1
|
Yotsomnuk P, Rajendran AP, Sundaram DNM, Morales LC, Kucharski C, Nasrullah M, Skolpap W, Jiang X, Gibson SB, Brandwein J, Uludağ H. Lipopolymers as the Basis of Non-Viral Delivery of Therapeutic siRNA Nanoparticles in a Leukemia (MOLM-13) Model. Biomolecules 2025; 15:115. [PMID: 39858509 PMCID: PMC11763671 DOI: 10.3390/biom15010115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Small interfering RNA (siRNA) therapy in acute myeloid leukemia (AML) is a promising strategy as the siRNA molecule can specifically target proteins involved in abnormal cell proliferation. The development of a clinically applicable method for delivering siRNA molecules is imperative due to the challenges involved in effectively delivering the siRNA into cells. We investigated the delivery of siRNA to AML MOLM-13 cells with the use of two lipid-substituted polyethyleneimines (PEIs), a commercially available reagent (Prime-Fect) and a recently reported reagent with improved lipid substitution (PEI1.2k-PHPA-Lin9). The siRNAs utilized in this study were targeting the oncogenes FLT3 and KMT2A::MLLT3. Both lipopolymers gave similar-size siRNA complexes (210-220 nm) with positive ζ-potentials (+17 to +25 mV). While the binding efficiency of both lipopolymers to siRNA were similar, PEI1.2k-PHPA-Lin9 complexes were more resistant to heparin-induced dissociation. The quantitative analysis of gene silencing performed by qPCR as well as immunostaining/flow cytometry indicated significant reduction in both FLT3 expression and FLT3 protein after specific siRNA delivery. The desired inhibition of cell growth was attained with both FLT3 and KMT2A::MLLT3 siRNAs, and the combination provided more potent effects in both cell growth and colony formation assays. Induction of apoptosis was confirmed after specific siRNA treatments using the Annexin V assay. Using Luc(+) MOLM-13 cells, the growth of the xenografted cells was shown to be retarded with Prime-Fect-delivered FLT3 siRNA, unlike the siRNA delivered with PEI1.2k-PHPA-Lin9. These results demonstrate the potential of designed lipopolymers in implementing RNAi (via delivery of siRNA) for inhibition of leukemia growth and provide evidence for the feasibility of targeting different oncogenes using siRNA-mediated therapy.
Collapse
MESH Headings
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- Humans
- Nanoparticles/chemistry
- Animals
- Cell Line, Tumor
- Polyethyleneimine/chemistry
- Mice
- Myeloid-Lymphoid Leukemia Protein/genetics
- Myeloid-Lymphoid Leukemia Protein/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
- fms-Like Tyrosine Kinase 3/antagonists & inhibitors
- Histone-Lysine N-Methyltransferase/genetics
- Histone-Lysine N-Methyltransferase/metabolism
- Polymers/chemistry
- Lipids/chemistry
- Apoptosis/drug effects
- Cell Proliferation/drug effects
Collapse
Affiliation(s)
- Panadda Yotsomnuk
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
- Department of Chemical Engineering, Faculty of Engineering, Thammasat University, Pathumthani 12120, Thailand
| | - Amarnath Praphakar Rajendran
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
| | - Daniel Nisakar Meenakshi Sundaram
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
| | - Luis Carlos Morales
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
| | - Cezary Kucharski
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
| | - Mohammad Nasrullah
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2H1, Canada
| | - Wanwisa Skolpap
- Department of Chemical Engineering, Faculty of Engineering, Thammasat University, Pathumthani 12120, Thailand
| | - Xiaoyan Jiang
- Department of Medical Genetics, Terry Fox Laboratory, British Columbia Cancer Research Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Spencer B. Gibson
- Department of Oncology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| | - Joseph Brandwein
- Division of Hematology, Department of Medicine, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2B7, Canada
| | - Hasan Uludağ
- Department of Chemical and Materials Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB T6G 1R1, Canada (M.N.)
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2H1, Canada
| |
Collapse
|
2
|
Sanati M, Figueroa-Espada CG, Han EL, Mitchell MJ, Yavari SA. Bioengineered Nanomaterials for siRNA Therapy of Chemoresistant Cancers. ACS NANO 2024; 18:34425-34463. [PMID: 39666006 DOI: 10.1021/acsnano.4c11259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Chemoresistance remains a long-standing challenge after cancer treatment. Over the last two decades, RNA interference (RNAi) has emerged as a gene therapy modality to sensitize cancer cells to chemotherapy. However, the use of RNAi, specifically small-interfering RNA (siRNA), is hindered by biological barriers that limit its intracellular delivery. Nanoparticles can overcome these barriers by protecting siRNA in physiological environments and facilitating its delivery to cancer cells. In this review, we discuss the development of nanomaterials for siRNA delivery in cancer therapy, current challenges, and future perspectives for their implementation to overcome cancer chemoresistance.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand 97178, Iran
| | - Christian G Figueroa-Espada
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Emily L Han
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, Pennsylvania 19104, United States
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Penn Institute for RNA Innovation, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Saber Amin Yavari
- Department of Orthopedics, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
| |
Collapse
|
3
|
Liu X, Jiang Q, Yin Y, Liang G. Phe-Phe-Based Macroscopic Supramolecular Hydrogel Construction Strategies and Biomedical Applications. CHEM & BIO ENGINEERING 2024; 1:664-677. [PMID: 39974324 PMCID: PMC11792915 DOI: 10.1021/cbe.4c00110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 02/21/2025]
Abstract
Since the phenylalanine (Phe) dipeptide moiety is referred to as an essential structure for building amyloid-β peptide from Alzheimer's disease, its wonderful assembly ability to form nanofibers has been extensively studied. Cross-linked Phe-Phe-based peptide nanofibers can construct networks, thus encapsulating the drugs to form supramolecular hydrogels. Recently, scientists have proposed a variety of Phe-Phe-based macroscopic supramolecular hydrogels and used them in biomedical applications. Therefore, we summarize the construction strategies of Phe-Phe-based macroscopic supramolecular hydrogels and list their represented biomedical applications (e.g., wound healing, eye protection, cancer therapy, etc.) since the birth of Phe-Phe-based supramolecular hydrogels. In addition, we present the perspectives and challenges of Phe-Phe-based macroscopic peptide hydrogels.
Collapse
Affiliation(s)
- Xiaoyang Liu
- State
Key Laboratory of Digital Medical Engineering, School of Biological
Science and Medical Engineering, Southeast
University, 2 Southeast University Road, Nanjing 211189, China
| | - Qiaochu Jiang
- State
Key Laboratory of Digital Medical Engineering, School of Biological
Science and Medical Engineering, Southeast
University, 2 Southeast University Road, Nanjing 211189, China
| | - Yun Yin
- Collaborative
Innovation Center of Tumor Marker Detection Technology, Equipment
and Diagnosis Therapy Integration in Universities of Shandong, Shandong
Province Key Laboratory of Detection Technology for Tumor Makers,
School of Chemistry and Chemical Engineering, Linyi University, Linyi 276005, China
| | - Gaolin Liang
- State
Key Laboratory of Digital Medical Engineering, School of Biological
Science and Medical Engineering, Southeast
University, 2 Southeast University Road, Nanjing 211189, China
- Handan
Norman Technology Company, Limited, Guantao 057750, China
| |
Collapse
|
4
|
Xue R, Pan Y, Xia L, Li J. Non-viral vectors combined delivery of siRNA and anti-cancer drugs to reverse tumor multidrug resistance. Biomed Pharmacother 2024; 178:117119. [PMID: 39142247 DOI: 10.1016/j.biopha.2024.117119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 08/16/2024] Open
Abstract
Multidrug resistance (MDR) of tumors is one of the main reasons for the failure of chemotherapy. Multidrug resistance refers to the cross-resistance of tumor cells to multiple antitumor drugs with different structures and mechanisms of action. Current strategies to reverse multidrug resistance in tumors include MDR inhibitors and RNAi technology. siRNA is a small molecule RNA that is widely used in RNAi technology and has the characteristics of being prepared in large quantities and chemically modified. However, siRNA is susceptible to degradation in vivo. The effect of siRNA therapy alone is not ideal, so siRNA and anticancer drugs are administered in combination to reverse the MDR of tumors. Non-viral vectors are now commonly used to deliver siRNA and anticancer drugs to tumor sites. This article will review the progress of siRNA and chemotherapeutic drug delivery systems and their mechanisms for reversing multidrug resistance.
Collapse
Affiliation(s)
- Renkai Xue
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Yanzhu Pan
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China
| | - Lijie Xia
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China.
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi 830046, China.
| |
Collapse
|
5
|
Cao F, Li Y, Ma F, Wu Z, Li Z, Chen ZS, Cheng X, Qin JJ, Dong J. Synthesis and evaluation of WK-X-34 derivatives as P-glycoprotein (P-gp/ABCB1) inhibitors for reversing multidrug resistance. RSC Med Chem 2024; 15:506-518. [PMID: 38389882 PMCID: PMC10880894 DOI: 10.1039/d3md00612c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 11/30/2023] [Indexed: 02/24/2024] Open
Abstract
The emergence of multidrug resistance (MDR) in malignant tumors is one of the leading threats encountered currently by many chemotherapeutic agents. A proposed strategy to overcome MDR is to disable the efflux function of P-glycoprotein (P-gp/ABCB1), a critical member of the ABC transporter family that significantly increases the efflux of various anticancer drugs from tumor cells. In this study, structural modification of a third-generation P-gp inhibitor WK-X-34 based on bioisosteric and fragment-growing strategies led to the discovery of the adamantane derivative PID-9, which exhibited the best MDR reversal activity (IC50 = 0.1338 μM, RF = 78.6) in this series, exceeding those of the reported P-gp inhibitors verapamil and WK-X-34. In addition, compared with WK-X-34, PID-9 showed decreased toxicity to cells. Furthermore, the mechanism studies revealed that the reversal activity of adamantane derivatives PID-5, PID-7, and PID-9 stemmed from the inhibition of P-gp efflux. These results indicated that compound PID-9 is the most effective P-gp inhibitor among them with low toxicity and high MDR reversal activity, which provided a fundamental structural reference for further discovery of novel, effective, and non-toxic P-gp inhibitors.
