1
|
Sivasankaran LG, Rahim S, Sreenivasan AT. Synthesis and Anticancer Activity Evaluation of Self- assembled Curcumin Loaded Gelatin - Oleic acid - Carboxymethyl Chitosan Nanoparticles on MCF-7 cells. Eur J Pharm Biopharm 2025; 211:114718. [PMID: 40216037 DOI: 10.1016/j.ejpb.2025.114718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/16/2025] [Accepted: 04/04/2025] [Indexed: 05/07/2025]
Abstract
Curcumin (CUR) is a natural herb with known anticancer effects against many malignancies such as breast cancer. However, CUR is poorly water-soluble and suffers from low bioavailability, and its delivery to breast cancer through oral administration is interrupted by early release and degradation before reaching the target site. So, this study aimed to develop an oral breast cancer-targeted drug delivery system (DDS) using a combination of biopolymers like mucoadhesive carboxymethyl chitosan, oleic acid and gelatin to create biopolymer-mediated nanoparticles (NPs) for the delivery of hydrophobic CUR. Gelatin-Oleic acid-Carboxymethyl chitosan (GOC) readily self-assembles into nanoparticles (GOCNPs) using the desolvation process. Such self-assembled DDS based on biopolymers without any crosslinkers for the preparation present controlled drug release and enhanced anticancer efficacy compared to existing systems. The prepared GOCNPs were characterized by FTIR, 1HNMR, XRD, SEM, HRTEM, DLS, and Zeta analysis. The DDS's maximum drug loading efficiency (DLE) and encapsulation efficiency (EE) values at pH 4.0 were 78.0 ± 2.34 % and 94.0 ± 2.8 %, respectively. The CUR-loaded GOCNPs shows a 90.0 ± 2.6 % CUR release at pH 5.5, while the release percentage of CUR at pH 7.4 was only 37.0 ± 1.06 %. According to the MTT data, CUR-GOCNPs show significant cytotoxicity to MCF-7 cells, showed a cell viability of 20.16 % towards MCF-7 cancer cells and loading of CUR to the GOCNPs notably increased its anticancer activity as the IC50 of CUR-GOCNPs was significantly lowered (6.880 µg/mL against MCF-7 in 24 h analysis). Studies on drug release kinetics, cytotoxicity, apoptosis, hemocompatibility, swelling, and in vivo pharmacokinetics were carried out to prove the effectiveness of the biopolymer-based nanoparticles developed as an effective oral delivery system for CUR. The future holds enormous possibilities for the clinical translation of prepared drug delivery system, as advances in controlled drug delivery continue to prove the design and capabilities of GOCNPs.
Collapse
Affiliation(s)
- Lekshmi Geetha Sivasankaran
- Department of Chemistry, Research Centre, School of Physical and Mathematical Sciences, University of Kerala, Kariavattom, Thiruvananthapuram, 695581, India
| | - Suriya Rahim
- Department of Chemistry, Research Centre, School of Physical and Mathematical Sciences, University of Kerala, Kariavattom, Thiruvananthapuram, 695581, India
| | - Anirudhan Thayyath Sreenivasan
- Department of Chemistry, Research Centre, School of Physical and Mathematical Sciences, University of Kerala, Kariavattom, Thiruvananthapuram, 695581, India.
| |
Collapse
|
2
|
Qin X, Yin P, Zhang Y, Su M, Chen F, Xu X, Zhao J, Gui Y, Guo H, Zhao C, Zhang Z. Self-assembled ordered AuNRs-modified electrodes for simultaneous determination of dopamine and topotecan with improved data reproducibility. Mikrochim Acta 2024; 191:350. [PMID: 38806865 DOI: 10.1007/s00604-024-06441-x] [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: 02/04/2024] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
Gold nanomaterials have been widely explored in electrochemical sensors due to their high catalytic property and good stability in multi-medium. In this paper, the reproducibility of the signal among batches of gold nanorods (AuNRs)-modified electrodes was investigated to improve the data stabilization and repeatability. Ordered and random self-assembled AuNRs-modified electrodes were used as electrochemical sensors for the simultaneous determination of dopamine (DA) and topotecan (TPC), with the aim of obtaining an improved signal stability in batches of electrodes and realizing the simultaneous determination of both substances. The morphology and structure of the assemblies were analyzed and characterized by UV-Vis spectra, scanning electron microscopy (SEM), transmission electron microscopy (TEM), and X-ray powder diffraction (XRD). Electrochemical studies showed that the ordered AuNRs/ITO electrodes have excellent signal reproducibility among several individuals due to the homogeneous mass transfer in the ordered arrangement of the AuNRs. Under the optimized conditions, the simultaneous detection results of DA and TPC showed good linearity in the ranges 1.75-45 μM and 1.5-40 μM, and the detection limits of DA and TPC were 0.06 μM and 0.17 μM, respectively. The results showed that the prepared ordered AuNR/ITO electrode had high sensitivity, long-term stability, and reproducibility for the simultaneous determination of DA and TPC, and it was expected to be applicable for real sample testing.
Collapse
Affiliation(s)
- Xiaoyun Qin
- School of Material and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450000, China
| | - Peijun Yin
- School of Material and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450000, China
| | - Yuhang Zhang
- School of Material and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450000, China
| | - Mingxing Su
- School of Material and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450000, China
| | - Fenghua Chen
- School of Material and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450000, China
| | - Xinru Xu
- Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, School of Science, Tianjin University, Tianjin, 300072, China
| | - Jianbo Zhao
- School of Material and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450000, China
| | - Yanghai Gui
- School of Material and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450000, China
| | - Huishi Guo
- School of Material and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou, 450000, China
| | - Chao Zhao
- Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, School of Science, Tianjin University, Tianjin, 300072, China
| | - Zhen Zhang
- Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, School of Science, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
3
|
Schmitt S, Machui P, Mai I, Herterich S, Wunder S, Cyprys P, Gerlach M, Ochtrop P, Hackenberger CP, Schumacher D, Helma J, Vogl AM, Kasper MA. Design and Evaluation of Phosphonamidate-Linked Exatecan Constructs for Highly Loaded, Stable, and Efficacious Antibody-Drug Conjugates. Mol Cancer Ther 2024; 23:199-211. [PMID: 37828728 PMCID: PMC10831470 DOI: 10.1158/1535-7163.mct-23-0359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 08/30/2023] [Accepted: 10/10/2023] [Indexed: 10/14/2023]
Abstract
Topoisomerase I (TOP1) Inhibitors constitute an emerging payload class to engineer antibody-drug conjugates (ADC) as next-generation biopharmaceutical for cancer treatment. Existing ADCs are using camptothecin payloads with lower potency and suffer from limited stability in circulation. With this study, we introduce a novel camptothecin-based linker-payload platform based on the highly potent camptothecin derivative exatecan. First, we describe general challenges that arise from the hydrophobic combination of exatecan and established dipeptidyl p-aminobenzyl-carbamate (PAB) cleavage sites such as reduced antibody conjugation yields and ADC aggregation. After evaluating several linker-payload structures, we identified ethynyl-phosphonamidates in combination with a discrete PEG24 chain to compensate for the hydrophobic PAB-exatecan moiety. Furthermore, we demonstrate that the identified linker-payload structure enables the construction of highly loaded DAR8 ADCs with excellent solubility properties. Head-to-head comparison with Enhertu, an approved camptothecin-based ADC, revealed improved target-mediated killing of tumor cells, excellent bystander killing, drastically improved linker stability in vitro and in vivo and superior in vivo efficacy over four tested dose levels in a xenograft model. Moreover, we show that ADCs based on the novel exatecan linker-payload platform exhibit antibody-like pharmacokinetic properties, even when the ADCs are highly loaded with eight drug molecules per antibody. This ADC platform constitutes a new and general solution to deliver TOP1 inhibitors with highest efficiency to the site of the tumor, independent of the antibody and its target, and is thereby broadly applicable to various cancer indications.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Christian P.R. Hackenberger
- Chemical Biology Department, Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Berlin, Germany
- Department of Chemistry, Humboldt Universität zu Berlin, Berlin, Germany
| | | | | | | | | |
Collapse
|
4
|
Williams KS, Secomb TW, El-Kareh AW. An autonomous mathematical model for the mammalian cell cycle. J Theor Biol 2023; 569:111533. [PMID: 37196820 DOI: 10.1016/j.jtbi.2023.111533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 04/04/2023] [Accepted: 05/10/2023] [Indexed: 05/19/2023]
Abstract
A mathematical model for the mammalian cell cycle is developed as a system of 13 coupled nonlinear ordinary differential equations. The variables and interactions included in the model are based on detailed consideration of available experimental data. A novel feature of the model is inclusion of cycle tasks such as origin licensing and initiation, nuclear envelope breakdown and kinetochore attachment, and their interactions with controllers (molecular complexes involved in cycle control). Other key features are that the model is autonomous, except for a dependence on external growth factors; the variables are continuous in time, without instantaneous resets at phase boundaries; mechanisms to prevent rereplication are included; and cycle progression is independent of cell size. Eight variables represent cell cycle controllers: the Cyclin D1-Cdk4/6 complex, APCCdh1, SCFβTrCP, Cdc25A, MPF, NuMA, the securin-separase complex, and separase. Five variables represent task completion, with four for the status of origins and one for kinetochore attachment. The model predicts distinct behaviors corresponding to the main phases of the cell cycle, showing that the principal features of the mammalian cell cycle, including restriction point behavior, can be accounted for in a quantitative mechanistic way based on known interactions among cycle controllers and their coupling to tasks. The model is robust to parameter changes, in that cycling is maintained over at least a five-fold range of each parameter when varied individually. The model is suitable for exploring how extracellular factors affect cell cycle progression, including responses to metabolic conditions and to anti-cancer therapies.
Collapse
Affiliation(s)
| | - Timothy W Secomb
- BIO5 Institute, University of Arizona, Tucson, AZ, USA; Department of Physiology, University of Arizona, Tucson, AZ, USA
| | | |
Collapse
|
5
|
Combined chemotherapy based on bioactive black phosphorus for pancreatic cancer therapy. J Control Release 2023; 354:889-901. [PMID: 36586672 DOI: 10.1016/j.jconrel.2022.12.054] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/23/2022] [Accepted: 12/26/2022] [Indexed: 01/02/2023]
Abstract
Pancreatic cancer is the most aggressive malignant tumor with difficulty in early diagnosis, very short survival time in advanced stage, and lack of effective treatment options. In this work, a novel combination chemotherapy strategy based on bioactive black phosphorus (BP) and gemcitabine (GEM) is developed for efficient treatment of pancreatic cancer. The combined cell cycle blockage in G2/M phase induced by BP and G0/G1 phase by GEM results in synergistic killing of pancreatic cancer cells with the combination index (CI) < 1. The iRGD modified zein nanoparticles co-loaded with BP quantum dots (BPQDs) and GEM are designed and prepared as a targeted nanoplatform (BP-GEM@NPs). After intravenous injection, the in vivo distribution and pharmacokinetics results demonstrate that BP-GEM@NPs shows excellent tumor targeting capability and significantly prolonged blood circulation time. The targeted co-delivery of BPQDs and GEM induces much more pancreatic tumor cell apoptosis and synergistically inhibits tumor growth in both subcutaneous xenograft and orthotopic models. Meanwhile, BP-GEM@NPs exhibit good biocompatibility without bring adverse effects. This work indicates the great potential of BP-GEM@NPs as a combination chemotherapy for pancreatic cancer and provides insights into development of biomedicine by exploring the intrinsic bioactivities of nanomaterials.
