1
|
Ferreira JCC, Gonçalves MST, Preto A, Sousa MJ. Anticancer Activity of Benzo[ a]phenoxazine Compounds Promoting Lysosomal Dysfunction. Cells 2024; 13:1385. [PMID: 39195273 PMCID: PMC11352945 DOI: 10.3390/cells13161385] [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: 07/04/2024] [Revised: 08/02/2024] [Accepted: 08/12/2024] [Indexed: 08/29/2024] Open
Abstract
Specific cancer therapy remains a problem to be solved. Breast and colorectal cancer are among the cancers with the highest prevalence and mortality rates. Although there are some therapeutic options, there are still few effective agents for those cancers, which constitutes a clinical problem that requires further research efforts. Lysosomes play an important role in cancer cells' survival, and targeting lysosomes has gained increased interest. In recent years, our team has been synthetizing and testing novel benzo[a]phenoxazine derivatives, as they have been shown to possess potent pharmacological activities. Here, we investigated the anticancer activity of three of the most potent derivatives from our library, C9, A36, and A42, on colorectal- and breast-cancer-derived cell lines, and compared this with the effect on non-neoplastic cell lines. We observed that the three compounds were selective for the cancer cells, namely the RKO colorectal cancer cell line and the MCF7 breast cancer cell line. In both models, the compounds reduced cell proliferation, cell survival, and cell migration, accumulated on the lysosome, and induced cell death accompanied by lysosomal membrane permeabilization (LMP), increasing the intracellular pH and ROS accumulation. Our results demonstrated that these compounds specifically target lysosomes from cancer cells, making them promising candidates as LMP inducers for cancer therapy.
Collapse
Affiliation(s)
- João Carlos Canossa Ferreira
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal; (A.P.); (M.J.S.)
- IBS-Institute of Science and Innovation for Bio-Sustainability, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
- Centre of Chemistry (CQUM), Department of Chemistry, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal;
| | - M. Sameiro T. Gonçalves
- Centre of Chemistry (CQUM), Department of Chemistry, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal;
| | - Ana Preto
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal; (A.P.); (M.J.S.)
- IBS-Institute of Science and Innovation for Bio-Sustainability, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| | - Maria João Sousa
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal; (A.P.); (M.J.S.)
- IBS-Institute of Science and Innovation for Bio-Sustainability, University of Minho, Campus of Gualtar, 4710-057 Braga, Portugal
| |
Collapse
|
2
|
Zhang C, Zheng J, Liu J, Li Y, Xing G, Zhang S, Chen H, Wang J, Shao Z, Li Y, Jiang Z, Pan Y, Liu X, Xu P, Wu W. Pan-cancer analyses reveal the molecular and clinical characteristics of TET family members and suggests that TET3 maybe a potential therapeutic target. Front Pharmacol 2024; 15:1418456. [PMID: 39104395 PMCID: PMC11298443 DOI: 10.3389/fphar.2024.1418456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 05/28/2024] [Indexed: 08/07/2024] Open
Abstract
The Ten-Eleven Translocation (TET) family genes are implicated in a wide array of biological functions across various human cancers. Nonetheless, there is a scarcity of studies that comprehensively analyze the correlation between TET family members and the molecular phenotypes and clinical characteristics of different cancers. Leveraging updated public databases and employing several bioinformatics analysis methods, we assessed the expression levels, somatic variations, methylation levels, and prognostic values of TET family genes. Additionally, we explored the association between the expression of TET family genes and pathway activity, tumor microenvironment (TME), stemness score, immune subtype, clinical staging, and drug sensitivity in pan-cancer. Molecular biology and cytology experiments were conducted to validate the potential role of TET3 in tumor progression. Each TET family gene displayed distinct expression patterns across at least ten detected tumors. The frequency of Single Nucleotide Variant (SNV) in TET genes was found to be 91.24%, primarily comprising missense mutation types, with the main types of copy number variant (CNV) being heterozygous amplifications and deletions. TET1 gene exhibited high methylation levels, whereas TET2 and TET3 genes displayed hypomethylation in most cancers, which correlated closely with patient prognosis. Pathway activity analysis revealed the involvement of TET family genes in multiple signaling pathways, including cell cycle, apoptosis, DNA damage response, hormone AR, PI3K/AKT, and RTK. Furthermore, the expression levels of TET family genes were shown to impact the clinical staging of tumor patients, modulate the sensitivity of chemotherapy drugs, and thereby influence patient prognosis by participating in the regulation of the tumor microenvironment, cellular stemness potential, and immune subtype. Notably, TET3 was identified to promote cancer progression across various tumors, and its silencing was found to inhibit tumor malignancy and enhance chemotherapy sensitivity. These findings shed light on the role of TET family genes in cancer progression and offer insights for further research on TET3 as a potential therapeutic target for pan-cancer.
Collapse
Affiliation(s)
- Chunyan Zhang
- Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin, China
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, Tianjin Fifth Central Hospital, Tianjin, China
- High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People’s Hospital, Huangnan Prefecture, Qinghai, China
| | - Jie Zheng
- Department of Pathology, Tianjin Fifth Central Hospital, Tianjin, China
| | - Jin Liu
- North China University of Science and Technology, Tangshan, Hebei, China
| | - Yanxia Li
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, Tianjin Fifth Central Hospital, Tianjin, China
- High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People’s Hospital, Huangnan Prefecture, Qinghai, China
| | - Guoqiang Xing
- Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin, China
| | - Shupeng Zhang
- Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin, China
| | - Hekai Chen
- Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin, China
| | - Jian Wang
- Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin, China
| | - Zhijiang Shao
- Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin, China
- High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People’s Hospital, Huangnan Prefecture, Qinghai, China
| | - Yongyuan Li
- Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin, China
- High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People’s Hospital, Huangnan Prefecture, Qinghai, China
| | - Zhongmin Jiang
- Department of Pathology, Tianjin Fifth Central Hospital, Tianjin, China
| | - Yingzi Pan
- Department of Ophthalmology, Peking University First Hospital, Beijing, China
| | - Xiaozhi Liu
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, Tianjin Fifth Central Hospital, Tianjin, China
- High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People’s Hospital, Huangnan Prefecture, Qinghai, China
| | - Ping Xu
- High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People’s Hospital, Huangnan Prefecture, Qinghai, China
- Department of Pharmacy, Tianjin Fifth Central Hospital, Tianjin, China
| | - Wenhan Wu
- Department of General Surgery, Tianjin Fifth Central Hospital, Tianjin, China
- Tianjin Key Laboratory of Epigenetics for Organ Development of Premature Infants, Tianjin Fifth Central Hospital, Tianjin, China
- High Altitude Characteristic Medical Research Institute, Huangnan Tibetan Autonomous Prefecture People’s Hospital, Huangnan Prefecture, Qinghai, China
- Department of General Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
3
|
Zaghmi A, Aybay E, Jiang L, Shang M, Steinmetz‐Späh J, Wermeling F, Kogner P, Korotkova M, Östling P, Jakobsson P, Seashore‐Ludlow B, Larsson K. High-content screening of drug combinations of an mPGES-1 inhibitor in multicellular tumor spheroids leads to mechanistic insights into neuroblastoma chemoresistance. Mol Oncol 2024; 18:317-335. [PMID: 37519014 PMCID: PMC10850797 DOI: 10.1002/1878-0261.13502] [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: 02/21/2023] [Revised: 06/09/2023] [Accepted: 07/28/2023] [Indexed: 08/01/2023] Open
Abstract
High-throughput drug screening enables the discovery of new anticancer drugs. Although monolayer cell cultures are commonly used for screening, their limited complexity and translational efficiency require alternative models. Three-dimensional cell cultures, such as multicellular tumor spheroids (MCTS), mimic tumor architecture and offer promising opportunities for drug discovery. In this study, we developed a neuroblastoma MCTS model for high-content drug screening. We also aimed to decipher the mechanisms underlying synergistic drug combinations in this disease model. Several agents from different therapeutic categories and with different mechanisms of action were tested alone or in combination with selective inhibition of prostaglandin E2 by pharmacological inhibition of microsomal prostaglandin E synthase-1 (mPGES-1). After a systematic investigation of the sensitivity of individual agents and the effects of pairwise combinations, GFP-transfected MCTS were used in a confirmatory screen to validate the hits. Finally, inhibitory effects on multidrug resistance proteins were examined. In summary, we demonstrate how MCTS-based high-throughput drug screening has the potential to uncover effective drug combinations and provide insights into the mechanism of synergy between an mPGES-1 inhibitor and chemotherapeutic agents.
Collapse
Affiliation(s)
- Ahlem Zaghmi
- Rheumatology Unit, Department of Medicine, SolnaKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Erdem Aybay
- Rheumatology Unit, Department of Medicine, SolnaKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Long Jiang
- Rheumatology Unit, Department of Medicine, SolnaKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Mingmei Shang
- Rheumatology Unit, Department of Medicine, SolnaKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Julia Steinmetz‐Späh
- Rheumatology Unit, Department of Medicine, SolnaKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Fredrik Wermeling
- Rheumatology Unit, Department of Medicine, SolnaKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Per Kogner
- Childhood Cancer Research Unit, Department of Women's and Children's HealthKarolinska InstitutetStockholmSweden
| | - Marina Korotkova
- Rheumatology Unit, Department of Medicine, SolnaKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Päivi Östling
- Department of Oncology‐Pathology, Science for Life LaboratoryKarolinska InstitutetStockholmSweden
| | - Per‐Johan Jakobsson
- Rheumatology Unit, Department of Medicine, SolnaKarolinska Institutet, Karolinska University HospitalStockholmSweden
| | - Brinton Seashore‐Ludlow
- Department of Oncology‐Pathology, Science for Life LaboratoryKarolinska InstitutetStockholmSweden
| | - Karin Larsson
- Rheumatology Unit, Department of Medicine, SolnaKarolinska Institutet, Karolinska University HospitalStockholmSweden
| |
Collapse
|
4
|
Bin Kanner Y, Teng QX, Ganoth A, Peer D, Wang JQ, Chen ZS, Tsfadia Y. Cytotoxicity and reversal effect of sertraline, fluoxetine, and citalopram on MRP1- and MRP7-mediated MDR. Front Pharmacol 2023; 14:1290255. [PMID: 38026953 PMCID: PMC10651738 DOI: 10.3389/fphar.2023.1290255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/13/2023] [Indexed: 12/01/2023] Open
Abstract
Cancer is one of the leading causes of death worldwide, and the development of resistance to chemotherapy drugs is a major challenge in treating malignancies. In recent years, researchers have focused on understanding the mechanisms of multidrug resistance (MDR) in cancer cells and have identified the overexpression of ATP-binding cassette (ABC) transporters, including ABCC1/MRP1 and ABCC10/MRP7, as a key factor in the development of MDR. In this study, we aimed to investigate whether three drugs (sertraline, fluoxetine, and citalopram) from the selective serotonin reuptake inhibitor (SSRI) family, commonly used as antidepressants, could be repurposed as inhibitors of MRP1 and MRP7 transporters and reverse MDR in cancer cells. Using a combination of in silico predictions and in vitro validations, we analyzed the interaction of MRP1 and MRP7 with the drugs and evaluated their ability to hinder cell resistance. We used computational tools to identify and analyze the binding site of these three molecules and determine their binding energy. Subsequently, we conducted experimental assays to assess cell viability when treated with various standard chemotherapies, both with and without the presence of SSRI inhibitors. Our results show that all three SSRI drugs exhibited inhibitory/reversal effects in the presence of chemotherapies on both MRP1-overexpressed cells and MRP7-overexpressed cells, suggesting that these medications have the potential to be repurposed to target MDR in cancer cells. These findings may open the door to using FDA-approved medications in combination therapy protocols to treat highly resistant malignancies and improve the efficacy of chemotherapy treatment. Our research highlights the importance of investigating and repurposing existing drugs to overcome MDR in cancer treatment.
