeted delivery of OXA by nanostructures is described here. The targeted delivery of OXA in cancer can be mediated by polymeric, metal, lipid and carbon nanostructures. The advantageous of these nanocarriers is that they enhance the accumulation of OXA in tumor and promote its cytotoxicity. Moreover, (nano)platforms mediate the co-delivery of OXA with drugs and genes in synergistic cancer therapy, overcoming OXA resistance and improving insights in cancer patient treatment in the future. Moreover, smart nanostructures, including pH-, redox-, light-, and thermo-sensitive nanostructures, have been designed for OXA delivery and cancer therapy. The application of nanoparticle-mediated phototherapy can increase OXA's potential in cancer suppression. All of these subjects and their clinical implications are discussed in the current review.
Collapse
Affiliation(s)
- Mohsen Bagheri
- Radiology Resident, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mahshid Daryab
- Department of Pharmaceutics, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyedeh Setareh Samaei
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sarah Gholami
- Young Researcher and Elite Club, Babol Branch, Islamic Azad University, Babol, Iran
| | - Parham Rahmanian
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Sadaf Dezfulian
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mahsa Eary
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Aryan Rezaee
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Peng Hu
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Alireza Khodaei Ardakan
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
10
|
Azarifar Z, Amini R, Tanzadehpanah H, Afshar S, Najafi R. In vitro co-delivery of 5-fluorouracil and all-trans retinoic acid by PEGylated liposomes for colorectal cancer treatment. Mol Biol Rep 2023; 50:10047-10059. [PMID: 37902908 DOI: 10.1007/s11033-023-08888-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/04/2023] [Indexed: 11/01/2023]
Abstract
BACKGROUND Single-target inhibitors have not been successful in cancer treatment due to the development of drug resistance. Nevertheless, therapeutic agents capable of simultaneously inhibiting multiple targets have revealed encouraging results in inducing apoptosis and overcoming drug resistance in cancerous cells. Here, we designed a composite liposomal nano-carrier co-loading 5-Fluorouracil (5-FU) with all-trans retinoic acid (ATRA) to assess anticancer efficacy of the combined drugs in colorectal cancer (CRC). METHODS A PEGylated liposomal nano-carrier with phospholipid/cholesterol/DSPE-PEG (2000) was synthesized by the thin film hydration technique for co-delivery of ATRA and 5-FU. After characterizing, the role of 5-FU and ATRA co-loaded liposomal nano-carrier in proliferation, epithelial-mesenchymal transition (EMT), apoptosis, and cancer stem cells (CSCs) were investigated by using colony forming and MTT assay, RT-qPCR and Annexin V/PI kit. RESULTS The average size of liposomes (LPs) was < 150 nm with uniform size distribution. Drug release analyses indicated that both ATRA and 5-FU could simultaneously release from LPs in a sustained release manner. The synergistic inhibitory effects of ATRA and 5-FU loaded in LPs were verified with a combination index of 0.43. Dual drug LPs showed the highest cytotoxicity, enhanced inhibition of cell proliferation, increased apoptotic potential, decreased CSCs, and attenuated EMT-associated biomarkers. Also, dual drug LPs decreased β-catenin gene expression more than other liposomal formulations. CONCLUSION These findings suggest that using LPs to achieve a synergistic effect of ATRA and 5-FU is an effectual approach to increase the therapeutic effect of 5-FU toward CRC cells.
Collapse
Affiliation(s)
- Zahra Azarifar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Razieh Amini
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saeid Afshar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Rezvan Najafi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
11
|
Liu B, Zhang J, Liu Z, Wang P, Zhang Y, He H, Yin T, Gou J, Tang X. Research on the preparation process of the cytarabine/daunorubicin dual-encapsulation liposome and its physicochemical properties and performances in vitro/vivo. Int J Pharm 2023; 646:123500. [PMID: 37820944 DOI: 10.1016/j.ijpharm.2023.123500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/20/2023] [Accepted: 10/08/2023] [Indexed: 10/13/2023]
Abstract
As the only Food and Drug Administration (FDA)-approved dual-encapsulation liposome injection for treating Acute myeloid leukemia (AML), CPX-351 outperforms the standard chemotherapy treatment "DA 7 + 3″ in terms of clinical effectiveness. Although research on dual-loaded liposomes has increased in recent years, little attention has been paid to their preparation, which can affect their quality, efficacy, and safety. This study explored various preparation processes to create the cytarabine/daunorubicin co-loaded liposome (the Cyt/Daun liposome) and eventually settled on two methods: the sequential loading approach, thin film hydration-extrusion-copper ion gradient, and the simultaneous encapsulation technique, copper ion gradient-concentration gradient. Different preparation methods resulted in different particle sizes and encapsulation efficiencies; the two aforementioned preparation processes generated dual-loaded liposomes with comparable physicochemical properties. The sequential encapsulation technique was selected for the subsequent research owing to its higher encapsulation efficiency prior to purification; the prepared Cyt/Daun liposomes had small and uniform particle size (108.6 ± 1.02 nm, Polydispersity index (PDI) 0.139 ± 0.01), negative charge (-(60.2 ± 1.15) mV), high drug encapsulation efficiency (Cyt 88.2 ± 0.24 %, Duan 94.2 ± 0.45 %) and good plasma stability. To improve its storage stability, the Cyt/Daun liposome was lyophilized (-40 °C for 4 h, maintained for 130 min, and dried for 1200 min) using sucrose-raffinose (mass ratio 7:3; glycolipid ratio 4:1, w/w) as a lyoprotectant. The lyophilized liposomes were purple cakes, redissolved rapidly with insignificant alterations in particle size and encapsulation efficiency, and possessed well storage stability. The pharmacokinetic and tissue distribution studies demonstrated that the Cyt/Daun liposome could achieve long circulation and maintain synergic proportions of drugs within 24 h, increasing the accumulation of drugs at tumor sites. Furthermore, the in vitro/in vivo pharmacodynamic studies confirmed its good anti-tumor activity and safety.
Collapse
Affiliation(s)
- Boyuan Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Jiaoyang Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Zixu Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Ping Wang
- School of Pharmacy, Jilin University, Changchun 130021, Jilin, PR China
| | - Yu Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Haibing He
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Tian Yin
- Faculty of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China
| | - Jingxin Gou
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China.
| | - Xing Tang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, PR China.
| |
Collapse
|
12
|
Wang Q, Xu W, Li Q, He C, Liu Y, Liu J, Wang R, Wu J, Xiang D, Chen C. Coaxial electrostatic spray-based preparation of localization missile liposomes on a microfluidic chip for targeted treatment of triple-negative breast cancer. Int J Pharm 2023; 643:123220. [PMID: 37437856 DOI: 10.1016/j.ijpharm.2023.123220] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/23/2023] [Accepted: 07/08/2023] [Indexed: 07/14/2023]
Abstract
Due to triple-negative breast cancer (TNBC) lacking specific targets for efficient therapies, nanoparticles have been widely developed to enhance efficacy and reduce the toxicity of chemotherapeutics. We prepared unique liposomes containing PTX and DOX by microfluidics-based coaxial electrostatic spray method, which have a uniform particle size, high drug loading capacity, and good stability. Meanwhile, the cRGD peptide was fused with the lipid membrane to form PTX/DOX@cRGD-Lipo, which played a GPS role in locating tumor neovascularization and further targeting TNBC cells where both overexpress αvβ3. The PTX/DOX@cRGD-Lipo showed synergistic anti-tumor activity of double drugs and enhanced tumor cell apoptosis. Fluorescence microscopy and flow cytometry showed that the co-loaded targeted liposomes could be effectively absorbed by MDA-MB-231 and 4T1 cells and then released the content. In addition, the PTX/DOX@cRGD-Lipo presented excellent targeting biodistribution in vivo and a higher tumor growth inhibition rate in the orthotopic tumor mouse model. All results suggested that the double drug-loaded targeted liposome could be a promising treatment modality for TNBC.
Collapse
Affiliation(s)
- Qian Wang
- Department of Pharmacy, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410000, China
| | - Wenjie Xu
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Qi Li
- Department of Pharmacy, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410000, China
| | - Chunpeng He
- Department of Pharmacy, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410000, China
| | - Yang Liu
- Department of Pharmacy, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410000, China
| | - Jia Liu
- Department of Pharmacy, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410000, China
| | - Rongrong Wang
- Hunan Institute for Drug, Bayi Road 60, Changsha 410000, China
| | - Junyong Wu
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410000, China
| | - Daxiong Xiang
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Changsha 410000, China.
| | - Chuanpin Chen
- Department of Pharmacy, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410000, China.
| |
Collapse
|
13
|
Li Z, Yu H, Liu C, Wang C, Zeng X, Yan J, Sun Y. Efficiency co-delivery of ellagic acid and oxygen by a non-invasive liposome for ameliorating diabetic retinopathy. Int J Pharm 2023; 641:122987. [PMID: 37207860 DOI: 10.1016/j.ijpharm.2023.122987] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/17/2023] [Accepted: 04/21/2023] [Indexed: 05/21/2023]
Abstract
Diabetic retinopathy (DR) is one of the serious complications of diabetes, which has become the fourth leading cause of vision loss worldwide. Current treatment of DR relies on intravitreal injections of antiangiogenic agents, which has made considerable achievements in reducing visual impairment. However, long-term invasive injections require advanced technology and can lead to poor patient compliance as well as the incidence of ocular complications including bleeding, endophthalmitis, retinal detachment and others. Hence, we developed non-invasive liposomes (EA-Hb/TAT&isoDGR-Lipo) for efficiency co-delivery of ellagic acid and oxygen, which can be administered intravenously or by eye drops. Among that, ellagic acid (EA), as an aldose reductase inhibitor, could remove excessive reactive oxygen species (ROS) induced by high glucose for preventing retinal cell apoptosis, as well as reduce retinal angiogenesis through the blockage of VEGFR2 signaling pathway; carried oxygen could ameliorate DR hypoxia, and further enhanced the anti-neovascularization efficacy. Our results showed that EA-Hb/TAT&isoDGR-Lipo not only effectively protected retinal cells from high glucose-induced damage, but also inhibited VEGF-induced vascular endothelial cells migration, invasion, and tube formation in vitro. In addition, in a hypoxic cell model, EA-Hb/TAT&isoDGR-Lipo could reverse retinal cell hypoxia, thereby reducing the expression of VEGF. Significantly, after being administered as an injection or eye drops, EA-Hb/TAT&isoDGR-Lipo obviously ameliorated the structure (central retinal thickness and retinal vascular network) of retina by eliminating ROS and down-regulating the expression of GFAP, HIF-1α, VEGF and p-VEGFR2 in a DR mouse model. In summary, EA-Hb/TAT&isoDGR-Lipo holds great potentials in improvement of DR, which provides a novel approach for the treatment of DR.