Collapse
Affiliation(s)
- Fei Cao
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou 310022 China
- College of Pharmaceutical Science, Zhejiang University of Technology Hangzhou 310032 China
| | - Yulong Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University Hangzhou 310053 China
| | - Furong Ma
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University Hangzhou 310053 China
| | - Zumei Wu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University Hangzhou 310053 China
| | - Zheshen Li
- College of Pharmacy and Health Sciences, St. John's University Queens NY 11439 USA
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University Queens NY 11439 USA
| | - Xiangdong Cheng
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou 310022 China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province Hangzhou 310022 China
| | - Jiang-Jiang Qin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou 310022 China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province Hangzhou 310022 China
| | - Jinyun Dong
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences Hangzhou 310022 China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province Hangzhou 310022 China
| |
Collapse
|
6
|
Liu X, Liu G, Mao Y, Luo J, Cao Y, Tan W, Li W, Yu H, Jia X, Li H. Engineering extracellular vesicles mimetics for targeted chemotherapy of drug-resistant ovary cancer. Nanomedicine (Lond) 2024; 19:25-41. [PMID: 38059464 DOI: 10.2217/nnm-2023-0289] [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] [Indexed: 12/08/2023] Open
Abstract
Aim: To develop nanocarriers for targeting the delivery of chemotherapeutics to overcome multidrug-resistant ovarian cancer. Materials & methods: Doxorubicin-loaded nanovesicles were obtained through serial extrusion, followed by loading of P-glycoprotein siRNA and folic acid. The targeting ability and anticancer efficacy of the nanovesicles were evaluated. Results: The doxorubicin-loaded nanovesicles showed a high production yield. The presence of P-glycoprotein siRNA and folic acid resulted in reversed drug resistance and tumor targeting. This nanoplatform tremendously inhibited the viability of multidrug-resistant ovarian cancer cells, which was able to target tumor tissue and suppress tumor growth without adverse effects. Conclusion: These bioengineered nanovesicles could serve as novel extracellular vesicles mimetics for chemotherapeutics delivery to overcome multidrug resistance.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity & Child Health Care Hospital, Nanjing, 210001, China
| | - Guangquan Liu
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity & Child Health Care Hospital, Nanjing, 210001, China
| | - Yinghua Mao
- Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China
| | - Jie Luo
- Department of Healthcare, General Hospital of Eastern Theater Command, Nanjing, 210002, China
| | - Yongping Cao
- Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China
| | - Weilong Tan
- Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China
| | - Wenhao Li
- Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China
| | - Huanhuan Yu
- Department of Clinical Pharmacy, General Hospital of Eastern Theater Command, Nanjing, 210002, China
| | - Xuemei Jia
- Department of Gynecology, Women's Hospital of Nanjing Medical University, Nanjing Maternity & Child Health Care Hospital, Nanjing, 210001, China
| | - Hong Li
- Centre for Diseases Prevention & Control of Eastern Theater, Nanjing, 210002, China
| |
Collapse
|
7
|
Zheng C, Zhu Z, Weng S, Zhang Q, Fu Y, Cai X, Liu Z, Shi Y. NOD2 silencing promotes cell apoptosis and inhibits drug resistance in chronic lymphocytic leukemia by inhibiting the NF-κB signaling pathway. J Biochem Mol Toxicol 2023; 37:e23510. [PMID: 37700718 DOI: 10.1002/jbt.23510] [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: 07/27/2022] [Revised: 02/14/2023] [Accepted: 08/17/2023] [Indexed: 09/14/2023]
Abstract
Recent years have witnessed increasing studies on the effect of epigenetic silencing of genes in the progression of chronic lymphocytic leukemia (CLL). This study investigates whether the nucleotide binding oligomerization domain containing 2 (NOD2) participates in the cell apoptosis and drug resistance of CLL cells. Cells were treated with adriamycin (ADR), etoposide, aclacinomycin and daunorubicin. After treatment, drug resistance and cell proliferation were examined to detect the inhibitory effect of ADR on cell proliferation; flow cytometry to identify ADR accumulation, the cell cycle distribution and apoptosis after transfection, and rhodamine 123 accumulation and efflux tests to assess P-glycoprotein (P-gp) function. NOD2 silencing or inhibition of the nuclear factor kappa-B (NF-κB) signaling pathway suppressed the multidrug resistance level in CLL, the inhibition rate, and cell proliferation caused by ADR at concentrations of approximately 0.25-1.5 μmol/L. Greater accumulation of ADR was observed in the CLL-AAT cell line than in the CLL-AAT/A02 cell line, but NOD2 silencing or inhibition of the NF-κB signaling pathway further increased the accumulation of ADR drugs in the CLL-AAT cell line and inhibited the drug efflux pump function of P-gp. Additionally, NOD2 silencing or NF-κB signaling pathway inhibition increased the apoptotic rate. The results of this study indicate that NOD2 promotes cell apoptosis and reduces the drug resistance of CLL by inhibiting the NF-κB signaling pathway.
Collapse
MESH Headings
- Humans
- NF-kappa B/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Drug Resistance, Neoplasm
- Signal Transduction
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Apoptosis
- ATP Binding Cassette Transporter, Subfamily B, Member 1
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- Nod2 Signaling Adaptor Protein/genetics
- Nod2 Signaling Adaptor Protein/metabolism
- Nod2 Signaling Adaptor Protein/pharmacology
Collapse
Affiliation(s)
- Cuiping Zheng
- Department of Haematology and Oncology, The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
- The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
| | - Zongsi Zhu
- Department of Haematology and Oncology, The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
| | - Shanshan Weng
- Department of Haematology and Oncology, The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
- The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
| | - Qikai Zhang
- The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
| | - Yixiao Fu
- The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
| | - Xiaoping Cai
- Department of Haematology and Oncology, The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
- The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
| | - Zhen Liu
- Department of Haematology and Oncology, The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
- The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
| | - Yuejian Shi
- Department of Haematology and Oncology, The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
- The Dingli Clinical Institute of Wenzhou Medical University & Wenzhou Central Hospital, Wenzhou, P.R. China
| |
Collapse
|
8
|
Kesharwani P, Chadar R, Sheikh A, Rizg WY, Safhi AY. CD44-Targeted Nanocarrier for Cancer Therapy. Front Pharmacol 2022; 12:800481. [PMID: 35431911 PMCID: PMC9008230 DOI: 10.3389/fphar.2021.800481] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/20/2021] [Indexed: 01/08/2023] Open
Abstract
Cluster of differentiation 44 (CD44) is a cell surface glycoprotein overexpressed in varieties of solid tumors including pancreatic, breast, ovary, brain, and lung cancers. It is a multi-structural glycoprotein of the cell surface which is majorly involved in cell proliferation, cell-to-cell interaction, cellular migration, inflammation, and generation of immune responses. Numerous studies focus on the development of nanocarriers for active targeting of the CD44 receptor to improve efficacy of targeting chemotherapy and achieve precise chemotherapy by defining the release, uptake, and accumulation of therapeutic agents. The CD44 receptor has a selective binding affinity towards hyaluronic and chondroitin sulfate (CS). Taking this into consideration, this review focused on the role of CD44 in cancer and its therapy using several nanocarriers such as polymeric/non-polymeric nanoparticles, dendrimer, micelles, carbon nanotubes, nanogels, nanoemulsions etc., for targeted delivery of several chemotherapeutic molecules and nucleic acid. This review also illuminates the role of hyaluronic acid (HA) in cancer therapy, interaction of HA with CD44, and various approaches to target CD44-overexpressed neoplastic cells.
Collapse
Affiliation(s)
- Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
- *Correspondence: Prashant Kesharwani,
| | - Rahul Chadar
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Afsana Sheikh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Waleed Y. Rizg
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Awaji Y Safhi
- Department of Pharmaceutics, Faculty of Pharmacy, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
9
|
Therapeutic delivery of siRNA with polymeric carriers to down-regulate STAT5A expression in high-risk B-cell acute lymphoblastic leukemia (B-ALL). PLoS One 2021; 16:e0251719. [PMID: 34157051 PMCID: PMC8219370 DOI: 10.1371/journal.pone.0251719] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 05/02/2021] [Indexed: 11/19/2022] Open
Abstract
Overexpression and persistent activation of STAT5 play an important role in the development and progression of acute lymphoblastic leukemia (ALL), the most common pediatric cancer. Small interfering RNA (siRNA)-mediated downregulation of STAT5 represents a promising therapeutic approach for ALL to overcome the limitations of current treatment modalities such as high relapse rates and poor prognosis. However, to effectively transport siRNA molecules to target cells, development of potent carriers is of utmost importance to surpass hurdles of delivery. In this study, we investigated the use of lipopolymers as non-viral delivery systems derived from low molecular weight polyethylenimines (PEI) substituted with lauric acid (Lau), linoleic acid (LA) and stearic acid (StA) to deliver siRNA molecules to ALL cell lines and primary samples. Among the lipid-substituted polymers explored, Lau- and LA-substituted PEI displayed excellent siRNA delivery to SUP-B15 and RS4;11 cells. STAT5A gene expression was downregulated (36-92%) in SUP-B15 and (32%) in RS4;11 cells using the polymeric delivery systems, which consequently reduced cell growth and inhibited the formation of colonies in ALL cells. With regard to ALL primary cells, siRNA-mediated STAT5A gene silencing was observed in four of eight patient cells using our leading polymeric delivery system, 1.2PEI-Lau8, accompanied by the significant reduction in colony formation in three of eight patients. In both BCR-ABL positive and negative groups, three of five patients demonstrated marked cell growth inhibition in both MTT and trypan blue exclusion assays using 1.2PEI-Lau8/siRNA complexes in comparison with their control siRNA groups. Three patient samples did not show any positive results with our delivery systems. Differential therapeutic responses to siRNA therapy observed in different patients could result from variable genetic profiles and patient-to-patient variability in delivery. This study supports the potential of siRNA therapy and the designed lipopolymers as a delivery system in ALL therapy.
Collapse
|
10
|
Wang J, Wu C, Qin X, Huang Y, Zhang J, Chen T, Wang Y, Ding Y, Yao Y. NIR-II light triggered nitric oxide release nanoplatform combined chemo-photothermal therapy for overcoming multidrug resistant cancer. J Mater Chem B 2021; 9:1698-1706. [PMID: 33495772 DOI: 10.1039/d0tb02626c] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The overexpression of P-glycoprotein (P-gp) in multidrug resistance (MDR) cancer cells increases the efflux of anticancer drugs thereby causing the failure of clinical chemotherapy. To address this obstacle, in this study, we rationally designed a near-infrared (NIR) light-responsive nitric oxide (NO) delivery nanoplatform for targeting the MDR tumors based on core-shell structured nanocomposites. The mesoporous silica shell provided abundant sites for modification of the NO donor, N-diazeniumdiolate, and tumor-targeting molecule, folic acid (FA), and enabled high encapsulation capacity for doxorubicin (DOX) loading. Under NIR light irradiation, the generation of NO gas can efficiently augment chemotherapeutic effects via the inhibition of P-gp expression. Simultaneously, the photothermal conversion agents of the Cu2-xSe core produce a large amount of heat for photothermal therapy (PTT). Finally, this combinational gas/chemo/PTT not only displays a superior and synergistic effect for overcoming MDR cancer, but also provides an efficient strategy to construct a multifunctional nano-drug delivery system with diversified therapeutic modalities.