Collapse
|
6
|
Bayoumi HM, Alkhatib MH. Incorporation of topotecan into sesame oil - Nanoemulsion potentiates its cytotoxic effect in cancer cells. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
7
|
Abstract
The paradigm of surface-expressed programmed death ligand 1 (PDL1) signalling to immune cell programmed death 1 (PD1) to inhibit antitumour immunity has helped to develop effective and revolutionary immunotherapies using antibodies blocking these cell-extrinsic interactions. The recent discovery of cancer cell-intrinsic PDL1 signals has broadened understanding of pathologic tumour PDL1 signal consequences that now includes control of tumour growth and survival pathways, stemness, immune effects, DNA damage responses and gene expression regulation. Many such effects are PD1-independent. These insights demonstrate that the prevailing cell-extrinsic PDL1 signalling paradigm is useful, but incomplete in important respects. This Perspective discusses historical and recent advances in understanding cancer cell-intrinsic PDL1 signals, mechanisms for signal controls and important immunopathologic consequences including resistance to cytotoxic agents, targeted small molecules and immunotherapies. Cancer cell-intrinsic PDL1 signals present novel drug discovery targets and also have potential as reliable treatment response biomarkers. Cancer cell-intrinsic PD1 signals and cell-intrinsic PDL1 signals in non-cancer cells are discussed briefly, as are PDL1 signals from soluble and vesicle-bound PDL1 and PDL1 isoforms. We conclude with suggestions for addressing the most pressing challenges and opportunities in this rapidly developing field.
Collapse
Affiliation(s)
- Anand V R Kornepati
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Ratna K Vadlamudi
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX, USA
- Department of Obstetrics and Gynecology, University of Texas Health San Antonio, San Antonio, TX, USA
- MD Anderson Cancer Center, University of Texas Health San Antonio, San Antonio, TX, USA
| | - Tyler J Curiel
- Graduate School of Biomedical Sciences, University of Texas Health San Antonio, San Antonio, TX, USA.
- MD Anderson Cancer Center, University of Texas Health San Antonio, San Antonio, TX, USA.
- Department of Medicine, University of Texas Health San Antonio, San Antonio, TX, USA.
| |
Collapse
|
8
|
Wu KM, Chi CW, Lai JCY, Chen YJ, Kou YR. TLC388 Induces DNA Damage and G2 Phase Cell Cycle Arrest in Human Non-Small Cell Lung Cancer Cells. Cancer Control 2020; 27:1073274819897975. [PMID: 32281394 PMCID: PMC7154561 DOI: 10.1177/1073274819897975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
TLC388, a camptothecin-derivative targeting topoisomerase I, is a potential anticancer drug. In this study, its effect on A549 and H838 human non-small cell lung cancer (NSCLC) cells was investigated. Cell viability and proliferation were determined by thiazolyl blue tetrazolium bromide and clonogenic assays, respectively, and cell cycle analysis and detection of phosphorylated histone H3 (Ser10) were performed by flow cytometry. γ-H2AX protein; G2/M phase-associated molecules ataxia-telangiectasia mutated (ATM), CHK1, CHK2, CDC25C, CDC2, and cyclin B1; and apoptosis were assessed with immunofluorescence staining, immunoblotting, and an annexin V assay, respectively. The effect of co-treatment with CHIR124 (a checkpoint kinase 1 [CHK1] inhibitor) was also studied. TLC388 decreased the viability and proliferation of cells of both NSCLC lines in a dose-dependent manner. TLC388 inhibited the viability of NSCLC cell lines with an estimated concentration of 50% inhibition (IC50), which was 4.4 and 4.1 μM for A549 and H838 cells, respectively, after 24 hours. Moreover, it resulted in the accumulation of cells at the G2/M phase and increased γ-H2AX levels in A549 cells. Levels of the G2 phase-related molecules phosphorylated ATM, CHK1, CHK2, CDC25C, and cyclin B1 were increased in TLC388-treated cells. CHIR124 enhanced the cytotoxicity of TLC388 toward A549 and H838 cells and induced apoptosis of the former. TLC388 inhibits NSCLC cell growth by inflicting DNA damage and activating G2/M checkpoint proteins that trigger G2 phase cell cycle arrest to enable DNA repair. CHIR124 enhanced the cytotoxic effect of TLC388 and induced apoptosis.
Collapse
Affiliation(s)
- Kun-Ming Wu
- Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei.,Chest Division, Department of Internal Medicine, MacKay Memorial Hospital, Taipei.,Mackay Junior College of Medicine, Nursing, and Management, Taipei
| | - Chih-Wen Chi
- Department of Medical Research, MacKay Memorial Hospital, New Taipei.,Department of Nursing, MacKay Medical College, New Taipei
| | | | - Yu-Jen Chen
- Mackay Junior College of Medicine, Nursing, and Management, Taipei.,Department of Medical Research, MacKay Memorial Hospital, New Taipei.,Department of Medical Research, China Medical University Hospital, Taichung.,Department of Radiation Oncology, MacKay Memorial Hospital, Taipei
| | - Yu Ru Kou
- Institute of Physiology, School of Medicine, National Yang-Ming University, Taipei
| |
Collapse
|
9
|
Hurley SK, Cutrone NM, Fath KR, Pajovich HT, Garcia J, Smith AM, Banerjee IA. Self-assembled phenylisoxazole-peptide hybrid assemblies and their interactions with breast and ovarian tumor cells. INT J POLYM MATER PO 2019. [DOI: 10.1080/00914037.2018.1525542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Sara K. Hurley
- Department of Chemistry, Fordham University, Bronx, NY, USA
| | | | - Karl R. Fath
- Department of Biology, Queens College, City University of New York, New York, NY, USA
| | | | - Jeremy Garcia
- Department of Biology, Queens College, City University of New York, New York, NY, USA
| | | | | |
Collapse
|
10
|
Sanhaji M, Göring J, Couleaud P, Aires A, Cortajarena AL, Courty J, Prina-Mello A, Stapf M, Ludwig R, Volkov Y, Latorre A, Somoza Á, Miranda R, Hilger I. The phenotype of target pancreatic cancer cells influences cell death by magnetic hyperthermia with nanoparticles carrying gemicitabine and the pseudo-peptide NucAnt. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 20:101983. [PMID: 30940505 DOI: 10.1016/j.nano.2018.12.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 12/17/2018] [Accepted: 12/26/2018] [Indexed: 12/22/2022]
Abstract
In this paper we show that conjugation of magnetic nanoparticles (MNPs) with Gemcitabine and/or NucAnt (N6L) fostered their internalization into pancreatic tumor cells and that the coupling procedure did not alter the cytotoxic potential of the drugs. By treating tumor cells (BxPC3 and PANC-1) with the conjugated MNPs and magnetic hyperthermia (43 °C, 60 min), cell death was observed. The two pancreatic tumor cell lines showed different reactions against the combined therapy according to their intrinsic sensitivity against Gemcitabine (cell death, ROS production, ability to activate ERK 1/2 and JNK). Finally, tumors (e.g. 3 mL) could be effectively treated by using almost 4.2 × 105 times lower Gemcitabine doses compared to conventional therapies. Our data show that this combinatorial therapy might well play an important role in certain cell phenotypes with low readiness of ROS production. This would be of great significance in distinctly optimizing local pancreatic tumor treatments.
Collapse
Affiliation(s)
- Mourad Sanhaji
- Institute for Diagnostic and Interventional Radiology, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
| | - Julia Göring
- Institute for Diagnostic and Interventional Radiology, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
| | - Pierre Couleaud
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Campus Universitario de Cantoblanco, Madrid, Spain; Unidad Asociada de Nanobiotecnología CNB-CSIC & IMDEA Nanociencia, Campus Universitario de Cantoblanco, Madrid, Spain
| | - Antonio Aires
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Campus Universitario de Cantoblanco, Madrid, Spain; Unidad Asociada de Nanobiotecnología CNB-CSIC & IMDEA Nanociencia, Campus Universitario de Cantoblanco, Madrid, Spain
| | - Aitziber L Cortajarena
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Campus Universitario de Cantoblanco, Madrid, Spain; Unidad Asociada de Nanobiotecnología CNB-CSIC & IMDEA Nanociencia, Campus Universitario de Cantoblanco, Madrid, Spain
| | - José Courty
- Laboratoire CRRET, Université Paris EST Créteil, 61 Avenue du Général de Gaulle, Créteil, France
| | - Adriele Prina-Mello
- Nanomedicine and Molecular Imaging group, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Marcus Stapf
- Institute for Diagnostic and Interventional Radiology, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
| | - Robert Ludwig
- Institute for Diagnostic and Interventional Radiology, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
| | - Yuri Volkov
- Nanomedicine and Molecular Imaging group, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Alfonso Latorre
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Campus Universitario de Cantoblanco, Madrid, Spain; Unidad Asociada de Nanobiotecnología CNB-CSIC & IMDEA Nanociencia, Campus Universitario de Cantoblanco, Madrid, Spain
| | - Álvaro Somoza
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Campus Universitario de Cantoblanco, Madrid, Spain; Unidad Asociada de Nanobiotecnología CNB-CSIC & IMDEA Nanociencia, Campus Universitario de Cantoblanco, Madrid, Spain
| | - Rodolfo Miranda
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Campus Universitario de Cantoblanco, Madrid, Spain; Unidad Asociada de Nanobiotecnología CNB-CSIC & IMDEA Nanociencia, Campus Universitario de Cantoblanco, Madrid, Spain
| | - Ingrid Hilger
- Institute for Diagnostic and Interventional Radiology, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
11
|
A novel nanocomposite based on gold nanoparticles loaded on acetylene black for electrochemical sensing of the anticancer drug topotecan in the presence of high concentration of uric acid. J Electroanal Chem (Lausanne) 2018. [DOI: 10.1016/j.jelechem.2018.07.031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
12
|
Yu CH, Chou CC, Tu HF, Huang WC, Ho YY, Khoo KH, Lee MS, Chang GD. Antibody-assisted target identification reveals afatinib, an EGFR covalent inhibitor, down-regulating ribonucleotide reductase. Oncotarget 2018; 9:21512-21529. [PMID: 29765556 PMCID: PMC5940374 DOI: 10.18632/oncotarget.25177] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 04/05/2018] [Indexed: 01/14/2023] Open
Abstract
Afatinib, used for the first-line treatment of non-small-cell lung carcinoma (NSCLC) patients with distinct epidermal growth factor receptor (EGFR) mutations, inactivates EGFR by mimicking ATP structure and forming a covalent adduct with EGFR. We developed a method to unravel potential targets of afatinib in NSCLC cells through immunoprecipitation of afatinib-labeling proteins with anti-afatinib antiserum and mass spectrometry analysis. Ribonucleotide reductase (RNR) is one of target proteins of afatinib revealed by this method. Treatment of afatinib at 10-100 nM potently inhibited intracellular RNR activity in an in vitro assay using permeabilized PC-9 cells (formerly known as PC-14). PC-9 cells treated with 10 μM afatinib displayed elevated markers of DNA damage. Long-term treatment of therapeutic concentrations of afatinib in PC-9 cells caused significant decrease in protein levels of RNR subunit M2 at 1-10 nM and RNR subunit M1 at 100 nM. EGFR-null Chinese hamster ovary (CHO) cells treated with afatinib also showed similar effects. Afatinib repressed the upregulation of RNR subunit M2 induced by gemcitabine. Covalent modification with afatinib resulting in inhibition and protein downregulation of RNR underscores the therapeutic and off-target effects of afatinib. Afatinib may serve as a lead compound of chemotherapeutic drugs targeting RNR. This method can be widely used in the identification of potential targets of other covalent drugs.