Collapse
Affiliation(s)
- Zhipeng Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Hongli Yu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Chaolong Liu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Changduo Wang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Xianhu Zeng
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Jianqin Yan
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao 266021, China.
| |
Collapse
|
14
|
Jiang Y, Jiang Y, Li M, Yu Q. Will nanomedicine become a good solution for the cardiotoxicity of chemotherapy drugs? Front Pharmacol 2023; 14:1143361. [PMID: 37214453 PMCID: PMC10194942 DOI: 10.3389/fphar.2023.1143361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 04/12/2023] [Indexed: 05/24/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide, and with the continuous development of life sciences and pharmaceutical technology, more and more antitumor drugs are being used in clinics to benefit cancer patients. However, the incidence of chemotherapy-induced cardiotoxicity has been continuously increasing, threatening patients' long-term survival. Cardio-oncology has become a research hot spot, and the combination of nanotechnology and biomedicine has brought about an unprecedented technological revolution. Nanomaterials have the potential to maximize the efficacy and reduce the side effects of chemotherapeutic drugs when used as their carriers, and several nano-formulations of frequently used chemotherapeutic drugs have already been approved for marketing. In this review, we summarize chemotherapeutic drugs that are highly associated with cardiotoxicity and evaluate the role of nano-delivery systems in reducing cardiotoxicity based on studies of their marketed or R&D nano-formulations. Some of the marketed chemotherapy drugs are combined with nano-delivery systems that can effectively deliver chemotherapy drugs to tumors and cannot easily penetrate the endothelial barrier of the heart, thus decreasing their distribution in the heart and reducing the cardiotoxicity to some extent. However, many chemotherapy nanomedicines that are marketed or in R&D have not received enough attention in determining their cardiotoxicity. In general, nanomedicine is an effective method to reduce the cardiotoxicity of traditional chemotherapy drugs. However, cardiovascular complications in cancer treatment are very complex diseases, requiring the application of multiple measures to achieve effective management and prevention.
Collapse
Affiliation(s)
- Yichuan Jiang
- Department of Pharmacy, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yueyao Jiang
- Department of Pharmacy, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Min Li
- Pharmacological Experiment Center, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Qian Yu
- Department of Pharmacy, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
15
|
Liu X, Liang S, Sang X, Chang L, Fu S, Yang H, Yang H, Liu Y, Zhang N. On-demand integrated nano-engager converting cold tumors to hot via increased DNA damage and dual immune checkpoint inhibition. Acta Pharm Sin B 2023; 13:1740-1754. [PMID: 37139406 PMCID: PMC10150160 DOI: 10.1016/j.apsb.2022.09.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/22/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022] Open
Abstract
Cancer immunotherapy has become a promising strategy. However, the effectiveness of immunotherapy is restricted in "cold tumors" characterized with insufficient T cells intratumoral infiltration and failed T cells priming. Herein, an on-demand integrated nano-engager (JOT-Lip) was developed to convert cold tumors to hot via "increased DNA damage and dual immune checkpoint inhibition" strategy. JOT-Lip was engineered by co-loading oxaliplatin (Oxa) and JQ1 into liposomes with T-cell immunoglobulin mucin-3 antibodies (Tim-3 mAb) coupled on the liposomal surface by metalloproteinase-2 (MMP-2)-sensitive linker. JQ1 inhibited DNA repair to increase DNA damage and immunogenic cell death (ICD) of Oxa, thus promoting T cells intratumoral infiltration. In addition, JQ1 inhibited PD-1/PD-L1 pathway, achieving dual immune checkpoint inhibition combining with Tim-3 mAb, thus effectively promoting T cells priming. It is demonstrated that JOT-Lip not only increased DNA damage and promoted the release of damage-associated molecular patterns (DAMPs), but also enhanced T cells intratumoral infiltration and promoted T cell priming, which successfully converted cold tumors to hot and showed significant anti-tumor and anti-metastasis effects. Collectively, our study provides a rational design of an effective combination regimen and an ideal co-delivery system to convert cold tumors to hot, which holds great potential in clinical cancer chemoimmunotherapy.
Collapse
Affiliation(s)
| | | | - Xiao Sang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Lili Chang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Shunli Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Han Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Huizhen Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| |
Collapse
|
16
|
Wang L, Wu Y, Weng T, Li X, Zhang X, Zhang Y, Yuan L, Zhang Y, Liu M. Binding of combined irinotecan and epicatechin to a pH-responsive DNA tetrahedron for controlled release and enhanced cytotoxicity. J Mol Struct 2023. [DOI: 10.1016/j.molstruc.2023.135323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
|
17
|
Dave R, Patel R, Patel M. Hybrid Lipid-Polymer Nanoplatform: A Systematic Review for Targeted Colorectal Cancer Therapy. Eur Polym J 2023. [DOI: 10.1016/j.eurpolymj.2023.111877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
18
|
Mahaki H, Mansourian M, Meshkat Z, Avan A, Shafiee MH, Mahmoudian RA, Ghorbani E, Ferns GA, Manoochehri H, Menbari S, Sheykhhasan M, Tanzadehpanah H. Nanoparticles Containing Oxaliplatin and the Treatment of Colorectal Cancer. Curr Pharm Des 2023; 29:3018-3039. [PMID: 37990895 DOI: 10.2174/0113816128274742231103063738] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/04/2023] [Accepted: 10/11/2023] [Indexed: 11/23/2023]
Abstract
BACKGROUND Colorectal cancer (CRC) is a highly widespread malignancy and ranks as the second most common cause of cancer-related mortality. OBJECTIVE Cancer patients, including those with CRC, who undergo chemotherapy, are often treated with platinum- based anticancer drugs such as oxaliplatin (OXA). Nevertheless, the administration of OXA is associated with a range of gastrointestinal problems, neuropathy, and respiratory tract infections. Hence, it is necessary to devise a potential strategy that can effectively tackle these aforementioned challenges. The use of nanocarriers has shown great potential in cancer treatment due to their ability to minimize side effects, target drugs directly to cancer cells, and improve drug efficacy. Furthermore, numerous studies have been published regarding the therapeutic efficacy of nanoparticles in the management of colorectal cancer. METHODS In this review, we present the most relevant nanostructures used for OXA encapsulation in recent years, such as solid lipid nanoparticles, liposomes, polysaccharides, proteins, silica nanoparticles, metal nanoparticles, and synthetic polymer-carriers. Additionally, the paper provides a summary of the disadvantages and limits associated with nanoparticles. RESULTS The use of different carriers for the delivery of oxaliplatin increased the efficiency and reduced the side effects of the drug. It has been observed that the majority of research investigations have focused on liposomes and polysaccharides. CONCLUSION This potentially auspicious method has the potential to enhance results and enhance the quality of life for cancer patients undergoing chemotherapy. However, additional investigation is required to ascertain the most suitable medium for the transportation of oxaliplatin and to assess its efficacy through clinical trials.
Collapse
Affiliation(s)
- Hanie Mahaki
- Vascular and Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Mansourian
- Vascular and Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Meshkat
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Australia
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
| | | | - Reihaneh Alsadat Mahmoudian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Ghorbani
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Hamed Manoochehri
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Shaho Menbari
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Laboratory Sciences, Faculty of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohsen Sheykhhasan
- Qom University of Medical Science and Health Services Mesenchymal Stem Cells Qom Iran
- Department of Mesenchymal Stem Cells, Qom University of Medical Science and Health Services, Qom, Iran
| | - Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
19
|
Kohan Hoosh Nejad A, Ahmad Panahi H, Keshmirizadeh E, Torabi Fard N. Fabrication of a pH-responsive drug delivery system based on the super-paramagnetic metal-organic framework for targeted delivery of oxaliplatin. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2082424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
| | - Homayon Ahmad Panahi
- Department of Chemistry, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | | | - Niloufar Torabi Fard
- Department of Chemistry, North Tehran Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
20
|
Valdez L, Cheng B, Gonzalez D, Rodriguez R, Campano P, Tsin A, Fang X. Combined treatment with niclosamide and camptothecin enhances anticancer effect in U87 MG human glioblastoma cells. Oncotarget 2022; 13:642-658. [PMID: 35548329 PMCID: PMC9084225 DOI: 10.18632/oncotarget.28227] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022] Open
Affiliation(s)
- Laura Valdez
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- These authors contributed equally to this work
| | - Benxu Cheng
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
- These authors contributed equally to this work
| | - Daniela Gonzalez
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Reanna Rodriguez
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Paola Campano
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Andrew Tsin
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| | - Xiaoqian Fang
- Department of Molecular Science, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX 78539, USA
| |
Collapse
|
21
|
Sun Y, Xie Y, Tang H, Ren Z, Luan X, Zhang Y, Zhu M, Lv Z, Bao H, Li Y, Liu R, Shen Y, Zheng Y, Pei J. In vitro and in vivo Evaluation of a Novel Estrogen-Targeted PEGylated Oxaliplatin Liposome for Gastric Cancer. Int J Nanomedicine 2022; 16:8279-8303. [PMID: 34992365 PMCID: PMC8712509 DOI: 10.2147/ijn.s340180] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 12/07/2021] [Indexed: 12/31/2022] Open
Abstract
Background Chemotherapy is still the main first-line treatment for advanced metastatic gastric cancer, but it has the limitations of serious side effects and drug resistance. Conventional liposome has been substantially used as drug carriers, but they lack targeting character with lower drug bioavailability in tumor tissues. Based on the above problems, a novel estrogen-targeted PEGylated liposome loaded with oxaliplatin (ES-SSL-OXA) was prepared to further improve the metabolic behavior, the safety profile, and the anti-tumor efficacy of oxaliplatin. Methods Four kinds of oxaliplatin (OXA) liposomes were prepared by film hydration method. The obtained formulations were characterized in terms of entrapment efficiency (EE), particle size, and so on by HPLC and DLS (dynamic light scanning). The morphology of ES-SSL-OXA was detected by transmission electron microscope (TEM). The in vitro and in vivo targeting effect of ES-SSL-OXA was verified by fluorescence microscopy and in vivo imaging system in gastric cancer cells (SGC-7901) and tumor-bearing athymic mice. The in vitro and in vivo antitumor efficacies of ES-SSL-OXA were investigated on SGC-7901 cells and athymic tumor-bearing mice. Pharmacokinetic, biodistribution, and acute toxicity tests of ES-SSL-OXA were performed on ICR mice. Results The ES-SSL-OXA exhibited an average particle size of about 153.37 nm with an encapsulation efficiency of 46.20% and low leakage rates at 4°C and 25°C. In vivo and in vitro targeting study confirmed that ES-SSL-OXA could effectively target the tumor site. The antitumor activity demonstrated the strongest inhibition in tumor growth of ES-SSL-OXA. Pharmacokinetics and acute toxicity study showed that ES-SSL-OXA could significantly improve the metabolic behavior and toxicity profile of oxaliplatin. Conclusion In this study, a novel estrogen-targeted long-acting liposomal formulation of OXA was successfully prepared. ES fragment effectively targeted the delivery system to tumor tissues which highly express estrogen receptor, providing a promising therapeutic method for gastric cancer in clinic.