Collapse
Affiliation(s)
- Jin Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, China.
| | - Canchen Wu
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, China.
| | - Xiru Qin
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, China.
| | - Youyou Huang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, China.
| | - Jianan Zhang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, China.
| | - Tingting Chen
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, China.
| | - Yang Wang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, China.
| | - Yue Ding
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, China.
| | - Yong Yao
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, Jiangsu 226019, China.
| |
Collapse
|
11
|
Tang Q, Yin D, Wang Y, Du W, Qin Y, Ding A, Li H. Cancer Stem Cells and Combination Therapies to Eradicate Them. Curr Pharm Des 2020; 26:1994-2008. [PMID: 32250222 DOI: 10.2174/1381612826666200406083756] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 02/13/2020] [Indexed: 12/23/2022]
Abstract
Cancer stem cells (CSCs) show self-renewal ability and multipotential differentiation, like normal stem or progenitor cells, and which proliferate uncontrollably and can escape the effects of drugs and phagocytosis by immune cells. Traditional monotherapies, such as surgical resection, radiotherapy and chemotherapy, cannot eradicate CSCs, however, combination therapy may be more effective at eliminating CSCs. The present review summarizes the characteristics of CSCs and several promising combination therapies to eradicate them.
Collapse
Affiliation(s)
- Qi Tang
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, China.,Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Dan Yin
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Yao Wang
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, China
| | - Wenxuan Du
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, China
| | - Yuhan Qin
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, China
| | - Anni Ding
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, China
| | - Hanmei Li
- College of Pharmacy and Biological Engineering, Chengdu University, Chengdu, China
| |
Collapse
|
12
|
Wu W, Chen M, Luo T, Fan Y, Zhang J, Zhang Y, Zhang Q, Sapin-Minet A, Gaucher C, Xia X. ROS and GSH-responsive S-nitrosoglutathione functionalized polymeric nanoparticles to overcome multidrug resistance in cancer. Acta Biomater 2020; 103:259-271. [PMID: 31846803 DOI: 10.1016/j.actbio.2019.12.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 12/27/2022]
Abstract
Multidrug resistance of cancer cells is one of the major obstacle for chemotherapeutic efficiency. Nitric oxide (NO) has raised the potential to overcome multidrug resistance (MDR) with low side effects. Herein, we report a reactive oxygen species (ROS) and glutathione (GSH) responsive nanoparticle for the delivery of NO prodrug such as S-nitrosoglutathione (GSNO), which was chemically conjugated to an amphiphilic block copolymer. The GSNO functionalized nanoparticles show high NO loading capacity, good stability and sustained NO release with specific GSH activated NO-releasing kinetics. Such GSNO functionalized nanoparticles delivered doxorubicin (DOX) in a ROS triggered manner and increased the intracellular accumulation of DOX. However, in normal healthy cells, showing physiological concentrations of ROS, these nanoparticles presented good biocompatibility. The present work indicated that these multifunctional nanoparticles can serve as effective co-delivery platforms of NO and DOX to selectively kill chemo-resistant cancer cells through increasing chemo-sensitivity. STATEMENT OF SIGNIFICANCE: In this work, we constructed nitric oxide donor (S-nitrosoglutathione, GSNO) functionalized amphiphilic copolymer (PEG-PPS-GSNO) to deliver doxorubicin (DOX). The developed PEG-PPS-GSNO@DOX nanoparticles presented high NO capacity, ROS triggered DOX release and GSH triggered NO release. Thus NO reversed the chemo-resistance in HepG2/ADR cells increasing intrcellular accumulation of DOX. Furthermore, these PEG-PPS-GSNO@DOX nanoparticles exhibited biocompatibility to healthy cells and toxicity to cancer cells, due to elevated ROS.
Collapse
|
13
|
Kazan HH, Urfali-Mamatoglu C, Yalcin GD, Bulut O, Sezer A, Banerjee S, Gunduz U. 15-LOX-1 has diverse roles in the resensitization of resistant cancer cell lines to doxorubicin. J Cell Physiol 2019; 235:4965-4978. [PMID: 31663148 DOI: 10.1002/jcp.29375] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 10/07/2019] [Indexed: 11/10/2022]
Abstract
Lipoxygenases (LOXs) are a family of enzymes that can oxygenate polyunsaturated fatty acids. As a member of the family, 15-lipoxygenase-1 (15-LOX-1) specifically metabolizes arachidonic acid and linoleic acid. 15-LOX-1 can affect physiological and pathophysiological events via regulation of the protein-lipid interactome, alterations in intracellular redox state and production of lipid metabolites that are involved in the induction and resolution of inflammation. Although several studies have shown that 15-LOX-1 has an antitumorigenic role in many different cancer models, including breast cancer, the role of the protein in cancer drug resistance has not been established yet. In this study, we, for the first time, aimed to show the potential role of 15-LOX-1 in acquired doxorubicin (DOX) resistance in MCF7 and HeLa cancer cell lines. Our results show that ALOX15 was transcriptionally downregulated in DOX-resistant cells compared with their drug-sensitive counterparts. Moreover, overexpression of ALOX15 in the drug-resistant cells resulted in resensitization of those cells to DOX in a cell-dependent manner. 15-LOX-1 expression could induce apoptosis by activating PPARγ and enhance the accumulation of DOX in drug-resistant MCF7 cells by altering cellular motility properties, and membrane dynamics. However, HeLa DOX cells did not show any of these effects but were susceptible to cell death when treated with 13(S)-HODE. These results underline the role and importance of 15-LOX-1 in cancer drug resistance, and points to novel mechanisms as a therapeutic approach to overcome cancer drug resistance.
Collapse
Affiliation(s)
- Hasan Huseyin Kazan
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | | | - Gizem Damla Yalcin
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Onur Bulut
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey.,Department of Molecular Biology and Genetics, Konya Food and Agriculture University, Konya, Turkey.,Research and Development Center for Diagnostic Kits (KIT-ARGEM), Konya Food and Agriculture University, Konya, Turkey
| | - Abdullah Sezer
- Department of Medical Genetics, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Sreeparna Banerjee
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| | - Ufuk Gunduz
- Department of Biological Sciences, Middle East Technical University, Ankara, Turkey
| |
Collapse
|
14
|
Guo D, Ji X, Peng F, Zhong Y, Chu B, Su Y, He Y. Photostable and Biocompatible Fluorescent Silicon Nanoparticles for Imaging-Guided Co-Delivery of siRNA and Doxorubicin to Drug-Resistant Cancer Cells. NANO-MICRO LETTERS 2019; 11:27. [PMID: 34137971 PMCID: PMC7770907 DOI: 10.1007/s40820-019-0257-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 03/06/2019] [Indexed: 05/06/2023]
Abstract
The development of effective and safe vehicles to deliver small interfering RNA (siRNA) and chemotherapeutics remains a major challenge in RNA interference-based combination therapy with chemotherapeutics, which has emerged as a powerful platform to treat drug-resistant cancer cells. Herein, we describe the development of novel all-in-one fluorescent silicon nanoparticles (SiNPs)-based nanomedicine platform for imaging-guided co-delivery of siRNA and doxorubicin (DOX). This approach enhanced therapeutic efficacy in multidrug-resistant breast cancer cells (i.e., MCF-7/ADR cells). Typically, the SiNP-based nanocarriers enhanced the stability of siRNA in a biological environment (i.e., medium or RNase A) and imparted the responsive release behavior of siRNA, resulting in approximately 80% down-regulation of P-glycoprotein expression. Co-delivery of P-glycoprotein siRNA and DOX led to > 35-fold decrease in the half maximal inhibitory concentration of DOX in comparison with free DOX, indicating the pronounced therapeutic efficiency of the resultant nanocomposites for drug-resistant breast cancer cells. The intracellular time-dependent release behaviors of siRNA and DOX were revealed through tracking the strong and stable fluorescence of SiNPs. These data provide valuable information for designing effective RNA interference-based co-delivery carriers.
Collapse
Affiliation(s)
- Daoxia Guo
- Laboratory of Nanoscale Biochemical Analysis, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Xiaoyuan Ji
- Laboratory of Nanoscale Biochemical Analysis, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Fei Peng
- Laboratory of Nanoscale Biochemical Analysis, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Yiling Zhong
- Laboratory of Nanoscale Biochemical Analysis, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Binbin Chu
- Laboratory of Nanoscale Biochemical Analysis, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China
| | - Yuanyuan Su
- Laboratory of Nanoscale Biochemical Analysis, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China.
| | - Yao He
- Laboratory of Nanoscale Biochemical Analysis, Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Soochow University, Suzhou, 215123, Jiangsu, People's Republic of China.
| |
Collapse
|
15
|
Li X, Mu P, Qiao H, Wen J, Deng Y. JNK-AKT-NF-κB controls P-glycoprotein expression to attenuate the cytotoxicity of deoxynivalenol in mammalian cells. Biochem Pharmacol 2018; 156:120-134. [DOI: 10.1016/j.bcp.2018.08.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/15/2018] [Indexed: 01/05/2023]
|
16
|
Huang C, Huang S, Li H, Li X, Li B, Zhong L, Wang J, Zou M, He X, Zheng H, Si X, Liao W, Liao Y, Yang L, Bin J. The effects of ultrasound exposure on P-glycoprotein-mediated multidrug resistance in vitro and in vivo. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:232. [PMID: 30231924 PMCID: PMC6149229 DOI: 10.1186/s13046-018-0900-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/04/2018] [Indexed: 12/22/2022]
Abstract
Background Multidrug resistance (MDR) is often responsible for the failure of chemotherapy treatment, and current strategies for cancer MDR are not adequately satisfying as to their efficacy and safety. In this study, we sought to determine the anti-MDR effects of ultrasound (US) irradiation and its underlying mechanisms against drug-resistance. Methods MDR variant MCF-7/ADR cell lines and endothelial cell lines were used to determine the appropriate ultrasound intensity for in vitro experiments. MCF-7/ADR cell and HEPG2/ADM cells were used to assess the anti-MDR effect of US irradiation. Intracellular adriamycin (ADM) accumulation, Cell viability, cell proliferation and cell apoptosis were evaluated after ADM + US treatment or ADM treatment alone. MCF-7/ADR xenograft mice were used to investigate the appropriate ultrasound intensity for in vivo experiments and its effect on the long-term prognosis. Underlining mechanisms by which ultrasound exposure reversing MDR phenotype were investigated both in vitro and in vivo. Results Combination of ADM and 0.74 W/cm2 US irradiation enhanced ADM intracellular concentration and nuclear accumulation in MCF-7/ADR and HEPG2/ADM cells, compared to those treated with ADM alone. Enhanced cellular ADM uptake and nuclei localization was associated with increased cytotoxicity of ADM to ADM-resistant cells, lower ADM-resistant cell viability and proliferative cell ratio, and higher apoptotic cell ratio. More importantly, US exposure increased the effectiveness of ADM to inhibit tumor growth in MCF-7/ADR xenograft mice. Mechanistically, US exposure promoted ADM accumulation in MDR cells mainly through down-regulation of P-glycoprotein (P-gp), which is dependent on US-induced intracellular reactive oxygen species (ROS) production. US-induced oxidative stress promoted miR-200c-3p and miR-34a-3p expression by forming miR-200c/34a/ZEB1 double-negative feedback loop. Finally, US-induced miR-200c/34a overexpression decreased P-gp expression and reversed MDR phenotype. Conclusion US irradiation could reverse MDR phenotype by activating ROS-ZEB1-miR200c/34a-P-gp signal pathway. Our findings offer a new and promising strategy for sensitizing cells to combat MDR and to improve the therapeutic index of chemotherapy. Electronic supplementary material The online version of this article (10.1186/s13046-018-0900-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chixiong Huang
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | | | - Hairui Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Xinzhong Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Bing Li
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Lintao Zhong
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Junfeng Wang
- Department of Gastroenterology, Guangdong Provincial Key Laboratory of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Meishen Zou
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Xiang He
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Hao Zheng
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Xiaoyun Si
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yulin Liao
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China
| | - Li Yang
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, People's Republic of China.