Collapse
Affiliation(s)
- Cheng-Han Yu
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Chi-Chi Chou
- Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Hsin-Fang Tu
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Wei-Chieh Huang
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Ya-Yeh Ho
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| | - Kay-Hooi Khoo
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan.,Institute of Biological Chemistry, Academia Sinica, Taipei 11529, Taiwan
| | - Ming-Shyue Lee
- Department of Biochemistry and Molecular Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Geen-Dong Chang
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
13
|
P-3F, a microtubule polymerization inhibitor enhances P53 stability through the change in localization of RPS27a. Int J Biochem Cell Biol 2017; 92:53-62. [DOI: 10.1016/j.biocel.2017.09.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 09/05/2017] [Accepted: 09/15/2017] [Indexed: 02/02/2023]
|
14
|
Zingoni A, Fionda C, Borrelli C, Cippitelli M, Santoni A, Soriani A. Natural Killer Cell Response to Chemotherapy-Stressed Cancer Cells: Role in Tumor Immunosurveillance. Front Immunol 2017; 8:1194. [PMID: 28993779 PMCID: PMC5622151 DOI: 10.3389/fimmu.2017.01194] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/08/2017] [Indexed: 12/13/2022] Open
Abstract
Natural killer (NK) cells are innate cytotoxic lymphoid cells that actively prevent neoplastic development, growth, and metastatic dissemination in a process called cancer immunosurveillance. An equilibrium between immune control and tumor growth is maintained as long as cancer cells evade immunosurveillance. Therapies designed to kill cancer cells and to simultaneously sustain host antitumor immunity are an appealing strategy to control tumor growth. Several chemotherapeutic agents, depending on which drugs and doses are used, give rise to DNA damage and cancer cell death by means of apoptosis, immunogenic cell death, or other forms of non-apoptotic death (i.e., mitotic catastrophe, senescence, and autophagy). However, it is becoming increasingly clear that they can trigger additional stress responses. Indeed, relevant immunostimulating effects of different therapeutic programs include also the activation of pathways able to promote their recognition by immune effector cells. Among stress-inducible immunostimulating proteins, changes in the expression levels of NK cell-activating and inhibitory ligands, as well as of death receptors on tumor cells, play a critical role in their detection and elimination by innate immune effectors, including NK cells. Here, we will review recent advances in chemotherapy-mediated cellular stress pathways able to stimulate NK cell effector functions. In particular, we will address how these cytotoxic lymphocytes sense and respond to different types of drug-induced stresses contributing to anticancer activity.
Collapse
Affiliation(s)
- Alessandra Zingoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Cinzia Fionda
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Cristiana Borrelli
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.,Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Marco Cippitelli
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| | - Angela Santoni
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy.,Neuromed I.R.C.C.S. - Istituto Neurologico Mediterraneo, Pozzilli, Italy
| | - Alessandra Soriani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
15
|
Al-Maleki AR, Loke MF, Lui SY, Ramli NSK, Khosravi Y, Ng CG, Venkatraman G, Goh KL, Ho B, Vadivelu J. Helicobacter pylori outer inflammatory protein A (OipA) suppresses apoptosis of AGS gastric cells in vitro. Cell Microbiol 2017; 19. [PMID: 28776327 DOI: 10.1111/cmi.12771] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 07/02/2017] [Accepted: 07/26/2017] [Indexed: 12/12/2022]
Abstract
Outer inflammatory protein A (OipA) is an important virulence factor associated with gastric cancer and ulcer development; however, the results have not been well established and turned out to be controversial. This study aims to elucidate the role of OipA in Helicobacter pylori infection using clinical strains harbouring oipA "on" and "off" motifs. Proteomics analysis was performed on AGS cell pre-infection and postinfection with H. pylori oipA "on" and "off" strains, using liquid chromatography/mass spectrometry. AGS apoptosis and cell cycle assays were performed. Moreover, expression of vacuolating cytotoxin A (VacA) was screened using Western blotting. AGS proteins that have been suggested previously to play a role or associated with gastric disease were down-regulated postinfection with oipA "off" strains comparing to oipA "on" strains. Furthermore, oipA "off" and ΔoipA cause higher level of AGS cells apoptosis and G0/G1 cell-cycle arrest than oipA "on" strains. Interestingly, deletion of oipA increased bacterial VacA production. The capability of H. pylori to induce apoptosis and suppress expression of proteins having roles in human disease in the absence of oipA suggests that strains not expressing OipA may be less virulent or may even be protective against carcinogenesis compared those expressing OipA. This potentially explains the higher incidence of gastric cancer in East Asia where oipA "on" strains predominates.
Collapse
Affiliation(s)
- Anis Rageh Al-Maleki
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Mun Fai Loke
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sook Yin Lui
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Nur Siti Khadijah Ramli
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Yalda Khosravi
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Chow Goon Ng
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gopinath Venkatraman
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Khean-Lee Goh
- Department of Medicine, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Bow Ho
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Singapore Precision Medicine Centre Pte Ltd, Singapore, Singapore
| | - Jamuna Vadivelu
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
16
|
Khosrawipour V, Diaz-Carballo D, Acikelli AH, Khosrawipour T, Falkenstein TA, Wu D, Zieren J, Giger-Pabst U. Cytotoxic effect of different treatment parameters in pressurized intraperitoneal aerosol chemotherapy (PIPAC) on the in vitro proliferation of human colonic cancer cells. World J Surg Oncol 2017; 15:43. [PMID: 28183319 PMCID: PMC5301439 DOI: 10.1186/s12957-017-1109-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 01/28/2017] [Indexed: 12/04/2022] Open
Abstract
Background Pressurized intraperitoneal aerosol chemotherapy (PIPAC) has been recently reported as a new approach for intraperitoneal chemotherapy (IPC). By means of a patented micropump, the liquid chemotherapy is delivered into the peritoneal cavity as an aerosol which is supposed to achieve “gas-like” distribution. However, recent data report that the fraction of the submicron aerosol (gas-like) is less than 3 vol% of the total amount of aerosolized chemotherapy. Until today, possible modifications of treatment parameters during PIPAC with the aim of improving therapeutic outcomes have not been studied yet. This study aims to establish an in vitro PIPAC model to explore the cytotoxic effect of the submicron aerosol fraction and to investigate the impact of different application parameters on the cytotoxic effect of PIPAC on human colonic cancer cells. Methods An in vitro model using HCT8 colon adenocarcinoma wild-type cells (HCT8WT) and multi-chemotherapy refractory subline (HCT8RT) was established. Different experimental parameters such as pressure, drug dosage, time exposure, and system temperature were monitored in order to search for the conditions with a higher impact on cell toxicity. Cell proliferation was determined by means of colorimetric MTT assay 48 h following PIPAC exposures. Results Standard operational parameters applied for PIPAC therapy depicted a cytotoxic effect of the submicron aerosol fraction generated by the PIPAC micropump. We also observed that increasing pressure significantly enhanced tumor cell toxicity in both wild-type and chemotherapy-resistant cells. A maximum of cytotoxicity was observed at 15 mmHg. Pressure >15 mmHg did not show additional cytotoxic effect on cells. Increased oxaliplatin dosage resulted in progressively higher cell toxicity as expected. However, in resistant cells, a significant effect was only found at higher drug concentrations. Neither an extension of exposure time nor an increase in temperature of the aerosolized chemotherapy solution added an improvement in cytotoxicity. Conclusions In this in vitro PIPAC model, the gas-like PIPAC aerosol fraction showed a cytotoxic effect which was enhanced by higher intra-abdominal pressure with a maximum at 15 mmHg. Similar findings were observed for drug dose escalation. A phase I dose escalation study is currently performed at our institution. However, increasing the intra-abdominal pressure might be a first and simple way to enhance the cytotoxic effect of PIPAC therapy which needs further clinical investigations. Electronic supplementary material The online version of this article (doi:10.1186/s12957-017-1109-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Veria Khosrawipour
- Department of General Surgery and Therapy Center for Peritonealcarcinomatosis, St. Mary's Hospital Herne, Ruhr University of Bochum, Hölkeskampring 40, 44625, Herne, Germany.,Basic Research Laboratory Department of Surgery, St. Mary's Hospital Herne, Ruhr University of Bochum, Herne, Germany
| | - David Diaz-Carballo
- Department of Hematology and Medical Oncology, St. Mary's Hospital Herne, Ruhr University Bochum, Herne, Germany
| | - Ali-Haydar Acikelli
- Department of Hematology and Medical Oncology, St. Mary's Hospital Herne, Ruhr University Bochum, Herne, Germany
| | - Tanja Khosrawipour
- Department of General Surgery and Therapy Center for Peritonealcarcinomatosis, St. Mary's Hospital Herne, Ruhr University of Bochum, Hölkeskampring 40, 44625, Herne, Germany. .,Basic Research Laboratory Department of Surgery, St. Mary's Hospital Herne, Ruhr University of Bochum, Herne, Germany.
| | - Thomas Albert Falkenstein
- Basic Research Laboratory Department of Surgery, St. Mary's Hospital Herne, Ruhr University of Bochum, Herne, Germany
| | - Dan Wu
- Department of General Surgery and Therapy Center for Peritonealcarcinomatosis, St. Mary's Hospital Herne, Ruhr University of Bochum, Hölkeskampring 40, 44625, Herne, Germany.,Basic Research Laboratory Department of Surgery, St. Mary's Hospital Herne, Ruhr University of Bochum, Herne, Germany
| | - Jürgen Zieren
- Department of General Surgery and Therapy Center for Peritonealcarcinomatosis, St. Mary's Hospital Herne, Ruhr University of Bochum, Hölkeskampring 40, 44625, Herne, Germany
| | - Urs Giger-Pabst
- Department of General Surgery and Therapy Center for Peritonealcarcinomatosis, St. Mary's Hospital Herne, Ruhr University of Bochum, Hölkeskampring 40, 44625, Herne, Germany.,Basic Research Laboratory Department of Surgery, St. Mary's Hospital Herne, Ruhr University of Bochum, Herne, Germany
| |
Collapse
|
17
|
Sun B, Gao L, Ahsan A, Chu P, Song Y, Li H, Zhang Z, Lin Y, Peng J, Song Z, Wang S, Tang Z. Anticancer effect of SZC015 on lung cancer cells through ROS-dependent apoptosis and autophagy induction mechanisms in vitro. Int Immunopharmacol 2016; 40:400-409. [PMID: 27697723 DOI: 10.1016/j.intimp.2016.09.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/31/2016] [Accepted: 09/24/2016] [Indexed: 12/16/2022]
Abstract
Oleanolic acid (OA) and its several derivatives possess various pharmacological activities, such as antitumor and anti-inflammation. In present study, anticancer effect of SZC015, an OA derivative, and its underlying mechanisms were investigated. We demonstrated that cell viability was significantly decreased in SZC015-treated lung cancer cells, but has less cytotoxicity in human bronchial epithelial cell line. Further investigation verified that apoptosis and autophagy induction and G0/G1 phase arrest were observed in SZC015-treated H322 cells. Mechanically, the level of Akt, p-Akt, p-IκBα, and total p65, the p-p65 in the cytoplasm and nucleus were suppressed by SZC015 in H322 cells, respectively. Inhibition of p65 nuclear translocation was also confirmed by immunofluorescence staining. In addition, co-treatment with chloroquine, an autophagy inhibitor, significantly inhibited SZC015-induced autophagy and enhanced SZC015-induced apoptotic cell death. Intracellular ROS was increased in a concentration-dependent manner, which could be prevented by N-Acetyl l-Cysteine, an ROS scavenger. Moreover, the level of Akt and procaspase-3 were increased, while the ratio of LC3 II/I was decreased. Taken together, our study demonstrates that the inhibitory effect of SZC015 against H322 cells is mediated by excessive ROS generation that could suppress Akt/NF-κB signaling pathway, which thereby leads to apoptotic and autophagic cell death.