Collapse
Affiliation(s)
- Yuxin Sun
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Yizhuo Xie
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Huan Tang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Zhihui Ren
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Xue Luan
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Yan Zhang
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Ming Zhu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Zhe Lv
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Han Bao
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Yan Li
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Rui Liu
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Yujia Shen
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Yucui Zheng
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| | - Jin Pei
- Department of Biopharmacy, School of Pharmaceutical Sciences, Jilin University, ChangChun, People's Republic of China
| |
Collapse
|
22
|
Farinha P, Pinho JO, Matias M, Gaspar MM. Nanomedicines in the treatment of colon cancer: a focus on metallodrugs. Drug Deliv Transl Res 2022; 12:49-66. [PMID: 33616870 DOI: 10.1007/s13346-021-00916-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2021] [Indexed: 02/06/2023]
Abstract
Worldwide, colon cancer (CC) represents the fourth most common type of cancer and the fifth major cause of cancer-associated deaths. Surgical resection is considered the standard therapeutic choice for CC in early stages. However, in latter stages of the disease, adjuvant chemotherapy is essential for an appropriate management of this pathology. Metal-based complexes displaying cytotoxic properties towards tumor cells emerge as potential chemotherapeutic options. One metallodrug, oxaliplatin, was already approved for clinical use, playing an important role in the treatment of CC patients. Unfortunately, most of the newly designed metal-based complexes exhibit lack of selectivity against cancer cells, low solubility and permeability, high dose-limiting toxicity, and emergence of resistances. Nanodelivery systems enable the incorporation of metallodrugs at adequate payloads, solving the above-referred drawbacks. Moreover, drug delivery systems, depending on their physicochemical properties, are able to release the incorporated material preferentially at affected tissues/organs, enhancing the therapeutic activity in vivo, with concomitant fewer side effects. In this review, the general features and therapeutic management of CC will be addressed, with a special focus on preclinical or clinical studies using metal-based compounds. Furthermore, the use of different nanodelivery systems will also be described as tools to potentiate the therapeutic index of metallodrugs for the management of CC.
Collapse
Affiliation(s)
- Pedro Farinha
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Jacinta O Pinho
- Faculty of Pharmacy, Research Institute for Medicines, iMed.ULisboa, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Mariana Matias
- Faculty of Pharmacy, Research Institute for Medicines, iMed.ULisboa, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal.
| | - M Manuela Gaspar
- Faculty of Pharmacy, Research Institute for Medicines, iMed.ULisboa, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal.
| |
Collapse
|
23
|
Ali A, Akhtar J, Ahmad U, Basheer AS, Jaiswal N, Jahan A. Armamentarium in drug delivery for colorectal cancer. Crit Rev Ther Drug Carrier Syst 2022; 40:1-48. [DOI: 10.1615/critrevtherdrugcarriersyst.2022039241] [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]
|
24
|
Arroyo-García CM, Quinteros D, Palma SD, Jiménez de los Santos CJ, Moyano JR, Rabasco AM, González-Rodríguez ML. Synergistic Effect of Acetazolamide-(2-hydroxy)propyl β-Cyclodextrin in Timolol Liposomes for Decreasing and Prolonging Intraocular Pressure Levels. Pharmaceutics 2021; 13:2010. [PMID: 34959292 PMCID: PMC8709067 DOI: 10.3390/pharmaceutics13122010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 11/16/2021] [Accepted: 11/20/2021] [Indexed: 11/16/2022] Open
Abstract
The purpose of this study was to design, for the first time, a co-loaded liposomal formulation (CLL) for treatment of glaucoma including timolol maleate (TM) in the lipid bilayer and acetazolamide (Acz)-(2-hydroxy)propyl β-cyclodextrin (HPβCD) complexes (AczHP) solubilized in the aqueous core of liposomes. Formulations with TM (TM-L) and AczHP (AczHP-L), separately, were also prepared and characterized. A preliminary study comprising the Acz/HPβCD complexes and their interaction with cholesterol (a component of the lipid bilayer) was realized. Then, a screening study on formulation factors affecting the quality of the product was carried out following the design of the experiment methodology. In addition, in vitro release and permeation studies and in vivo lowering intraocular pressure (IOP) studies were performed. The results of the inclusion complexation behavior, characterization, and binding ability of Acz with HPβCD showed that HPβCD could enhance the water solubility of Acz despite the weak binding ability of the complex. Ch disturbed the stability and solubility parameters of Acz due to the fact of its competence by CD; thus, Chems (steroid derivative) was selected for further liposome formulation studies. The optimization of the lipid bilayer composition (DDAB, 0.0173 mmol and no double loading) and the extrusion as methods to reduce vesicle size were crucial for improving the physico-chemical properties and encapsulation efficiency of both drugs. In vitro release and permeation studies demonstrated that the CLL formulation showed improvement in in vitro drug release and permeation compared to the liposomal formulations with a single drug (TM-L and AczHP-L) and the standard solutions (TM-S and AczHP-S). CLL showed high efficacy in reducing and prolonging IOP, suggesting that the synergistic effect of TM and Acz on aqueous humor retention and the presence of this cyclodextrin and liposomes as permeation enhancers are responsible for the success of this strategy of co-loading for glaucoma therapy.
Collapse
Affiliation(s)
- Carmen M. Arroyo-García
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González, 2, 41012 Sevilla, Spain; (C.M.A.-G.); (C.J.J.d.l.S.); (J.R.M.); (A.M.R.)
| | - Daniela Quinteros
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Universidad Nacional de Córdoba, Ciudad Universitaria, Córdoba 5000, Argentina; (D.Q.); (S.D.P.)
- Departamento de Farmacia, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, Córdoba 5000, Argentina
| | - Santiago D. Palma
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), CONICET, Universidad Nacional de Córdoba, Ciudad Universitaria, Córdoba 5000, Argentina; (D.Q.); (S.D.P.)
- Departamento de Farmacia, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Ciudad Universitaria, Córdoba 5000, Argentina
| | - Cesáreo J. Jiménez de los Santos
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González, 2, 41012 Sevilla, Spain; (C.M.A.-G.); (C.J.J.d.l.S.); (J.R.M.); (A.M.R.)
| | - José R. Moyano
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González, 2, 41012 Sevilla, Spain; (C.M.A.-G.); (C.J.J.d.l.S.); (J.R.M.); (A.M.R.)
| | - Antonio M. Rabasco
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González, 2, 41012 Sevilla, Spain; (C.M.A.-G.); (C.J.J.d.l.S.); (J.R.M.); (A.M.R.)
| | - María Luisa González-Rodríguez
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, Universidad de Sevilla, C/Prof. García González, 2, 41012 Sevilla, Spain; (C.M.A.-G.); (C.J.J.d.l.S.); (J.R.M.); (A.M.R.)
| |
Collapse
|
25
|
Zhang L, Zhang S, Jiang M, Lu L, Ding Y, Ma N, Zhao Y, Xuchen S, Zhang N. Novel Timosaponin AIII-Based Multifunctional Liposomal Delivery System for Synergistic Therapy Against Hepatocellular Carcinoma Cancer. Int J Nanomedicine 2021; 16:5531-5550. [PMID: 34429598 PMCID: PMC8379713 DOI: 10.2147/ijn.s313759] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction As high cholesterol level has been reported to be associated with cancer cell growth and cholesterol is vulnerable to oxidation, the conventional liposomes including cholesterol in the formulation seem to be challenged. Timosaponin AIII (TAIII), as a steroid saponin from Anemarrhena asphodeloides Bunge, possesses a similar structure with cholesterol and exhibits a wide range of antitumor activities, making it possible to develop a TAIII-based liposome where TAIII could potentially stabilize the phospholipid bilayer as a substitution of cholesterol and work as a chemotherapeutic drug as well. Meanwhile, TAIII could enhance the uptake of doxorubicin hydrochloride (DOX) in human hepatocellular carcinoma (HCC) cells and exhibit synergistic effect. Thus, we designed a novel thermally sensitive multifunctional liposomal system composed of TAIII and lipids to deliver DOX for enhanced HCC treatment. Methods The synergistic effects of DOX and TAIII were explored on HCC cells and the tumor inhibition rate of TAIII-based liposomes carrying DOX was evaluated on both subcutaneous and orthotopic transplantation tumor models. TAIII-based multifunctional liposomes were characterized. Results Synergistic HCC cytotoxicity was achieved at molar ratios of 1:1, 1:2 and 1:4 of DOX/TAIII. TAIII-based liposomes carrying a low DOX dose of 2 mg/kg exhibited significantly enhanced antitumor activity than 5 mg/kg of DOX without detected cardiotoxicity on both subcutaneous and orthotopic transplantation tumor models. TAIII-based liposomes were characterized with smaller size than cholesterol liposomes but exhibited favorable stability. Mild hyperthermia generated by laser irradiation accelerated the release of DOX and TAIII from liposomes at tumor site, and cell permeability of TAIII enhanced uptake of DOX in HCC cells. Conclusion The innovative application of TAIII working as bilayer stabilizer and chemotherapeutic drug affords a stable multifunctional liposomal delivery system for synergistic therapy against HCC, which may be referred for the development of other types of saponins with similar property.
