1
|
Ligasová A, Piskláková B, Friedecký D, Koberna K. A new technique for the analysis of metabolic pathways of cytidine analogues and cytidine deaminase activities in cells. Sci Rep 2023; 13:20530. [PMID: 37993628 PMCID: PMC10665361 DOI: 10.1038/s41598-023-47792-4] [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: 10/15/2023] [Accepted: 11/18/2023] [Indexed: 11/24/2023] Open
Abstract
Deoxycytidine analogues (dCas) are widely used for the treatment of malignant diseases. They are commonly inactivated by cytidine deaminase (CDD), or by deoxycytidine monophosphate deaminase (dCMP deaminase). Additional metabolic pathways, such as phosphorylation, can substantially contribute to their (in)activation. Here, a new technique for the analysis of these pathways in cells is described. It is based on the use of 5-ethynyl 2'-deoxycytidine (EdC) and its conversion to 5-ethynyl 2'-deoxyuridine (EdU). Its use was tested for the estimation of the role of CDD and dCMP deaminase in five cancer and four non-cancer cell lines. The technique provides the possibility to address the aggregated impact of cytidine transporters, CDD, dCMP deaminase, and deoxycytidine kinase on EdC metabolism. Using this technique, we developed a quick and cheap method for the identification of cell lines exhibiting a lack of CDD activity. The data showed that in contrast to the cancer cells, all the non-cancer cells used in the study exhibited low, if any, CDD content and their cytidine deaminase activity can be exclusively attributed to dCMP deaminase. The technique also confirmed the importance of deoxycytidine kinase for dCas metabolism and indicated that dCMP deaminase can be fundamental in dCas deamination as well as CDD. Moreover, the described technique provides the possibility to perform the simultaneous testing of cytotoxicity and DNA replication activity.
Collapse
Affiliation(s)
- Anna Ligasová
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic.
| | - Barbora Piskláková
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic
- Laboratory of Inherited Metabolic Disorders, Department of Clinical Chemistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - David Friedecký
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic
- Laboratory of Inherited Metabolic Disorders, Department of Clinical Chemistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Karel Koberna
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic.
| |
Collapse
|
2
|
Jang Y, Shin JS, Lee MK, Jung E, An T, Kim UI, Kim K, Kim M. Comparison of Antiviral Activity of Gemcitabine with 2'-Fluoro-2'-Deoxycytidine and Combination Therapy with Remdesivir against SARS-CoV-2. Int J Mol Sci 2021; 22:ijms22041581. [PMID: 33557278 PMCID: PMC7915419 DOI: 10.3390/ijms22041581] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 01/28/2021] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the coronavirus disease 2019 (COVID-19) pandemic. The virus still spreads globally through human-to-human transmission. Nevertheless, there are no specific treatments clinically approved. This study aimed to compare antiviral activity of gemcitabine and its analogue 2′-fluoro-2′-deoxycytidine (2FdC) against SARS-CoV-2 as well as cytotoxicity in vitro. Fluorescent image-based antiviral assays revealed that gemcitabine was highly potent, with a 50% effective concentration (EC50) of 1.2 μM, more active than the well-known nucleoside monophosphate remdesivir (EC50 = 35.4 μM). In contrast, 2FdC was marginally active (EC50 = 175.2 μM). For all three compounds, the 50% cytotoxic concentration (CC50) values were over 300 μM toward Vero CCL-81 cells. Western blot and quantitative reverse-transcription polymerase chain reaction analyses verified that gemcitabine blocked viral protein expression in virus-infected cells, not only Vero CCL-81 cells but also Calu-3 human lung epithelial cells in a dose-dependent manner. It was found that gemcitabine has a synergistic effect when combined with remdesivir. This report suggests that the difluoro group of gemcitabine is critical for the antiviral activity and that its combination with other evaluated antiviral drugs, such as remdesivir, could be a desirable option to treat SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yejin Jang
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (Y.J.); (J.S.S.); (M.K.L.); (E.J.); (T.A.)
| | - Jin Soo Shin
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (Y.J.); (J.S.S.); (M.K.L.); (E.J.); (T.A.)
| | - Myoung Kyu Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (Y.J.); (J.S.S.); (M.K.L.); (E.J.); (T.A.)
| | - Eunhye Jung
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (Y.J.); (J.S.S.); (M.K.L.); (E.J.); (T.A.)
| | - Timothy An
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (Y.J.); (J.S.S.); (M.K.L.); (E.J.); (T.A.)