Collapse
Affiliation(s)
- Yuval Bin Kanner
- George S. Wise Faculty of Life Sciences, The School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Assaf Ganoth
- Department of Physical Therapy, Sackler Faculty of Medicine, School of Health Professions, Tel Aviv University, Tel Aviv, Israel
- Reichman University, Herzliya, Israel
| | - Dan Peer
- Laboratory of Precision NanoMedicine, George S. Wise Faculty of Life Sciences, Shmunis School for Biomedicine and Cancer Research, Tel Aviv University, Tel Aviv, Israel
- Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Department of Materials Sciences and Engineering, Iby and Aladar Fleischman Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- Cancer Biology Research Center, Tel Aviv University, Tel Aviv, Israel
| | - Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, New York, NY, United States
| | - Yossi Tsfadia
- George S. Wise Faculty of Life Sciences, The School of Neurobiology, Biochemistry and Biophysics, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
5
|
Kudo N, Kouno R, Shibayama Y. SLC25A40 Facilitates Anticancer Drug Resistance in Human Leukemia K562 Cells. Biol Pharm Bull 2023; 46:1304-1309. [PMID: 37407483 DOI: 10.1248/bpb.b23-00293] [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] [Indexed: 07/07/2023]
Abstract
The chronic myelogenous leukemia cell line, K562/ADM is derived from the K562 cell line, which is resistant to doxorubicin (alias, adriamycin: ADM). P-glycoprotein levels are significantly higher in K562/ADM cells than in K562 cells. The overexpression of p-glycoprotein has been shown to cause drug resistance. Therefore, the present study investigated a novel mechanism underlying the drug resistance of K562/ADM cells. A gene ontology analysis demonstrated that the expression of solute carrier (SLC)-mediated transmembrane transport genes was significantly higher in K562/ADM cells than in K562 cells. The expression level of a member of the SLC family, SLC25A40 was higher in K562/ADM cells than in K562 cells. SLC25A40 is located near the ABCB1 gene. A real-time PCR analysis showed that the expression of SLC25A40, ABCB4, and ADAM22 was up-regulated. These genes are located close to SLC25A40. The down-regulation of SLC25A40 significantly decreased the mitochondrial concentration of glutathione and cell proliferation. Collectively, the present results demonstrated that the expression of SLC25A40 was up-regulated in K562/ADM cells, which enhanced to cell proliferation, and that the expression of SLC25A40 affected drug resistance to ADM.
Collapse
Affiliation(s)
- Nodoka Kudo
- Department of Drug Formulation, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Rikuma Kouno
- Department of Drug Formulation, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Yoshihiko Shibayama
- Department of Drug Formulation, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| |
Collapse
|
6
|
Miao L, Liu Y, Ali NM, Dong Y, Zhang B, Cui X. Bufalin serves as a pharmaceutic that mitigates drug resistance. Drug Metab Rev 2023:1-10. [PMID: 37114332 DOI: 10.1080/03602532.2023.2206065] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/29/2023]
Abstract
Intrinsic or acquired drug resistance of tumor cells is the main cause of tumor chemotherapy failure and tumor-related death. Bufalin (BF) is the main active monomer component extracted from the Traditional Chinese Medicine Toad venom (secretions of glands behind the ears and epidermis of bufo gargarizans and Bufo Melanostictus Schneider). It is a cardiotonic steroid with broad-spectrum anti-cancer effects and has been widely used against various malignant tumors in clinical practice. Pharmacological studies also found that BF has the effect of reversing drug resistance, which provides a new perspective for the application of Traditional Chinese Medicine as a chemosensitizer in cancer therapy. This article provides an extensive search and summary of published research on mitigating drug resistance to BF and reviews its potential mechanisms.
Collapse
Affiliation(s)
- Linxuan Miao
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Ying Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
- Department of Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, P.R. China
| | - Nasra Mohamoud Ali
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Yan Dong
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Bin Zhang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Xiaonan Cui
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| |
Collapse
|
7
|
Olguin JE, Mendoza-Rodriguez MG, Sanchez-Barrera CA, Terrazas LI. Is the combination of immunotherapy with conventional chemotherapy the key to increase the efficacy of colorectal cancer treatment? World J Gastrointest Oncol 2023; 15:251-267. [PMID: 36908325 PMCID: PMC9994043 DOI: 10.4251/wjgo.v15.i2.251] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 11/03/2022] [Accepted: 01/10/2023] [Indexed: 02/14/2023] Open
Abstract
Colorectal cancer (CRC) is among the most prevalent and deadly neoplasms worldwide. According to GLOBOCAN predictions, its incidence will increase from 1.15 million CRC cases in 2020 to 1.92 million cases in 2040. Therefore, a better understanding of the mechanisms involved in CRC development is necessary to improve strategies focused on reducing the incidence, prevalence, and mortality of this oncological pathology. Surgery, chemotherapy, and radiotherapy are the main strategies for treating CRC. The conventional chemotherapeutic agent utilized throughout the last four decades is 5-fluorouracil, notwithstanding its low efficiency as a single therapy. In contrast, combining 5-fluorouracil therapy with leucovorin and oxaliplatin or irinotecan increases its efficiency. However, these treatments have limited and temporary solutions and aggressive side effects. Additionally, most patients treated with these regimens develop drug resistance, which leads to disease progression. The immune response is considered a hallmark of cancer; thus, the use of new strategies and methodologies involving immune molecules, cells, and transcription factors has been suggested for CRC patients diagnosed in stages III and IV. Despite the critical advances in immunotherapy, the development and impact of immune checkpoint inhibitors on CRC is still under investigation because less than 25% of CRC patients display an increased 5-year survival. The causes of CRC are diverse and include modifiable environmental factors (smoking, diet, obesity, and alcoholism), individual genetic mutations, and inflammation-associated bowel diseases. Due to these diverse causes, the solutions likely cannot be generalized. Interestingly, new strategies, such as single-cell multiomics, proteomics, genomics, flow cytometry, and massive sequencing for tumor microenvironment analysis, are beginning to clarify the way forward. Thus, the individual mechanisms involved in developing the CRC microenvironment, their causes, and their consequences need to be understood from a genetic and immunological perspective. This review highlighted the importance of altering the immune response in CRC. It focused on drugs that may modulate the immune response and show specific efficacy and contrasted with evidence that immunosuppression or the promotion of the immune response is the answer to generating effective treatments with combined chemotherapeutic drugs.
Collapse
Affiliation(s)
- Jonadab E Olguin
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-degenerativas, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Monica G Mendoza-Rodriguez
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - C Angel Sanchez-Barrera
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| | - Luis I Terrazas
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-degenerativas, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Estado de Mexico, Mexico
| |
Collapse
|
8
|
Ulker D, Ozyurt R, Erkasap N, Butun V. Magnetic Targeting of 5-Fluorouracil-Loaded Liposome-Nanogels for In Vivo Breast Cancer Therapy and the Cytotoxic Effects on Liver and Kidney. AAPS PharmSciTech 2022; 23:289. [DOI: 10.1208/s12249-022-02438-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 10/03/2022] [Indexed: 02/06/2023] Open
|
9
|
Igaz N, Bélteky P, Kovács D, Papp C, Rónavári A, Szabó D, Gácser A, Kónya Z, Kiricsi M. Functionalized Mesoporous Silica Nanoparticles for Drug-Delivery to Multidrug-Resistant Cancer Cells. Int J Nanomedicine 2022; 17:3079-3096. [PMID: 35859731 PMCID: PMC9293248 DOI: 10.2147/ijn.s363952] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 07/06/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Multidrug resistance is a common reason behind the failure of chemotherapy. Even if the therapy is effective, serious adverse effects might develop due to the low specificity and selectivity of antineoplastic agents. Mesoporous silica nanoparticles (MSNs) are promising materials for tumor-targeting and drug-delivery due to their small size, relatively inert nature, and extremely large specific surfaces that can be functionalized by therapeutic and targeting entities. We aimed to create a fluorescently labeled MSN-based drug-delivery system and investigate their internalization and drug-releasing capability in drug-sensitive MCF-7 and P-glycoprotein-overexpressing multidrug-resistant MCF-7 KCR cancer cells. METHODS AND RESULTS To track the uptake and subcellular distribution of MSNs, particles with covalently coupled red fluorescent Rhodamine B (RhoB) were produced (RhoB@MSNs). Both MCF-7 and MCF-7 KCR cells accumulated a significant amount of RhoB@MSNs. The intracellular RhoB@MSN concentrations did not differ between sensitive and multidrug-resistant cells and were kept at the same level even after cessation of RhoB@MSN exposure. Although most RhoB@MSNs resided in the cytoplasm, significantly more RhoB@MSNs co-localized with lysosomes in multidrug-resistant cells compared to sensitive counterparts. To examine the drug-delivery capability of these particles, RhoB@Rho123@MSNs were established, where RhoB-functionalized nanoparticles carried green fluorescent Rhodamine 123 (Rho123) - a P-glycoprotein substrate - as cargo within mesopores. Significantly higher Rho123 fluorescence intensity was detected in RhoB@Rho123@MSN-treated multidrug-resistant cells than in free Rho123-exposed counterparts. The exceptional drug-delivery potential of MSNs was further verified using Mitomycin C (MMC)-loaded RhoB@MSNs (RhoB@MMC@MSNs). Exposures to RhoB@MMC@MSNs significantly decreased the viability not only of drug-sensitive but of multidrug-resistant cells and the elimination of MDR cells was significantly more robust than upon free MMC treatments. CONCLUSION The efficient delivery of Rho123 and MMC to multidrug-resistant cells via MSNs, the amplified and presumably prolonged intracellular drug concentration, and the consequently enhanced cytotoxic effects envision the enormous potential of MSNs to defeat multidrug-resistant cancer.
Collapse
Affiliation(s)
- Nóra Igaz
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| | - Péter Bélteky
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, Hungary
| | - Dávid Kovács
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
- Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Inserm, CNRS, Valbonne, France
| | - Csaba Papp
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, University of Szeged, Szeged, Hungary
| | - Andrea Rónavári
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, Hungary
| | - Diána Szabó
- Department of Oto-Rhino-Laryngology and Head & Neck Surgery, Szeged, Hungary
| | - Attila Gácser
- HCEMM-USZ Fungal Pathogens Research Group, Department of Microbiology, University of Szeged, Szeged, Hungary
| | - Zoltán Kónya
- Department of Applied and Environmental Chemistry, University of Szeged, Szeged, Hungary
- Eötvös Loránd Research Network, Reaction Kinetics and Surface Chemistry Research Group, Szeged, Hungary
| | - Mónika Kiricsi
- Department of Biochemistry and Molecular Biology, University of Szeged, Szeged, Hungary
| |
Collapse
|
10
|
Novel benzo[4,5]thiazolo[2,3-C][1,2,4]triazoles: Design, synthesis, anticancer evaluation, kinase profiling and molecular docking study. J Mol Struct 2021. [DOI: 10.1016/j.molstruc.2021.131138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
11
|
Kobayashi M, Yonezawa A, Takasawa H, Nagao Y, Iguchi K, Endo S, Ikari A, Matsunaga T. Development of cisplatin resistance in breast cancer MCF7 cells by up-regulating aldo-keto reductase 1C3 expression, glutathione synthesis, and proteasomal proteolysis. J Biochem 2021; 171:97-108. [PMID: 34676395 DOI: 10.1093/jb/mvab117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/18/2021] [Indexed: 02/04/2023] Open
Abstract
Cisplatin (CDDP) is widely prescribed for the treatment of various cancers including bladder cancers, whereas its clinical use for breast cancer chemotherapy is restricted owing to easy acquisition of the chemoresistance. Here, we established a highly CDDP-resistant variant of human breast cancer MCF7 cells and found that procuring the resistance aberrantly elevates the expression of aldo-keto reductase (AKR) 1C3. Additionally, MCF7 cell sensitivity to CDDP was decreased and increased by overexpression and knockdown, respectively, of AKR1C3, clearly inferring that the enzyme plays a crucial role in acquiring the CDDP resistance. The CDDP-resistant cells suppressed the formation of cytotoxic reactive aldehydes by CDDP treatment, and the suppressive effects were almost completely abolished by pretreating with AKR1C3 inhibitor. The resistant cells also exhibited the elevated glutathione amount and 26S proteasomal proteolytic activities, and their CDDP sensitivity was significantly augmented by pretreatment with an inhibitor of glutathione synthesis or proteasomal proteolysis. Moreover, the combined treatment with inhibitors of AKR1C3, glutathione synthesis, and/or proteasomal proteolysis potently overcame the CDDP resistance and docetaxel cross-resistance. Taken together, our results suggest that the combination of inhibitors of AKR1C3, glutathione synthesis, and/or proteasomal proteolysis is effective as an adjuvant therapy to enhance CDDP sensitivity of breast cancer cells.