| | - Jianping Bin
- State Key Laboratory of Organ Failure Research, Department of Cardiology, Nanfang Hospital, Southern Medical University, 1838 Guangzhou Avenue North, Guangzhou, 510515, China.
| |
Collapse
|
17
|
Zhao CY, Cheng R, Yang Z, Tian ZM. Nanotechnology for Cancer Therapy Based on Chemotherapy. Molecules 2018; 23:E826. [PMID: 29617302 PMCID: PMC6017446 DOI: 10.3390/molecules23040826] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 03/27/2018] [Accepted: 03/29/2018] [Indexed: 02/07/2023] Open
Abstract
Chemotherapy has been widely applied in clinics. However, the therapeutic potential of chemotherapy against cancer is seriously dissatisfactory due to the nonspecific drug distribution, multidrug resistance (MDR) and the heterogeneity of cancer. Therefore, combinational therapy based on chemotherapy mediated by nanotechnology, has been the trend in clinical research at present, which can result in a remarkably increased therapeutic efficiency with few side effects to normal tissues. Moreover, to achieve the accurate pre-diagnosis and real-time monitoring for tumor, the research of nano-theranostics, which integrates diagnosis with treatment process, is a promising field in cancer treatment. In this review, the recent studies on combinational therapy based on chemotherapy will be systematically discussed. Furthermore, as a current trend in cancer treatment, advance in theranostic nanoparticles based on chemotherapy will be exemplified briefly. Finally, the present challenges and improvement tips will be presented in combination therapy and nano-theranostics.
Collapse
Affiliation(s)
| | | | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Zhong-Min Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
18
|
Contino M, Guglielmo S, Perrone MG, Giampietro R, Rolando B, Carrieri A, Zaccaria D, Chegaev K, Borio V, Riganti C, Zabielska-Koczywąs K, Colabufo NA, Fruttero R. New tetrahydroisoquinoline-based P-glycoprotein modulators: decoration of the biphenyl core gives selective ligands. MEDCHEMCOMM 2018; 9:862-869. [PMID: 30108975 DOI: 10.1039/c8md00075a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 03/30/2018] [Indexed: 01/25/2023]
Abstract
P-glycoprotein (P-gp, MDR1) is a membrane transporter expressed in several regions of our body. It plays a crucial defense role as it mediates the efflux of hundreds of potentially toxic substances. However, P-gp is one of the main causes of failure in cancer chemotherapy, as a number of chemotherapeutic agents are P-gp substrates. Another interesting implication concerns the correlation between P-gp expression impairment and the onset of several central nervous system pathologies such as Alzheimer's and Parkinson's diseases. In view of these considerations, in the present study, a new series of P-gp modulators have been designed, synthesized and evaluated for their activity towards P-gp and two other sister proteins (BCRP and MRP1). The compounds, structurally correlated to the potent but non-selective P-gp inhibitor MC70 [4'-(6,7-dimethoxy-3,4-dihydro-1H-isoquinolin-2-ylmethyl)biphenyl-4-ol], proved fairly selective towards P-gp, with a potency in the micromolar range. Compounds 5a, 5d and 12d proved capable of restoring doxorubicin toxicity in resistant cancer cells.
Collapse
Affiliation(s)
- Marialessandra Contino
- Dipartimento di Farmacia-Scienze del Farmaco , Universita' degli Studi di Bari "Aldo Moro" , Via Orabona 4 , 70125 Bari , Italy .
| | - Stefano Guglielmo
- Dipartimento di Scienza e Tecnologia del Farmaco , Universita' degli Studi di Torino , Via P. Giuria 9 , 10125 Torino , Italy .
| | - Maria Grazia Perrone
- Dipartimento di Farmacia-Scienze del Farmaco , Universita' degli Studi di Bari "Aldo Moro" , Via Orabona 4 , 70125 Bari , Italy .
| | - Roberta Giampietro
- Dipartimento di Farmacia-Scienze del Farmaco , Universita' degli Studi di Bari "Aldo Moro" , Via Orabona 4 , 70125 Bari , Italy .
| | - Barbara Rolando
- Dipartimento di Scienza e Tecnologia del Farmaco , Universita' degli Studi di Torino , Via P. Giuria 9 , 10125 Torino , Italy .
| | - Antonio Carrieri
- Dipartimento di Farmacia-Scienze del Farmaco , Universita' degli Studi di Bari "Aldo Moro" , Via Orabona 4 , 70125 Bari , Italy .
| | - Daniele Zaccaria
- Dipartimento di Farmacia-Scienze del Farmaco , Universita' degli Studi di Bari "Aldo Moro" , Via Orabona 4 , 70125 Bari , Italy .
| | - Konstantin Chegaev
- Dipartimento di Scienza e Tecnologia del Farmaco , Universita' degli Studi di Torino , Via P. Giuria 9 , 10125 Torino , Italy .
| | - Vanessa Borio
- Dipartimento di Scienza e Tecnologia del Farmaco , Universita' degli Studi di Torino , Via P. Giuria 9 , 10125 Torino , Italy .
| | - Chiara Riganti
- Dipartimento di Oncologia , Università degli Studi di Torino , via Santena 5/bis , 10126 Torino , Italy
| | - Katarzyna Zabielska-Koczywąs
- Department of Small Animal Diseases with Clinic , Faculty of Veterinary Medicine , Warsaw University of Life Sciences , Nowoursynowska 159c , 02-776 , Warsaw , Poland
| | - Nicola A Colabufo
- Dipartimento di Farmacia-Scienze del Farmaco , Universita' degli Studi di Bari "Aldo Moro" , Via Orabona 4 , 70125 Bari , Italy . .,Biofordrug s.r.l. , Spin-off dell'Università degli Studi di Bari "A. Moro" , Via Orabona 4 , 70125 Bari , Italy
| | - Roberta Fruttero
- Dipartimento di Scienza e Tecnologia del Farmaco , Universita' degli Studi di Torino , Via P. Giuria 9 , 10125 Torino , Italy .
| |
Collapse
|
19
|
Waghray D, Zhang Q. Inhibit or Evade Multidrug Resistance P-Glycoprotein in Cancer Treatment. J Med Chem 2017; 61:5108-5121. [PMID: 29251920 DOI: 10.1021/acs.jmedchem.7b01457] [Citation(s) in RCA: 267] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multidrug resistance (MDR) is a major cause of failure in cancer chemotherapy. P-glycoprotein (P-gp), a promiscuous drug efflux pump, has been extensively studied for its association with MDR due to overexpression in cancer cells. Several P-gp inhibitors or modulators have been investigated in clinical trials in hope of circumventing MDR, with only limited success. Alternative strategies are actively pursued, such as the modification of existing drugs, development of new drugs, or combination of novel drug delivery agents to evade P-gp-dependent efflux. Despite the importance and numerous studies, these efforts have mostly been undertaken without a priori knowledge of how drugs interact with P-gp at the molecular level. This review highlights and discusses progress toward and challenges impeding drug development for inhibiting or evading P-gp in the context of our improved understanding of the structural basis and mechanism of P-gp-mediated MDR.
Collapse
Affiliation(s)
- Deepali Waghray
- Department of Integrative Structural and Computational Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| | - Qinghai Zhang
- Department of Integrative Structural and Computational Biology , The Scripps Research Institute , La Jolla , California 92037 , United States
| |
Collapse
|
20
|
A polycation coated liposome as efficient siRNA carrier to overcome multidrug resistance. Colloids Surf B Biointerfaces 2017; 159:427-436. [PMID: 28826111 DOI: 10.1016/j.colsurfb.2017.08.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 07/22/2017] [Accepted: 08/06/2017] [Indexed: 01/12/2023]
Abstract
Multidrug resistance (MDR) is one of the important factors that impede effective chemotherapy against cancer. Codelivery of MDR1 siRNA (silencing ABCB1 gene) and anticancer drug can greatly inhibit tumor proliferation. Here in this work, we synthesized poly(diallyldimethylammonium chloride) (PDADMAC) coated liposome formula as siMDR1 carrier (AL-PDAD-RNA) and applied it to reverse doxorubicin resistance of OVCAR8/ADR cells. The AL-PDAD-RNA can load siRNA effectively and release siRNA under physiological conditions, leading to improved tumor inhibition than free DOX without siRNA treatment. Meanwhile, the gene silencing effect of AL-PDAD-RNA was shown to be comparable to that of commercial transfection agent lipofectamine, but with less toxicity. The main novelty of this work is to offer a new type of siRNA carrier (PDADMAC coated liposome, AL-PDAD), which is simple-structured, highly-effective and non-toxic. Therefore, we anticipate that PDADMAC-coated liposomes would be very promising in the application of other siRNA delivery or even plasmid delivery.
Collapse
|
21
|
Liu J, Li J, Liu N, Guo N, Gao C, Hao Y, Chen L, Zhang X. In vitro studies of phospholipid-modified PAMAM-siMDR1 complexes for the reversal of multidrug resistance in human breast cancer cells. Int J Pharm 2017; 530:291-299. [PMID: 28619457 DOI: 10.1016/j.ijpharm.2017.06.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/08/2017] [Accepted: 06/10/2017] [Indexed: 02/05/2023]
Abstract
The application of RNAi therapeutics is promising in combating several major human diseases including malignant tumors. However, this approach is limited due to its delivery barriers. In this study, we designed a new carrier system loaded with a functional siRNA targeting MDR1 gene to reverse multi-drug resistance (MDR) in human breast cancer MCF-7/ADR cells. Phospholipid-modified PAMAM-siMDR1 complexes were designed on the external decoration of polyamidoamine (PAMAM) with phospholipid (PL) and the electrostatical interaction between PAMAM and siMDR1 to form hybrid nanocomplexes (PL-dendriplexes). Compared with siMDR1 and dendriplexes (PAMAM-siMDR1), this delivery system represented higher gene silencing efficiency, enhanced cellular uptake of siMDR1, decreased p-gp expression, raised cellular accumulation of doxorubicin (DOX) and inhibited the tumor cell migration. Moreover, the siMDR1 loaded PL-dendriplexes worked synergistically with paclitaxel (PTX) for treating MDR, leading to increased cell apoptosis and cell phase regulation. Overall, this study shows that the PL-dendriplexes hold great promise in reversing the drug-resistance in MCF-7/ADR cells.