Collapse
Affiliation(s)
- Bin Sun
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Lei Gao
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Anil Ahsan
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Peng Chu
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Yanlin Song
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Hailong Li
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Zonghui Zhang
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Yuan Lin
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Jinyong Peng
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China
| | - Zhicheng Song
- College of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China
| | - Shisheng Wang
- College of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China.
| | - Zeyao Tang
- Pharmacology Department, Dalian Medical University, 9 West Section, South Road of Lvshun, Dalian, China.
| |
Collapse
|
18
|
Kolokotroni E, Dionysiou D, Veith C, Kim YJ, Sabczynski J, Franz A, Grgic A, Palm J, Bohle RM, Stamatakos G. In Silico Oncology: Quantification of the In Vivo Antitumor Efficacy of Cisplatin-Based Doublet Therapy in Non-Small Cell Lung Cancer (NSCLC) through a Multiscale Mechanistic Model. PLoS Comput Biol 2016; 12:e1005093. [PMID: 27657742 PMCID: PMC5033576 DOI: 10.1371/journal.pcbi.1005093] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 08/01/2016] [Indexed: 11/30/2022] Open
Abstract
The 5-year survival of non-small cell lung cancer patients can be as low as 1% in advanced stages. For patients with resectable disease, the successful choice of preoperative chemotherapy is critical to eliminate micrometastasis and improve operability. In silico experimentations can suggest the optimal treatment protocol for each patient based on their own multiscale data. A determinant for reliable predictions is the a priori estimation of the drugs’ cytotoxic efficacy on cancer cells for a given treatment. In the present work a mechanistic model of cancer response to treatment is applied for the estimation of a plausible value range of the cell killing efficacy of various cisplatin-based doublet regimens. Among others, the model incorporates the cancer related mechanism of uncontrolled proliferation, population heterogeneity, hypoxia and treatment resistance. The methodology is based on the provision of tumor volumetric data at two time points, before and after or during treatment. It takes into account the effect of tumor microenvironment and cell repopulation on treatment outcome. A thorough sensitivity analysis based on one-factor-at-a-time and latin hypercube sampling/partial rank correlation coefficient approaches has established the volume growth rate and the growth fraction at diagnosis as key features for more accurate estimates. The methodology is applied on the retrospective data of thirteen patients with non-small cell lung cancer who received cisplatin in combination with gemcitabine, vinorelbine or docetaxel in the neoadjuvant context. The selection of model input values has been guided by a comprehensive literature survey on cancer-specific proliferation kinetics. The latin hypercube sampling has been recruited to compensate for patient-specific uncertainties. Concluding, the present work provides a quantitative framework for the estimation of the in-vivo cell-killing ability of various chemotherapies. Correlation studies of such estimates with the molecular profile of patients could serve as a basis for reliable personalized predictions. Less than 14% of medically treated patients with locally advanced and metastatic non-small cell lung cancer are expected to be alive 5 years after diagnosis. Standard therapeutic strategies include the administration of two drugs in combination, aiming at shrinking the tumor before surgery and improving overall survival. Knowing the sensitivity profile of each patient to different treatment strategies at diagnosis may help choose the most appropriate ones. We develop a methodology for the quantitative estimation of the cytotoxic efficacy of cisplatin-based doublets on cancer cells by applying a simulation model of cancer progression and response. The model incorporates the proliferation cycle, quiescence, differentiation and loss of tumor cells. We evaluate the effect of in vivo microenvironment of real tumors, as expressed by measurable tumor proliferation kinetics, such as how fast the tumor grows, the percentage of cells that are actively dividing, the resistance of stem cells, etc. on treatment outcome so as to derive more accurate estimates. A literature survey guides the selection of values. The methodology is applied to a real clinical dataset of patients. Correlation studies between the derived cytotoxicities and the patients’ molecular profile could lead to predictions of treatment response at the time of diagnosis.
Collapse
Affiliation(s)
- Eleni Kolokotroni
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, National Technical University of Athens, Athens, Greece
| | - Dimitra Dionysiou
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, National Technical University of Athens, Athens, Greece
| | - Christian Veith
- Institute of Pathology, University of Saarland, Homburg (Saar), Germany
| | - Yoo-Jin Kim
- Institute of Pathology, University of Saarland, Homburg (Saar), Germany
| | | | | | - Aleksandar Grgic
- Department of Nuclear Medicine, University of Saarland, Homburg (Saar), Germany
| | - Jan Palm
- Department of Radiotherapy and Radiation Oncology, University of Saarland, Homburg (Saar), Germany
| | - Rainer M. Bohle
- Institute of Pathology, University of Saarland, Homburg (Saar), Germany
| | - Georgios Stamatakos
- In Silico Oncology and In Silico Medicine Group, Institute of Communication and Computer Systems, National Technical University of Athens, Athens, Greece
- * E-mail:
| |
Collapse
|
19
|
Wang X, Tanaka M, Krstin S, Peixoto HS, Moura CCDM, Wink M. Cytoskeletal interference - A new mode of action for the anticancer drugs camptothecin and topotecan. Eur J Pharmacol 2016; 789:265-274. [PMID: 27474470 DOI: 10.1016/j.ejphar.2016.07.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 07/26/2016] [Accepted: 07/26/2016] [Indexed: 11/24/2022]
Abstract
The anticancer drugs camptothecin (CPT) and topotecan (TPT) are known DNA topoisomerase I inhibitors which cause DNA damage and lead to cell death. In this study we provide evidence that CPT and TPT also interfere with the elements of cytoskeleton - microtubules and actin filaments which could be partly responsible for their cytotoxic properties. CPT and TPT apparently affected microtubule structures in living cells (Hela and U2OS) and inhibited tubulin polymerization in vitro with IC50 values of 74.57±9.96µM and 121.55±58.68µM, respectively. TPT significantly affected the nucleation and growth phase during the microtubule assembly in vitro, whereas the mode of action of CPT was different in that it specifically affected the 'tread milling' of polymerized microtubules. Cell cycle effects of CPT and TPT varied with their concentrations. CPT and TPT induced G2/M arrest and promoted the population to 76.94±11.20% and 83.91±2.43% at a concentration of 9.4nM and 46.9nM, respectively. As the concentration increased, cells were blocked in S phase with a dose-dependent reduction in G2/M population. In addition, CPT and TPT exhibited a certain effect on actin filaments by reducing the mass of actin filaments. The interactions of CPT and TPT with microtubules and actin filaments present new insights into their modes of action.
Collapse
Affiliation(s)
- Xiaojuan Wang
- Department of Biology, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany.
| | - Mine Tanaka
- Department of Biology, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany.
| | - Sonja Krstin
- Department of Biology, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany.
| | - Herbenya Silva Peixoto
- Department of Biology, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany.
| | - Carina Carneiro de Melo Moura
- Department of Biology, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany.
| | - Michael Wink
- Department of Biology, Institute of Pharmacy and Molecular Biotechnology, Heidelberg University, Heidelberg, Germany.
| |
Collapse
|
20
|
Sarin H. Conserved molecular mechanisms underlying the effects of small molecule xenobiotic chemotherapeutics on cells. Mol Clin Oncol 2015; 4:326-368. [PMID: 26998284 DOI: 10.3892/mco.2015.714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 12/08/2015] [Indexed: 12/14/2022] Open
Abstract
For proper determination of the apoptotic potential of chemoxenobiotics in synergism, it is important to understand the modes, levels and character of interactions of chemoxenobiotics with cells in the context of predicted conserved biophysical properties. Chemoxenobiotic structures are studied with respect to atom distribution over molecular space, the predicted overall octanol-to-water partition coefficient (Log OWPC; unitless) and molecular size viz a viz van der Waals diameter (vdWD). The Log OWPC-to-vdWD (nm-1 ) parameter is determined, and where applicable, hydrophilic interacting moiety/core-to-vdWD (nm-1 ) and lipophilic incorporating hydrophobic moiety/core-to-vdWD (nm-1 ) parameters of their part-structures are determined. The cellular and sub-cellular level interactions of the spectrum of xenobiotic chemotherapies have been characterized, for which a classification system has been developed based on predicted conserved biophysical properties with respect to the mode of chemotherapeutic effect. The findings of this study are applicable towards improving the effectiveness of existing combination chemotherapy regimens and the predictive accuracy of personalized cancer treatment algorithms as well as towards the selection of appropriate novel xenobiotics with the potential to be potent chemotherapeutics for dendrimer nanoparticle-based effective transvascular delivery.
Collapse
Affiliation(s)
- Hemant Sarin
- Freelance Investigator in Translational Science and Medicine, Charleston, WV 25314, USA
| |
Collapse
|
21
|
Okita R, Wolf D, Yasuda K, Maeda A, Yukawa T, Saisho S, Shimizu K, Yamaguchi Y, Oka M, Nakayama E, Lundqvist A, Kiessling R, Seliger B, Nakata M. Contrasting Effects of the Cytotoxic Anticancer Drug Gemcitabine and the EGFR Tyrosine Kinase Inhibitor Gefitinib on NK Cell-Mediated Cytotoxicity via Regulation of NKG2D Ligand in Non-Small-Cell Lung Cancer Cells. PLoS One 2015; 10:e0139809. [PMID: 26439264 PMCID: PMC4595469 DOI: 10.1371/journal.pone.0139809] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 09/17/2015] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Several cytotoxic anticancer drugs inhibit DNA replication and/or mitosis, while EGFR tyrosine kinase inhibitors inactivate EGFR signalling in cancer cell. Both types of anticancer drugs improve the overall survival of the patients with non-small-cell lung cancer (NSCLC), although tumors often become refractory to this treatment. Despite several mechanisms by which the tumors become resistant having been described the effect of these compounds on anti-tumor immunity remains largely unknown. METHODS This study examines the effect of the cytotoxic drug Gemcitabine and the EGFR tyrosine kinase inhibitor Gefitinib on the expression of NK group 2 member D (NKG2D) ligands as well as the sensitivity of NSCLC cells to the NK-mediated lysis. RESULTS We demonstrate that Gemcitabine treatment leads to an enhanced expression, while Gefitinib downregulated the expression of molecules that act as key ligands for the activating receptor NKG2D and promote NK cell-mediated recognition and cytolysis. Gemcitabine activated ATM and ATM- and Rad-3-related protein kinase (ATR) pathways. The Gemcitabine-induced phosphorylation of ATM as well as the upregulation of the NKG2D ligand expression could be blocked by an ATM-ATR inhibitor. In contrast, Gefitinib attenuated NKG2D ligand expression. Silencing EGFR using siRNA or addition of the PI3K inhibitor resulted in downregulation of NKG2D ligands. The observations suggest that the EGFR/PI3K pathway also regulates the expression of NKG2D ligands. Additionally, we showed that both ATM-ATR and EGFR regulate MICA/B via miR20a. CONCLUSION In keeping with the effect on NKG2D expression, Gemcitabine enhanced NK cell-mediated cytotoxicity while Gefitinib attenuated NK cell killing in NSCLC cells.