Collapse
Affiliation(s)
- Mio Kobayashi
- Laboratory of Biochemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Ayano Yonezawa
- Laboratory of Biochemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Hiroaki Takasawa
- Laboratory of Biochemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Yukino Nagao
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, 5-6-1 Mitahora-higashi, Gifu 502-8585, Japan
| | - Kazuhiro Iguchi
- Laboratory of Community Pharmacy, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, 1-25-4 Daigaku-nishi, Gifu 501-1196, Japan
| | - Toshiyuki Matsunaga
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, 5-6-1 Mitahora-higashi, Gifu 502-8585, Japan
| |
Collapse
|
12
|
Zhu X, Xue D, Liu J, Dong F, Li Y, Liu Y. Nodal is involved in chemoresistance of renal cell carcinoma cells via regulation of ABCB1. J Cancer 2021; 12:2041-2049. [PMID: 33754002 PMCID: PMC7974526 DOI: 10.7150/jca.52092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/26/2020] [Indexed: 11/05/2022] Open
Abstract
Renal cell carcinoma (RCC) is the third most frequent malignancy within urological oncology. Understanding mechanisms of chemoresistance in RCC cell is important for therapy and drug development. We established cisplatin (CDDP) resistant RCC cells by treating cells with increasing concentrations of CDDP. Nodal, an important embryonic morphogen, was increased in RCC/CDDP cells. Targeted inhibition of Nodal via its siRNA or neutralization antibody restored sensitivity of RCC resistant cells to CDDP treatment. It was due to that si-Nodal can decrease expression of P-glycoprotein (P-gp, encoded by ABCB1), one important ATP-binding cassette (ABC) membrane transporter for drug efflux. si-Nodal can decrease the transcription and promoter activity of ABCB1. Mechanistically, si-Nodal can decrease the phosphorylation of p65, which can bind to the promoter of ABCB1 and then trigger its transcription. Further, CDDP treatment decreased the expression of Nodal in culture medium of RCC cells. Collectively, we found that Nodal can regulate chemoresistance of RCC cells via regulating transcription of ABCB1.
Collapse
Affiliation(s)
- Xingwang Zhu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Dongwei Xue
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Jia Liu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Fengming Dong
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Yongzhi Li
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| | - Yili Liu
- Department of Urology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China
| |
Collapse
|
13
|
Wang C, Wei X, Shao G. Functional Doxorubicin-Loaded Omega-3 Unsaturated Fatty Acids Nanoparticles in Reversing Hepatocellular Carcinoma Multidrug Resistance. Med Sci Monit 2021; 27:e927727. [PMID: 33524008 PMCID: PMC7863563 DOI: 10.12659/msm.927727] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Background This study investigated a nanoparticle drug delivery system to reverse multidrug resistance (MDR) and assessed its anticancer efficacy in hepatocellular carcinoma (HCC). Material/Methods Docosahexaenoic acid (DHA) was used as the functional excipient and doxorubicin (DOX) as the chemotherapeutic drug to synthesize DOX nanoparticles (DOX-nano). The human HCC cell line HepG2 was used for experiments. HepG2/DOX, HepG2+DOX, HepG2+DOX-nano, HepG2/DOX+DOX, and HepG2/DOX+DOX-nano groups cells were treated with DOX or DOX-nano (5 μg/mL). Nude mice bearing a HepG2/DOX xenograft were divided into model, DOX, vector-nano, and DOX-nano groups and injected with saline, DOX reagent, vector-nano, and DOX-nano (2 mg/kg), respectively. Next, cytotoxicity, cellular uptake, cell apoptosis and migration, fluorescence imaging, TUNEL assay, and tumor inhibition effects were assessed in vitro and in vivo. Furthermore, expression of MDR-related proteins was also detected using western blotting. Results Fluorescence imaging showed that the DOX uptake in the DOX-nano-treated group was the strongest in the HCC cells or tumors. Cell apoptosis was significantly increased in DOX-nano-treated HepG2/DOX cells and tumors, and cell migration was significantly inhibited in the DOX-nano-treated HepG2/DOX cells compared with the other groups. The tumor inhibitory rate in DOX-nano-injected tumors was also significantly higher than in other groups. The expression of breast cancer resistance protein, B-cell lymphoma 2, lung resistance protein, multidrug resistance protein, and protein kinase C alpha was significantly decreased in DOX-nano-treated HepG2/DOX cells and xenograft tumors. Significantly better antitumor and MDR-reversing effects were also observed in the HepG2+DOX group compared with the HepG2/DOX group. Conclusions This study revealed the potential efficacy of a DOX-nano drug delivery system for the treatment of HCC, using HepG2/DOX cells and nude mice bearing HepG2/DOX xenografts.
Collapse
Affiliation(s)
- Chunlei Wang
- Pharmaceutical Preparation Section, Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China (mainland).,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China (mainland)
| | - Xiaoyan Wei
- Pharmaceutical Preparation Section, Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China (mainland).,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China (mainland)
| | - Guoliang Shao
- Pharmaceutical Preparation Section, Cancer Hospital of The University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China (mainland).,Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
14
|
D’yakonov VA, Tuktarova RA, Dzhemileva LU, Ishmukhametova SR, Dzhemilev UM. Synthesis and Anticancer Activity of Hybrid Molecules Based on Lithocholic and (5 Z,9 Z)-Tetradeca-5,9-dienedioic Acids Linked via Mono(di,tri,tetra)ethylene Glycol and α,ω-Diaminoalkane Units. Pharmaceuticals (Basel) 2021; 14:ph14020084. [PMID: 33498764 PMCID: PMC7911507 DOI: 10.3390/ph14020084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 01/25/2023] Open
Abstract
For the first time, hybrid molecules were synthesized on the basis of lithocholic and (5Z,9Z)-1,14-tetradeca-5,9-dienedicarboxylic acids, obtained in two stages using the homo-cyclomagnesiation reaction of 2-(hepta-5,6-diene-1-yloxy)tetrahydro-2H-pyran at the key stage. The resulting hybrid molecules containing 5Z,9Z-dienoic acids are of interest as novel synthetic biologically active precursors to create modern drugs for the treatment of human oncological diseases. The synthesized hybrid molecules were found to exhibit extremely high in vitro inhibitory activity against human topoisomerase I, which is 2-4 times higher than that of camptothecin, a known topoisomerase I inhibitor. Using flow cytometry and fluorescence microscopy, it was first shown that these new molecules are efficient apoptosis inducers in HeLa, U937, Jurkat, K562, and Hek293 cell cultures. In addition, the results of investigations into the effect of the synthesized acids on mitochondria and studies of possible DNA damage in Jurkat tumor cells are also presented.
Collapse
|
15
|
Lucianò AM, Perciballi E, Fiore M, Del Bufalo D, Tata AM. The Combination of the M2 Muscarinic Receptor Agonist and Chemotherapy Affects Drug Resistance in Neuroblastoma Cells. Int J Mol Sci 2020; 21:ijms21228433. [PMID: 33182656 PMCID: PMC7697391 DOI: 10.3390/ijms21228433] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/04/2020] [Accepted: 11/06/2020] [Indexed: 12/15/2022] Open
Abstract
One of the major limits of chemotherapy is depending on the ability of the cancer cells to elude and adapt to different drugs. Recently, we demonstrated how the activation of the M2 muscarinic receptor could impair neuroblastoma cell proliferation. In the present paper, we investigate the possible effects mediated by the preferential M2 receptor agonist arecaidine propargyl ester (APE) on drug resistance in two neuroblastoma cell lines, SK-N-BE and SK-N-BE(2C), a sub-clone presenting drug resistance. In both cell lines, we compare the expression of the M2 receptor and the effects mediated by the M2 agonist APE on cell cycle, demonstrating a decreased percentage of cells in S phase and an accumulation of SK-N-BE cells in G1 phase, while the APE treatment of SK-N-BE(2C) cells induced a block in G2/M phase. The withdrawal of the M2 agonist from the medium shows that only the SK-N-BE(2C) cells are able to rescue cell proliferation. Further, we demonstrate that the co-treatment of low doses of APE with doxorubicin or cisplatin significantly counteracts cell proliferation when compared with the single treatment. Analysis of the expression of ATP-binding cassette (ABC) efflux pumps demonstrates the ability of the M2 agonist to downregulate their expression and that this negative modulation may be dependent on N-MYC decreased expression induced by the M2 agonist. Our data demonstrate that the combined effect of low doses of conventional drugs and the M2 agonist may represent a new promising therapeutic approach in neuroblastoma treatment, in light of its significant impact on drug resistance and the possible reduction in the side effects caused by high doses of chemotherapy drugs.
Collapse
Affiliation(s)
- Anna Maria Lucianò
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (A.M.L.); (E.P.)
| | - Elisa Perciballi
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (A.M.L.); (E.P.)
| | - Mario Fiore
- Institute of Molecular Biology and Pathology, CNR, 00185 Rome, Italy;
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Ada Maria Tata
- Department of Biology and Biotechnologies Charles Darwin, Sapienza University of Rome, 00185 Rome, Italy; (A.M.L.); (E.P.)
- Research Centre of Neurobiology Daniel Bovet, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
16
|
Zhong W, Zhang X, Zhao M, Wu J, Lin D. Advancements in nanotechnology for the diagnosis and treatment of multiple myeloma. Biomater Sci 2020; 8:4692-4711. [PMID: 32779645 DOI: 10.1039/d0bm00772b] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Multiple myeloma (MM), known as a tumor of plasma cells, is not only refractory but also has a high relapse rate, and is the second-most common hematologic tumor after lymphoma. It is often accompanied by multiple osteolytic damage, hypercalcemia, anemia, and renal insufficiency. In terms of diagnosis, conventional detection methods have many limitations, such as it is invasive and time-consuming and has low accuracy. Measures to change these limitations are urgently needed. At the therapeutic level, although the survival of MM continues to prolong with the advent of new drugs, MM remains incurable and has a high recurrence rate. With the development of nanotechnology, nanomedicine has become a powerful way to improve the current diagnosis and treatment of MM. In this review, the research progress and breakthroughs of nanomedicine in MM will be presented. Meanwhile, both superiorities and challenges of nanomedicine were discussed. As a new idea for the diagnosis and treatments of MM, nanomedicine will play a very important role in the research field of MM.
Collapse
Affiliation(s)
- Wenhao Zhong
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, P.R. China.
| | | | | | | | | |
Collapse
|
17
|
Bhatia R, Sharma A, Narang RK, Rawal RK. Recent Nanocarrier Approaches for Targeted Drug Delivery in Cancer Therapy. Curr Mol Pharmacol 2020; 14:350-366. [PMID: 32744982 DOI: 10.2174/1874467213666200730114943] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 01/16/2023]
Abstract
Cancer is one of the most serious health concerns in the 21st century whose prevalence is beyond boundaries and can affect any organ of the human body. The conventional chemotherapeutic treatment strategies lack specificity to tumors and are associated with toxic effects on the immune system and other organ systems. In the past decades, there has been continuous progress in the development of smart nanocarrier systems for target-specific delivery of drugs against a variety of tumors, including intracellular gene-specific targeting. These nanocarriers are able to recognize the tumor cells and deliver the therapeutic agent in fixed proportions, causing no or very less harm to healthy cells. Nanosystems have modified physicochemical properties, improved bioavailability, and long retention in blood, which enhances their potency. A huge number of nanocarrier based formulations have been developed and are in clinical trials. Nanocarrier systems include polymeric micelles, liposomes, dendrimers, carbon nanotubes, gold nanoparticles, etc. Recent advancements in nanocarrier systems include mesoporous silica nanoparticles (MSNs), metal organic frameworks, and quantum dots. In the present review, various nanocarrier based drug delivery systems, along with their applications in the management of cancer, have been described with special emphasis on MSNs.
Collapse
Affiliation(s)
- Rohit Bhatia
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Ferozepur G.T. Road, Moga-142 001, Punjab, India
| | - Amit Sharma
- Department of Pharmaceutics, ISF College of Pharmacy, Ferozepur G.T. Road, Moga-142 001, Punjab, India
| | - Raj K Narang
- Department of Pharmaceutics, ISF College of Pharmacy, Ferozepur G.T. Road, Moga-142 001, Punjab, India
| | - Ravindra K Rawal
- Department of Chemistry, Maharishi Markandeshwar (Deemed to be University), Mullana-133207, Haryana, India
| |
Collapse
|
18
|
Cancer Stem Cells in Soft-Tissue Sarcomas. Cells 2020; 9:cells9061449. [PMID: 32532153 PMCID: PMC7349510 DOI: 10.3390/cells9061449] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/06/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
Soft tissue sarcomas (STS) are a rare group of mesenchymal solid tumors with heterogeneous genetic profiles and clinical features. Systemic chemotherapy is the backbone treatment for advanced STS; however, STS frequently acquire resistance to standard therapies, which highlights the need to improve treatments and identify novel therapeutic targets. Increases in the knowledge of the molecular pathways that drive sarcomas have brought to light different molecular alterations that cause tumor initiation and progression. These findings have triggered a breakthrough of targeted therapies that are being assessed in clinical trials. Cancer stem cells (CSCs) exhibit mesenchymal stem cell (MSC) features and represent a subpopulation of tumor cells that play an important role in tumor progression, chemotherapy resistance, recurrence and metastasis. In fact, CSCs phenotypes have been identified in sarcomas, allied to drug resistance and tumorigenesis. Herein, we will review the published evidence of CSCs in STS, discussing the molecular characteristic of CSCs, the commonly used isolation techniques and the new possibilities of targeting CSCs as a way to improve STS treatment and consequently patient outcome.