Collapse
Affiliation(s)
- Jing Liu
- Collaborative Innovation Center for Biotherapy, Tsinghua University, Beijing 100084, China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Jun Li
- School of Life Science, Jiangsu Normal University, Xuzhou, Jiangsu 221116, China.
| | - Nan Liu
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Nana Guo
- Department of Gynaecology and Obstetrics, PLA Navy General Hospital, Beijing 100037, China
| | - Chen Gao
- College of Life Science, Hebei Normal University, Shijiazhuang, Hebei 050024, China
| | - Yanli Hao
- School of Medicine, Tsinghua University, Beijing 100084, China
| | - Lei Chen
- Department of Gynaecology and Obstetrics, PLA Navy General Hospital, Beijing 100037, China
| | - Xiaoning Zhang
- Collaborative Innovation Center for Biotherapy, Tsinghua University, Beijing 100084, China; School of Medicine, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
22
|
Nucleic acid combinations: A new frontier for cancer treatment. J Control Release 2017; 256:153-169. [DOI: 10.1016/j.jconrel.2017.04.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 12/19/2022]
|
23
|
Rui M, Qu Y, Gao T, Ge Y, Feng C, Xu X. Simultaneous delivery of anti-miR21 with doxorubicin prodrug by mimetic lipoprotein nanoparticles for synergistic effect against drug resistance in cancer cells. Int J Nanomedicine 2016; 12:217-237. [PMID: 28115844 PMCID: PMC5221799 DOI: 10.2147/ijn.s122171] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The development of drug resistance in cancer cells is one of the major obstacles to achieving effective chemotherapy. We hypothesized that the combination of a doxorubicin (Dox) prodrug and microRNA (miR)21 inhibitor might show synergistic antitumor effects on drug-resistant breast cancer cells. In this study, we aimed to develop new high-density lipoprotein-mimicking nanoparticles (HMNs) for coencapsulation and codelivery of this potential combination. Dox was coupled with a nuclear localization signal (NLS) peptide to construct a prodrug (NLS-Dox), thereby electrostatically condensing miR21 inhibitor (anti-miR21) to form cationic complexes. The HMNs were formulated by shielding these complexes with anionic lipids and Apo AI proteins. We have characterized that the coloaded HMNs had uniformly dispersed distribution, favorable negatively charged surface, and high coencapsulation efficiency. The HMN formulation effectively codelivered NLS-Dox and anti-miR21 into Dox-resistant breast cancer MCF7/ADR cells and wild-type MCF7 cells via a high-density-lipoprotein receptor-mediated pathway, which facilitated the escape of Pgp drug efflux. The coloaded HMNs consisting of NLS-Dox/anti-miR21 demonstrated greater cytotoxicity with enhanced intracellular accumulation in resistant MCF7/ADR cells compared with free Dox solution. The reversal of drug resistance by coloaded HMNs might be attributed to the suppression of miR21 expression and the related antiapoptosis network. Furthermore, the codelivery of anti-miR21 and NLS-Dox by HMNs showed synergistic antiproliferative effects in MCF7/ADR-bearing nude mice, and was more effective in tumor inhibition than other drug formulations. These data suggested that codelivery of anti-miR21 and chemotherapeutic agents by HMNs might be a promising strategy for antitumor therapy, and could restore the drug sensitivity of cancer cells, alter intracellular drug distribution, and ultimately enhance chemotherapeutic effects.
Collapse
Affiliation(s)
- Mengjie Rui
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, People's Republic of China
| | - Yang Qu
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, People's Republic of China
| | - Tong Gao
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, People's Republic of China
| | - Yanru Ge
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, People's Republic of China
| | - Chunlai Feng
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, People's Republic of China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, People's Republic of China
| |
Collapse
|
24
|
Yao C, Wang P, Li X, Hu X, Hou J, Wang L, Zhang F. Near-Infrared-Triggered Azobenzene-Liposome/Upconversion Nanoparticle Hybrid Vesicles for Remotely Controlled Drug Delivery to Overcome Cancer Multidrug Resistance. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:9341-9348. [PMID: 27578301 DOI: 10.1002/adma.201503799] [Citation(s) in RCA: 224] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 07/22/2016] [Indexed: 05/21/2023]
Abstract
Overcoming multidrug resistance is achieved by developing a novel drugdelivery-system paradigm based on azobenzene liposome and phosphatidylcholine-modified upconversion nanoparticle (UCNP) hybrid vesicles for controlled drug release using a nearinfrared (NIR) laser. Upon 980 nm light irradiation, the reversible photoisomerization of the azobenzene derivatives by simultaneous UV and visible light emitted from the UCNPs makes it possible to realize NIR-triggered release of the chemotherapeutic drug doxorubicin.
Collapse
Affiliation(s)
- Chi Yao
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Peiyuan Wang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Xiaomin Li
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Xiaoyu Hu
- School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, P. R. China
| | - Junli Hou
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai, 200433, P. R. China
| | - Leyong Wang
- School of Chemistry and Chemical Engineering, Nanjing University, Nanjing, 210093, P. R. China
| | - Fan Zhang
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Collaborative Innovation Center of Chemistry for Energy Materials, Fudan University, Shanghai, 200433, P. R. China
| |
Collapse
|
25
|
Jones SK, Merkel OM. Tackling breast cancer chemoresistance with nano-formulated siRNA. Gene Ther 2016; 23:821-828. [PMID: 27648580 DOI: 10.1038/gt.2016.67] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 07/25/2016] [Accepted: 09/13/2016] [Indexed: 12/11/2022]
Abstract
Breast cancer is the leading cancer diagnosed in women and the second leading cause of cancer-related deaths in women. Current limitations to standard chemotherapy in the clinic are extensively researched, including problems arising from repeated treatments with the same drugs. The phenomenon that cancer cells become resistant toward certain chemo drugs is called chemotherapy resistance. In this review, we are focusing on nanoformulation of siRNA for the fight against breast cancer chemoresistance.
Collapse
Affiliation(s)
- S K Jones
- Department of Oncology, Wayne State University, Detroit, MI, USA
| | - O M Merkel
- Department of Oncology, Wayne State University, Detroit, MI, USA.,Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA.,Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-Universität München, München, Germany
| |
Collapse
|
26
|
Lee SJ, Kim MJ, Kwon IC, Roberts TM. Delivery strategies and potential targets for siRNA in major cancer types. Adv Drug Deliv Rev 2016; 104:2-15. [PMID: 27259398 DOI: 10.1016/j.addr.2016.05.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 02/24/2016] [Accepted: 05/15/2016] [Indexed: 02/08/2023]
Abstract
Small interfering RNA (siRNA) has gained attention as a potential therapeutic reagent due to its ability to inhibit specific genes in many genetic diseases. For many years, studies of siRNA have progressively advanced toward novel treatment strategies against cancer. Cancer is caused by various mutations in hundreds of genes including both proto-oncogenes and tumor suppressor genes. In order to develop siRNAs as therapeutic agents for cancer treatment, delivery strategies for siRNA must be carefully designed and potential gene targets carefully selected for optimal anti-cancer effects. In this review, various modifications and delivery strategies for siRNA delivery are discussed. In addition, we present current thinking on target gene selection in major tumor types.
Collapse
|
27
|
Park J, Park J, Pei Y, Xu J, Yeo Y. Pharmacokinetics and biodistribution of recently-developed siRNA nanomedicines. Adv Drug Deliv Rev 2016; 104:93-109. [PMID: 26686832 DOI: 10.1016/j.addr.2015.12.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 10/26/2015] [Accepted: 12/03/2015] [Indexed: 02/07/2023]
Abstract
Small interfering RNA (siRNA) is a promising drug candidate, expected to have broad therapeutic potentials toward various diseases including viral infections and cancer. With recent advances in bioconjugate chemistry and carrier technology, several siRNA-based drugs have advanced to clinical trials. However, most cases address local applications or diseases in the filtering organs, reflecting remaining challenges in systemic delivery of siRNA. The difficulty in siRNA delivery is in large part due to poor circulation stability and unfavorable pharmacokinetics and biodistribution profiles of siRNA. This review describes the pharmacokinetics and biodistribution of siRNA nanomedicines, focusing on those reported in the past 5years, and their pharmacological effects in selected disease models such as hepatocellular carcinoma, liver infections, and respiratory diseases. The examples discussed here will provide an insight into the current status of the art and unmet needs in siRNA delivery.
Collapse
|
28
|
Yang SD, Zhu WJ, Zhu QL, Chen WL, Ren ZX, Li F, Yuan ZQ, Li JZ, Liu Y, Zhou XF, Liu C, Zhang XN. Binary-copolymer system base on low-density lipoprotein-coupled N-succinyl chitosan lipoic acid micelles for co-delivery MDR1 siRNA and paclitaxel, enhances antitumor effects via reducing drug. J Biomed Mater Res B Appl Biomater 2016; 105:1114-1125. [PMID: 27008163 DOI: 10.1002/jbm.b.33636] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 01/04/2016] [Accepted: 01/31/2016] [Indexed: 12/13/2022]
Abstract
The development of effective and stable carriers of small interfering RNA (siRNA) is important for treating cancer with multidrug resistance (MDR). We developed a new gene and drug co-delivery system and checked its characteristics. Low-density lipoprotein (LDL) was coupled with N-succinyl chitosan (NSC) Lipoic acid (LA) micelles and co-delivered MDR1 siRNA and paclitaxel (PTX-siRNA/LDL-NSC-LA) to enhance antitumor effects by silencing the MDR gene of tumors (Li et al., Adv Mater 2014;26:8217-8224). In our study, we developed a new type of containing paclitaxel-loaded micelles and siRNA-loaded LDL nanoparticle. This "binary polymer" is pH and reduction dual-sensitive core-crosslinked micelles. PTX-siRNA/LDL-NSC-LA had an average particle size of (171.6 ± 6.42) nm, entrapment efficiency of (93.92 ± 1.06) %, and drug-loading amount of (12.35% ± 0.87) %. In vitro, MCF-7 cells, high expressed LDL receptor, were more sensitive to this delivery system than to taxol® and cell activity was inhibited significantly. Fluorescence microscopy showed that PTX-siRNA/LDL-NSC-LA was uptaken very conveniently and played a key role in antitumor activity. PTX-siRNA/LDL-NSC-LA protected the siRNA from degradation by macrophage phagocytosis and evidently down-regulated the level of mdr1 mRNA as well as the expression of P-gp. We tested the target ability of PTX-siRNA/LDL-NSC-LA in vivo in tumor-bearing nude mice. Results showed that this system could directly deliver siRNA and PTX to cancer cells. Thus, new co-delivering siRNA and antitumor drugs should be explored for solving MDR in cancer. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 1114-1125, 2017.