Collapse
Affiliation(s)
- Riki Okita
- Department of General Thoracic Surgery, Kawasaki Medical School, Kurashiki, Japan
- Department of Oncology and Pathology, Immune and Gene Therapy Laboratory, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| | - Diana Wolf
- Institute of Medical Immunology, Martin Luther University, Halle-Wittenberg, Halle, Germany
| | - Koichiro Yasuda
- Department of General Thoracic Surgery, Kawasaki Medical School, Kurashiki, Japan
| | - Ai Maeda
- Department of General Thoracic Surgery, Kawasaki Medical School, Kurashiki, Japan
| | - Takuro Yukawa
- Department of General Thoracic Surgery, Kawasaki Medical School, Kurashiki, Japan
| | - Shinsuke Saisho
- Department of General Thoracic Surgery, Kawasaki Medical School, Kurashiki, Japan
| | - Katsuhiko Shimizu
- Department of General Thoracic Surgery, Kawasaki Medical School, Kurashiki, Japan
| | | | - Mikio Oka
- Department of Respiratory Medicine, Kawasaki Medical School, Kurashiki, Japan
| | - Eiichi Nakayama
- Faculty of Health and Welfare, Kawasaki University of Medical Welfare, Kurashiki, Japan
| | - Andreas Lundqvist
- Department of Oncology and Pathology, Immune and Gene Therapy Laboratory, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Rolf Kiessling
- Department of Oncology and Pathology, Immune and Gene Therapy Laboratory, Cancer Center Karolinska, Karolinska Institutet, Stockholm, Sweden
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University, Halle-Wittenberg, Halle, Germany
| | - Masao Nakata
- Department of General Thoracic Surgery, Kawasaki Medical School, Kurashiki, Japan
| |
Collapse
|
22
|
Liposomes as multicompartmental carriers for multidrug delivery in anticancer chemotherapy. Drug Deliv Transl Res 2015; 1:66-75. [PMID: 25787890 DOI: 10.1007/s13346-010-0007-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
A new PEGylated liposomal formulation containing both gemcitabine (GEM) and paclitaxel (PTX) was investigated in order to realize an innovative multidrug carrier (MDC) to test on human cancer cells. The MDC in question was realized by the liposome extrusion method. Photocorrelation spectroscopy was used for the physicochemical characterization of the vesicular carriers. In vitro cytotoxicity was studied through MTT testing. The contemporary presence of the two antitumoral compounds induced no destabilization phenomena in the liposomal structure. The extrusion method provided vesicles with mean sizes of ∼100 nm and a zeta-potential of ∼ -10 mV. The liposomal MDC showed a high drug loading capacity (∼90% and ∼80% for GEM and PTX, respectively) as well as a controlled release of the active compounds over a 24-h period. Cell viability testing on Michigan Cancer Foundation-7 human breast cancer cells evidenced the MDC as having a stronger cytotoxic effect with respect to the active compounds tested in free and liposomal formulations, both as single molecules and in association. Flow cytometry furnished evidence of the synergistic in vitro antitumoral action between the GEM and PTX co-encapsulated the liposomal MDC. This formulation may offer even more advantages in in vivo testing in terms of drug pharmacokinetic, biodistribution, and antitumoral efficacy for the treatment of breast cancer, as compared to past formulations.
Collapse
|
23
|
Li B, Gao MH, Chu XM, Teng L, Lv CY, Yang P, Yin QF. The synergistic antitumor effects of all-trans retinoic acid and C-phycocyanin on the lung cancer A549 cells in vitro and in vivo. Eur J Pharmacol 2015; 749:107-14. [PMID: 25617793 DOI: 10.1016/j.ejphar.2015.01.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 01/14/2015] [Accepted: 01/15/2015] [Indexed: 01/05/2023]
Abstract
The anticancer effects and mechanism of all-trans retinoic acid (ATRA), C-phycocyanin (C-PC) or ATRA+C-PC on the growth of A549 cells were studied in in vitro and in vivo experiments. The effects of C-PC and ATRA on the growth of A549 cells were determined. The expression of CDK-4 and caspase-3, and the cellular apoptosis levels were detected. The tumor model was established by subcutaneous injection of A549 cells to the left axilla of the NU/NU mice. The weights of tumor and the spleen were tested. The viabilities of T-cells and spleen cells, TNF levels, the expression of Bcl-2 protein and Cyclin D1 gene were examined. Results showed both C-PC and ATRA could inhibit the growth of tumor cells in vivo and in vitro. ATRA+C-PC cooperatively showed a higher antitumor activity. The dosage of ATRA was reduced when it was administered with C-PC together, and the toxicity was reduced as well. ATRA+C-PC could decrease CDK-4 but increase caspase-3 protein expression level and induce cell apoptosis. ATRA alone could lower the activities of T lymphocytes and spleen weights, but the combination with C-PC could effectively promote viability of T cells and spleen. C-PC+ATRA could up-regulate TNF, and down-regulate Bcl-2 and Cyclin D1 gene. The combination might inhibit tumor growth by inhibiting the progress of cell cycle, inducing cell apoptosis and enhancing the body immunity.
Collapse
Affiliation(s)
- Bing Li
- Department of Biology, Medical College of Qingdao University, Qingdao 266021, China.
| | - Mei-Hua Gao
- Department of Immunology, Medical College of Qingdao University, Qingdao 266021, China.
| | - Xian-Ming Chu
- Department of Cardiology, The Affiliated Hospital of Medical College of Qingdao University, Qingdao 266021, China.
| | - Lei Teng
- Department of Biology, Medical College of Qingdao University, Qingdao 266021, China.
| | - Cong-Yi Lv
- Department of Biology, Medical College of Qingdao University, Qingdao 266021, China.
| | - Peng Yang
- Department of Biology, Medical College of Qingdao University, Qingdao 266021, China.
| | - Qi-Feng Yin
- Department of Biology, Medical College of Qingdao University, Qingdao 266021, China.
| |
Collapse
|
24
|
Yang F, Li B, Chu XM, Lv CY, Xu YJ, Yang P. Molecular mechanism of inhibitory effects of C-phycocyanin combined with all-trans-retinoic acid on the growth of HeLa cells in vitro. Tumour Biol 2014; 35:5619-28. [PMID: 24563337 DOI: 10.1007/s13277-014-1744-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 02/10/2014] [Indexed: 01/06/2023] Open
Abstract
We studied the effects of all-trans-retinoic acid (ATRA), C-phycocyanin (C-PC), or ATRA+C-PC on the growth of cervical cells (HeLa cells), cell cycle distribution, and apoptosis. The anticancer mechanism of the drug combination was revealed. MTT assay was adopted to determine the effects of C-PC and ATRA on the growth of HeLa cells. The expression quantities of cyclin-dependent kinase (CDK) 4, cyclin D1, Bcl-2, caspase-3, and CD59 were determined by in situ hybridization, immunofluorescence, immunohistochemistry staining, Western blot, and RT-PCR. TUNEL assay was adopted to determine the cellular apoptosis levels. Both C-PC and ATRA could inhibit the growth of HeLa cells, and the combination of ATRA+C-PC functioned cooperatively to induce apoptosis in HeLa cells. The dosage of ATRA was reduced when it cooperated with C-PC to reduce the toxicity. ATRA treated with C-PC could induce more cell cycle arrests than the single drug used by decrease in cyclin D1 and CDK4 expression. The combination of the two drugs could upregulate caspase-3 and downregulate the Bcl-2 gene and induce cell apoptosis. Moreover, the combination therapy has an important immunological significance in decreased expression of the CD59 protein. Singly, C-PC or ATRA could inhibit the growth of HeLa cells, and the effects of treatment were further enhanced in the combination group. In combination with C-PC, the dosage of ATRA was effectively reduced. The C-PC + ATRA combination might take effect by inhibiting the progress of the cell cycle, inducing cell apoptosis and promoting complement-mediated cytolysis.
Collapse
Affiliation(s)
- Fan Yang
- Department of Biology, Medical College of Qingdao University, 38 Dengzhou Road, Qingdao, 266021, People's Republic of China,
| | | | | | | | | | | |
Collapse
|
25
|
Li R, Cui B, Li Y, Zhao C, Jia N, Wang C, Wu Y, Wen A. A new synthetic Cu(II) compound, [Cu3(p-3-bmb)2Cl4·(CH3OH)2]n, inhibits tumor growth in vivo and in vitro. Eur J Pharmacol 2014; 724:77-85. [DOI: 10.1016/j.ejphar.2013.12.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 12/02/2013] [Accepted: 12/04/2013] [Indexed: 12/26/2022]
|
26
|
Abstract
Gemcitabine is a pyrimidine antimetabolite which has shown activity in metastatic breast cancer both as a single agent, but also in various combination regimens. It is characterized by a unique mechanism of action which includes cytotoxic self-potentiation, masked DNA chain termination and potent inhibition of DNA repair. The clinical application of gemcitabine is supported by a favorable toxicity profile. In metastatic breast cancer, several Phase II trials document the activity of gemcitabine in pretreated and unpretreated patients. In a single Phase III trial performed in elderly patients not pretreated, gemcitabine was inferior to epirubicin. High activity has, however, been obtained by the combination of gemcitabine with taxanes such as paclitaxel or docetaxel. In a randomized trial performed in anthracycline-pretreated patients, the combined use of gemcitabine and paclitaxel induced a significant improvement not only of response rate and time to disease progression, but also caused a significant increase in quality of life and survival when compared with paclitaxel alone. The combination of gemcitabine with vinorelbine and cisplatin has been validated in numerous Phase II trials and promises reliable activity in anthracycline- and/or taxane-pretreated patients. Triple-agent regimens such as gemcitabine/epirubicin/paclitaxel provided consistently high response rates in Phase II trials, but failed to show superiority over the 5-fluorouracil/epirubicin/cyclophosphamide regimen in a randomized Phase III trial. Based on high response rates and pathological complete remission rates achieved by preoperative induction therapy with gemcitabine/epirubicin/taxane regimens, ongoing trials focus on the incorporation of gemcitabine into neoadjuvant and adjuvant regimens.
Collapse
Affiliation(s)
- Volker Heinemann
- Medical Clinic III, Klinikum Grosshadern, Marchioninistrasse 15, 81377, Munich, Germany.
| |
Collapse
|
27
|
Combination of fenretinide and indole-3-carbinol results in synergistic cytotoxic activity inducing apoptosis against human breast cancer cells in vitro. Anticancer Drugs 2013; 24:577-86. [PMID: 23542749 DOI: 10.1097/cad.0b013e328360a921] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The outcome in patients with breast cancer is not satisfactory to date, although new chemotherapy regimens have been introduced in clinics. Therefore, novel approaches are required for better management of patients with breast cancer. In this study, we tested the cytotoxic activity of a new combination of fenretinide, a synthetic retinoid, with indole-3-carbinol, a natural product present in vegetables such as broccoli and cabbage, against MCF-7 (estrogen receptor-positive) and MDA-MB-231 (estrogen receptor-negative) cell lines. It has been found that the combination resulted in more powerful cytotoxic activity, by induction of apoptosis, compared with that when they were used singly. In conclusion, this novel combination warrants in-vivo experiments to elucidate its possible use in the treatment of breast cancer.
Collapse
|
28
|
Ghosh RD, Banerjee K, Das S, Ganguly A, Chakraborty P, Sarkar A, Chatterjee M, Choudhuri SK. A novel manganese complex, Mn-(II) N-(2-hydroxy acetophenone) glycinate overcomes multidrug-resistance in cancer. Eur J Pharm Sci 2013; 49:737-47. [PMID: 23665413 DOI: 10.1016/j.ejps.2013.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 02/11/2013] [Accepted: 05/01/2013] [Indexed: 01/08/2023]
Abstract
Multidrug resistance (MDR) remains a significant problem for effective cancer chemotherapy. In spite of considerable advances in drug discovery, most of the cancer cases still stay incurable because of resistance to chemotherapy. We synthesized a novel, Mn (II) complex (chelate), viz., manganese N-(2-hydroxy acetophenone) glycinate (MnNG) that exhibits considerable efficacy to overcome drug resistant cancer. The antiproliferative activity of MnNG was studied on doxorubicin resistant and sensitive human T lymphoblastic leukemia cells (CEM/ADR 5000 and CCRF/CEM). MnNG induced apoptosis significantly in CEM/ADR 5000 cells probably through generation of reactive oxygen species. Moreover, intraperitoneal (i.p.) application of MnNG at non-toxic doses caused significant increase in the life-span of Swiss albino mice bearing sensitive and doxorubicin resistant subline of Ehrlich ascites carcinoma cells.