Collapse
|
19
|
Zhang HH, Zhao LD, Zuo P, Yin BC, Ye BC. A telomerase-responsive nanoprobe with theranostic properties in tumor cells. Talanta 2020; 215:120898. [PMID: 32312443 DOI: 10.1016/j.talanta.2020.120898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/01/2020] [Accepted: 03/03/2020] [Indexed: 12/11/2022]
Abstract
Multidrug resistance (MDR) is the main cause of treatment failure in clinical cancer chemotherapy due to the presence of P-glycoproteins (P-gp), which widely exist in stubborn drug-resistant tumor membranes and actively pump drugs from inside the tumor cell to the outside. In this study, we report a novel telomerase-responsive nanoprobe with theranostic properties for inhibiting P-gp expression and reversing MDR by gene silencing. This nanoprobe is composed of an AuNP assembled with telomerase primer, antisense oligonucleotide (ASO), and doxorubicin (Dox). When the designed nanoprobe is uptaken by the MDR cancer cells, the Dox and ASO are specifically released due to the extension of telomerase primer triggered by telomerase. The released ASO specifically hybridizes with multidrug resistance 1 (MDR1) mRNA sequence, which encodes the P-gp. As a result, the expression of P-gp is inhibited and the efflux of Dox is prevented with reduced MDR in cancerous cells. The results demonstrate that the nanoprobe based on telomerase switching for drug release and gene silencing, can both target cancer cells for delivering drugs and overcome the effect of efflux pumps. This work presents a novel paradigm for theranostics of MDR cancer and enhances the efficacy of chemotherapeutics.
Collapse
Affiliation(s)
- He-Hua Zhang
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Li-Dong Zhao
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Peng Zuo
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Bin-Cheng Yin
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China; Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China; School of Chemistry and Chemical Engineering, Shihezi University, Shihezi, 832000, Xinjiang, China.
| | - Bang-Ce Ye
- Lab of Biosystem and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China; Institute of Engineering Biology and Health, Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, Zhejiang, China; School of Chemistry and Chemical Engineering, Shihezi University, Shihezi, 832000, Xinjiang, China.
| |
Collapse
|
20
|
Pinto V, Bergantim R, Caires HR, Seca H, Guimarães JE, Vasconcelos MH. Multiple Myeloma: Available Therapies and Causes of Drug Resistance. Cancers (Basel) 2020; 12:E407. [PMID: 32050631 PMCID: PMC7072128 DOI: 10.3390/cancers12020407] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 02/03/2020] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) is the second most common blood cancer. Treatments for MM include corticosteroids, alkylating agents, anthracyclines, proteasome inhibitors, immunomodulatory drugs, histone deacetylase inhibitors and monoclonal antibodies. Survival outcomes have improved substantially due to the introduction of many of these drugs allied with their rational use. Nonetheless, MM patients successively relapse after one or more treatment regimens or become refractory, mostly due to drug resistance. This review focuses on the main drugs used in MM treatment and on causes of drug resistance, including cytogenetic, genetic and epigenetic alterations, abnormal drug transport and metabolism, dysregulation of apoptosis, autophagy activation and other intracellular signaling pathways, the presence of cancer stem cells, and the tumor microenvironment. Furthermore, we highlight the areas that need to be further clarified in an attempt to identify novel therapeutic targets to counteract drug resistance in MM patients.
Collapse
Affiliation(s)
- Vanessa Pinto
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- FCTUC–Faculty of Science and Technology of the University of Coimbra, 3030-790 Coimbra, Portugal
| | - Rui Bergantim
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital São João, 4200-319 Porto, Portugal
- Clinical Hematology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Hugo R. Caires
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - Hugo Seca
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
| | - José E. Guimarães
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Clinical Hematology, Hospital São João, 4200-319 Porto, Portugal
- Clinical Hematology, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - M. Helena Vasconcelos
- i3S–Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal; (V.P.); (R.B.); (H.R.C.); (H.S.); (J.E.G.)
- Cancer Drug Resistance Group, IPATIMUP–Institute of Molecular Pathology and Immunology of the University of Porto, 4200-135 Porto, Portugal
- Department of Biological Sciences, FFUP-Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
21
|
Al-Malky HS, Osman AMM, Damanhouri ZA, Alkreathy HM, Al Aama JY, Ramadan WS, Al Qahtani AA, Al Mahdi HB. Modulation of doxorubicin-induced expression of the multidrug resistance gene in breast cancer cells by diltiazem and protection against cardiotoxicity in experimental animals. Cancer Cell Int 2019; 19:191. [PMID: 31367189 PMCID: PMC6657176 DOI: 10.1186/s12935-019-0912-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
Background Doxorubicin (DOX) is one of the most important anticancer agents used in treating breast cancer. However, chronic cardiotoxicity and multidrug resistance limit the chemotherapeutic use of DOX. Methods This study aimed to evaluate the capability of calcium channel blocker diltiazem (DIL) to reverse DOX resistance in breast cancer MCF-7 cells and to confer protection against DOX-induced cardiotoxicity in Wistar rats. For this purpose, we explored the effects of DOX on cell cycle phase distribution and expression of ABCB1, FOXO3a, and p53 genes in the presence and absence of DIL (20 μg/ml) and studied the ability of DIL to prevent DOX-induced cardiotoxicity after a single injection of DOX (15 mg/kg) in male Wister rats. Results We found that compared with DOX alone treatment, DIL + DOX treatment down regulated the ABCB1 gene expression by > fourfold but up regulated the FOXO3a and p53 genes expression by 1.5 fold. DIL treatment conferred protection against DOX-induced cardiotoxicity, as indicated by a decrease in the levels of the cardiac enzyme creatine kinase MB and malondialdehyde and an increase in the total antioxidant capacity and glutathione peroxidase levels. These biochemical results were further confirmed by the histopathological investigation of cardiac cells, which showed normal cardiac cells with central vesicular nuclei and prevention of DOX-induced disruption of normal cardiac architecture in the DIL to DOX group. Conclusions Taken together, our results indicate that DIL treatment can reverse the resistance of breast cancer cells to the therapeutic effects of DOX and can protect against DOX-induced cardiotoxicity in rats.
Collapse
Affiliation(s)
- Hamdan S Al-Malky
- Pharmacology Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia
| | - Abdel-Moneim M Osman
- Pharmacology Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia.,2Pharmacology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - Huda M Alkreathy
- Pharmacology Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia
| | - Jumana Y Al Aama
- Department of Genetic Medicine, Faculty of Medicine, KAU, Jeddah, Saudi Arabia.,Princess Aljawhara Center of Excellence in Research of Hereditary Disorders, KAU, Jeddah, Saudi Arabia
| | - Wafaa S Ramadan
- Anatomy Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia.,6Anatomy Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ali A Al Qahtani
- Pharmacology Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia
| | - Hadiah B Al Mahdi
- Department of Genetic Medicine, Faculty of Medicine, KAU, Jeddah, Saudi Arabia.,Princess Aljawhara Center of Excellence in Research of Hereditary Disorders, KAU, Jeddah, Saudi Arabia
| |
Collapse
|
22
|
Rajendrakumar SK, Venu A, Revuri V, George Thomas R, Thirunavukkarasu GK, Zhang J, Vijayan V, Choi SY, Lee JY, Lee YK, Jeong YY, Park IK. Hyaluronan-Stabilized Redox-Sensitive Nanoassembly for Chemo-Gene Therapy and Dual T1/T2 MR Imaging in Drug-Resistant Breast Cancer Cells. Mol Pharm 2019; 16:2226-2234. [PMID: 30924664 DOI: 10.1021/acs.molpharmaceut.9b00189] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tailoring combinatorial therapies along with real-time monitoring strategies has been the major focus of overcoming multidrug resistance in cancer. However, attempting to develop a multifunctional nanoplatform in a single construct leads to compromising therapeutic outcomes. Herein, we developed a simple, theranostic nanoassembly containing a hyaluronic acid-stabilized redox-sensitive (HART) polyethylenimine polyplex composed of a doxorubicin (DOX) intercalated Bcl-2 shRNA encoded plasmid along with a green-synthesized hausmannite (Mn3O4) and hematite (Fe3O4) nanoparticle (GMF). The highly stable HART nanoassembly has enhanced CD44-mediated intracellular uptake along with hyaluronidase (hylase) and redox-responsive drug-gene release. With Bcl-2 gene silencing induced by the successful delivery of HART in multidrug-resistant MCF7 breast cancer cells, the synergistic cytotoxic effect of Bcl-2 silencing and DOX was achieved. In addition, the HART nanoassembly containing GMF exhibited excellent dual MRI contrast (T1/T2) by reducing artifact signals. Overall, the HART nanoassembly with its enhanced theranostic properties has the potential to improve the therapeutic efficacy in future preclinical and clinical trials.
Collapse
Affiliation(s)
- Santhosh Kalash Rajendrakumar
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University , Chonnam National University Medical School , Gwangju 61469 , Republic of Korea
| | - Akhil Venu
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University , Chonnam National University Medical School , Gwangju 61469 , Republic of Korea
| | - Vishnu Revuri
- Department of Green Bio-Engineering , Korea National University of Transportation , Chungju 27469 , Republic of Korea
| | - Reju George Thomas
- Department of Radiology , Chonnam National University Hwasun Hospital , Hwasun , Jeollanam-Do 58128 , Republic of Korea
| | | | - Jun Zhang
- Department of Biomedical Sciences , Chonnam National University Medical School , Hwasun , Jeollanam-Do 58128 , Republic of Korea
| | - Veena Vijayan
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University , Chonnam National University Medical School , Gwangju 61469 , Republic of Korea
| | - Seok-Yong Choi
- Department of Biomedical Sciences , Chonnam National University Medical School , Hwasun , Jeollanam-Do 58128 , Republic of Korea
| | | | - Yong-Kyu Lee
- Department of Green Bio-Engineering , Korea National University of Transportation , Chungju 27469 , Republic of Korea
| | - Yong Yeon Jeong
- Department of Radiology , Chonnam National University Hwasun Hospital , Hwasun , Jeollanam-Do 58128 , Republic of Korea
| | - In-Kyu Park
- Department of Biomedical Science and BK21 PLUS Center for Creative Biomedical Scientists at Chonnam National University , Chonnam National University Medical School , Gwangju 61469 , Republic of Korea
| |
Collapse
|
23
|
Abstract
Background In clinical practice, many patients become multidrug resistant during chemotherapy, resulting in reduced or no healing effect. Therefore, the present study focused on bufalin, which is extracted from a traditional Chinese medicine named Chan Su (Venenum bufonis). We assessed the effect of bufalin in reversing K562/A02 cell drug resistance and inducing apoptosis, and explored the possible mechanism by which bufalin induces K562/A02 cell apoptosis. Material/Methods We used flow cytometry to evaluate intracellular ADM concentration, and RT-PCR and Western blot analysis were used to assess the effect of nuclear factor erythroid-2-related factor 2 (Nrf2) bufalin-related resistance gene expression. We used MTT and flow cytometry to detect apoptosis, and RT-PCR and Western blot were used to detect endoplasmic reticulum stress and apoptosis gene action. Results We found that bufalin can increase the concentration of Adriamycin (ADM) in K562/A02 cells by inhibiting the expression of Nrf2 and related drug resistance factors. The results showed that bufalin induced apoptosis of K562/A02 cells by the IRE1α/TRAF2/JNK/caspase-12 pathway. Conclusions These results suggest bufalin can reverse drug resistance in K562/A02 cells and that it induces apoptosis of K562/A02 cells by the IRE1α/TRAF2/JNK/caspase-12 pathway.