Collapse
Affiliation(s)
- Shu-Di Yang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, People's Republic of China
| | - Wen-Jing Zhu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, People's Republic of China
| | - Qiao-Ling Zhu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, People's Republic of China.,Nanjing Gulou Hospital, Nanjing, 210029, People's Republic of China
| | - Wei-Liang Chen
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, People's Republic of China
| | - Zhao-Xiang Ren
- Jiangsu Key Laboratory for Translational Research and Therapy for Neuropsycho-disoders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, People's Republic of China
| | - Fang Li
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, People's Republic of China
| | - Zhi-Qiang Yuan
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, People's Republic of China
| | - Ji-Zhao Li
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, People's Republic of China
| | - Yang Liu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, People's Republic of China
| | - Xiao-Feng Zhou
- College of Radiological Medicine and Protection, Soochow University, Suzhou, 215123, People's Republic of China.,Changshu Hospital of Traditional Chinese Medicine, Changshu, 215500, People's Republic of China
| | - Chun Liu
- The Hospital of Suzhou People's Hospital Affiliated to Nanjing Medical University, Suzhou, 215000, People's Republic of China
| | - Xue-Nong Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, People's Republic of China
| |
Collapse
|
29
|
Sigg SJ, Postupalenko V, Duskey JT, Palivan CG, Meier W. Stimuli-Responsive Codelivery of Oligonucleotides and Drugs by Self-Assembled Peptide Nanoparticles. Biomacromolecules 2016; 17:935-45. [PMID: 26871486 DOI: 10.1021/acs.biomac.5b01614] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Ever more emerging combined treatments exploiting synergistic effects of drug combinations demand smart, responsive codelivery carriers to reveal their full potential. In this study, a multifunctional stimuli-responsive amphiphilic peptide was designed and synthesized to self-assemble into nanoparticles capable of co-bearing and -releasing hydrophobic drugs and antisense oligonucleotides for combined therapies. The rational design was based on a hydrophobic l-tryptophan-d-leucine repeating unit derived from a truncated sequence of gramicidin A (gT), to entrap hydrophobic cargo, which is combined with a hydrophilic moiety of histidines to provide electrostatic affinity to nucleotides. Stimuli-responsiveness was implemented by linking the hydrophobic and hydrophilic sequence through an artificial amino acid bearing a disulfide functional group (H3SSgT). Stimuli-responsive peptides self-assembled in spherical nanoparticles in sizes (100-200 nm) generally considered as preferable for drug delivery applications. Responsive peptide nanoparticles revealed notable nucleotide condensing abilities while maintaining the ability to load hydrophobic cargo. The disulfide cleavage site introduced in the peptide sequence induced responsiveness to physiological concentrations of reducing agent, serving to release the incorporated molecules. Furthermore, the peptide nanoparticles, singly loaded or coloaded with boron-dipyrromethene (BODIPY) and/or antisense oligonucleotides, were efficiently taken up by cells. Such amphiphilic peptides that led to noncytotoxic, reduction-responsive nanoparticles capable of codelivering hydrophobic and nucleic acid payloads simultaneously provide potential toward combined treatment strategies to exploit synergistic effects.
Collapse
Affiliation(s)
- Severin J Sigg
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Viktoriia Postupalenko
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Jason T Duskey
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Cornelia G Palivan
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| | - Wolfgang Meier
- Department of Chemistry, University of Basel , Klingelbergstrasse 80, CH-4056 Basel, Switzerland
| |
Collapse
|
30
|
Huang Z, Zhang S, Shen Y, Liu W, Long J, Zhou S. Influence of MDR1 methylation on the curative effect of interventional embolism chemotherapy for cervical cancer. Ther Clin Risk Manag 2016; 12:217-23. [PMID: 26929635 PMCID: PMC4760654 DOI: 10.2147/tcrm.s95453] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Multi-drug resistance (MDR) is the main cause of tumor failure to chemotherapy. This study aims to explore the influence of MDR1 methylation on curative effect of interventional embolism chemotherapy for cervical cancer. Methods Sixty-seven patients with cervical cancer receiving embolism chemotherapy were selected, and 45 normal cervical tissues were included as a control. Immunohistochemistry was used to detect the level of P-glycoprotein (P-gp) in cervical cancer, and to make an analysis compared with normal tissues. The methylation status of the MDR1 gene promoter region 16 CpG units was analyzed by using kilobase-specific cracking and matrix-assisted laser desorption ionization time of flight mass spectrometry. Results The results indicated that the positive expression rates of P-gp were 0% (0/45) in normal cervical tissue, and 61.19% (41/67) and 77.61% (52/67) before and after interventional embolism chemotherapy in cervical cancer tissues, respectively. There were significant differences compared with normal cervical tissues (χ2=4.2523, 0.0392). The positive expression rate of P-gp before chemotherapy was negatively correlated with efficacy of chemotherapy (r=−0.340, P=0.005). Methylation rate of 13 CpG units in normal tissues was significantly greater than cervical tissues (P<0.05). In cervical cancer tissue, methylation rate of six CpG units before interventional embolism chemotherapy was higher than after chemotherapy, but that of one CpG unit was lower than after chemotherapy (P<0.05). The methylation rate of one CpG unit with effective chemotherapy before chemotherapy was significantly higher than ineffective chemotherapy (P<0.05), and the other CpG units were similar (P>0.05). Conclusion P-gp expression level coded by MDR1, methylation status of partial MDR1 gene promoter regions CpG island, is closely related to the efficacy of interventional embolism chemotherapy for cervical cancer before the operation.
Collapse
Affiliation(s)
- Zhi Huang
- Department of Interventional Radiology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, People's Republic of China
| | - Shuai Zhang
- Department of Interventional Radiology, Cancer Hospital of Guizhou Medical University, Guiyang, People's Republic of China
| | - Yaping Shen
- Department of Interventional Radiology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, People's Republic of China
| | - Weixin Liu
- Department of Interventional Radiology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, People's Republic of China
| | - Jipu Long
- Department of Interventional Radiology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, People's Republic of China
| | - Shi Zhou
- Department of Interventional Radiology, The Affiliated Baiyun Hospital of Guizhou Medical University, Guizhou Medical University, Guiyang, People's Republic of China
| |
Collapse
|
31
|
Lu S, Ding Y, Cui W, Pan R, Xu W, Chen P. Supramolecular peptide amphiphile based nanocarrier for pH-triggered Dox release, overcoming drug resistance. RSC Adv 2016. [DOI: 10.1039/c6ra21389h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Self-assembled peptide amphiphile–doxorubicin conjugates showed pH-triggered drug release and ability to combat the drug resistance in cancer cells.
Collapse
Affiliation(s)
- Sheng Lu
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology
- University of Waterloo
- Waterloo
- Canada
| | - Yong Ding
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology
- University of Waterloo
- Waterloo
- Canada
| | - Weijia Cui
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology
- University of Waterloo
- Waterloo
- Canada
| | - Ran Pan
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology
- University of Waterloo
- Waterloo
- Canada
| | - Wen Xu
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology
- University of Waterloo
- Waterloo
- Canada
| | - P. Chen
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology
- University of Waterloo
- Waterloo
- Canada
| |
Collapse
|
32
|
Sobot D, Mura S, Couvreur P. How can nanomedicines overcome cellular-based anticancer drug resistance? J Mater Chem B 2016; 4:5078-5100. [DOI: 10.1039/c6tb00900j] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This review discusses the mechanisms of anticancer drug resistance according to its cellular level of action and outlines the nanomedicine-based strategies adopted to overcome it.
Collapse
Affiliation(s)
- Dunja Sobot
- Institut Galien Paris-Sud
- UMR 8612
- CNRS
- Univ. Paris-Sud
- Université Paris-Saclay
| | - Simona Mura
- Institut Galien Paris-Sud
- UMR 8612
- CNRS
- Univ. Paris-Sud
- Université Paris-Saclay
| | - Patrick Couvreur
- Institut Galien Paris-Sud
- UMR 8612
- CNRS
- Univ. Paris-Sud
- Université Paris-Saclay
| |
Collapse
|
33
|
Mechanisms of Drug Resistance in Veterinary Oncology- A Review with an Emphasis on Canine Lymphoma. Vet Sci 2015; 2:150-184. [PMID: 29061939 PMCID: PMC5644636 DOI: 10.3390/vetsci2030150] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 07/22/2015] [Accepted: 08/04/2015] [Indexed: 01/09/2023] Open
Abstract
Drug resistance (DR) is the major limiting factor in the successful treatment of systemic neoplasia with cytotoxic chemotherapy. DR can be either intrinsic or acquired, and although the development and clinical implications are different, the underlying mechanisms are likely to be similar. Most causes for DR are pharmacodynamic in nature, result from adaptations within the tumor cell and include reduced drug uptake, increased drug efflux, changes in drug metabolism or drug target, increased capacity to repair drug-induced DNA damage or increased resistance to apoptosis. The role of active drug efflux transporters, and those of the ABC-transporter family in particular, have been studied extensively in human oncology and to a lesser extent in veterinary medicine. Methods reported to assess ABC-transporter status include detection of the actual protein (Western blot, immunohistochemistry), mRNA or ABC-transporter function. The three major ABC-transporters associated with DR in human oncology are ABCB1 or P-gp, ABCC1 or MRP1, and ABCG2 or BCRP, and have been demonstrated in canine cell lines, healthy dogs and dogs with cancer. Although this supports a causative role for these ABC-transporters in DR cytotoxic agents in the dog, the relative contribution to the clinical phenotype of DR in canine cancer remains an area of debate and requires further prospective studies.