Collapse
Affiliation(s)
- Ruma Dey Ghosh
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, Kolkata, India
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Moysan E, Bastiat G, Benoit JP. Gemcitabine versus Modified Gemcitabine: a review of several promising chemical modifications. Mol Pharm 2012; 10:430-44. [PMID: 22978251 DOI: 10.1021/mp300370t] [Citation(s) in RCA: 168] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Gemcitabine, an anticancer agent which acts against a wide range of solid tumors, is known to be rapidly deaminated in blood to the inactive metabolite 2',2'-difluorodeoxyuridine and to be rapidly excreted by the urine. Moreover, many cancers develop resistance against this drug, such as loss of transporters and kinases responsible for the first phosphorylation step. To increase its therapeutic levels, gemcitabine is administered at high doses (1000 mg/m(2)) causing side effects (neutropenia, nausea, and so forth). To improve its metabolic stability and cytotoxic activity and to limit the phenomena of resistance many alternatives have emerged, such as the synthesis of prodrugs. Modifying an anticancer agent is not new; paclitaxel or ara-C has been subjected to such changes. This review summarizes the various chemical modifications that can be found in the 4-(N)- and 5'-positions of gemcitabine. They can provide (i) a protection against deamination, (ii) a better storage and (iii) a prolonged release in the cell, (iv) a possible use in the case of deoxycytidine kinase deficiency, and (v) transporter deficiency. These new gemcitabine-based sysems have the potential to improve the clinical outcome of a chemotherapy strategy.
Collapse
Affiliation(s)
- Elodie Moysan
- LUNAM Université -Micro et Nanomédecines Biomimétiques, F-49933 Angers, France
| | | | | |
Collapse
|
30
|
Zheng X, Cui XX, Gao Z, Verano M, Huang MT, Liu Y, Rabson AB, Conney AH. Effects of 12-O-tetradecanoylphorbol-13-acetate in combination with gemcitabine on Panc-1 pancreatic cancer cells cultured in vitro or Panc-1 tumors grown in immunodeficient mice. Int J Oncol 2012; 41:2269-75. [PMID: 23041978 DOI: 10.3892/ijo.2012.1651] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 08/17/2012] [Indexed: 11/06/2022] Open
Abstract
In the present study, the effects of 12-O-tetra-decanoylphorbol-13-acetate (TPA) alone or in combination with gemcitabine on the growth of Panc-1 pancreatic cancer cells cultured in vitro or grown in NCr immunodeficient nude mice were investigated. Combinations of TPA and gemcitabine synergi-stically inhibited the growth and induced apoptosis in Panc-1 cells. The combination of TPA (0.16 nM) and gemcitabine (0.5 µM) induced a marked increase in phosphorylated c-Jun NH2-terminal kinase (JNK) in the Panc-1 cells. In animal experiments, NCr nude mice with established Panc-1 tumors received daily intraperitoneal (i.p.) injections of TPA (50 ng/g body weight/day) or gemcitabine (0.5 µg/g body weight/day) alone or in combination for 26 days. Treatment with daily i.p. injections of low doses of TPA or gemcitabine alone had a modest inhibitory effect on the growth of the tumors. However, the combination of low doses of TPA and gemcitabine more potently inhibited the growth of Panc-1 tumors than either agent used individually. Treatment with TPA or gemcitabine alone or in combination did not affect the body weight of the animals. Clinical trials with TPA alone or in combination with gemcitabine on patients with pancreatic cancer are warranted in order to confirm our results.
Collapse
Affiliation(s)
- Xi Zheng
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
SL-01, an oral gemcitabine derivative, inhibited human cancer growth more potently than gemcitabine. Toxicol Appl Pharmacol 2012; 262:293-300. [DOI: 10.1016/j.taap.2012.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2012] [Revised: 04/27/2012] [Accepted: 05/11/2012] [Indexed: 11/20/2022]
|
32
|
Kongpetch S, Kukongviriyapan V, Prawan A, Senggunprai L, Kukongviriyapan U, Buranrat B. Crucial role of heme oxygenase-1 on the sensitivity of cholangiocarcinoma cells to chemotherapeutic agents. PLoS One 2012; 7:e34994. [PMID: 22514698 PMCID: PMC3325916 DOI: 10.1371/journal.pone.0034994] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 03/09/2012] [Indexed: 01/03/2023] Open
Abstract
Cancer cells acquire drug resistance via various mechanisms including enhanced cellular cytoprotective and antioxidant activities. Heme oxygenase-1 (HO-1) is a key enzyme exerting potent cytoprotection, cell proliferation and drug resistance. We aimed to investigate roles of HO-1 in human cholangiocarcinoma (CCA) cells for cytoprotection against chemotherapeutic agents. KKU-100 and KKU-M214 CCA cell lines with high and low HO-1 expression levels, respectively, were used to evaluate the sensitivity to chemotherapeutic agents, gemcitabine (Gem) and doxorubicin. Inhibition of HO-1 by zinc protoporphyrin IX (ZnPP) sensitized both cell types to the cytotoxicity of chemotherapeutic agents. HO-1 gene silencing by siRNA validated the cytoprotective effect of HO-1 on CCA cells against Gem. Induction of HO-1 protein expression by stannous chloride enhanced the cytoprotection and suppression of apoptosis caused by anticancer agents. The sensitizing effect of ZnPP was associated with increased ROS formation and loss of mitochondrial transmembrane potential, while Gem alone did not show any effects. A ROS scavenger, Tempol, abolished the sensitizing effect of ZnPP on Gem. Combination of ZnPP and Gem enhanced the release of cytochrome c and increased p21 levels. The results show that HO-1 played a critical role in cytoprotection in CCA cells against chemotherapeutic agents. Targeted inhibition of HO-1 may be a strategy to overcome drug resistance in chemotherapy of bile duct cancer.
Collapse
Affiliation(s)
- Sarinya Kongpetch
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Veerapol Kukongviriyapan
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand
| | - Auemduan Prawan
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand
| | - Laddawan Senggunprai
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- Liver Fluke and Cholangiocarcinoma Research Center, Khon Kaen University, Khon Kaen, Thailand
| | - Upa Kukongviriyapan
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Benjaporn Buranrat
- Faculty of Pharmaceutical Sciences, University of Phayao, Phayao, Thailand
| |
Collapse
|
33
|
Yang S, Liu J, Chen Y, Jiang J. Reversal effect of Tween-20 on multidrug resistance in tumor cells in vitro. Biomed Pharmacother 2012; 66:187-94. [PMID: 22440896 DOI: 10.1016/j.biopha.2011.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 10/31/2011] [Indexed: 10/14/2022] Open
Abstract
Multidrug resistance (MDR) is a major barrier for chemotherapy of many cancers. Non-ionic surfactants have great potential to reverse the MDR by preventing onset or delay progression of the carcinogenic process. However, the role of Tween-20 in the development of MDR remains unknown. The aim of this study was to explore the reversal effect and potential mechanism of Tween-20 on tumor cells in vitro. Alamar Blue assay was used to examine the reversal index of Tween-20 to vincristine (VCR), doxorubicin (DOX) and 5-fluorouracil (5-FU) in KBv200, HepG2/R and Bel-7402/5-FU, respectively. Morphological change was determined by Gimsa and Hoechst 33258 staining. The acumulation of DOX was confirmed by spectrofluorimetric assay. Cell cycle analysis was performed using flow cytometry. The mRNA and protein expression levels of MDR were assessed by semiquantitative RT-PCR and dot blot, respectively. The results showed that Tween-20 at concentrations of 0.0025%, 0.005%, 0.01% had little cytotoxicity. When combined with the cancer drugs, it significantly promoted the sensitivity of MDR cells. Fluorescence staining confirmed that the percentage of apoptotic cell increased when combined with Tween-20. This notion was further supported by the observation that Tween-20 treatment potentiated VIN-induced G2/M arrest of the cell cycle. Furthermore, Tween-20 treatment increased significantly intracellular accumulation of DOX. RT-PCR and dot blot revealed that Tween-20 could downregulate the expression of MDR and P-glycoprotein. Low concentrations of Tween-20 can efficiently reverse the multidrug resistance phenotype by enhancing accumulation of the anticancer drugs. The potential mechanism may be via inhibiting the multidrug-resistant gene expression.
Collapse
Affiliation(s)
- Shouhui Yang
- Key Laboratory of Biotechnology for Medicinal Plants of Jiangsu Province, Xuzhou Normal University, 101 Shanghai Road, Xuzhou, China
| | | | | | | |
Collapse
|
34
|
Chougule MB, Patel A, Sachdeva P, Jackson T, Singh M. Enhanced anticancer activity of gemcitabine in combination with noscapine via antiangiogenic and apoptotic pathway against non-small cell lung cancer. PLoS One 2011; 6:e27394. [PMID: 22102891 PMCID: PMC3216931 DOI: 10.1371/journal.pone.0027394] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Accepted: 10/16/2011] [Indexed: 11/28/2022] Open
Abstract
Background The aim of this investigation was to evaluate the anticancer activity of Noscapine (Nos) and Gemcitabine (Gem) combination (NGC) against non-small cell lung cancer (NSCLC) and to elucidate the underlying mechanism of action. Methods Isobolographic method was used to calculate combination index values from cytotoxicity data. In vitro antiangiogenic and apoptotic activity of Nos, Gem and NGC was evaluated. For in vivo studies, female athymic Nu/nu mice were xenografted with H460 tumors and the efficacy of Nos, Gem, or NGC was determined. Protein expressions by immunohistochemical staining were evaluated in harvested tumor tissues. Results The CI values (<0.59) were suggestive of synergistic behavior between Nos and Gem. NGC treatment showed significantly inhibited tube formation and increased percentage of apoptotic cells. NGC, Gem and Nos treatment reduced tumor volume by 82.9±4.5 percent, 39.4±5.8 percent and 34.2±5.7 percent respectively. Specifically, NGC treatment decreased expression cell survival proteins; VEGF, CD31 staining and microvessel density and enhanced DNA fragmentation and cleaved caspase 3 levels compared to single agent treated and control groups. Conclusion Nos potentiated the anticancer activity of Gem in an additive to synergistic manner against lung cancer via antiangiogenic and apoptotic pathways. These findings suggest potential benefit for use of NGC chemotherapy for treatment of lung cancer.
Collapse
Affiliation(s)
- Mahavir B. Chougule
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Hawaii, Hilo, Hawaii, United States of America
| | - Apurva Patel
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, United States of America
| | - Pratik Sachdeva
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, United States of America
| | - Tanise Jackson
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, United States of America
| | - Mandip Singh
- Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida, United States of America
- * E-mail:
| |
Collapse
|
35
|
Bildstein L, Pili B, Marsaud V, Wack S, Meneau F, Lepêtre-Mouelhi S, Desmaële D, Bourgaux C, Couvreur P, Dubernet C. Interaction of an amphiphilic squalenoyl prodrug of gemcitabine with cellular membranes. Eur J Pharm Biopharm 2011; 79:612-20. [DOI: 10.1016/j.ejpb.2011.07.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2010] [Revised: 06/27/2011] [Accepted: 07/08/2011] [Indexed: 01/18/2023]
|
36
|
Aljuffali IA, Mock JN, Costyn LJ, Nguyen H, Nagy T, Cummings BS, Arnold RD. Enhanced antitumor activity of low-dose continuous administration schedules of topotecan in prostate cancer. Cancer Biol Ther 2011; 12:407-20. [PMID: 21709443 DOI: 10.4161/cbt.12.5.15950] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
PURPOSE The objective of this study was to determine the antitumor effects of alternate dosing schedules of topotecan in prostate cancer. RESULTS A concentration-dependent increase in cytotoxicity was observed in PC-3 and LNCaP cells after topotecan treatment using conventional and metronomic protocols. A significant increase in potency (2.4-18 fold, after 72 hr) was observed following metronomic dosing compared to conventional dosing administration in both cell lines. Metronomic dosing also increased the percentage of PC-3 cells in the G2/M, compared to control, but did not alter LNCaP cell cycle distribution. Metronomic dosing increased p21 protein expression in LNCaP and PC-3 cells compared to conventional dosing. The observed in vitro activity was confirmed using an in vivo model of human prostate cancer. Metronomic dosing and continuous infusion decreased tumor volume significantly (p < 0.05) compared to control and conventional topotecan treatment, but had no effect on tumor vascular staining. METHODS The cytotoxicity of topotecan after conventional or metronomic dosing was determined by examining cellular morphology, mitochondrial enzymatic activity (MTT), total cellular protein (SRB), annexin V and propidium iodine (PI) staining, cell cycle and western blot analysis in human prostate cancer cell lines (PC-3 and LNCaP) and the effects metronomic or continuous infusion on tumor growth in an in vivo tumor xenograft model. CONCLUSIONS These data support the hypothesis that low-dose continuous administration of topotecan increases potency compared to conventional dosing in prostate cancer. These data also suggest the novel finding that the enhanced antitumor activity of topotecan following low-dose exposure correlates to alterations in cell cycle and increased p21 expression.