Collapse
Affiliation(s)
- Ying Xie
- Department of Blood Transfusion, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Xu Yan
- Department of Orthopedics, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| | - Ling Sun
- Department of Hematology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China (mainland)
| |
Collapse
|
24
|
Hossen S, Hossain MK, Basher M, Mia M, Rahman M, Uddin MJ. Smart nanocarrier-based drug delivery systems for cancer therapy and toxicity studies: A review. J Adv Res 2019; 15:1-18. [PMID: 30581608 PMCID: PMC6300464 DOI: 10.1016/j.jare.2018.06.005] [Citation(s) in RCA: 568] [Impact Index Per Article: 94.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 06/21/2018] [Accepted: 06/23/2018] [Indexed: 02/06/2023] Open
Abstract
Nonspecific distribution and uncontrollable release of drugs in conventional drug delivery systems (CDDSs) have led to the development of smart nanocarrier-based drug delivery systems, which are also known as Smart Drug Delivery Systems (SDDSs). SDDSs can deliver drugs to the target sites with reduced dosage frequency and in a spatially controlled manner to mitigate the side effects experienced in CDDSs. Chemotherapy is widely used to treat cancer, which is the second leading cause of death worldwide. Site-specific drug delivery led to a keen interest in the SDDSs as an alternative to chemotherapy. Smart nanocarriers, nanoparticles used to carry drugs, are at the focus of SDDSs. A smart drug delivery system consists of smart nanocarriers, targeting mechanisms, and stimulus techniques. This review highlights the recent development of SDDSs for a number of smart nanocarriers, including liposomes, micelles, dendrimers, meso-porous silica nanoparticles, gold nanoparticles, super paramagnetic iron-oxide nanoparticles, carbon nanotubes, and quantum dots. The nanocarriers are described in terms of their structures, classification, synthesis and degree of smartness. Even though SDDSs feature a number of advantages over chemotherapy, there are major concerns about the toxicity of smart nanocarriers; therefore, a substantial study on the toxicity and biocompatibility of the nanocarriers has been reported. Finally, the challenges and future research scope in the field of SDDSs are also presented. It is expected that this review will be widely useful for those who have been seeking new research directions in this field and for those who are about to start their studies in smart nanocarrier-based drug delivery.
Collapse
Affiliation(s)
- Sarwar Hossen
- Department of Physics, Khulna Govt. Mahila College, National University, Gazipur 1704, Bangladesh
| | - M. Khalid Hossain
- Institute of Electronics, Atomic Energy Research Establishment, Bangladesh Atomic Energy Commission, Dhaka 1349, Bangladesh
| | - M.K. Basher
- Institute of Electronics, Atomic Energy Research Establishment, Bangladesh Atomic Energy Commission, Dhaka 1349, Bangladesh
| | - M.N.H. Mia
- Institute of Electronics, Atomic Energy Research Establishment, Bangladesh Atomic Energy Commission, Dhaka 1349, Bangladesh
| | - M.T. Rahman
- Department of Materials Science and Engineering, University of Rajshahi, Rajshahi 6205, Bangladesh
| | - M. Jalal Uddin
- Department of Radio Sciences and Engineering, KwangWoon University, Seoul 01897, Republic of Korea
| |
Collapse
|
25
|
Histone deacetylase 2 regulates the doxorubicin (Dox) resistance of hepatocarcinoma cells and transcription of ABCB1. Life Sci 2018; 216:200-206. [PMID: 30465789 DOI: 10.1016/j.lfs.2018.11.043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 11/10/2018] [Accepted: 11/19/2018] [Indexed: 01/06/2023]
Abstract
Histone deacetylases (HDACs) can regulate cell-cycle, differentiation, and apoptosis of hepatocarcinoma (HCC) cells, while their roles in drug sensitivity remain unclear. Our results showed that the expression of HDAC2 was significantly increased in HCC doxorubicin (Dox) resistant cells as compared with their corresponding control cells. Over expression of HDAC2 can increase the cell viability and decrease the Dox sensitivity. Kaplan-Meier Plotter assay revealed that HCC patients with higher levels of HDAC2 had significantly poor prognosis than that of the lower expression patients. Mechanistically studies revealed that HDAC2 can regulate the transcription of ABCB1 via directly binding with its promoter and increasing its expression in Dox resistant HCC cells. Knockdown of HDAC2 significantly inhibited the expression of ABCB1. Co-immunoprecipitation revealed that HDAC2 can bind with c-fos, an important transcription factor of ABCB1, in HCC/Dox cells. Knockdown of c-Fos decreased the binding between HDAC2 and promoter of ABCB1 in HCC/Dox cells. Collectively, our data revealed that HDAC2 can regulate Dox sensitivity of HCC cells and the transcription of ABCB1.
Collapse
|
26
|
Xiao H, He J, Li X, Li B, Zhang L, Wang Y, Cheng D, Shuai X. Polymeric nanovesicles as simultaneous delivery platforms with doxorubicin conjugation and elacridar encapsulation for enhanced treatment of multidrug-resistant breast cancer. J Mater Chem B 2018; 6:7521-7529. [PMID: 32254754 DOI: 10.1039/c8tb01829d] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Multidrug resistance (MDR) is one of the major obstacles hindering the successful chemotherapy of cancer. Overexpression of drug efflux transporters such as P-glycoprotein (P-gp) is an important factor responsible for MDR. In this study, a novel copolymer methoxy-poly(ethylene glycol)-poly[(N-(6-hydroxyhexyl)-g-doxorubicin-l-aspartamide)-(β-benzyl-l-aspartate)] (mPEG-P[Asp(HPA-g-DOX)-BLA)] was synthesized and utilized to assemble into nanovesicles with hydrophilic P-gp inhibitor elacridar hydrochloride (Ela) encapsulated into the aqueous lumen. Doxorubicin (DOX) was covalently conjugated onto the polymer chain via a pH-cleavable amide linkage, leading to a pH responsive DOX release as well as disintegration of the nanovesicles in the lysosome of tumor cells. In vitro studies demonstrated that the DOX and Ela co-delivered nanovesicles showed superior cytotoxicity and enhanced anti-tumor properties as compared to single DOX-delivery nanosystems in MCF-7/ADR cancer, which was attributed to the P-gp bioactivity inhibition as investigated by a cell immunofluorescence assay. In vivo studies showed that the polymeric nanovesicles effectively accumulated at the tumor site and the co-delivered DOX and Ela effectively suppressed the MCF-7/ADR tumor growth. All the results indicated that the acid-liable nanovesicles had a synergistic effect to enhance antitumor efficacy for multidrug-resistant breast cancer treatment.
Collapse
Affiliation(s)
- Hong Xiao
- PCFM Lab of Ministry of Education, School of Materials Science and Engineering, Sun Yat-sen University, Guangzhou 510275, China.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Xiong H, Ni J, Jiang Z, Tian F, Zhou J, Yao J. Intracellular self-disassemble polysaccharide nanoassembly for multi-factors tumor drug resistance modulation of doxorubicin. Biomater Sci 2018; 6:2527-2540. [DOI: 10.1039/c8bm00570b] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Drug efflux induced by multidrug resistance (MDR) overexpression, as well as secondary drug resistance caused by subtoxic drug microenvironments as a result of inefficient drug release of nanoscopic drug carriers in tumor cells, are major bottlenecks for chemotherapy.
Collapse
Affiliation(s)
- Hui Xiong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Jiang Ni
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Zhijie Jiang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Fengchun Tian
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Jing Yao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Druggability of Biopharmaceuticals
- Department of Pharmaceutics
- China Pharmaceutical University
- Nanjing 210009
- China
| |
Collapse
|
28
|
de la Puente P, Azab AK. Nanoparticle delivery systems, general approaches, and their implementation in multiple myeloma. Eur J Haematol 2017; 98:529-541. [PMID: 28208215 DOI: 10.1111/ejh.12870] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2017] [Indexed: 12/25/2022]
Abstract
Multiple myeloma (MM) is a hematological malignancy that remains incurable, with relapse rates >90%. The main limiting factor for the effective use of chemotherapies in MM is the serious side effects caused by these drugs. The emphasis in cancer treatment has shifted from cytotoxic, non-specific chemotherapies to molecularly targeted and rationally designed therapies showing greater efficacy and fewer side effects. Traditional chemotherapy has shown several disadvantages such as lack of targeting capabilities, systemic toxicity, and side effects; low therapeutic index, as well as most anticancer drugs, has poor water solubility. Nanoparticle delivery systems (NPs) are capable of targeting large doses of chemotherapies into the target area while sparing healthy tissues, overcoming the limitations of traditional chemotherapy. Here, we review the current state of the art in nanoparticle-based strategies designed to treat MM. Many nanoparticle delivery systems have been studied for myeloma using non-targeted NPs (liposomes, polymeric NPs, and inorganic NPs), triggered NPs, as well as targeted NPs (VLA-4, ABC drug transporters, bone microenvironment targeting). The results in preclinical and clinical studies are promising; however, there remains much to be learned in the emerging field of nanomedicine in myeloma.
Collapse
Affiliation(s)
- Pilar de la Puente
- Cancer Biology Division, Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| | - Abdel Kareem Azab
- Cancer Biology Division, Department of Radiation Oncology, Washington University in Saint Louis School of Medicine, St. Louis, MO, USA
| |
Collapse
|
29
|
Pluronic Nanotechnology for Overcoming Drug Resistance. BIOACTIVITY OF ENGINEERED NANOPARTICLES 2017. [DOI: 10.1007/978-981-10-5864-6_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
30
|
Yang D, Wang T, Su Z, Xue L, Mo R, Zhang C. Reversing Cancer Multidrug Resistance in Xenograft Models via Orchestrating Multiple Actions of Functional Mesoporous Silica Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2016; 8:22431-22441. [PMID: 27420116 DOI: 10.1021/acsami.6b04885] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
A multistimuli responsive drug delivery system (DDS) based on sulfhydryl and amino-cofunctionalized mesoporous silica nanoparticles (SH/NH2-MSNs) has been developed, in which the multifunctional hyaluronic acid (HA) derivatives were grafted onto the SH/NH2-MSNs by disulfide bonds for targeting delivery, controlling drug release and reversing multidrug resistance (MDR). The doxorubicin (Dox) loaded multifunctional HA derivatives modified mesoporous silica nanoparticles (Dox/HHS-MSNs) were enzyme and redox sensitive, which could respond to the intracellular stimuli of hyaluronidase (HAase) and glutathione (GSH) successively and prevent drug leakage before reaching the tumor tissues. The cellular uptake experiments showed that Dox/HHS-MSNs were vulnerable to be endocytosed into the Dox-resistant human breast adenocarcinoma (MCF-7/ADR) cells, efficiently realized the endolysosomal escape and remained in the cytoplasm. Because of orchestrating multiple actions above including active targeting, endolysosomal escape and efficient multilevel drug release, Dox/HHS-MSNs could induce the strongest apoptosis and cytotoxicity of MCF-7/ADR cells. Furthermore, a series of in vivo studies on MCF-7/ADR tumor-bearing xenograft mouse models demonstrated that Dox/HHS-MSNs possessed the enhanced tumor-targeting capacity and the best therapeutic efficacy to reverse cancer MDR.
Collapse
Affiliation(s)
- Debin Yang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University , Nanjing 210009, China
| | - Tingfang Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University , Nanjing 210009, China
| | - Zhigui Su
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University , Nanjing 210009, China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University , Nanjing 210009, China
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University , Nanjing 210009, China
| | - Can Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University , Nanjing 210009, China
| |
Collapse
|
31
|
Yang C, Xiong F, Dou J, Xue J, Zhan X, Shi F, Li M, Wu S, Luo S, Zhang T, Zhang Y, Ming J, Gu N. Target therapy of multiple myeloma by PTX-NPs and ABCG2 antibody in a mouse xenograft model. Oncotarget 2016; 6:27714-24. [PMID: 26314844 PMCID: PMC4695020 DOI: 10.18632/oncotarget.4663] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/06/2015] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) remains to be an incurable disease. The purpose of this study was to evaluate the effect of ABCG2 monoclonal antibody (McAb) combined with paclitaxel (PTX) conjugated with Fe3O4 nanoparticles (NPs) on MM progressed from cancer stem cells (CSCs)in non-obese-diabetic/severe-combined-immunodeficiency (NOD/SCID) mouse model. Mice were injected with MM CSCs as marked by CD138−CD34− phenotypes through tail veins. The developed MM mice were examined by micro-computer tomography scanning, ultrasonography and enzyme-linked immunosorbent analysis. These mice were then intravenously treated with different combinations of NPs, PTX, McAb, PTX-NPs and melphalan/prednisone once a week for four weeks. The injected mice developed characteristic MM-associated syndromes, including lytic bone lesions, renal damages and proteinuria. All the treated mice showed decrease in bone lesions, renal damages and anemia but increase in apoptosis compared with the mice treated with NPs only. In particular, the treatment with ABCG2 McAb plus PTX-NPs induced the strongest therapeutic response and had an efficacy even better than that of melphalan/prednisone, a conventional regimen for MM patients. These data suggest that PTX-NPs with ABCG2 McAb can be developed into potential treatment regimens for patients with relapsed/refractory MM.