Collapse
|
34
|
Kong LL, Zhuang XM, Yang HY, Yuan M, Xu L, Li H. Inhibition of P-glycoprotein Gene Expression and Function Enhances Triptolide-induced Hepatotoxicity in Mice. Sci Rep 2015; 5:11747. [PMID: 26134275 PMCID: PMC4488747 DOI: 10.1038/srep11747] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 06/04/2015] [Indexed: 11/29/2022] Open
Abstract
Triptolide (TP) is the major active principle of Tripterygium wilfordii Hook f. and very effective in treatment of autoimmune diseases. However, TP induced hepatotoxicity limited its clinical applications. Our previous study found that TP was a substrate of P-glycoprotein and its hepatobiliary clearance was markedly affected by P-gp modulation in sandwich-cultured rat hepatocytes. In this study, small interfering RNA (siRNA) and specific inhibitor tariquidar were used to investigate the impact of P-gp down regulation on TP-induced hepatotoxicity. The results showed that when the function of P-gp was inhibited by mdr1a-1 siRNA or tariquidar, the systemic and hepatic exposures of TP were significantly increased. The aggravated hepatotoxicity was evidenced with the remarkably lifted levels of serum biomarkers (ALT and AST) and pathological changes in liver. The other toxicological indicators (MDA, SOD and Bcl-2/Bax) were also significantly changed by P-gp inhibition. The data analysis showed that the increase of TP exposure in mice was quantitatively correlated to the enhanced hepatotoxicity, and the hepatic exposure was more relevant to the toxicity. P-gp mediated clearance played a significant role in TP detoxification. The risk of herb-drug interaction likely occurs when TP is concomitant with P-gp inhibitors or substrates in clinic.
Collapse
Affiliation(s)
- Ling-Lei Kong
- 1] State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China [2] Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Xiao-Mei Zhuang
- 1] State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China [2] Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Hai-Ying Yang
- 1] State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China [2] Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Mei Yuan
- 1] State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China [2] Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Liang Xu
- 1] State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China [2] Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Hua Li
- 1] State Key Laboratory of Toxicology and Medical Countermeasures, Beijing 100850, China [2] Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| |
Collapse
|
35
|
Essex S, Navarro G, Sabhachandani P, Chordia A, Trivedi M, Movassaghian S, Torchilin VP. Phospholipid-modified PEI-based nanocarriers for in vivo siRNA therapeutics against multidrug-resistant tumors. Gene Ther 2015; 22:257-266. [PMID: 25354685 PMCID: PMC4352110 DOI: 10.1038/gt.2014.97] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Revised: 09/05/2014] [Accepted: 09/17/2014] [Indexed: 12/17/2022]
Abstract
Multidrug resistance (MDR) mediated by P-glycoprotein overexpression in solid tumors is a major factor in the failure of many forms of chemotherapy. Here we evaluated phospholipid-modified, low-molecular-weight polyethylenimine (DOPE-PEI) nanocarriers for intravenous delivery of anti-P-pg siRNA to tumors with the final goal of modulating MDR in breast cancer. First, we studied the biodistribution of DOPE-PEI nanocarriers and the effect of PEG coating in a subcutaneous breast tumor model. Four hours postinjection, PEGylated carriers showed an 8% injected dose (ID) accumulation in solid tumor via the enhanced permeability and retention effect and 22% ID in serum due to a prolonged, PEG-mediated circulation. Second, we established the therapeutic efficacy and safety of DOPE-PEI/siRNA-mediated P-gp downregulation in combination with doxorubicin (Dox) chemotherapy in MCF-7/MDR xenografts. Weekly injection of siRNA nanopreparations and Dox for up to 5 weeks sensitized the tumors to otherwise non-effective doses of Dox and decreased the tumor volume by threefold vs controls. This therapeutic improvement in response to Dox was attributed to the significant, sequence-specific P-gp downregulation in excised tumors mediated by the DOPE-PEI formulations.
Collapse
Affiliation(s)
- Sean Essex
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Gemma Navarro
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Pooja Sabhachandani
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Aabha Chordia
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Malav Trivedi
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Sara Movassaghian
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| | - Vladimir P. Torchilin
- Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
36
|
Cort A, Ozben T. Natural Product Modulators to Overcome Multidrug Resistance In Cancer. Nutr Cancer 2015; 67:411-23. [DOI: 10.1080/01635581.2015.1002624] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Aysegul Cort
- Department of Biochemistry, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| | - Tomris Ozben
- Department of Biochemistry, Faculty of Medicine, Akdeniz University, Antalya, Turkey
| |
Collapse
|
37
|
Cancer-targeted MDR-1 siRNA delivery using self-cross-linked glycol chitosan nanoparticles to overcome drug resistance. J Control Release 2014; 198:1-9. [PMID: 25481438 DOI: 10.1016/j.jconrel.2014.11.019] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 11/18/2014] [Indexed: 02/07/2023]
Abstract
P-glycoprotein (Pgp) mediated multi-drug resistance (MDR) is a major cause of failure in chemotherapy. In this study, small interfering RNA (siRNA) for Pgp down-regulation was delivered to tumors to overcome MDR in cancer. To achieve an efficient siRNA delivery in vivo, self-polymerized 5'-end thiol-modified siRNA (poly-siRNA) was incorporated in tumor targeting glycol chitosan nanoparticles. Pgp-targeted poly-siRNA (psi-Pgp) and thiolated glycol chitosan polymers (tGC) formed stable nanoparticles (psi-Pgp-tGC NPs), and the resulting nanoparticles protected siRNA molecules from enzymatic degradation. The psi-Pgp-tGC NPs could release functional siRNA molecules after cellular delivery, and they were able to facilitate siRNA delivery to Adriamycin-resistant breast cancer cells (MCF-7/ADR). After intravenous administration, the psi-Pgp-tGC NPs accumulated in MCF-7/ADR tumors and down-regulated P-gp expression to sensitize cancer cells. Consequently, chemo-siRNA combination therapy significantly inhibited tumor growth without systemic toxicity. These psi-Pgp-tGC NPs showed great potential as a supplementary therapeutic agent for drug-resistant cancer.
Collapse
|
38
|
Misra R, Das M, Sahoo BS, Sahoo SK. Reversal of multidrug resistance in vitro by co-delivery of MDR1 targeting siRNA and doxorubicin using a novel cationic poly(lactide-co-glycolide) nanoformulation. Int J Pharm 2014; 475:372-84. [PMID: 25178825 DOI: 10.1016/j.ijpharm.2014.08.056] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 08/22/2014] [Accepted: 08/27/2014] [Indexed: 12/22/2022]
Abstract
Over expression of drug efflux transporters such as P-glycoprotein (P-gp) cumulatively leading to multidrug resistance (MDR) embodies a major hindrance for successful cancer therapy. A paradigm nanomedicinal approach involving an anticancer drug and modulators of drug resistance within one multifunctional nanocarrier-based delivery system represent an ideal modality for the treatment of MDR. In this regards, we have developed a cationic polymeric nanoparticulate system loaded with MDR1-siRNA and doxorubicin. Results indicated augmented synergistic effect of combinational nanoformulation in overcoming MDR in MCF-7/ADR cells. Therefore, the above regime could be a promising co-delivery system for effective therapy of drug resistant breast cancer.
Collapse
Affiliation(s)
- Ranjita Misra
- Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Orissa, India
| | - Manasi Das
- Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Orissa, India
| | - Bhabani Sankar Sahoo
- Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Orissa, India
| | - Sanjeeb K Sahoo
- Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Orissa, India.
| |
Collapse
|
39
|
Shen J, Wang Q, Hu Q, Li Y, Tang G, Chu PK. Restoration of chemosensitivity by multifunctional micelles mediated by P-gp siRNA to reverse MDR. Biomaterials 2014; 35:8621-34. [PMID: 25002258 DOI: 10.1016/j.biomaterials.2014.06.035] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2014] [Accepted: 06/17/2014] [Indexed: 01/29/2023]
Abstract
One of the main obstacles in tumor therapy is multiple drug resistance (MDR) and an underlying mechanism of MDR is up-regulation of the transmembrane ATP-binding cassette (ABC) transporter proteins, especially P-glycoprotein (P-gp). In the synergistic treatment of siRNA and anti-cancer drug doxorubicin, it is crucial that both the siRNA and doxorubicin are simultaneously delivered to the tumor cells and the siRNA can fleetly down-regulate P-g before doxorubicin inactivates the P-gp and is pumped out. Herein, a type of micelles comprising a polycationic PEI-CyD shell to condense the siRNA and hydrophobic core to package doxorubicin is reported. The structure of the polymer is determined by (1)H NMR, FT-IR, DSC, and XRD and the micelles are characterized by DLS, 2D-NOESY NMR, and TEM to study the self-assembly of the micelles with siRNA and drugs. In vitro studies demonstrate controlled release and temporal enhancement of the therapeutic efficacy of P-gp siRNA and doxorubicin. Release of siRNA down-regulates the mRNA and protein levels of P-gp in the MCF-7/ADR cell lines effectively and the accumulated doxorubicin facilitates apoptosis of the cells to reverse MDR. Moreover, in vivo research reveals that the siRNA and doxorubicin loaded micelles induce tumor cell apoptosis and inhibit the growth of MDR tumor. The western blotting and RT-PCR results illustrate that the synergistic treatment of siRNA and doxorubicin leads to efficient reduction of the P-gp expression as well as cell apoptotic induction in MDR tumors at a small dosage of 0.5 mg/kg. The micelles have large clinical potential in drug/RNAi synergistic treatment via restoration of the chemosensitivity in MDR cancer therapy.
Collapse
Affiliation(s)
- Jie Shen
- Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou 310028, PR China; Department of Physics & Materials Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Qiwen Wang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou 310028, PR China; Department of Cardiology, The First Affiliated Hospital, Zhejiang University, School of Medicine, Hangzhou 310003, PR China
| | - Qida Hu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, PR China
| | - Yongbing Li
- Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou 310028, PR China; Department of Physics & Materials Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Guping Tang
- Institute of Chemical Biology and Pharmaceutical Chemistry, Zhejiang University, Hangzhou 310028, PR China; Department of Physics & Materials Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China.
| | - Paul K Chu
- Department of Physics & Materials Science, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China.
| |
Collapse
|
40
|
Vargas JR, Stanzl EG, Teng NNH, Wender PA. Cell-penetrating, guanidinium-rich molecular transporters for overcoming efflux-mediated multidrug resistance. Mol Pharm 2014; 11:2553-65. [PMID: 24798708 PMCID: PMC4123947 DOI: 10.1021/mp500161z] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
![]()
Multidrug resistance (MDR) is a major
cause of chemotherapy failure
in the clinic. Drugs that were once effective against naïve
disease subsequently prove ineffective against recurrent disease,
which often exhibits an MDR phenotype. MDR can be attributed to many
factors; often dominating among these is the ability of a cell to
suppress or block drug entry through upregulation of membrane-bound
drug efflux pumps. Efflux pumps exhibit polyspecificity, recognizing
and exporting many different types of drugs, especially those whose
lipophilic nature contributes to residence in the membrane. We have
developed a general strategy to overcome efflux-based resistance.