Collapse
Affiliation(s)
- Lijuan Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Shengan Zhang
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Min Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Lu Lu
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Yue Ding
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Ninghui Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Yuan Zhao
- Center of Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Sihan Xuchen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Nailian Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| |
Collapse
|
26
|
Li Z, Wang F, Li Y, Wang X, Lu Q, Wang D, Qi C, Li C, Li Z, Lian B, Tian G, Gao Z, Zhang B, Wu J. Combined anti-hepatocellular carcinoma therapy inhibit drug-resistance and metastasis via targeting "substance P-hepatic stellate cells-hepatocellular carcinoma" axis. Biomaterials 2021; 276:121003. [PMID: 34273686 DOI: 10.1016/j.biomaterials.2021.121003] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 06/21/2021] [Accepted: 07/01/2021] [Indexed: 12/13/2022]
Abstract
Peripheral nerves have emerged as the important components in tumor microenvironment (TME), which could activate hepatic stellate cells (HSCs) by secreting substance P (SP), leading to hepatocellular carcinoma (HCC) invasion and metastasis. Herein, we proposed a novel anti-HCC concept of blocking "SP-HSCs-HCC" axis for omnidirectional inhibition of HCC development. To pursue this aim, the novel CAP/GA-sHA-DOX NPs were developed for targeted co-delivery of capsaicin (CAP) and doxorubicin (DOX) using glycyrrhetinic acid (GA) modified sulfated-HA (sHA) as nanocarriers. Among that, CAP could inhibit the activation of HSCs as an inhibitor of SP. Notably, to real mimic "SP-HSCs-HCC" axis for in vitro and in vivo evaluation, both "SP + LX-2+BEL-7402" co-cultured cell model and "SP + m-HSC + H22" co-implantation mice model were attempted for the first time. Furthermore, in vivo anti-HCC effects were performed in three different tumor-bearing models: subcutaneous implantation of H22 or "SP + m-HSC + H22", intravenous injection of H22 for lung metastasis, and orthotopic implantation of H22 for primary HCC. Our results showed that CAP/GA-sHA-DOX NPs could be efficiently taken up by tumor cells and activated HSCs (aHSCs) simultaneously, and effectively inhibit tumor drug-resistance and migration by blocking SP-induced HSCs activation. In addition, CAP/GA-sHA-DOX NPs exhibited low ECM deposition, less tumor angiogenesis, and superior in vivo anti-HCC effects. The anti-HCC mechanisms revealed that CAP/GA-sHA-DOX NPs could down-regulate the expression level of Vimentin and P-gp, reverse epithelial-mesenchymal transition (EMT) of tumor cells. In brief, the nano-sized combination therapy based on GA-sHA-DOX polymers could effectively inhibit drug-resistance and metastasis of HCC by blocking "SP-HSCs-HCC" axis, which provides a promising approach for cancer therapy.
Collapse
Affiliation(s)
- Zhipeng Li
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Fangqing Wang
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Yanying Li
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Xiaoxue Wang
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Qiao Lu
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Di Wang
- School of Nursing, Weifang Medical University, PR China
| | - Cuiping Qi
- School of Nursing, Weifang Medical University, PR China
| | - Chenglei Li
- School of Pharmacy, Weifang Medical University, Weifang, 261053, PR China
| | - Zhaohuan Li
- School of Pharmacy, Weifang Medical University, Weifang, 261053, PR China
| | - Bo Lian
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Guixiang Tian
- School of Bioscience and Technology, Weifang Medical University, PR China
| | - Zhiqin Gao
- School of Bioscience and Technology, Weifang Medical University, PR China.
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang, 261053, PR China.
| | - Jingliang Wu
- School of Bioscience and Technology, Weifang Medical University, PR China.
| |
Collapse
|
27
|
Li C, Li Z, Gong X, Liu J, Zheng T, Wang F, Wu J, Zhang B. Acidic tumor microenvironment-sensitive liposomes enhance colorectal cancer therapy by acting on both tumor cells and cancer-associated fibroblasts. NANOSCALE 2021; 13:10509-10525. [PMID: 34096957 DOI: 10.1039/d1nr01506k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cancer-associated fibroblasts (CAFs) play a crucial role in facilitating tumor invasion and metastasis, which act as the "soil" in the tumor microenvironment (TME). Accordingly, it would be a promising strategy to enhance the antitumor effect by killing both tumor cells and CAFs simultaneously. Herein, novel TME acid-responsive liposomes for co-delivery of IRI and 398 (IRI&398-s-LPs) were developed, in which the rapid release of both drugs could be triggered under acidic conditions. Notably, a CT-26/3T3 cell co-culture system was used to mimic the real TME both in vitro and in vivo. Cellular immunofluorescence revealed that IRI&398-s-LPs could efficiently decrease the activation of CAFs. In vitro cytotoxicity evaluation demonstrated that IRI&398-s-LPs exhibited higher cytotoxicity than the other liposomal formulations in the CT-26 and CT-26/3T3 cell co-culture system. In vivo NIRF imaging showed that the IRI&398-s-LPs could increase drug accumulation in the tumor sites. Furthermore, IRI&398-s-LPs not only presented superior in vivo anti-tumor activity in CT-26 bearing BALB/c mice, but also enhanced the effect in CT-26/3T3 cell bearing mice with decreased collagen and CAF biomarker expression. Furthermore, IRI&398-s-LPs also presented superior anti-metastatic efficiency in a lung metastasis model. These results indicated that this combinational strategy for eliminating both tumor cells and CAFs provides a new approach for cancer therapy, and the prepared TME-responsive liposomes for co-delivery of drugs hold promising clinical application prospects.
Collapse
Affiliation(s)
- Chenglei Li
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 260153, China.
| | - Zhaohuan Li
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 260153, China.
| | - Xue Gong
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, China.
| | - Jianhao Liu
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 260153, China.
| | - Tingyue Zheng
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 260153, China.
| | - Fangqing Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, China.
| | - Jingliang Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, China.
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 260153, China.
| |
Collapse
|
28
|
Self-targeted polymersomal co-formulation of doxorubicin, camptothecin and FOXM1 aptamer for efficient treatment of non-small cell lung cancer. J Control Release 2021; 335:369-388. [PMID: 34058270 DOI: 10.1016/j.jconrel.2021.05.039] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 12/25/2022]
Abstract
In spite of huge developments in cancer treatment, versatile combinational formulations of different chemotherapeutic agents to enhance anticancer activity while reducing systemic toxicity still remains a challenge. In this regard, in the current study, an amphiphilic hyaluronic acid-b-polycaprolactone diblock copolymer was synthesized using "click chemistry". The synthesized copolymer was self-assembled to form polymersomal structures for co-encapsulation of hydrophilic doxorubicin (DOX) and hydrophobic camptothecin (CPT) in their interior aqueous compartment and their bilayer, respectively with 1:10 and 1:1 ratios. The prepared polymersomal combinational formulation surrounded by hyaluronic acid brush as hydrophilic segment, could provide active targeting of the system against CD44 marker expressed on the surface of cancerous cells. The hyaluronic acid shell could also provide flexible chemistry for the conjugation of therapeutic FOXM1-specific DNA aptamer (Forkhead Box M1; against transcription factor FOXM1) on the surface of polymersomes in order to further suppress cancerous cell proliferation. The obtained results demonstrated that the prepared co-formulation provided sustained, controlled release of the entrapped drugs during 200 h. In vitro cytotoxicity experiments on non-small cell lung cancer, A549 and SK-MES-1 cell lines, demonstrated that the co-formulation of DOX and CPT provided synergistic effect and significantly higher cytotoxicity in comparison with free drugs. The cytotoxicity experiment also indicated that the aptamer conjugation on the co-formulations surface could significantly increase the cytotoxicity and induce apoptosis in combination therapy on both A549 and SK-MES-1 cell lines while aptamer-conjugated blank NPs did not show any cytotoxicity which emphasizes on the sensitization capability of the FOXM1 DNA aptamer against non-small cell lung cancer. Furthermore, it was shown that the co-formulation with or without aptamer renders the formulation specific tumor accumulation in vivo 24 h post-administration, assisting the combination synergy observed in vitro to be translated to in vivo antitumor efficacy. This combinatorial delivery platform strongly offers a novel approach for the synergistic controlled transportation of several chemotherapeutics for the treatment of non-small cell lung cancer.
Collapse
|
29
|
Li Y, Jia F, Deng X, Wang X, Lu J, Shao L, Cui X, Pan Z, Wu Y. Combinatorial miRNA-34a replenishment and irinotecan delivery via auto-fluorescent polymeric hybrid micelles for synchronous colorectal cancer theranostics. Biomater Sci 2021; 8:7132-7144. [PMID: 33150879 DOI: 10.1039/d0bm01579b] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The synergistic combination of microRNA (miRNA) modulation and chemotherapy has emerged as an effective strategy to combat cancer. Irinotecan (IRI) is a potent antitumor chemotherapeutic in clinical practice and has been used for treating various malignant tumors, including colorectal cancer (CRC). However, IRI is not effective for advanced CRC or metastatic behavior. Herein, novel polymeric hybrid micelles were engineered based on two different amphiphilic copolymers, polyethyleneimine-poly(d,l-lactide) (PEI-PLA) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy (polyethyleneglycol) (DSPE-PEG), in which IRI and a tumor suppressive microRNA-34a (miR-34a) gene were efficiently co-loaded (MINPs) to achieve a chemo-miRNA combination therapy against CRC. MINPs were successfully constructed by two-step film dispersion and electrostatic interaction methods. IRI and miR-34a could be efficaciously encapsulated as MINPs and transferred to CRC cells. After encapsulation, MINPs would then upregulate miR-34a expression and regulate miR-34a-related downstream genes, which in turn led to enhanced cell cytotoxicity and apoptosis ratios. MINPs presented an excitation-dependent multi-wavelength emission feature due to the intrinstic fluorescence properties of PEI-PLA and could be utilized for in vitro/vivo imaging. According to the in vivo experimental results, MINPs possess the great characteristic of accumulating in situ in a tumor site and lightening it after intravenous administration. Furthermore, MINPs presented extraordinary antitumor efficacy owing to the combined therapy effects of IRI and miR-34a with good biocompability. Overall, our findings validated MINPs-mediated miR-34a replenishment and IRI co-delivery to serve as an effective theranostic platform and provided an innovative horizon for combining chemo-gene therapy against CRC.
Collapse
Affiliation(s)
- Yunhao Li
- Department of General Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, P. R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Cheraga N, Ouahab A, Shen Y, Huang NP. Characterization and Pharmacokinetic Evaluation of Oxaliplatin Long-Circulating Liposomes. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5949804. [PMID: 33987441 PMCID: PMC8079196 DOI: 10.1155/2021/5949804] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 03/20/2021] [Accepted: 04/07/2021] [Indexed: 11/18/2022]
Abstract
The clinical efficacy of Oxaliplatin (L-OHP) is potentially limited by dose-dependent neurotoxicity and high partitioning to erythrocytes in vivo. Long-circulating liposomes could improve the pharmacokinetic profile of L-OHP and thus enhance its therapeutic efficacy and reduce its toxicity. The purpose of this study was to prepare L-OHP long-circulating liposomes (L-OHP PEG lip) by reverse-phase evaporation method (REV) and investigate their pharmacokinetic behavior based on total platinum in rat plasma using atomic absorption spectrometry (AAS). A simple and a sensitive AAS method was developed and validated to determine the total platinum originated from L-OHP liposomes in plasma. Furthermore, long-circulating liposomes were fully characterized in vitro and showed great stability when stored at 4°C for one month. The results showed that the total platinum in plasma of L-OHP long-circulating liposomes displayed a biexponential pharmacokinetic profile with five folds higher bioavailability and longer distribution half-life compared to L-OHP solution. Thus, long-circulating liposomes prolonged L-OHP circulation time and may present a potential candidate for its tumor delivery. Conclusively, the developed AAS method could serve as a reference to investigate the pharmacokinetic behavior of total platinum in biological matrices for other L-OHP delivery systems.