Collapse
Affiliation(s)
- Cuiping Yang
- Department of Pathogenic Biology and Immunology, School of Medicine & Collaborative Innovation Center of Suzhou NanoScience and Technology, Southeast University, Nanjing 210009, China
| | - Fei Xiong
- School of Biological Science & Medical Engineering & Collaborative Innovation Center of Suzhou NanoScience and Technology, Southeast University, Nanjing 210096, China
| | - Jun Dou
- Department of Pathogenic Biology and Immunology, School of Medicine & Collaborative Innovation Center of Suzhou NanoScience and Technology, Southeast University, Nanjing 210009, China
| | - Jun Xue
- Department of Hematology, Affiliated Nanjing First Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Xi Zhan
- The Center for Vascular and Inflammatory Diseases, Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Fangfang Shi
- Department of Oncology, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Miao Li
- Department of Pathogenic Biology and Immunology, School of Medicine & Collaborative Innovation Center of Suzhou NanoScience and Technology, Southeast University, Nanjing 210009, China
| | - Songyan Wu
- Department of Pathogenic Biology and Immunology, School of Medicine & Collaborative Innovation Center of Suzhou NanoScience and Technology, Southeast University, Nanjing 210009, China
| | - Shouhua Luo
- School of Biological Science & Medical Engineering & Collaborative Innovation Center of Suzhou NanoScience and Technology, Southeast University, Nanjing 210096, China
| | - Tianzhu Zhang
- School of Biological Science & Medical Engineering & Collaborative Innovation Center of Suzhou NanoScience and Technology, Southeast University, Nanjing 210096, China
| | - Yu Zhang
- School of Biological Science & Medical Engineering & Collaborative Innovation Center of Suzhou NanoScience and Technology, Southeast University, Nanjing 210096, China
| | - Ji Ming
- School of Biological Science & Medical Engineering & Collaborative Innovation Center of Suzhou NanoScience and Technology, Southeast University, Nanjing 210096, China
| | - Ning Gu
- School of Biological Science & Medical Engineering & Collaborative Innovation Center of Suzhou NanoScience and Technology, Southeast University, Nanjing 210096, China
| |
Collapse
|
32
|
Ye P, Xing H, Lou F, Wang K, Pan Q, Zhou X, Gong L, Li D. Histone deacetylase 2 regulates doxorubicin (Dox) sensitivity of colorectal cancer cells by targeting ABCB1 transcription. Cancer Chemother Pharmacol 2016; 77:613-21. [DOI: 10.1007/s00280-016-2979-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 01/26/2016] [Indexed: 02/07/2023]
|
33
|
Zhou XQ, Sun Q, Jiang L, Li ST, Gu W, Tian JL, Liu X, Yan SP. Synthesis, characterization, DNA/BSA interactions and anticancer activity of achiral and chiral copper complexes. Dalton Trans 2016; 44:9516-27. [PMID: 25919814 DOI: 10.1039/c5dt00931f] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Six novel copper(ii) complexes of [CuCl]ClO4 (), [Cu(acac)]PF6 (), [CuCl]2(PF6)2 (), [CuCl]2(PF6)2 (), [Cu(acac)]PF6 () and [Cu(acac)]PF6 (), ( = 1-naphthyl-N,N-[bis(2-pyridyl)methyl]amine, = R/S-1-naphthyl-N,N-[bis(2-pyridyl)methyl]ethanamine, acac = diacetone) were synthesized to serve as artificial nucleases. All complexes were structurally characterized using X-ray crystallography. The crystal structures showed the presence of distorted square-planar CuLCl (, and ) and distorted tetragonal-pyramidal CuL(acac) (, and ) geometry. The interaction of these complexes with calf thymus DNA (CT-DNA) was researched by means of several spectroscopy methods, which indicated that the complexes were bound to CT-DNA by an intercalation binding mode. DNA cleavage experiments revealed that the complexes exhibited remarkable DNA cleavage activities in the presence of H2O2, and single oxygen ((1)O2) or hydroxyl radicals may serve as the major cleavage active species. In particular, the in vitro cytotoxicity of the complexes on four human cancer cell lines (HeLa, MCF-7, Bel-7404 and HepG-2) demonstrated that the six compounds had broad-spectrum anti-cancer activity with low IC50 values. The stronger cytotoxicity and DNA cleavage activity of the chiral enantiomers compared with chiral analogues verified the influence of chirality on the antitumor activity of complexes. Meanwhile, the protein binding ability was revealed by quenching of tryptophan emission with the addition of complexes using BSA as a model protein. The results indicated that the quenching mechanism of BSA by the complexes was a static process.
Collapse
Affiliation(s)
- Xue-Quan Zhou
- College of Chemistry, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Nankai University, Tianjin 300071, China.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Cheng X, Qiu N, Yang J, Liu H, Wen J, Wang W, Wang Z, Chen L. Preparation, Characterization, and In Vivo Study of 7-Ethyl-14-Aminocamptothecin-Loaded Poly(Ethylene Glycol)2000-Poly(Lactic Acid)2000 Polymeric Micelles Against H460 Human Nonsmall Cell Lung Carcinoma. J Pharm Sci 2015; 104:3934-3942. [DOI: 10.1002/jps.24613] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Revised: 07/23/2015] [Accepted: 07/24/2015] [Indexed: 12/23/2022]
|
35
|
Walther W, Kobelt D, Bauer L, Aumann J, Stein U. Chemosensitization by diverging modulation by short-term and long-term TNF-α action on ABCB1 expression and NF-κB signaling in colon cancer. Int J Oncol 2015; 47:2276-85. [PMID: 26463665 DOI: 10.3892/ijo.2015.3189] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 08/19/2015] [Indexed: 11/05/2022] Open
Abstract
Multidrug resistance (MDR) is a major cause for cancer chemotherapy failure. Among the numerous strategies to overcome persistent action of proinflammatory cytokines, such as tumor necrosis factor α (TNF-α) permits downregulation of MDR-associated genes, including ATP-binding cassette, subfamily B 1 gene (ABCB1). A key regulator of ABCB1 expression is the transcription factor nuclear factor κ light chain enhancer (NF-κB)/p65. We analyzed diverging short- and long-term effects of TNF-α regarding modulation of NF-κB/p65 signaling and ABCB1 expression in colon cancer cells. Highly resistant ABCB1 overexpressing human HCT15 colorectal carcinoma cells were subjected to short- (30-120 min) or long-term (24-96 h) TNF-α treatment. TNF-α mediated modulation of ABCB1 expression was analyzed by real-time RT-PCR and western blot analysis. The TNF-mediated chemosensitization was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cytotoxicity assay. The involvement of TNF receptors and of NF-κB/p65 signaling was analyzed by western blot analysis, ABCB1 promoter analysis and electrophoretic mobility shift assay (EMSA). The study revealed, that long-term, but not short-term TNF-α treatment leads to TNF-receptor 1 (TNFR1) mediated downregulation of ABCB1 resulting in sensitization towards drug treatment. It dampens NF-κB/p65 activation and nuclear factor of κ light polypeptide gene enhancer in B-cells inhibitor α (IκBα) resynthesis, associated with reduced nuclear accumulation of NF-κB/p65 and reduced binding to its consensus sequence in the ABCB1 promoter. The study reveals the diverging effects of short- or long-term TNF-α action and provides novel insights on downregulation of ABCB1 expression by TNF-mediated repression of NF-κB signaling.
Collapse
Affiliation(s)
- Wolfgang Walther
- Experimental and Clinical Research Center, Charité, University Medicine Berlin and the Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| | - Dennis Kobelt
- Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| | - Lisa Bauer
- Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| | - Jutta Aumann
- Experimental and Clinical Research Center, Charité, University Medicine Berlin and the Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| | - Ulrike Stein
- Experimental and Clinical Research Center, Charité, University Medicine Berlin and the Max-Delbrück-Center for Molecular Medicine, D-13125 Berlin, Germany
| |
Collapse
|
36
|
Assanhou AG, Li W, Zhang L, Xue L, Kong L, Sun H, Mo R, Zhang C. Reversal of multidrug resistance by co-delivery of paclitaxel and lonidamine using a TPGS and hyaluronic acid dual-functionalized liposome for cancer treatment. Biomaterials 2015; 73:284-95. [PMID: 26426537 DOI: 10.1016/j.biomaterials.2015.09.022] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 09/11/2015] [Accepted: 09/14/2015] [Indexed: 12/13/2022]
Abstract
Multidrug resistance (MDR) remains the primary issue in cancer therapy, which is characterized by the overexpressed P-glycoprotein (P-gp)-included efflux pump or the upregulated anti-apoptotic proteins. In this study, a D-alpha-tocopheryl poly (ethylene glycol 1000) succinate (TPGS) and hyaluronic acid (HA) dual-functionalized cationic liposome containing a synthetic cationic lipid, 1,5-dioctadecyl-N-histidyl-L-glutamate (HG2C18) was developed for co-delivery of a small-molecule chemotherapeutic drug, paclitaxel (PTX) with a chemosensitizing agent, lonidamine (LND) to treat the MDR cancer. It was demonstrated that the HG2C18 lipid contributes to the endo-lysosomal escape of the liposome following internalization for efficient intracellular delivery. The TPGS component was confirmed able to elevate the intracellular accumulation of PTX by inhibiting the P-gp efflux, and to facilitate the mitochondrial-targeting of the liposome. The intracellularly released LND suppressed the intracellular ATP production by interfering with the mitochondrial function for enhanced P-gp inhibition, and additionally, sensitized the MDR breast cancer (MCF-7/MDR) cells to PTX for promoted induction of apoptosis through a synergistic effect. Functionalized with the outer HA shell, the liposome preferentially accumulated at the tumor site and showed a superior antitumor efficacy in the xenograft MCF-7/MDR tumor mice models. These findings suggest that this dual-functional liposome for co-delivery of a cytotoxic drug and an MDR modulator provides a promising strategy for reversal of MDR in cancer treatment.
Collapse
Affiliation(s)
- Assogba G Assanhou
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China; UFR Pharmacie, Faculté des Sciences de la Santé, Université d'Abomey-Calavi, 01 BP 188 Cotonou, Benin; Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Wenyuan Li
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, VIC 3052, Australia
| | - Lei Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Hongbin Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China
| | - Ran Mo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China.
| | - Can Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Drug Discovery, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
37
|
Zhang Z, Shao Z, Xiong L, Yang S. Inhibition of autophagy enhances cisplatin-induced apoptosis in the MG63 human osteosarcoma cell line. Oncol Lett 2015; 10:2941-2946. [PMID: 26722268 DOI: 10.3892/ol.2015.3692] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 08/17/2015] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a significant catabolic process that allows the renewal of intracellular organelles, through which cells are able to maintain homeostasis. In addition, autophagy may be associated with the carcinogenesis of osteosarcoma (OS). Cisplatin (CDDP) is an alkylating agent that is commonly used as an anticancer therapy. However, the pathways underlying the effects of CDDP remain to be elucidated. The present study demonstrated that 3-methyladenine (3-MA), an inhibitor of autophagy, was able to increase the proliferation inhibition ratios of MG63 human OS cells when used in combination with CDDP. Furthermore, MG63 cells produced significantly more microtubule-associated protein light chain 3II (LC3II), a widely used marker for monitoring autophagy, following CDDP treatment. Treatment with 3-MA was observed to inhibit these changes. Similarly, MG63 cells co-treated with 3-MA and CDDP demonstrated increased sensitivity to CDDP-induced apoptosis, compared with those exposed to CDDP alone. The present study revealed variation in the expression of LC3II and caspase-3 activity following treatment with certain drugs. The results of the present study suggest that CDDP may be capable of inducing apoptosis and autophagy, and that autophagy may be able to inhibit apoptosis in MG63 cells. Therefore, downregulation of autophagy may increase the chemotherapeutic sensitivity of MG63 cells to CDDP.