This strategy involves conjugating a known drug that succumbs to efflux-mediated
resistance to a cell-penetrating molecular transporter, specifically,
the cell-penetrating peptide (CPP), d-octaarginine. The resultant
conjugates are discrete single entities (not particle mixtures) and
highly water-soluble. They rapidly enter cells, are not substrates
for efflux pumps, and release the free drug only after cellular entry
at a rate controlled by linker design and favored by target cell chemistry.
This general strategy can be applied to many classes of drugs and
allows for an exceptionally rapid advance to clinical testing, especially
of drugs that succumb to resistance. The efficacy of this strategy
has been successfully demonstrated with Taxol in cellular and animal
models of resistant cancer and with ex vivo samples from patients
with ovarian cancer. Next generation efforts in this area will involve
the extension of this strategy to other chemotherapeutics and other
MDR-susceptible diseases.
Collapse
Affiliation(s)
- Jessica R Vargas
- Departments of Chemistry and Chemical and Systems Biology, Stanford University , Stanford, California 94305, United States
| | | | | | | |
Collapse
|
41
|
Emerging integrated nanohybrid drug delivery systems to facilitate the intravenous-to-oral switch in cancer chemotherapy. J Control Release 2014; 176:94-103. [DOI: 10.1016/j.jconrel.2013.12.030] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/24/2013] [Accepted: 12/24/2013] [Indexed: 12/31/2022]
|
42
|
Reversal of ATP-binding cassette drug transporter activity to modulate chemoresistance: why has it failed to provide clinical benefit? Cancer Metastasis Rev 2013; 32:211-27. [PMID: 23093326 DOI: 10.1007/s10555-012-9402-8] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Enhanced drug extrusion from cells due to the overexpression of the ATP-binding cassette (ABC) drug transporters inhibits the cytotoxic effects of structurally diverse and mechanistically unrelated anticancer agents and is a major cause of multidrug resistance (MDR) of human malignancies. Multiple compounds can suppress the activity of these efflux transporters and sensitize resistant tumor cells, but despite promising preclinical and early clinical data, they have yet to find a role in oncologic practice. Based on the knowledge of the structure, function, and distribution of MDR-related ABC transporters and the results of their preclinical and clinical evaluation, we discuss probable reasons why these inhibitors have not improved the outcome of therapy for cancer patients. We also outline new MDR-reversing strategies that directly target ABC transporters or circumvent relevant signaling pathways.
Collapse
|
43
|
Saneja A, Khare V, Alam N, Dubey RD, Gupta PN. Advances in P-glycoprotein-based approaches for delivering anticancer drugs: pharmacokinetic perspective and clinical relevance. Expert Opin Drug Deliv 2013; 11:121-38. [DOI: 10.1517/17425247.2014.865014] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
44
|
Minko T, Rodriguez-Rodriguez L, Pozharov V. Nanotechnology approaches for personalized treatment of multidrug resistant cancers. Adv Drug Deliv Rev 2013; 65:1880-95. [PMID: 24120655 DOI: 10.1016/j.addr.2013.09.017] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 09/28/2013] [Accepted: 09/30/2013] [Indexed: 12/14/2022]
Abstract
The efficacy of chemotherapy is substantially limited by the resistance of cancer cells to anticancer drugs that fluctuates significantly in different patients. Under identical chemotherapeutic protocols, some patients may receive relatively ineffective doses of anticancer agents while other individuals obtain excessive amounts of drugs that induce severe adverse side effects on healthy tissues. The current review is focused on an individualized selection of drugs and targets to suppress multidrug resistance. Such selection is based on the molecular characteristics of a tumor from an individual patient that can potentially improve the treatment outcome and bring us closer to an era of personalized medicine.
Collapse
|
45
|
Yin Q, Shen J, Zhang Z, Yu H, Li Y. Reversal of multidrug resistance by stimuli-responsive drug delivery systems for therapy of tumor. Adv Drug Deliv Rev 2013; 65:1699-715. [PMID: 23611952 DOI: 10.1016/j.addr.2013.04.011] [Citation(s) in RCA: 294] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 02/01/2013] [Accepted: 04/13/2013] [Indexed: 12/15/2022]
Abstract
Multidrug resistance (MDR) is a major obstacle to successful cancer therapy, especially for chemotherapy. The new drug delivery system (DDS) provides promising approaches to reverse MDR, for which the poor cellular uptake and insufficient intracellular drug release remain rate-limiting steps for reaching the drug concentration level within the therapeutic window. Stimulus-coupled drug delivery can control the drug-releasing pattern temporally and spatially, and improve the accumulation of chemotherapeutic agents at targeting sites. In this review, the applications of DDS which is responsive to different types of stimuli in MDR cancer therapy is introduced, and the design, construction, stimuli-sensitivity and the effect to reverse MDR of the stimuli-responsive DDS are discussed.
Collapse
|
46
|
Patel NR, Pattni BS, Abouzeid AH, Torchilin VP. Nanopreparations to overcome multidrug resistance in cancer. Adv Drug Deliv Rev 2013; 65:1748-62. [PMID: 23973912 DOI: 10.1016/j.addr.2013.08.004] [Citation(s) in RCA: 257] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 08/02/2013] [Indexed: 01/08/2023]
Abstract
Multidrug resistance is the most widely exploited phenomenon by which cancer eludes chemotherapy. Broad variety of factors, ranging from the cellular ones, such as over-expression of efflux transporters, defective apoptotic machineries, and altered molecular targets, to the physiological factors such as higher interstitial fluid pressure, low extracellular pH, and formation of irregular tumor vasculature are responsible for multidrug resistance. A combination of various undesirable factors associated with biological surroundings together with poor solubility and instability of many potential therapeutic small & large molecules within the biological systems and systemic toxicity of chemotherapeutic agents has necessitated the need for nano-preparations to optimize drug delivery. The physiology of solid tumors presents numerous challenges for successful therapy. However, it also offers unique opportunities for the use of nanotechnology. Nanoparticles, up to 400 nm in size, have shown great promise for carrying, protecting and delivering potential therapeutic molecules with diverse physiological properties. In this review, various factors responsible for the MDR and the use of nanotechnology to overcome the MDR, the use of spheroid culture as well as the current technique of producing microtumor tissues in vitro are discussed in detail.
Collapse
|
47
|
Effective response of doxorubicin-sensitive and -resistant breast cancer cells to combinational siRNA therapy. J Control Release 2013; 172:219-228. [DOI: 10.1016/j.jconrel.2013.08.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 08/10/2013] [Accepted: 08/12/2013] [Indexed: 12/16/2022]
|
48
|
Polymeric delivery of siRNA for dual silencing of Mcl-1 and P-glycoprotein and apoptosis induction in drug-resistant breast cancer cells. Cancer Gene Ther 2013; 20:169-77. [DOI: 10.1038/cgt.2013.8] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
49
|
Tay Z, Eng RJ, Sajiki K, Lim KK, Tang MY, Yanagida M, Chen ES. Cellular robustness conferred by genetic crosstalk underlies resistance against chemotherapeutic drug doxorubicin in fission yeast. PLoS One 2013; 8:e55041. [PMID: 23365689 PMCID: PMC3554685 DOI: 10.1371/journal.pone.0055041] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Accepted: 12/18/2012] [Indexed: 11/28/2022] Open
Abstract
Doxorubicin is an anthracycline antibiotic that is among one of the most commonly used chemotherapeutic agents in the clinical setting. The usage of doxorubicin is faced with many problems including severe side effects and chemoresistance. To overcome these challenges, it is important to gain an understanding of the underlying molecular mechanisms with regards to the mode of action of doxorubicin. To facilitate this aim, we identified the genes that are required for doxorubicin resistance in the fission yeast Schizosaccharomyces pombe. We further demonstrated interplay between factors controlling various aspects of chromosome metabolism, mitochondrial respiration and membrane transport. In the nucleus we observed that the subunits of the Ino80, RSC, and SAGA complexes function in the similar epistatic group that shares significant overlap with the homologous recombination genes. However, these factors generally act in synergistic manner with the chromosome segregation regulator DASH complex proteins, possibly forming two major arms for regulating doxorubicin resistance in the nucleus. Simultaneous disruption of genes function in membrane efflux transport or the mitochondrial respiratory chain integrity in the mutants defective in either Ino80 or HR function resulted in cumulative upregulation of drug-specific growth defects, suggesting a rewiring of pathways that synergize only when the cells is exposed to the cytotoxic stress. Taken together, our work not only identified factors that are required for survival of the cells in the presence of doxorubicin but has further demonstrated that an extensive molecular crosstalk exists between these factors to robustly confer doxorubicin resistance.
Collapse
Affiliation(s)
- Zoey Tay
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- National University Health System, Singapore
| | - Ru Jun Eng
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- National University Health System, Singapore
| | - Kenichi Sajiki
- G0 Cell Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Kim Kiat Lim
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- National University Health System, Singapore
| | - Ming Yi Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- National University Health System, Singapore
| | - Mitsuhiro Yanagida
- G0 Cell Unit, Okinawa Institute of Science and Technology, Okinawa, Japan
| | - Ee Sin Chen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- National University Health System, Singapore
| |
Collapse
|
50
|
Xia CQ, Smith PG. Drug efflux transporters and multidrug resistance in acute leukemia: therapeutic impact and novel approaches to mediation. Mol Pharmacol 2012; 82:1008-21. [PMID: 22826468 DOI: 10.1124/mol.112.079129] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Multidrug resistance (MDR), which is mediated by multiple drug efflux ATP-binding cassette (ABC) transporters, is a critical issue in the treatment of acute leukemia, with permeability glycoprotein (P-gp), multidrug resistance-associated protein 1, and breast cancer resistance protein (i.e., ABCG2) consistently being shown to be key effectors of MDR in cell line studies. Studies have demonstrated that intrinsic MDR can arise as a result of specific gene expression profiles and that drug-induced overexpression of P-gp and other MDR proteins can result in acquired resistance, with multiple ABC transporters having been shown to be overexpressed in cell lines selected for resistance to multiple drugs used to treat acute leukemia. Furthermore, numerous anticancer drugs, including agents commonly used for the treatment of acute leukemia (e.g., doxorubicin, vincristine, mitoxantrone, and methotrexate), have been shown to be P-gp substrates or to be susceptible to efflux mediated by other MDR proteins, and multiple clinical studies have demonstrated associations between P-gp or other MDR protein expression and responses to therapy or survival rates in acute leukemia. Here we review the importance of MDR in cancer, with a focus on acute leukemia, and we highlight the need for rapid accurate assessment of MDR status for optimal treatment selection. We also address the latest research on overcoming MDR, from inhibition of P-gp and other MDR proteins through various approaches (including direct antagonism and gene silencing) to the design of novel agents or novel delivery systems for existing therapeutic agents, to evade cellular efflux.
Collapse
Affiliation(s)
- Cindy Q Xia
- Millennium Pharmaceuticals, Inc., Cambridge, MA 02139, USA.
| | | |
Collapse
|