Collapse
Affiliation(s)
- Ibrahim A Aljuffali
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, GA USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Nagoor NH, Shah Jehan Muttiah N, Soon Lim C, In LLA, Mohammad K, Awang K. Regulation of apoptotic effects by erythrocarpine E, a cytotoxic limonoid from Chisocheton erythrocarpus in HSC-4 human oral cancer cells. PLoS One 2011; 6:e23661. [PMID: 21858194 PMCID: PMC3157399 DOI: 10.1371/journal.pone.0023661] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 07/22/2011] [Indexed: 12/20/2022] Open
Abstract
The aim of this study was to determine the cytotoxic and apoptotic effects of erythrocarpine E (CEB4), a limonoid extracted from Chisocheton erythrocarpus on human oral squamous cell carcinoma. Based on preliminary dimethyl-2-thiazolyl-2,5-diphenyl-2H-tetrazolium bromide (MTT) assays, CEB4 treated HSC-4 cells demonstrated a cytotoxic effect and inhibited cell proliferation in a time and dose dependent manner with an IC(50) value of 4.0±1.9 µM within 24 h of treatment. CEB4 was also found to have minimal cytotoxic effects on the normal cell line, NHBE with cell viability levels maintained above 80% upon treatment. Annexin V-fluorescein isothiocyanate (FITC), poly-ADP ribose polymerase (PARP) cleavage and DNA fragmentation assay results showed that CEB4 induces apoptosis mediated cell death. Western blotting results demonstrated that the induction of apoptosis by CEB4 appeared to be mediated through regulation of the p53 signalling pathway as there was an increase in p53 phosphorylation levels. CEB4 was also found to up-regulate the pro-apoptotic protein, Bax, while down-regulating the anti-apoptotic protein, Bcl-2, suggesting the involvement of the intrinsic mitochondrial pathway. Reduced levels of initiator procaspase-9 and executioner caspase-3 zymogen were also observed following CEB4 exposure, hence indicating the involvement of cytochrome c mediated apoptosis. These results demonstrate the cytotoxic and apoptotic ability of erythrocarpine E, and suggest its potential development as a cancer chemopreventive agent.
Collapse
Affiliation(s)
- Noor Hasima Nagoor
- Faculty of Science, Institute of Biological Science (Genetics and Molecular Biology), University Malaya, Kuala Lumpur, Malaysia
| | - Norliza Shah Jehan Muttiah
- Faculty of Science, Institute of Biological Science (Genetics and Molecular Biology), University Malaya, Kuala Lumpur, Malaysia
| | - Chong Soon Lim
- Department of Chemistry, Faculty of Science, Centre for Natural Product Research and Drug Discovery (CENAR), University Malaya, Kuala Lumpur, Malaysia
| | - Lionel L. A. In
- Faculty of Science, Institute of Biological Science (Genetics and Molecular Biology), University Malaya, Kuala Lumpur, Malaysia
| | - Khalit Mohammad
- Department of Pharmacy, Faculty of Medicine, University Malaya, Kuala Lumpur, Malaysia
| | - Khalijah Awang
- Department of Chemistry, Faculty of Science, Centre for Natural Product Research and Drug Discovery (CENAR), University Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
38
|
Ghosh RD, Das S, Ganguly A, Banerjee K, Chakraborty P, Sarkar A, Chatterjee M, Nanda A, Pradhan K, Choudhuri SK. An in vitro and in vivo study of a novel zinc complex, zinc N-(2-hydroxyacetophenone)glycinate to overcome multidrug resistance in cancer. Dalton Trans 2011; 40:10873-84. [PMID: 21717020 DOI: 10.1039/c1dt10501a] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Multiple drug resistance (MDR) remains a major clinical challenge for cancer treatment. P-glycoprotein is the major contributor and they exceed their role in the chemotherapy resistance of most of the malignancies. Attempts in several preclinical and clinical studies to reverse the MDR phenomenon by using MDR modulators have not yet generated promising results. In the present study, a co-ordination complex of zinc viz., Zn N-(2-hydroxyacetophenone)glycinate (ZnNG) has been synthesized, characterized and its antitumour activity was tested in vitro against drug sensitive and resistant human T-lymphoblastic leukemic cell lines (CCRF/CEM and CEM/ADR5000 respectively) and in vivo against Ehrlich ascites carcinoma (EAC) implanted in female Swiss albino mice. To evaluate the cytotoxic potential of ZnNG, we used sensitive CCRF/CEM and drug resistant CEM/ADR 5000 cell lines in vitro. Moreover, ZnNG also has the potential ability to reverse the multidrug resistance phenotype in drug resistant CEM/ADR 5000 cell line and induces apoptosis in combination with vinblastine. ZnNG remarkably increases the life span of Swiss albino mice bearing sensitive and doxorubicin resistant subline of EAC in presence and in absence of doxorubicin. In addition, intraperitoneal application of ZnNG in mice does not show any systemic toxicity in preliminary trials in normal mice. To conclude, a novel metal chelate of zinc viz., ZnNG, may be a promising therapeutic agent against sensitive as well as drug resistant cancers.
Collapse
Affiliation(s)
- Ruma Dey Ghosh
- Department of In Vitro Carcinogenesis and Cellular Chemotherapy, Chittaranjan National Cancer Institute, 37, S.P. Mukherjee Road, 700 026, Kolkata, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Sung V, Richard N, Brady H, Maier A, Kelter G, Heise C. Histone deacetylase inhibitor MGCD0103 synergizes with gemcitabine in human pancreatic cells. Cancer Sci 2011; 102:1201-7. [PMID: 21375679 DOI: 10.1111/j.1349-7006.2011.01921.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Histone deacetylase inhibitors are a group of recently developed compounds that modulate cell growth and survival. We evaluated the effects of the histone deacetylase inhibitor MGCD0103 on growth of pancreatic carcinoma models following single agent treatment and in combination with gemcitabine. MGCD0103 inhibited tumor cell growth and acted synergistically with gemcitabine to enhance its cytotoxic effects. Gene expression analysis identified the cell cycle pathway as one of the most highly modulated gene groups. Our data suggest that MGCD0103 + gemcitabine might be an effective treatment for gemcitabine-refractory pancreatic cancer.
Collapse
|
40
|
Li R, Chen WC, Pang XQ, Tian WY, Zhang XG. Influence of sCD40L on gastric cancer cell lines. Mol Biol Rep 2011; 38:5459-64. [DOI: 10.1007/s11033-011-0702-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2010] [Accepted: 03/01/2011] [Indexed: 01/30/2023]
|
41
|
Zhao H, Zhang M, Zhao L, Ge YK, Sheng J, Shi W. Changes of constituents and activity to apoptosis and cell cycle during fermentation of tea. Int J Mol Sci 2011; 12:1862-75. [PMID: 21673927 PMCID: PMC3111638 DOI: 10.3390/ijms12031862] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 02/24/2011] [Accepted: 02/28/2011] [Indexed: 11/16/2022] Open
Abstract
Tea is believed to be beneficial for health, and the effects of the fermentation process on its contributions to apoptosis and cell cycle arrest of gastric cancer cells have not been completely investigated. In this study, the chemical components in green tea, black tea and pu-erh tea aqueous extracts were analyzed and compared. The polysaccharide and caffeine levels were substantially higher in the fermented black tea and pu-erh tea, while the polyphenol level was higher in the unfermented green tea. Hence, a treatment of tea aqueous extract and the components, which are emerging as promising anticancer agents, were pursued to determine whether this treatment could lead to enhance apoptosis and cell cycle arrest. In the human gastric cancer cell line SGC-7901, the cell viability and flow cytometry analysis for apoptotic cells indicated effects in a dose-dependent inhibition manner for the three tea treatment groups. The apoptosis rates were found to be elevated after 48 h of treatment with 31.2, 125, and 500 μg/mL of green tea extract, the higher catechins content may be involved in the mechanism. Cell cycle was arrested in S phase in the fermented black tea and pu-erh tea, and the populations were significantly decreased in G2/M phases, possibly due to the oxidation of tea polyphenols, which causes an increase of theabrownins. CCC-HEL-1 normal cells were not sensitive to tea extract. These findings suggest that the fermentation process causes changes of the compounds which might be involved in the changes of cell proliferation inhibition, apoptosis induction and cell cycle arrest.
Collapse
Affiliation(s)
- Hang Zhao
- Key Laboratory for Molecular Enzymology & Engineering, the Ministry of Education, Jilin University, Changchun 130012, China; E-Mails: (H.Z.); (M.Z.); (L.Z.); (Y.-K.G.)
- College of Life Science, Jilin University, Changchun 130012, China
| | - Min Zhang
- Key Laboratory for Molecular Enzymology & Engineering, the Ministry of Education, Jilin University, Changchun 130012, China; E-Mails: (H.Z.); (M.Z.); (L.Z.); (Y.-K.G.)
- College of Life Science, Jilin University, Changchun 130012, China
| | - Lu Zhao
- Key Laboratory for Molecular Enzymology & Engineering, the Ministry of Education, Jilin University, Changchun 130012, China; E-Mails: (H.Z.); (M.Z.); (L.Z.); (Y.-K.G.)
- College of Life Science, Jilin University, Changchun 130012, China
| | - Ya-kun Ge
- Key Laboratory for Molecular Enzymology & Engineering, the Ministry of Education, Jilin University, Changchun 130012, China; E-Mails: (H.Z.); (M.Z.); (L.Z.); (Y.-K.G.)
- College of Life Science, Jilin University, Changchun 130012, China
| | - Jun Sheng
- College of Life Science, Jilin University, Changchun 130012, China
- Yunnan Research Centre for Advance Tea Processing, Yunnan Agricultural University, Kunming 650201, China
- Authors to whom correspondence should be addressed; E-Mails: (J.S.); (W.S.); Tel.: +86-431-85155216; Fax: +86-431-85155200
| | - Wei Shi
- Key Laboratory for Molecular Enzymology & Engineering, the Ministry of Education, Jilin University, Changchun 130012, China; E-Mails: (H.Z.); (M.Z.); (L.Z.); (Y.-K.G.)