Collapse
Affiliation(s)
- Zhicai Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Zengwu Shao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Liming Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Shuhua Yang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
38
|
Mao D, Che J, Han S, Zhao H, Zhu Y, Zhu H. RNAi-mediated knockdown of the CLN3 gene inhibits proliferation and promotes apoptosis in drug-resistant ovarian cancer cells. Mol Med Rep 2015; 12:6635-41. [PMID: 26299671 PMCID: PMC4626189 DOI: 10.3892/mmr.2015.4238] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 06/22/2015] [Indexed: 01/15/2023] Open
Abstract
CLN3 is a recently identified anti-apoptotic gene, which has been demonstrated to be highly expressed in a diverse range of cancer cell lines, including ovarian cancer. In the present study, RNA interference, mediated by a lentivirus expressing CLN3 short hairpin RNA (shRNA) was utilized to knockdown the expression of CLN3 in the A2780 human ovarian cancer cell line, and its cisplatin-resistant and carboplatin-resistant sublines, A2780/DDP and A2780/CBP cells. It was revealed that the mRNA and protein expression levels of CLN3 were significantly reduced in the CLN3-specific shRNA-transduced cells, compared with the untransduced and control shRNA-transduced cells. In addition, specific knockdown of CLN3 in these cells inhibited cell proliferation and led to cell cycle arrest at the G0/G1 phase, with eventual apoptosis. CLN3 knockdown caused increases in the levels of Bax, FAX, cleaved-caspase 3, cleaved-caspase 8 and cleaved-RARP, but decreased the level of Bcl-2. Finally, it was observed that CLN3 depletion markedly reduced the half maximum inhibitory concentration in the A2780/DDP and A2780/CBP cells. Taken together, these data suggested that CLN3 is involved in tumorigenesis and drug resistance in ovarian cancer, and may serve as a promising therapeutic target for its treatment.
Collapse
Affiliation(s)
- Dongwei Mao
- Department of Gynaecology and Obstetrics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Jianhua Che
- Department of Gynaecology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150037, P.R. China
| | - Shiyu Han
- Department of Gynaecology and Obstetrics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Honghui Zhao
- Department of Gynaecology and Obstetrics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Yumei Zhu
- Department of Gynaecology and Obstetrics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hong Zhu
- Department of Gynaecology and Obstetrics, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
39
|
Cao Z, Zhu W, Wang W, Zhang C, Xu M, Liu J, Feng ST, Jiang Q, Xie X. Stable cerasomes for simultaneous drug delivery and magnetic resonance imaging. Int J Nanomedicine 2014; 9:5103-16. [PMID: 25395848 PMCID: PMC4227624 DOI: 10.2147/ijn.s66919] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Magnetic liposomes have been frequently used as nanocarriers for targeted drug delivery and magnetic resonance imaging in recent years. Despite great potentials, their morphological/structural instability in the physiological environment still remains an intractable challenge for clinical applications. In this study, stable hybrid liposomal cerasomes (ie, liposomes partially coated with silica) which can co-encapsulate Fe3O4 nanoparticles and the anticancer drug paclitaxel were developed using thin film hydration method. Compared with the drug loaded liposomes, the paclitaxel-loaded magnetic cerasomes (PLMCs) exhibited much higher storage stability and better sustained release behavior. Cellular uptake study showed that the utilization of an external magnetic field significantly facilitated the internalization of PLMCs into cancer cells, resulting in potentiated drug efficacy of killing tumor cells. The T2 relaxivity (r2) of our PLMCs was much higher than that of free Fe3O4 nanoparticles, suggesting increased sensitivity in T2-weighted imaging. Given its excellent biocompatibility also shown in the study, such dual functional PLMC is potentially a promising nanosystem for effective cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Zhong Cao
- Department of Biomedical Engineering, College of Engineering, Guangzhou, People's Republic of China
| | - Wenjian Zhu
- Department of Biomedical Engineering, College of Engineering, Guangzhou, People's Republic of China
| | - Wei Wang
- Department of Medical Ultrasonics, The First Affiliated Hospital of Sun Yat-sen University, Institute of Diagnostic and Interventional Ultrasound, Guangzhou, People's Republic of China
| | - Chunyang Zhang
- Department of Biomedical Engineering, College of Engineering, Guangzhou, People's Republic of China
| | - Ming Xu
- Department of Medical Ultrasonics, The First Affiliated Hospital of Sun Yat-sen University, Institute of Diagnostic and Interventional Ultrasound, Guangzhou, People's Republic of China
| | - Jie Liu
- Department of Biomedical Engineering, College of Engineering, Guangzhou, People's Republic of China
| | - Shi-Ting Feng
- Department of Radiology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Qing Jiang
- Department of Biomedical Engineering, College of Engineering, Guangzhou, People's Republic of China
| | - Xiaoyan Xie
- Department of Medical Ultrasonics, The First Affiliated Hospital of Sun Yat-sen University, Institute of Diagnostic and Interventional Ultrasound, Guangzhou, People's Republic of China
| |
Collapse
|
40
|
Neumann W, Crews BC, Sárosi MB, Daniel CM, Ghebreselasie K, Scholz MS, Marnett LJ, Hey-Hawkins E. Conjugation of cisplatin analogues and cyclooxygenase inhibitors to overcome cisplatin resistance. ChemMedChem 2014; 10:183-92. [PMID: 25318459 DOI: 10.1002/cmdc.201402353] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Indexed: 12/17/2022]
Abstract
Cyclooxygenase (COX) is an enzyme involved in tumorigenesis and is associated with tumor cell resistance against platinum-based antitumor drugs. Cisplatin analogues were conjugated with COX inhibitors (indomethacin, ibuprofen) to study the synergistic effects that were previously observed in combination treatments. The conjugates ensure concerted transport of both drugs into cells, and subsequent intracellular cleavage enables a dual-action mode. Whereas the platinum(II) complexes showed cytotoxicities similar to those of cisplatin, the platinum(IV) conjugates revealed highly increased cytotoxic activities and were able to completely overcome cisplatin-related resistance. Although some of the complexes are potent COX inhibitors, the conjugates appear to execute their cytotoxic action via COX-independent mechanisms. Instead, the increased lipophilicity and kinetic inertness of the conjugates seem to facilitate cellular accumulation of the platinum drugs and thus improve the efficacy of the antitumor agents. These conjugates are important tools for the elucidation of the direct influence of COX inhibitors on platinum-based anticancer drugs in tumor cells.
Collapse
Affiliation(s)
- Wilma Neumann
- Institut für Anorganische Chemie, Universität Leipzig, Johannisallee 29, 04103 Leipzig (Germany)
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Hamilton G, Rath B. A short update on cancer chemoresistance. Wien Med Wochenschr 2014; 164:456-60. [PMID: 25249024 DOI: 10.1007/s10354-014-0311-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/28/2014] [Indexed: 12/16/2022]
Abstract
Chemotherapeutic interventions in cancer patients are limited by the appearance of chemoresistance. For instance, advanced lung and ovarian cancer patients relapse invariably after few cycles of platinum-based chemotherapy. Disseminated tumors are characterized by genetic instability/heterogeneity, thus containing or generating a repertoire of resistant subpopulations. At the cellular level, altered drug uptake, efflux, and metabolization, as well as modifications of drug targets, increased repair, and decreased cell death complement the limited perfusion and adverse hypoxic/acidic extracellular conditions at the tumor level in retaining cancer cell viability. Similarly, targeted therapy is rendered ineffective by mutations of the specific target protein within a few months or years of administration. Assessment of the expression profiles of resistant tumor cells revealed extensive changes in numerous pathways affecting hundreds of genes. Therefore, reversal of drug resistance will require individual profiles of drug resistance mediators and the combination of several specific drugs, targeting critical components to provide new therapeutic options.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Ludwig Boltzmann Cluster of Translational Oncology, c/o Balderichgasse 26/13, 11170, Vienna, Austria,
| | | |
Collapse
|
42
|
CAO BO, CHEN HONG, GAO YING, NIU CONG, ZHANG YUAN, LI LING. CIP-36, a novel topoisomerase II-targeting agent, induces the apoptosis of multidrug-resistant cancer cells in vitro. Int J Mol Med 2014; 35:771-6. [DOI: 10.3892/ijmm.2015.2068] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 01/08/2015] [Indexed: 11/05/2022] Open
|
43
|
Yang C, Xiong F, Wang J, Dou J, Chen J, Chen D, Zhang Y, Luo S, Gu N. Anti-ABCG2 monoclonal antibody in combination with paclitaxel nanoparticles against cancer stem-like cell activity in multiple myeloma. Nanomedicine (Lond) 2014; 9:45-60. [DOI: 10.2217/nnm.12.216] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Aim: To investigate the effects of anti-ABCG2 monoclonal antibodies (mAbs) in combination with paclitaxel iron oxide nanoparticles (PTX-NPs) on CD138-CD34- multiple myeloma (MM) cancer stem cells (CSCs) in JJN3 cells. Materials & methods: PTX-NPs were prepared using the hydrophobic interaction of the polyoxypropylene chain and oleic acid on the surface of iron oxide NPs and were targeted to the ABCG2 transporter overexpressing MM CSCs with mAbs. Results: The data showed that MM CSCs have strong drug resistance and tumorigenicity compared with non-MM CSCs. PTX-NPs combined with mAbs led to a significant reduction in the tumor volume, a visible alleviation of lytic bone lesions and a markedly increased survival rate in contrast to using a single agent in MM CSCs when it was transplanted to nonobese diabetic/severe combined immunodeficiency mice. Conclusion: This study is the first to report on the anti-MM CSC activity by PTX-NPs as a single agent or used together with anti-ABCG2 mAbs to treat MM. These findings provide a rationale for future clinical trials. Original submitted 18 June 2012; Revised submitted 29 November 2012; Published online 27 March 2013
Collapse
Affiliation(s)
- Cuiping Yang
- Department of Pathogenic Biology & Immunology, Medical School, Southeast University, Nanjing 210009, China
| | - Fei Xiong
- Jiangsu Laboratory for Biomaterials & Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jing Wang
- Department of Gynecology & Obstetrics, Zhongda Hospital, Southeast University, Nanjing 210009, China
| | - Jun Dou
- Department of Pathogenic Biology & Immunology, Medical School, Southeast University, Nanjing 210009, China
| | - Junsong Chen
- Department of Pathogenic Biology & Immunology, Medical School, Southeast University, Nanjing 210009, China
| | - Dengyu Chen
- Department of Pathogenic Biology & Immunology, Medical School, Southeast University, Nanjing 210009, China
| | - Yu Zhang
- Jiangsu Laboratory for Biomaterials & Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Shouhua Luo
- Jiangsu Laboratory for Biomaterials & Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Ning Gu
- Jiangsu Laboratory for Biomaterials & Devices, School of Biological Science & Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
44
|
Cao Z, Yue X, Li X, Dai Z. Stabilized magnetic cerasomes for drug delivery. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2013; 29:14976-83. [PMID: 24188471 DOI: 10.1021/la401965a] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Doxorubicin hydrochloride (DOX)-loaded magnetic cerasomes (DLMCs) were successfully constructed by loading both hydrophobic Fe3O4 nanoparticles (NPs) and antitumor drug DOX into the aqueous interior of cerasomes via facile one-step construction. A possible explanation is that the hydrophobic Fe3O4 NPs can be trapped inside the aqueous core of cerasomes through the formation of an intermediate Fe3O4/micelle complex. It was found that the loading content of Fe3O4 in DLMCs could reach the maximum at a Fe3O4/lipid molar ratio of 4:1. Moreover, DLMCs demonstrated high superparamagnetism and responded strongly to magnetic fields. In addition, DLMCs had a high encapsulation efficiency of 43.4 ± 4.7% and a high drug loading content of 3.2 ± 1.3%. In comparison to drug-loaded liposomes, DLMCs exhibited higher storage stability and better sustained release behavior. A cellular uptake study showed that the use of an external magnetic field enables a rapid and efficient uptake of DLMCs by cancer cells, resulting in higher capability to kill tumor cells than non-magnetic drug-loaded cerasomes. This study suggests that magnetic cerasome offers a potential and effective drug carrier for anticancer applications.
Collapse
Affiliation(s)
- Zhong Cao
- Department of Biomedical Engineering, College of Engineering, Peking University , Beijing 100871, People's Republic of China
| | | | | | | |
Collapse
|
45
|
Induction of apoptosis through caspase-independent or caspase-9-dependent pathway in mouse and human osteosarcoma cells by a new nitroxyl spin-labeled derivative of podophyllotoxin. Apoptosis 2013; 18:727-38. [PMID: 23430060 DOI: 10.1007/s10495-013-0819-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Previous study has found that a new nitroxyl spin-labeled derivative of podophyllotoxin, 4-[4"-(2",2",6",6"-tetramethyl-1"-piperidinyloxy)amino]-4'-demethyl-epipodophyllotoxin (GP7), can induce apoptosis in human leukemia cells. However, there have been no studies about the effects of GP7 on osteosarcoma (OS) cells. Here, we observed the anti-OS effects of GP7 in mouse and human OS cells with the comparison of etoposide. GP7 and etoposide inhibited the proliferation of a panel of mouse and human OS cells in a concentration- or time-dependent manner, and the inhibitory effect of GP7 on the proliferation of mouse LM8 or human U2OS cells was 1.28- or 1.35-fold higher than that of etoposide. GP7 or etoposide augmented the anti-OS effects of methotrexate, adriamycin, cisplatin, or their combination, and the combined inhibitory effects of GP7 with MTX on the proliferation of LM8 cells was higher than those of etoposide with MTX. GP7 arrested the cell cycle in S phase but etoposide in G(2)/M phase. GP7 or etoposide induced sub-G(1) peak, apoptotic DNA fragmentation, activations of caspase-3, -8, -9, and DNA fragmentation factor, downregulation of Bcl-2 and Bcl-xL, upregulation of Bax and Bak, and cytochrome-c release from mitochondria in both mouse and human OS cells. GP7 or etoposide also induced endonuclease G translocation from mitochondria into cytosol in mouse cells. GP7- or etoposide-induced apoptotic DNA fragmentation of human OS cells was inhibited by the pan caspase inhibitor and caspase-9 inhibitor, not by caspase-8 inhibitor whereas it was not inhibited by the pan caspase inhibitor in mouse OS cells. Our findings indicate that GP7 is effective against mouse and human OS cells in vitro. The apoptotic DNA fragmentation in mouse OS cells may be mediated by caspase-independent pathway with the involvement of endonuclease G whereas in human OS cells by caspase-9-dependent pathway downstream of the cytochrome-c-initiated caspase cascade.