Collapse
Affiliation(s)
- Nihad Cheraga
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Ammar Ouahab
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Yan Shen
- Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Ning-Ping Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| |
Collapse
|
31
|
Du C, Li S, Li Y, Galons H, Guo N, Teng Y, Zhang Y, Li M, Yu P. F7 and topotecan co-loaded thermosensitive liposome as a nano-drug delivery system for tumor hyperthermia. Drug Deliv 2021; 27:836-847. [PMID: 32508162 PMCID: PMC8216433 DOI: 10.1080/10717544.2020.1772409] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In order to enhance the targeting efficiency and reduce anti-tumor drug’s side effects, topotecan (TPT) and F7 were co-loaded in thermosensitive liposomes (F7-TPT-TSL), which show enhanced permeability and retention in tumors, as well as local controlled release by heating in vitro. TPT is a water-soluble inhibitor of topoisomerase I that is converted to an inactive carboxylate structure under physiological conditions (pH 7.4). F7 is a novel drug significantly resistant to cyclin-dependent kinase but its use was restricted by its high toxicity. F7-TPT-TSL had excellent particle distribution (about 103 nm), high entrapment efficiency (>95%), obvious thermosensitive property, and good stability. Confocal microscopy demonstrated specific higher accumulation of TSL in tumor cells. MTT proved F7-TPT-TSL/H had strongest cell lethality compared with other formulations. Then therapeutic efficacy revealed synergism of TPT and F7 co-loaded in TSL, together with hyperthermia. Therefore, the F7-TPT-TSL may serve as a promising system for temperature triggered cancer treatment.
Collapse
Affiliation(s)
- Chunyang Du
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Shuangshuang Li
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Yuan Li
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Hervé Galons
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China.,Institut Parisien de Chimie Moléculaire, UMR CNRS 8232, Paris, France
| | - Na Guo
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Yuou Teng
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Yongmin Zhang
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China.,Institut Parisien de Chimie Moléculaire, UMR CNRS 8232, Paris, France
| | - Mingyuan Li
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Peng Yu
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| |
Collapse
|
32
|
Fumoto S, Nishida K. Co-delivery Systems of Multiple Drugs Using Nanotechnology for Future Cancer Therapy. Chem Pharm Bull (Tokyo) 2021; 68:603-612. [PMID: 32611997 DOI: 10.1248/cpb.c20-00008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cancer treatments have improved significantly during the last decade but are not yet satisfactory. Combination therapy is often administered to improve efficacy and safety. Drug delivery systems can also improve efficacy and safety. To control the spatiotemporal distribution of drugs, nanotechnology involving liposomes, solid lipid nanoparticles, and polymeric micelles has been developed. Co-delivery systems of multiple drugs are a promising approach to combat cancer. Synergistic effects and reduced side effects are expected from the use of co-delivery systems. In this review, we summarize various co-delivery systems for multiple drugs, including small-molecule drugs, nucleic acids, genes, and proteins. Co-delivery of drugs with different properties is relatively difficult, but some researchers have succeeded in developing such co-delivery systems. Environment-responsive carrier designs can control the release of cargos. Although their preparation is more complicated than that of mono-delivery systems, co-delivery systems can simplify clinical procedures and improve patient QOL.
Collapse
Affiliation(s)
| | - Koyo Nishida
- Graduate School of Biomedical Sciences, Nagasaki University
| |
Collapse
|
33
|
Sun S, Du X, Fu M, Khan AR, Ji J, Liu W, Zhai G. Galactosamine-modified PEG-PLA/TPGS micelles for the oral delivery of curcumin. Int J Pharm 2021; 595:120227. [PMID: 33484915 DOI: 10.1016/j.ijpharm.2021.120227] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 12/19/2020] [Accepted: 01/04/2021] [Indexed: 12/18/2022]
Abstract
In this study, galactosamine-modified poly(ethylene glycol)-poly(lactide) (Gal-PEG-PLA) polymers were synthesized and Gal-PEG-PLA/D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) micelles named as GPP micelles were designed to promote the oral absorption of a hydrophobic drug, curcumin (CUR). CUR-loaded Gal-PEG-PLA/TPGS micelles (CUR@GPP micelles) were fabricated using the thin-film dispersion method. CUR@GPP micelles had a size of about 100 nm, a near-neutral zeta potential, drug loading (DL) of 14.6%, and sustained release properties. GPP micelles with high Gal density (GPP3 micelles) were superior in facilitating uptake in epithelial cells and improving intestinal permeation. In situ intestinal absorption studies suggested that the jejunum and ileum were the best absorption segments in the intestinal tract. Additionally, biodistribution results revealed that GPP3 micelles could be remarkably taken up by the jejunum and ileum. Pharmacokinetics revealed that the maximum plasma concentration (Cmax) and the area under the plasma concentration-time curve from 0 to 24 h (AUC0-24) for CUR@GPP3 micelles were both significantly increased, and that the relative bioavailability of CUR@GPP3 micelles to CUR-loaded mPEG-PLA/TPGS micelles (CUR@PP micelles) was 258.8%. Furthermore, CUR-loaded micelles could reduce damage to the liver and intestinal tissues. This study highlights the importance of Gal content in the design of targeting nanocarrier Gal-modified micelles, which have broad prospects for oral delivery of hydrophobic drugs. Therefore, they could serve as a promising candidate for targeted delivery to the liver.
Collapse
Affiliation(s)
- Siyu Sun
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan 250012, China
| | - Xiyou Du
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan 250012, China
| | - Manfei Fu
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan 250012, China
| | - Abdur Rauf Khan
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan 250012, China
| | - Jianbo Ji
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan 250012, China
| | - Weidong Liu
- Department of Pharmacy, Linyi People's Hospital Affiliated to Shandong University, Linyi 276003, China.
| | - Guangxi Zhai
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan 250012, China.
| |
Collapse
|
34
|
Lian B, Wei H, Pan R, Sun J, Zhang B, Wu J, Li X, Tian G. Galactose Modified Liposomes for Effective Co-Delivery of Doxorubicin and Combretastatin A4. Int J Nanomedicine 2021; 16:457-467. [PMID: 33488080 PMCID: PMC7816220 DOI: 10.2147/ijn.s283793] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/01/2020] [Indexed: 12/28/2022] Open
Abstract
Background Tumor angiogenesis plays a crucial role in tumor development, and recent efforts have been focused on combining proapoptotic and antiangiogenic activities to enhance antitumor therapy. Methods In this study, galactose-modified liposomes (Gal-LPs) were prepared for co-delivery of doxorubicin (DOX) and combretastatin A4 phosphate (CA4P). The co-cultured system composed of BEL-7402 and human umbilical vein endothelial cells (HUVEC) cells was established to effectively evaluate in vitro anti-tumor activity through cell viability and cell migration assay. Furthermore, both in vivo bio-distribution and anti-hepatoma effect of DOX&CA4P/Gal-LPs were investigated on H22 tumor cell-bearing mice. Results The results showed that DOX&CA4P/Gal-LPs were spherical with a mean particle size of 143 nm, and could readily be taken up by BEL-7402 cells. Compared with a mixture of free DOX and CA4P, the DOX&CA4P/Gal-LPs were more effective in inhibiting cell migration and exhibited stronger cytotoxicity against BEL-7402 cells alone or a co-cultured system. The in vitro studies showed that the co-cultured system was a more effective model to evaluate the anti-tumor activity of combination therapy. Moreover, DOX&CA4P/Gal-LPs exhibited a greater anti-hepatoma effect than other drug formulations, indicating that Gal-LPs could promote drug accumulation in the tumor region and improve the anti-tumor activity. Conclusion Gal-LPs co-loaded with chemotherapeutic and antiangiogenic drugs are a promising strategy for anti-hepatoma therapy.
Collapse
Affiliation(s)
- Bo Lian
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Hua Wei
- Department of Endocrinology, ShouGuang Peoples' Hospital, Weifang 262700, People's Republic of China
| | - Ruiyan Pan
- School of Pharmacy, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Jingui Sun
- Department of Oncology, ShouGuang Peoples' Hospital, Weifang 262700, People's Republic of China
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Jingliang Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Xiujie Li
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, People's Republic of China
| | - Guixiang Tian
- School of Bioscience and Technology, Weifang Medical University, Weifang 261053, People's Republic of China
| |
Collapse
|
35
|
Li Y, Xu P, He D, Xu B, Tu J, Shen Y. Long-Circulating Thermosensitive Liposomes for the Targeted Drug Delivery of Oxaliplatin. Int J Nanomedicine 2020; 15:6721-6734. [PMID: 32982229 PMCID: PMC7494235 DOI: 10.2147/ijn.s250773] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 08/07/2020] [Indexed: 12/17/2022] Open
Abstract
Introduction Oxaliplatin (L-OHP) is a well-known third-generation platinum anticancer drug with severe systemic- and neuro-toxicity. The main objective of the current research was to develop a targeted long-circulating thermosensitive smart-release liposome (LCTL) system for better therapeutic efficacy and less toxicity. Methods The reverse-phase evaporation method (REV) was used to prepare L-OHP loaded LCTL (L-OHP/LCTL). The physical characteristics were evaluated including encapsulation efficiency (EE), size, zeta potential and stability. The release behavior, cytotoxicity and in vivo evaluation were also carried out. Results EE of LCTL was around 25% with a uniform size distribution, and LCTL achieved almost complete release at 42°C while it was only 10% at 37°C. Moreover, the LCTL showed significantly higher cytotoxicity at 42°C than that at 37°C. The in vivo results indicated LCTL could target tumors and enhance retention for more than 24 h, thereby enhancing anti-tumor efficacy on 4T1-bearing mice. Discussion These results indicated that LCTL not only possessed a prolonged circulation time but it also enhanced accumulation and achieved selective release at the tumor sites. Conclusively, LCTL could serve as a promising carrier for oxaliplatin delivery to treat solid tumors.