- College of Life Science, Jilin University, Changchun 130012, China
- Authors to whom correspondence should be addressed; E-Mails: (J.S.); (W.S.); Tel.: +86-431-85155216; Fax: +86-431-85155200
| |
Collapse
|
42
|
Retention of the in vitro radiosensitizing potential of gemcitabine under anoxic conditions, in p53 wild-type and p53-deficient non-small-cell lung carcinoma cells. Int J Radiat Oncol Biol Phys 2011; 80:558-66. [PMID: 21377279 DOI: 10.1016/j.ijrobp.2010.12.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2010] [Revised: 12/08/2010] [Accepted: 12/17/2010] [Indexed: 11/23/2022]
Abstract
PURPOSE Whereas radiosensitization by gemcitabine is well studied under normal oxygen conditions, little is known about its radiosensitizing potential under reduced oxygen conditions. Therefore, the present study evaluated the impact of anoxia on gemcitabine-mediated radiosensitization. METHODS AND MATERIALS The clonogenic assay was performed in three isogenic A549 cell lines differing in p53 status (24 h, 0-15 nM gemcitabine, 0-8 Gy irradiation, normoxia vs. anoxia). Using radiosensitizing conditions, cells were collected for cell cycle analysis and apoptosis detection. RESULTS Whereas wild-type p53 A549-LXSN cells were more sensitive to radiation than p53-deficient A549-E6 cells, both cell lines showed similar radiosensitization by gemcitabine under normoxia and anoxia. Independent of p53 functionality, gemcitabine was able to overcome anoxia-induced G(0/1) arrest and established an (early) S phase block in normoxic and anoxic cells. The percentage early and late apoptotic/necrotic cells increased with the gemcitabine/radiation combination, with a significant difference between A549-LXSN and A549-E6. CONCLUSIONS This study is the first to show that gemcitabine retains its radiosensitizing potential under low oxygen conditions. Although radiosensitization was observed in both p53 wild-type and p53-deficient cells, p53 status might influence induction of apoptosis after gemcitabine/radiation treatment, whereas no effect on cell cycle progression was noticed.
Collapse
|
43
|
Iwata K, Aizawa K, Sakai S, Jingami S, Fukunaga E, Yoshida M, Hamada A, Saito H. The Relationship between Treatment Time of Gemcitabine and Development of Hematologic Toxicity in Cancer Patients. Biol Pharm Bull 2011; 34:1765-8. [DOI: 10.1248/bpb.34.1765] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Kazufumi Iwata
- Department of Pharmacy, Kumamoto Red Cross Hospital
- Department of Pharmacy, Kumamoto University Hospital
| | - Keiji Aizawa
- Department of Pharmacy, Kumamoto Red Cross Hospital
| | - Saori Sakai
- Department of Pharmacy, Kumamoto Red Cross Hospital
| | | | | | - Minoru Yoshida
- Department of Medical Oncology, Kumamoto Red Cross Hospital
| | | | | |
Collapse
|
44
|
A phase I study of topotecan and gemcitabine in advanced solid tumors. Invest New Drugs 2010; 29:1390-4. [PMID: 20574789 DOI: 10.1007/s10637-010-9480-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2010] [Accepted: 06/15/2010] [Indexed: 10/19/2022]
Abstract
PURPOSE Gemcitabine and topotecan are commonly used anti-tumor agents with a wide spectrum of activity in vitro and in vivo. A phase I trial of a combination of these two agents was initiated based on the premise that both gemcitabine and topotecan cause DNA damage and interfere with DNA repair by different mechanisms. Synergism has been demonstrated in vitro when gemcitabine and other topoisomerase I inhibitors have been combined. PATIENTS AND METHODS Seventeen patients with advanced solid tumors signed consent and were treated on this study with at least one cycle. Treatment consisted of gemcitabine at doses of 400 to 625 mg/m(2) days 1 and 5 in combination with topotecan at doses of 0.8 to 1 mg/m(2) given on days 2 through 5 every 21 days. RESULTS The dose limiting toxicities of granulocytopenia and thrombocytopenia were reached at the highest dose level of gemcitabine 625 mg/m(2) and topotecan 1 mg/m(2). A diffuse skin rash was also seen in four treated patients and responded well to treatment with steroids. One partial response and seven stable disease were seen as best response in 16 evaluable patients. CONCLUSION The combination of gemcitabine and topotecan was found to be tolerable with interesting preliminary activity. The recommended phase II dose for this combination is gemcitabine at 500 mg/m(2) on days 1 and 5 with topotecan at 0.8 mg/m(2) on days 2 to 5.
Collapse
|
45
|
In vitro study of the anti-cancer effects of artemisone alone or in combination with other chemotherapeutic agents. Cancer Chemother Pharmacol 2010; 67:569-77. [DOI: 10.1007/s00280-010-1355-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2010] [Accepted: 04/30/2010] [Indexed: 11/25/2022]
|
46
|
Guo X, Xu B, Pandey S, Goessl E, Brown J, Armesilla AL, Darling JL, Wang W. Disulfiram/copper complex inhibiting NFkappaB activity and potentiating cytotoxic effect of gemcitabine on colon and breast cancer cell lines. Cancer Lett 2009; 290:104-13. [PMID: 19782464 DOI: 10.1016/j.canlet.2009.09.002] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2009] [Revised: 08/30/2009] [Accepted: 09/02/2009] [Indexed: 10/20/2022]
Abstract
Most of the gemcitabine (dFdC) resistant cell lines manifested high NFkappaB activity. The NFkappaB activity can be induced by dFdC and 5-FU exposure. The chemosensitizing effect of disulfiram (DS), an anti-alcoholism drug and NFkappaB inhibitor, and copper (Cu) on the chemoresistant cell lines was examined. The DS/Cu complex significantly enhanced the cytotoxicity of dFdC (resistant cells: 12.2-1085-fold) and completely reversed the dFdC resistance in the resitant cell lines. The dFdC-induced NFkappaB activity was markedly inhibited by DS/Cu complex. The data from this study indicated that DS may be used in clinic to improve the therapeutic effect of dFdC in breast and colon cancer patients.
Collapse
Affiliation(s)
- Xiaoxia Guo
- Research Institute in Healthcare Science, University of Wolverhampton, UK
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Background: The aim of this study was to evaluate the radiosensitising effect of gemcitabine, in terms of cell-cycle progression, induction of apoptosis, and to investigate the molecular events regulating apoptosis. Methods: Tumour cells were treated with gemcitabine, radiation, or the combination. 0–72 h after treatment, cells were collected for cell-cycle analysis and apoptosis determination. Caspase 8 and 9, Bid and tBid expression were determined by western blot. The mitochondrial membrane potential was determined using flow cytometry. An RT2Profiler PCR Array for human apoptotic genes was performed after the combination or TRAIL treatment. Results: Gemcitabine and radiation resulted in an early S-phase block immediately after treatment, after which the cells moved synchronously through the cell cycle. When cell-cycle distribution returned to pre-treatment levels, an increased induction of apoptosis was observed with activation of caspase 8 and 9 and a reduction of the mitochondrial membrane potential. Gene expression after treatment with radiosensitising conditions was comparable with expression after the TRAIL treatment. Conclusion: A role for the cell-cycle perturbations and the induction of apoptosis could be attributed to the radiosensitising effect of gemcitabine. Apoptosis induction was comparable with the apoptotic pathway observed after the TRAIL treatment, that is the involvement of the extrinsic apoptosis pathway.
Collapse
|
48
|
Mine T, Matsueda S, Li Y, Tokumitsu H, Gao H, Danes C, Wong KK, Wang X, Ferrone S, Ioannides CG. Breast cancer cells expressing stem cell markers CD44+ CD24 lo are eliminated by Numb-1 peptide-activated T cells. Cancer Immunol Immunother 2009; 58:1185-94. [PMID: 19048252 PMCID: PMC2726795 DOI: 10.1007/s00262-008-0623-1] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2008] [Accepted: 10/29/2008] [Indexed: 12/22/2022]
Abstract
Cancer stem cells (CSC) are resistant to chemo- and radiotherapy. To eliminate cells with phenotypic markers of CSC-like we characterized: (1) expression of CD44, CD24, CD133 and MIC-A/B (NKG2 receptors) in breast (MCF7) and ovarian (SK-OV-3) cells resistant to gemcitabine (GEM), paclitaxel (PTX) and 5-fluorouracil (5-FU) and (2) their elimination by Numb- and Notch-peptide activated CTL. The number of cells in all populations with the luminal CSC phenotype [epithelial specific antigen(+) (ESA) CD44(hi) CD24(lo), CD44(hi) CD133(+), and CD133(+) CD24(lo)] increased in drug-resistant MCF7 and SK-OV-3 cells. Similarly, the number of cells with expressed MIC-A/B increased 4 times in drug-resistant tumor cells compared with drug-sensitive cells. GEM(Res) MCF7 cells had lower levels of the Notch-1-extracellular domain (NECD) and Notch trans-membrane intracellular domain (TMIC) than GEM(Sens) MCF7. The levels of Numb, and Numb-L-[P]-Ser(265) were similar in GEM(Res) and GEM(Sens) MCF7 cells. Only the levels of Numb-L (long)-Ser(295) decreased slightly. This finding suggests that Notch-1 cleavage to TMIC is inhibited in GEM(Res) MCF7 cells. PBMC activated by natural immunogenic peptides Notch-1 (2112-2120) and Numb-1 (87-95) eliminated NICD(positive), CD24(hi) CD24(lo) MCF7 cells. It is likely that the immunogenic Numb-1 peptide in MCF7 cells originated from Numb, [P]-lated by an unknown kinase, because staurosporine but not wortmannin and MAPK-inhibitors decreased peptide presentation. Numb and Notch are antagonistic proteins which degrade each other to stop and activate cell proliferation, respectively. Their peptides are presented alternatively. Targeting both antagonistic proteins should be useful to prevent metastases in patients whose tumors are resistant to conventional treatments.
Collapse
MESH Headings
- Anticarcinogenic Agents/pharmacology
- Biomarkers, Tumor/immunology
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/immunology
- Breast Neoplasms/metabolism
- CD24 Antigen/immunology
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Cell Line, Tumor
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/physiology
- Female
- GPI-Linked Proteins
- Humans
- Hyaluronan Receptors/immunology
- Immunotherapy, Active
- Immunotherapy, Adoptive
- Intercellular Signaling Peptides and Proteins/immunology
- Intercellular Signaling Peptides and Proteins/metabolism
- Interferon-gamma/biosynthesis
- Interferon-gamma/immunology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/immunology
- Leukocytes, Mononuclear/metabolism
- Lymphocyte Activation
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Neoplastic Stem Cells/immunology
- Neoplastic Stem Cells/metabolism
- Nerve Tissue Proteins/immunology
- Nerve Tissue Proteins/metabolism
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/metabolism
- Peptides/immunology
- Receptor, Notch1/immunology
- Receptor, Notch1/metabolism
Collapse
Affiliation(s)
- Takashi Mine
- Department of Gynecologic Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Adema AD, van der Born K, Honeywell RJ, Peters GJ. Cell cycle effects and increased adduct formation by temozolomide enhance the effect of cytotoxic and targeted agents in lung cancer cell lines. J Chemother 2009; 21:338-346. [PMID: 19567356 DOI: 10.1179/joc.2009.21.3.338] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Temozolomide (TMZ) exerts its cytotoxic effects by methylating guanine in DNA, resulting in a mismatch with thymine. We studied possible enhancement of the cytotoxic activity of several other targeted drugs in four lung cancer cell lines by TMZ. the data are in relation to O(6)-alkylguanine-DNA-alkyltransferase (AGT) expression, gene methylation, cell cycle distribution and adduct formation. Synergism/additivity was found with O(6)-BG), gemcitabine, lonafarnib and paclitaxel, but not with platinum analogs and topoisomerase-inhibitors. O(6)-BG enhanced TMZ-induced accumulation in the G2/m-phase by increasing formation and retention of the O(6)-methyldeoxyguanosine adducts. TMZ combinations with drugs showing a different individual effect on the cell cycle (e.g. gemcitabine-induced S-phase) were most effective. The results show that O(6)-BG enhanced the TMZ effect in all cell lines. TMZ enhanced the cytotoxicity of gemcitabine, paclitaxel and lonafarnib in most cell lines, possibly by affecting the cell cycle, supporting possible application of TMZ in the treatment of lung cancer.
Collapse
Affiliation(s)
- A D Adema
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | |
Collapse
|
50
|
WAN ZAINON WMN, BORG M, HIGGS B, YEOH E, KOTASEK D, PARNIS F, HILLENBAND E, HAMZAH S. Gemcitabine-induced radiation recall in the treatment of pancreatic cancer. Asia Pac J Clin Oncol 2009. [DOI: 10.1111/j.1743-7563.2009.01187.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|