Collapse
|
46
|
Pathophysiological roles of aldo-keto reductases (AKR1C1 and AKR1C3) in development of cisplatin resistance in human colon cancers. Chem Biol Interact 2012; 202:234-42. [PMID: 23165153 DOI: 10.1016/j.cbi.2012.09.024] [Citation(s) in RCA: 84] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 09/25/2012] [Accepted: 09/26/2012] [Indexed: 01/06/2023]
Abstract
Cisplatin (cis-diamminedichloroplatinum, CDDP) is widely used for treatment of patients with solid tumors formed in various organs including the lung, prostate and cervix, but is much less sensitive in colon and breast cancers. One major factor implicated in the ineffectiveness has been suggested to be acquisition of the CDDP resistance. Here, we established the CDDP-resistant phenotypes of human colon HCT15 cells by continuously exposing them to incremental concentrations of the drug, and monitored expressions of aldo-keto reductases (AKRs) 1A1, 1B1, 1B10, 1C1, 1C2 and 1C3. Among the six AKRs, AKR1C1 and AKR1C3 are highly induced with the CDDP resistance. The resistance lowered the sensitivity toward cellular damages evoked by oxidative stress-derived aldehydes, 4-hydroxy-2-nonenal and 4-oxo-2-nonenal that are detoxified by AKR1C1 and AKR1C3. Overexpression of AKR1C1 or AKR1C3 in the parental HCT15 cells mitigated the cytotoxicity of the aldehydes and CDDP. Knockdown of both AKR1C1 and AKR1C3 in the resistant cells or treatment of the cells with specific inhibitors of the AKRs increased the sensitivity to CDDP toxicity. Thus, the two AKRs participate in the mechanism underlying the CDDP resistance probably via detoxification of the aldehydes resulting from enhanced oxidative stress. The resistant cells also showed an enhancement in proteolytic activity of proteasome accompanied by overexpression of its catalytic subunits (PSMβ9 and PSMβ10). Pretreatment of the resistant cells with a potent proteasome inhibitor Z-Leu-Leu-Leu-al augmented the CDDP sensitization elicited by the AKR inhibitors. Additionally, the treatment of the cells with Z-Leu-Leu-Leu-al and the AKR inhibitors induced the expressions of the two AKRs and proteasome subunits. Collectively, these results suggest the involvement of up-regulated AKR1C1, AKR1C3 and proteasome in CDDP resistance of colon cancers and support a chemotherapeutic role for their inhibitors.
Collapse
|
47
|
Tseng LM, Liu CY, Chang KC, Chu PY, Shiau CW, Chen KF. CIP2A is a target of bortezomib in human triple negative breast cancer cells. Breast Cancer Res 2012; 14:R68. [PMID: 22537901 PMCID: PMC3446403 DOI: 10.1186/bcr3175] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Revised: 04/16/2012] [Accepted: 04/26/2012] [Indexed: 11/10/2022] Open
Abstract
Introduction Triple negative breast cancer (TNBC) is very aggressive and currently has no specific therapeutic targets, such as hormone receptors or human epidermal growth factor receptor type 2 (HER2); therefore, prognosis is poor. Bortezomib, a proteasome inhibitor, may exert efficacy in TNBC through its multiple cellular effects. Here, we tested the efficacy of bortezomib and examined the drug mechanism in breast cancer cells. Methods Five breast cancer cell lines: TNBC HCC-1937, MDA-MB-231, and MDA-MB-468; HER2-overexpressing MDA-MB-453; and estrogen receptor positive MCF-7 were used for in vitro studies. Apoptosis was examined by both flow cytometry and Western Blot. Signal transduction pathways in cells were assessed by Western Blot. Gene silencing was done by small interfering RNA (siRNA). In vivo efficacy of bortezomib was tested in nude mice with breast cancer xenografts. Immunohistochemical study was performed on tumor tissues from patients with TNBC. Results Bortezomib induced significant apoptosis, which was independent of its proteasome inhibition, in the three TNBC cell lines, but not in MDA-MB-453 or MCF-7 cells. Furthermore, cancerous inhibitor of protein phosphatase 2A (CIP2A), a cellular inhibitor of protein phosphatase 2A (PP2A), mediated the apoptotic effect of bortezomib. We showed that bortezomib inhibited CIP2A in association with p-Akt downregulation in a dose- and time-dependent manner in all sensitive TNBC cells, whereas no alterations in CIP2A expression and p-Akt were noted in bortezomib-resistant cells. Overexpression of CIP2A upregulated p-Akt and protected MDA-MB-231 and MDA-MB-468 cells from bortezomib-induced apoptosis, whereas silencing CIP2A by siRNA overcame the resistance to bortezomib-induced apoptosis in MCF-7 cells. In addition, bortezomib downregulated CIP2A mRNA but did not affect the degradation of CIP2A protein. Furthermore, bortezomib exerted in vivo antitumor activity in HCC-1937 xenografted tumors, but not in MCF-7 tumors. Bortezomib downregulated CIP2A expression in the HCC-1937 tumors but not in the MCF-7 tumors. Importantly, CIP2A expression is readily detectable in tumor samples from TNBC patients. Conclusions CIP2A is a major determinant mediating bortezomib-induced apoptosis in TNBC cells. CIP2A may thus be a potential therapeutic target in TNBC.
Collapse
Affiliation(s)
- Ling-Ming Tseng
- Department of Surgery, Taipei Veterans General Hospital, No. 201 Sec. 2 Shih-Pai Road, Taipei 112, Taiwan
| | | | | | | | | | | |
Collapse
|
48
|
Wink M, Ashour ML, El-Readi MZ. Secondary Metabolites from Plants Inhibiting ABC Transporters and Reversing Resistance of Cancer Cells and Microbes to Cytotoxic and Antimicrobial Agents. Front Microbiol 2012; 3:130. [PMID: 22536197 PMCID: PMC3332394 DOI: 10.3389/fmicb.2012.00130] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 03/19/2012] [Indexed: 12/14/2022] Open
Abstract
Fungal, bacterial, and cancer cells can develop resistance against antifungal, antibacterial, or anticancer agents. Mechanisms of resistance are complex and often multifactorial. Mechanisms include: (1) Activation of ATP-binding cassette (ABC) transporters, such as P-gp, which pump out lipophilic compounds that have entered a cell, (2) Activation of cytochrome p450 oxidases which can oxidize lipophilic agents to make them more hydrophilic and accessible for conjugation reaction with glucuronic acid, sulfate, or amino acids, and (3) Activation of glutathione transferase, which can conjugate xenobiotics. This review summarizes the evidence that secondary metabolites (SM) of plants, such as alkaloids, phenolics, and terpenoids can interfere with ABC transporters in cancer cells, parasites, bacteria, and fungi. Among the active natural products several lipophilic terpenoids [monoterpenes, diterpenes, triterpenes (including saponins), steroids (including cardiac glycosides), and tetraterpenes] but also some alkaloids (isoquinoline, protoberberine, quinoline, indole, monoterpene indole, and steroidal alkaloids) function probably as competitive inhibitors of P-gp, multiple resistance-associated protein 1, and Breast cancer resistance protein in cancer cells, or efflux pumps in bacteria (NorA) and fungi. More polar phenolics (phenolic acids, flavonoids, catechins, chalcones, xanthones, stilbenes, anthocyanins, tannins, anthraquinones, and naphthoquinones) directly inhibit proteins forming several hydrogen and ionic bonds and thus disturbing the 3D structure of the transporters. The natural products may be interesting in medicine or agriculture as they can enhance the activity of active chemotherapeutics or pesticides or even reverse multidrug resistance, at least partially, of adapted and resistant cells. If these SM are applied in combination with a cytotoxic or antimicrobial agent, they may reverse resistance in a synergistic fashion.
Collapse
Affiliation(s)
- Michael Wink
- Institute of Pharmacy and Molecular Biotechnology, Heidelberg University Heidelberg, Germany
| | | | | |
Collapse
|
49
|
Sun L, Morris LM, Luo J, Mackey LV, Leslie JS, Franko-Tobin LG, Fuselier JA, LePage KT, Coy DH. Application of human pancreatic carcinoid BON cells for receptor-targeted drug development. J Drug Target 2010; 19:666-74. [PMID: 21083509 DOI: 10.3109/1061186x.2010.531728] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
In our previous study, we found that several tumor cell lines displayed high receptor-specific binding affinity, one of which, the human pancreatic carcinoid BON cell line, demonstrates high affinity binding of the bombesin (BN) and somatostatin (SST) receptor-specific ligands. In the present study, BON cells, as a representative model, were further applied to evaluate various peptide analogs and cytotoxic receptor-targeted peptide conjugates. We observed quick ligand-receptor internalization in BON cells as well as high binding affinity. Furthermore, BON cells have high expression of multidrug resistance-associated genes (MDR1) and show camptothecin (CPT) resistance. Various receptor-specific cytotoxic conjugates were synthesized and evaluated in the BON cell model via in vitro and in vivo studies. We found that all the tested conjugates displayed potent antitumor ability in xenografts. Especially, the CPT conjugates, CPT-SST, and CPT-BN, are most likely to increase sensitivity to CPT-resistant BON cells. Our findings suggest that appropriately defined tumor cell lines may provide physiologically relevant cell-based evaluations of novel peptide analogs and receptor-targeted chemotherapeutics.
Collapse
Affiliation(s)
- Lichun Sun
- Department of Medicine, Peptide Research Laboratories, Tulane Health Sciences Center, New Orleans, LA 70112-2699, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Syl611, a novel semisynthetic taxane derivative, reverses multidrug resistance by p-glycoprotein inhibition and facilitating inward transmembrane action. Cancer Chemother Pharmacol 2010; 66:851-9. [PMID: 20052473 DOI: 10.1007/s00280-009-1229-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2009] [Accepted: 12/22/2009] [Indexed: 10/20/2022]
Abstract
PURPOSE To investigate the reversal mechanisms of a novel semisynthetic taxane derivative, Syl611. Syl611 is a structurally modified compound from Sinenxan A, and the chemical structure is entirely new. It was found to significantly increase paclitaxel-induced cytotoxicity in drug-resistant cells, while presenting a low level of cytotoxicity. METHODS The in vitro cytotoxic and MDR-reversing activities of the Syl611 were determined by MTT assays. The cytotoxicity enhancement of paclitaxel was performed using the acridine orange/ethidium bromide double staining. Rhodamine 123 accumulation and retention assay in KB/V cells, Caco-2 monolayer model were used to find mechanism of action. RESULTS The cytotoxicity of Syl611 was wondrously lower in all tested cell lines than that of paclitaxel. Cytotoxicity enhancement from Syl611 was dramatically higher than that of verapamil of the same concentration (10 muM): the reversal fold index for A549/Paclitaxel, KB/V, and Bel7402/5-FU were 45.95, 73.56, and 107.13 (Syl611) and 11.36, 23.92, and 70.42 (verapamil). AO/EB double staining assay equally showed that Syl611 could enhance the cytotoxicity induced by paclitaxel. Furthermore, Syl611 could also increase the intracellular accumulation of Rhodamine 123 in KB/V cells without affecting P-gp's expression, and this accumulation was reversible. In bidirectional permeability assay, Syl611 increased the permeability of paclitaxel but decreased the net secretory of paclitaxel. CONCLUSIONS Syl611 is an effective and potential agent in reversing multidrug resistance (MDR) by multiple actions, which attributed to p-glycoprotein inhibition and drug permeability enhancement.
Collapse
|