Collapse
Affiliation(s)
- Yanan Li
- China Pharmaceutical University, Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Nanjing 210009, People's Republic of China
| | - Pengcheng Xu
- China Pharmaceutical University, Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Nanjing 210009, People's Republic of China
| | - Dongsheng He
- China Pharmaceutical University, Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Nanjing 210009, People's Republic of China
| | - Bohui Xu
- School of Pharmacy, Nantong University, Nantong 226001, People's Republic of China
| | - Jiasheng Tu
- China Pharmaceutical University, Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Nanjing 210009, People's Republic of China
| | - Yan Shen
- China Pharmaceutical University, Center for Research Development and Evaluation of Pharmaceutical Excipients and Generic Drugs, Nanjing 210009, People's Republic of China
| |
Collapse
|
36
|
Alavi M, Webster TJ. Nano liposomal and cubosomal formulations with platinum-based anticancer agents: therapeutic advances and challenges. Nanomedicine (Lond) 2020; 15:2399-2410. [PMID: 32945246 DOI: 10.2217/nnm-2020-0199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Nephrotoxicity, neurotoxicity and multidrug resistance in tumor cells can result from platinum-based anticancer (PBA) agents which can be reduced by nano formulations. Recently, novel formulations based on liposomes and cubosomes have been described as efficient strategies to overcome nephrotoxicity, ototoxicity, neurotoxicity, cardiotoxicity, hematological toxicities, hepatotoxicity and gastrointestinal toxicity as well as multidrug resistance. The co-delivery of anticancer agents concomitant with PBAs via biocompatible and biodegradable smart liposomes and cubosomes can augment therapeutic results of chemotherapy as well as radiotherapy owing to their high accessibility of surface and internal modification. For this purpose, surface, bilayer or core sections of these formulations can be functionalized by pure PBAs or modified PBAs. In this review, recent significant advances and challenges related to various liposomal and cubosomal formulations of PBA are presented in order to emphasize suitable formulations for anticancer applications with critical thoughts provided on how the field can progress.
Collapse
Affiliation(s)
- Mehran Alavi
- Nanobiotechnology Laboratory, Biology Department, Faculty of Science, Razi University, Kermanshah, Iran
| | - Thomas J Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
37
|
Hu Y, Zhang Y, Wang X, Jiang K, Wang H, Yao S, Liu Y, Lin YZ, Wei G, Lu W. Treatment of Lung Cancer by Peptide-Modified Liposomal Irinotecan Endowed with Tumor Penetration and NF-κB Inhibitory Activities. Mol Pharm 2020; 17:3685-3695. [PMID: 32816496 DOI: 10.1021/acs.molpharmaceut.0c00052] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Current chemotherapy for lung cancer achieved limited efficacy due to poor tumor targeting and tissue penetration. Another obstacle in the therapy is activated nuclear factor-κB (NF-κB) in tumor cells, which plays a crucial role in promotion of antiapoptosis and drug resistance. In this study, we utilized a multifunctional liposome loaded with irinotecan and surface modified with a cell-permeable NF-κB inhibitor (CB5005), for treatment of non-small-cell lung carcinoma. CB5005 downregulated the level of NF-κB-related protein in the nuclei of A549 cells, and increased cellular uptake of the modified liposomes. In vivo antitumor activity in mice bearing A549 xenografts revealed that modification with CB5005 significantly improved the tumor inhibition rate of irinotecan. Immunohistochemical assays showed that the tumors treated with CB5005-modified liposomes possessed the most apoptotic cells and the lowest level of p50 in the cell nuclei. These results strongly suggest that antitumor efficacy of the irinotecan liposomes can be enhanced by tumor-penetrating and NF-κB-inhibiting functions of CB5005. Consequently, CB5005-modified liposomes provide a possible synergistic therapy for lung cancer, and would also be appropriate for other types of tumors associated with elevated NF-κB activity.
Collapse
Affiliation(s)
- Yang Hu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Yanyu Zhang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Xiaoyi Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Kuan Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Huan Wang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Shengyu Yao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Yu Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Yao-Zhong Lin
- Celtek Bioscience, LLC, 2550 Meridian Boulevard, Suite 200, Franklin, Tennessee 37067, United States
| | - Gang Wei
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.,The Institutes of Integrative Medicine of Fudan University, Shanghai 200040, China.,Shanghai Engineering Research Center of ImmunoTherapeutics, Shanghai 201203, China
| | - Weiyue Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, Department of Pharmaceutics, School of Pharmacy, Fudan University, 826 Zhangheng Road, Shanghai 201203, China.,The Institutes of Integrative Medicine of Fudan University, Shanghai 200040, China
| |
Collapse
|
38
|
Wang R, Yang Y, Yang M, Yuan D, Huang J, Chen R, Wang H, Hu L, Di L, Li J. Synergistic inhibition of metastatic breast cancer by dual-chemotherapy with excipient-free rhein/DOX nanodispersions. J Nanobiotechnology 2020; 18:116. [PMID: 32847586 PMCID: PMC7449082 DOI: 10.1186/s12951-020-00679-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 08/17/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The management of metastatic cancer remains a major challenge in cancer therapy worldwide. The targeted delivery of chemotherapeutic drugs through rationally designed formulations is one potential therapeutic option. Notably, excipient-free nanodispersions that are entirely composed of pharmaceutically active molecules have been evaluated as promising candidates for the next generation of drug formulations. Formulated from the self-assembly of drug molecules, these nanodispersions enable the safe and effective delivery of therapeutic drugs to local disease lesions. Here, we developed a novel and green approach for preparing nanoparticles via the self-assembly of rhein (RHE) and doxorubicin (DOX) molecules, named RHE/DOX nanoparticles (RD NPs); this assembly was associated with the interaction force and did not involve any organic solvents. RESULTS According to molecular dynamics (MD) simulations, DOX molecules tend to assemble around RHE molecules through intermolecular forces. This intermolecular retention of DOX was further improved by the nanosizing effect of RD NPs. Compared to free DOX, RD NPs exerted a slightly stronger inhibitory effect on 4T1 cells in the scratch healing assay. As a dual drug-loaded nanoformulation, the efficacy of RD NPs against tumor cells in vitro was synergistically enhanced. Compared to free DOX, the combination of DOX and RHE in nanoparticles exerted a synergistic effect with a combination index (CI) value of 0.51 and showed a stronger ability to induce cell apoptosis. Furthermore, the RD NP treatment not only effectively suppressed primary tumor growth but also significantly inhibited tumor metastasis both in vitro and in vivo, with a better safety profile. CONCLUSIONS The generation of pure nanodrugs via a self-assembly approach might hold promise for the development of more efficient and novel excipient-free nanodispersions, particularly for two small molecular antitumor drugs that potentially exert synergistic antiproliferative effects on metastatic breast cancer.
Collapse
Affiliation(s)
- Ruoning Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Yujie Yang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Mengmeng Yang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Dandan Yuan
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
| | - Jinyu Huang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Rui Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
| | - Honglan Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Lihong Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, Nanjing, China
| | - Liuqing Di
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China
| | - Junsong Li
- School of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, China.
- Jiangsu Engineering Research Center for Efficient Delivery System of TCM, Nanjing, China.
| |
Collapse
|
39
|
Study on different particle sizes of DOX-loaded mixed micelles for cancer therapy. Colloids Surf B Biointerfaces 2020; 196:111303. [PMID: 32798988 DOI: 10.1016/j.colsurfb.2020.111303] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/15/2020] [Accepted: 07/31/2020] [Indexed: 12/17/2022]
Abstract
Nano-based drug delivery systems have been widely applied in cancer therapy, among that, particle sizes may affect the delivery efficiency of nanocarriers. The purpose of this study was to evaluate the potential impacts of particle size on tumor therapy, in consideration of this, lipid/glycocholic acid mixed micelles (LGs) were designed as the model nanocarriers. Doxorubicin (DOX) loaded LGs with two different particle sizes at around 10 nm and 100 nm, respectively, were successfully prepared by controlling the ratio of EPC to GAH. In vitro release study showed that the release behaviors of DOX in mixed micelles with two different particle sizes was basically consistent and showed sustained release. DOX-LGs at 10 nm exhibited higher cellular uptake capacity, compared with DOX-LGs at 100 nm. Besides, in vivo NIFR imaging also demonstrated that DOX-LGs at 10 nm had more accumulation in tumor site. Furthermore, DOX-LGs at 10 nm presented both higher in vitro cytotoxicity and superior in vivo antitumor activity than that of 100 nm. In vivo safety evaluations showed that the mixed micelles had lower toxicities than free DOX solution formulations. These results indicated that the nanoparticles with smaller particle size could improve the profiles in cellular uptake, tumor accumulation as well as anti-tumor efficacy, which would provide a theoretical principle for the design of nanoparticles.
Collapse
|
40
|
Li J, Zheng L, Wang R, Sun D, Liang S, Wu J, Liu Y, Tian X, Li T, Yang Y, Han L. Synergistic Combination of Sodium Aescinate-Stabilized, Polymer-Free, Twin-Like Nanoparticles to Reverse Paclitaxel Resistance. Int J Nanomedicine 2020; 15:5839-5853. [PMID: 32848393 PMCID: PMC7428345 DOI: 10.2147/ijn.s259432] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The development of paclitaxel (PTX) resistance seriously restricts its clinical efficacy. An attractive option for combating resistance is inhibiting the expression of P-glycoprotein (P-gp) in tumor cells. We have reported that flavokawain A (FKA) inhibited P-gp protein expression in PTX-resistant A549 (A549/T) cells, indicating that FKA combined with PTX may reverse PTX resistance. However, due to the variable pharmacokinetics of FKA and PTX, the conventional cocktail combination in clinics may cause uncertainty of treatment efficacy in vivo. MATERIALS AND METHODS To synergistically elevate the anti-cancer activity of PTX and FKA in vivo, the national medical products administration (NMPA) approved sodium aescinate (Aes) was utilized to stabilize hydrophobic PTX and FKA to form polymer-free twin like PTX-A nanoparticles (NPs) and FKA-A NPs. RESULTS The resulting nanoparticles prepared simply by nanoprecipitation possessed similar particle size, good stability and ultrahigh drug loadings of up to 50%. With the aid of Aes, these two drugs accumulated in tumor tissue by passive targeting and were efficiently taken up by A549/T cells; this resulted in significant suppression of tumor growth in A549/T homograft mice at a low PTX dose (2.5 mg·kg-1). Synergistic effects and reversed PTX resistance were achieved by the combination of PTX-A NPs and FKA-A NPs by inhibiting P-gp expression in tumor cells. CONCLUSION Using NMPA-approved Aes to prepare twin-like nanoparticles without introducing any new materials provides an efficient platform for combination chemotherapy and clinical translation.
Collapse
Affiliation(s)
- Juan Li
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Lei Zheng
- Department of Pharmacy, Shandong Provincial Third Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250031, People’s Republic of China
| | - Rongmei Wang
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Deqing Sun
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Shuang Liang
- School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250012, People’s Republic of China
| | - Jing Wu
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Yongqing Liu
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Xiaona Tian
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| | - Tingting Li
- Department of Pharmaceutical Sciences, University of Connecticut, Storrs, CT, USA
| | - Yang Yang
- China National Center for Biotechnology Development, Beijing100039, People’s Republic of China
| | - Leiqiang Han
- Department of Clinical Pharmacy, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong250033, People’s Republic of China
| |
Collapse
|
41
|
Preparation and Evaluation of Intraperitoneal Long-Acting Oxaliplatin-Loaded Multi-Vesicular Liposomal Depot for Colorectal Cancer Treatment. Pharmaceutics 2020; 12:pharmaceutics12080736. [PMID: 32764318 PMCID: PMC7466130 DOI: 10.3390/pharmaceutics12080736] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 12/21/2022] Open
Abstract
Colorectal cancer with peritoneal metastasis has a poor prognosis because of inadequate responses to systemic chemotherapy. Cytoreductive surgery followed by intraperitoneal (IP) chemotherapy using oxaliplatin has attracted attention; however, the short half-life of oxaliplatin and its rapid clearance from the peritoneal cavity limit its clinical application. Here, a multivesicular liposomal (MVL) depot of oxaliplatin was prepared for IP administration, with an expected prolonged effect. After optimization, a combination of phospholipids, cholesterol, and triolein was used based on its ability to produce MVL depots of monomodal size distribution (1–20 µm; span 1.99) with high entrapment efficiency (EE) (92.16% ± 2.17%). An initial burst release followed by a long lag phase of drug release was observed for the MVL depots system in vitro. An in vivo pharmacokinetic study mimicking the early postoperative IP chemotherapy regimen in rats showed significantly improved bioavailability, and the mean residence time of oxaliplatin after IP administration revealed that slow and continuous erosion of the MVL particles yielded a sustained drug release. Thus, oxaliplatin-loaded MVL depots presented in this study have potential for use in the treatment of colorectal cancer.
Collapse
|
42
|
Yousefi Aldashi S, Saffari Z, Ebrahimi Shahmabadi H, Akbarzadeh A. In Vitro Assessment of Magnetic Liposomal Paclitaxel Nanoparticles as a Potential Carrier for the Treatment of Ovarian Cancer. Adv Pharm Bull 2020; 11:267-273. [PMID: 33880348 PMCID: PMC8046399 DOI: 10.34172/apb.2021.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/29/2020] [Accepted: 08/05/2020] [Indexed: 01/12/2023] Open
Abstract
Purpose: This study aimed to evaluate the role of magnetic liposome nanoparticles (ML NPs) as a carrier for paclitaxel (PTX) for the treatment of ovarian cancer in vitro. Methods: Magnetic NPs (MNPs) were synthesized by chemical co-precipitation method. The resulting NPs were characterized in terms of size, size distribution, zeta potential, drug encapsulation efficiency (EE), drug release pattern, and cytotoxicity effects. Results: The size and zeta potential of PTX-PEG-L and PTX-PEG-ML NPs were determined to be 296, 198 nm; -20, and -19 mV, respectively. Also, their drug encapsulation efficiencies were determined to be 97% and 96%, respectively. It was found that PTX-PEG-ML NPs, compared to PTX-PEG-L NPs, caused a reduction (11%) in the rate of drug release. The cytotoxicity of the drug-loaded NPs was assessed using 3-[4,5-dimethylthiazole-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay against human ovarian epithelial cancer (A2780CP) cells, and the results demonstrated that PTX-PEG-ML NPs caused higher cytotoxicity (by 14%) compared to PTX-PEG-L NPs (IC50: 1.88 ± 0.09 and 2.142 ± 0.1 µM, respectively). Conclusion: Overall, the results of this study suggest that PTX-PEG-ML NPs could be considered as a therapeutic candidate for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Sara Yousefi Aldashi
- Islamic Azad University Faculty of Technical and Engineering, Science and Research Branch, Tehran, Iran
| | - Zahra Saffari
- Department of Pilot Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Hasan Ebrahimi Shahmabadi
- Department of Microbiology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Azim Akbarzadeh
- Department of Pilot Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
43
|
He K, Liu J, Gao Y, Hao Y, Yang X, Huang G. Preparation and Evaluation of Stearylamine-Bearing Pemetrexed Disodium-Loaded Cationic Liposomes In Vitro and In Vivo. AAPS PharmSciTech 2020; 21:193. [PMID: 32661922 DOI: 10.1208/s12249-019-1586-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/20/2019] [Indexed: 01/10/2023] Open
Abstract
Pemetrexed disodium (PMX) stands out in the treatment of non-small cell lung cancer (NSCLC), but with short half-life and toxic side effects. This study was to design cationic liposomes for targeting delivery PMX to the lungs. The PMX cationic liposome was prepared by thin-film hydration using stearylamine (SA) as the positive component of charge-regulating charge. Then, the PMX cationic liposome (SA-PMX-Lips) was characterized by particle size, morphology, entrapment efficiency (EE), and drug loading (DL). Finally, the drug release behavior in vitro, the pharmacokinetic study, and tissue distribution of SA-PMX-Lips were evaluated separately, with PMX solution (PMX-Sol) and PMX liposome (PMX-Lips) as the control. According to results, SA-PMX-Lips were spherical and the particle size was 219.7 ± 4.97 nm with a narrow polydispersity index (PDI) (0.231 ± 0.024) and a positive zeta potential 22.2 ± 0.52 mV. Its EE was 92.39 ± 1.94% and DL was 9.15 ± 0.07%. The results of in vitro and in vivo experiments showed that SA-PMX-Lips released slowly, prolonged retention time and increased the value of AUC. More notably, SA-PMX-Lips could improve the accumulation of drugs in the lungs and the relative uptake rate (Re) was 2.35 in the lungs, which indicated its lung targeting. In summary, SA-PMX-Lips showed the potential for the effective delivery of PMX and the treatment of NSCLC.
Collapse
|
44
|
Mu W, Chu Q, Liu Y, Zhang N. A Review on Nano-Based Drug Delivery System for Cancer Chemoimmunotherapy. NANO-MICRO LETTERS 2020; 12:142. [PMID: 34138136 PMCID: PMC7770879 DOI: 10.1007/s40820-020-00482-6] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 06/11/2020] [Indexed: 05/11/2023]
Abstract
Although notable progress has been made on novel cancer treatments, the overall survival rate and therapeutic effects are still unsatisfactory for cancer patients. Chemoimmunotherapy, combining chemotherapeutics and immunotherapeutic drugs, has emerged as a promising approach for cancer treatment, with the advantages of cooperating two kinds of treatment mechanism, reducing the dosage of the drug and enhancing therapeutic effect. Moreover, nano-based drug delivery system (NDDS) was applied to encapsulate chemotherapeutic agents and exhibited outstanding properties such as targeted delivery, tumor microenvironment response and site-specific release. Several nanocarriers have been approved in clinical cancer chemotherapy and showed significant improvement in therapeutic efficiency compared with traditional formulations, such as liposomes (Doxil®, Lipusu®), nanoparticles (Abraxane®) and micelles (Genexol-PM®). The applications of NDDS to chemoimmunotherapy would be a powerful strategy for future cancer treatment, which could greatly enhance the therapeutic efficacy, reduce the side effects and optimize the clinical outcomes of cancer patients. Herein, the current approaches of cancer immunotherapy and chemoimmunotherapy were discussed, and recent advances of NDDS applied for chemoimmunotherapy were further reviewed.
Collapse
Affiliation(s)
- Weiwei Mu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Qihui Chu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, People's Republic of China.
| |
Collapse
|
45
|
Pang X, Liang S, Wang T, Yu S, Yang R, Hou T, Liu Y, He C, Zhang N. Engineering Thermo-pH Dual Responsive Hydrogel for Enhanced Tumor Accumulation, Penetration, and Chemo-Protein Combination Therapy. Int J Nanomedicine 2020; 15:4739-4752. [PMID: 32753862 PMCID: PMC7342477 DOI: 10.2147/ijn.s253990] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/01/2020] [Indexed: 01/20/2023] Open
Abstract
Purpose Combined chemotherapeutic drug and protein drug has been a widely employed strategy for tumor treatment. To realize both tumor accumulation and deep tumor penetration for drugs with different pharmacokinetics, we propose a structure-transformable, thermo-pH dual responsive co-delivery system to co-load granzyme B/docetaxel (GrB/DTX). Methods Thermo-sensitive hydrogels based on diblock copolymers (mPEG-b-PELG) were synthesized through ring opening polymerization. GrB/DTX mini micelles (GDM) was developed by co-loading these two drugs in pH-sensitive mini micelles, and the GDM-incorporated thermo-sensitive hydrogel (GDMH) was constructed. The thermo-induced gelation behavior of diblock copolymers and the physiochemical properties of GDMH were characterized. GDMH degradation and deep tumor penetration of released mini micelles were confirmed. The pH-sensitive disassembly and lysosomal escape abilities of released mini micelles were evaluated. In vitro cytotoxicity was studied using MTT assays and the in vivo antitumor efficacy study was evaluated in B16-bearing C57BL/6 mice. Results GDMH was gelatinized at body temperature and can be degraded by proteinase to release mini micelles. The mini micelles incorporated in GDMH can achieve deep tumor penetration and escape from lysosomes to release GrB and DTX. MTT results showed that maximum synergistic antitumor efficacy of GrB and DTX was observed at mass ratio of 1:100. Our in vivo antitumor efficacy study showed that GDMH inhibited tumor growth in the subcutaneous tumor model and in the post-surgical recurrence model. Conclusion The smart-designed transformable GDMH can facilitate tumor accumulation, deep tumor penetration, and rapid drug release to achieve synergistic chemo-protein therapy.
Collapse
Affiliation(s)
- Xiuping Pang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Shuang Liang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Tianqi Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Shuangjiang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| | - Rui Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Teng Hou
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| | - Chaoliang He
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan, Shandong Province 250012, People's Republic of China
| |
Collapse
|
46
|
Improved efficacy of doxorubicin delivery by a novel dual-ligand-modified liposome in hepatocellular carcinoma. Cancer Lett 2020; 489:163-173. [PMID: 32592729 DOI: 10.1016/j.canlet.2020.06.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/08/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023]
Abstract
Liposomes have been widely used as drug carriers in both biomedical research and for clinical applications, allowing the stabilisation of therapeutic compounds and overcoming obstacles to cellular and tissue uptake. However, liposomes still have low targeting efficiency, resulting in insufficient killing of tumour cells and unnecessary damage to normal cells. In this study, glycyrrhetinic acid (GA) and peanut agglutinin (PNA) were used as ligands to prepare dual-ligand-modified doxorubicin-loaded liposomes (DOX-GA/PNA-Lips) to enhance the targeting accuracy and efficacy of drug delivery against malignant liver cancer. PNA and GA modification enhanced the binding ability of liposomes to liver cancer cells, leading to excellent tissue and cell targeting of DOX-GA/PNA-Lips. DOX-GA/PNA-Lips showed an effective anti-tumour effect in vivo and in vitro, with its targeted delivery facilitating attenuation of the toxic side effects of DOX. These results demonstrated that dual-ligand-modified liposomes may provide an effective strategy for the treatment of hepatocellular carcinoma.
Collapse
|
47
|
Fu S, Liang S, Jiang D, Yang R, Zhang Z, Chang L, Zhang X, Liu Y, Zhang N. Gas-blasting nanocapsules to accelerate carboplatin lysosome release and nucleus delivery for prostate cancer treatment. Asian J Pharm Sci 2020; 16:192-202. [PMID: 33995613 PMCID: PMC8105516 DOI: 10.1016/j.ajps.2020.05.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/04/2020] [Accepted: 05/19/2020] [Indexed: 12/24/2022] Open
Abstract
To improve therapeutic effect and reduce severely side effects of carboplatin (CBP), the gas-generating nanocapsules were developed to accelerate CBP lysosome release and nucleus delivery. CBP/SB-NC was prepared by co-loading CBP and NaHCO3 (SB) in nanocapsules using w/o/w emulsification solvent evaporation. They exhibited vesicle-like spherical morphology, uniform particle size and negative zeta potential. Reaching the tumor site with a relatively high concentration is the first step for CBP delivery and the results showed that CBP/SB-NC could effectively increase drug accumulation at tumor site. After that, the drug delivery carriers need to be internalized into tumor cells and the in vitro cellular uptake ability results showed CBP/SB-NC could be internalized into RM-1 cells more efficient than CBP solution. After internalized by RM-1 cells, the gas-blasting release process was tested in acid environment. It was demonstrated that 5 mg/ml NaHCO3 was optimal to achieve pH-responsive gas-blasting release. In vitro release results showed that CBP significantly rapid release in acid environment (pH 5.0) compared to neutral pH (pH 7.4) (P < 0.05). Meanwhile, TEM and the change of the concentration of H+ results exhibited that the explosion of CBP/SB5-NC was more easily happened in lysosome acid environment (pH 5.0). The blasting release can accelerate CBP lysosome release to cytoplasm. Furthermore, the nucleus delivery results showed CBP/SB5-NC can promote pH-triggered rapid nucleus delivery. And the results of Pt-DNA adduct assay showed that the binding efficiency between CBP and DNA of CBP/SB5-NC was higher than CBP solution. At last, in vitro and in vivo anti-tumor efficacy proved that CBP/SB5-NC could enhance anti-tumor activity for prostate cancer therapy. CBP/SB5-NC also showed superior safety in vitro and in vivo by hemolysis assay and histopathological study. All of the results demonstrate that CBP/SB5-NC would be an efficient gas-blasting release formulation to enhance prostate cancer treatment.
Collapse
Affiliation(s)
- Shunli Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Shuang Liang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Dandan Jiang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Rui Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Zipeng Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Lili Chang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xinke Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
48
|
Li ZP, Tian GX, Jiang H, Pan RY, Lian B, Wang M, Gao ZQ, Zhang B, Wu JL. Liver-Targeting and pH-Sensitive Sulfated Hyaluronic Acid Mixed Micelles for Hepatoma Therapy. Int J Nanomedicine 2019; 14:9437-9452. [PMID: 31819442 PMCID: PMC6896933 DOI: 10.2147/ijn.s214528] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
Background The tumor-targeting ability and pH-sensitive properties of intelligent drug delivery systems are crucial for effective drug delivery and anti-tumor therapy. Methods In this study, sHA-DOX/HA-GA mixed micelles were designed with the following properties: sulfated hyaluronic acid (sHA) was synthesized to block cell migration by inhibiting HAase; sHA-DOX conjugates were synthesized via pH-sensitive hydrazone bond to realize DOX-sensitive release. The introduction of HA-GA conjugate could improve active-targeting ability and cellular uptake. Results The results showed that the mixed micelles possessed a nearly spherical shape, nanoscale particle size (217.70±0.89 nm), narrow size distribution (PDI=0.07±0.04), negative zeta potential (-31.87±0.61 mV) and pH-dependent DOX release. In addition, the sHA-DOX/HA-GA micelles exhibited concentration-dependent cytotoxicities against liver carcinoma cells (HepG2) and HeLa cells, and were shown to be effectively taken up by HepG2 cells by confocal microscopy analysis. Furthermore, the in vivo anti-tumor study showed that mixed micelles had a superior anti-tumor effect compared to that of free DOX. Further evidence obtained from the hematoxylin-eosin staining and immunohistochemistry analysis also demonstrated that sHA-DOX/HA-GA exhibited stronger tumor inhibition and lower systemic toxicity than free DOX. Conclusion The sHA-DOX/HA-GA mixed micelles could be a potential drug delivery system for anti-hepatoma therapy.
Collapse
Affiliation(s)
- Zhi-Peng Li
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Gui-Xiang Tian
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Hong Jiang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Rui-Yan Pan
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Bo Lian
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Min Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Zhi-Qin Gao
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Bo Zhang
- School of Pharmacy, Weifang Medical University, Weifang, Shandong, People's Republic of China
| | - Jing-Liang Wu
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, People's Republic of China
| |
Collapse
|
49
|
Wang T, Zhang J, Hou T, Yin X, Zhang N. Selective targeting of tumor cells and tumor associated macrophages separately by twin-like core-shell nanoparticles for enhanced tumor-localized chemoimmunotherapy. NANOSCALE 2019; 11:13934-13946. [PMID: 31305839 DOI: 10.1039/c9nr03374b] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Tumor associated macrophage (TAM)-based immunotherapy has been presented as a promising strategy in cancer therapy. The combination of TAM-based immunotherapy with sorafenib (SF) could be conceivably quite more effective in hepatocellular carcinoma (HCC) treatment. A co-delivery system was superior in improving the co-accumulation of two drugs in tumor tissues for chemoimmunotherapy, while in the case of selective targeting of separated cells such as tumor cells and immune cells, a novel targeted co-delivery strategy was badly required. In this study, twin-like core-shell nanoparticles (TCN) were developed for synchronous biodistribution and separated cell targeting delivery of SF and TAM re-polarization agents IMD-0354 to cancer cells and TAM to enhance tumor-localized chemoimmunotherapy, respectively. First of all, SF loaded cationic lipid-based nanoparticles (SF-CLN) and mannose-modified IMD-0354 loaded cationic lipid-based nanoparticles (M-IMD-CLN) were prepared, respectively. SF on the surface of SF-CLN and mannose on the M-IMD-CLN were regarded as targeting ligands for selective targeting delivery of SF-CLN and M-IMD-CLN to cancer cells and TAM separately. Then, pH-responsive charge reversal polymer O-carboxymethyl-chitosan (CMCS) was coated on the SF-CLN and M-IMD-CLN to obtain twin-like CMCS/SF-CLN and CMCS/M-IMD-CLN, respectively. The results of cellular uptake assay on Hepa1-6 cells and RAW 264.7 cells in vitro, respectively, as well as the results of tumor tissue distribution of SF and IMD-0354 in vivo suggested that CMCS/SF-CLN and CMCS/M-IMD-CLN exhibited similar properties in vitro and synchronous biodistribution in vivo, and were efficient at separated cell targeting delivery. What's more, the results of antitumor efficiency in vivo and phenotype analysis of TAM in tumor tissues proved that CMCS/SF-CLN and CMCS/M-IMD-CLN exhibited superior synergistic antitumor efficacy and M2-type TAM polarization ability compared with SF treatment in Hepa1-6 tumor bearing mice. Consequently, TCN which was the combination of co-administration and nano-drug delivery systems has great potential to be used in tumor-localized chemoimmunotherapy in clinics.
Collapse
Affiliation(s)
- Tianqi Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, People's Republic of China.
| | - Jing Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, People's Republic of China.
| | - Teng Hou
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, People's Republic of China.
| | - Xiaolan Yin
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, People's Republic of China.
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 Wenhua Xi Road, Jinan, Shandong Province 250012, People's Republic of China.
| |
Collapse
|
50
|
Schneible JD, Singhal A, Lilova RL, Hall CK, Grafmüller A, Menegatti S. Tailoring the Chemical Modification of Chitosan Hydrogels to Fine-Tune the Release of a Synergistic Combination of Chemotherapeutics. Biomacromolecules 2019; 20:3126-3141. [PMID: 31310515 DOI: 10.1021/acs.biomac.9b00707] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Combination chemotherapy with a defined ratio and sequence of drug release is a clinically established and effective route to treat advanced solid tumors. In this context, a growing body of literature demonstrates the potential of hydrogels constructed with chemically modified polysaccharides as depots for controlled release of chemotherapeutics. Identifying the appropriate modification in terms of physicochemical properties of the functional group and its degree of substitution (χ) to achieve the desired release profile for multiple drugs is, however, a complex multivariate problem. To address this issue, we have developed a computational toolbox that models the migration of a drug pair through a hydrated network of polysaccharide chains modified with hydrophobic moieties. In this study, we chose doxorubicin (DOX) and Gemcitabine (GEM) as model drugs, as their synergistic effect against breast cancer has been thoroughly investigated, and chitosan as the model polymer. Our model describes how the modification of chitosan chains with acetyl, butanoyl, and heptanoyl moieties at different values χ governs both the structure of the hydrogel network and drug migration through it. Our experimental data confirm the in silico predictions for both single- and dual-drug release and, most notably, the counterintuitive inversion of release vs χ that occurs when switching from a single- to a dual-drug system. Consensus between predicted and experimental data indicates that acetyl modifications (χ = 32-42%) and butanoyl modifications (χ = 19-24%) provide synergistic GEM/DOX release molar ratios (i.e., 5-10). Collectively, these results demonstrate the potential of this model in guiding the design of chemotherapeutic hydrogels to combat cancer.
Collapse
Affiliation(s)
- John D Schneible
- Department of Chemical and Biomolecular Engineering , North Carolina State University , Raleigh , North Carolina 27695 , United States
| | - Ankush Singhal
- Department of Theory and Biosystems , Max Planck Institute for Colloids and Interfaces , Potsdam 14476 , Germany
| | - Radina L Lilova
- Department of Chemical and Biomolecular Engineering , North Carolina State University , Raleigh , North Carolina 27695 , United States
| | - Carol K Hall
- Department of Chemical and Biomolecular Engineering , North Carolina State University , Raleigh , North Carolina 27695 , United States
| | - Andrea Grafmüller
- Department of Theory and Biosystems , Max Planck Institute for Colloids and Interfaces , Potsdam 14476 , Germany
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering , North Carolina State University , Raleigh , North Carolina 27695 , United States
| |
Collapse
|