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Korea
| | - Uk-Il Kim
- Research and Development Center, ST Pharm Co., Ltd., Seoul 01694, Korea;
| | - Kyungjin Kim
- Research and Development Center, ST Pharm Co., Ltd., Seoul 01694, Korea;
- Correspondence: (K.K.); (M.K.)
| | - Meehyein Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon 34114, Korea; (Y.J.); (J.S.S.); (M.K.L.); (E.J.); (T.A.)
- Graduate School of New Drug Discovery and Development, Chungnam National University, Daejeon 34134, Korea
- Correspondence: (K.K.); (M.K.)
| |
Collapse
|
3
|
Bjånes TK, Jordheim LP, Schjøtt J, Kamceva T, Cros-Perrial E, Langer A, Ruiz de Garibay G, Kotopoulis S, McCormack E, Riedel B. Intracellular Cytidine Deaminase Regulates Gemcitabine Metabolism in Pancreatic Cancer Cell Lines. Drug Metab Dispos 2020; 48:153-158. [PMID: 31871136 PMCID: PMC11022907 DOI: 10.1124/dmd.119.089334] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 12/11/2019] [Indexed: 04/19/2024] Open
Abstract
Cytidine deaminase (CDA) is a determinant of in vivo gemcitabine elimination kinetics and cellular toxicity. The impact of CDA activity in pancreatic ductal adenocarcinoma (PDAC) cell lines has not been elucidated. We hypothesized that CDA regulates gemcitabine flux through its inactivation and activation pathways in PDAC cell lines. Three PDAC cell lines (BxPC-3, MIA PaCa-2, and PANC-1) were incubated with 10 or 100 µM gemcitabine for 60 minutes or 24 hours, with or without tetrahydrouridine, a CDA inhibitor. Extracellular inactive gemcitabine metabolite (dFdU) and intracellular active metabolite (dFdCTP) were quantified with liquid chromatography tandem mass spectrometry. Cellular expression of CDA was assessed with real-time PCR and Western blot. Gemcitabine conversion to dFdU was extensive in BxPC-3 and low in MIA PaCa-2 and PANC-1, in accordance with their respective CDA expression levels. CDA inhibition was associated with low or undetectable dFdU in all three cell lines. After 24 hours gemcitabine incubation, dFdCTP was highest in MIA PaCa-2 and lowest in BxPC-3. CDA inhibition resulted in a profound dFdCTP increase in BxPC-3 but not in MIA PaCa-2 or PANC-1. dFdCTP concentrations were not higher after exposure to 100 versus 10 µM gemcitabine when CDA activities were low (MIA PaCa-2 and PANC-1) or inhibited (BxPC-3). The results suggest a regulatory role of CDA for gemcitabine activation in PDAC cells but within limits related to the capacity in the activation pathway in the cell lines. SIGNIFICANCE STATEMENT: The importance of cytidine deaminase (CDA) for cellular gemcitabine toxicity, linking a lower activity to higher toxicity, is well described. An underlying assumption is that CDA, by inactivating gemcitabine, limits the amount available for the intracellular activation pathway. Our study is the first to illustrate this regulatory role of CDA in pancreatic ductal adenocarcinoma cell lines by quantifying intracellular and extracellular gemcitabine metabolite concentrations.
Collapse
Affiliation(s)
- Tormod K Bjånes
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Lars Petter Jordheim
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Jan Schjøtt
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Tina Kamceva
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Emeline Cros-Perrial
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Anika Langer
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Gorka Ruiz de Garibay
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Spiros Kotopoulis
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Emmet McCormack
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| | - Bettina Riedel
- Section of Clinical Pharmacology, Department of Medical Biochemistry and Pharmacology (T.K.B., J.S., T.K., B.R.) and National Centre for Ultrasound in Gastroenterology (S.K.), Haukeland University Hospital, Bergen, Norway; Department of Clinical Science, Faculty of Medicine (T.K.B., J.S., A.L., G.R.G., E.M., B.R.), Centre for Cancer Biomarkers, Department of Clinical Science (A.L., G.R.G., E.M.), and Department of Clinical Medicine (S.K.), University of Bergen, Bergen, Norway; Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France (L.P.J., E.C.-P.); and Phoenix Solutions AS, Oslo, Norway (S.K.)
| |
Collapse
|
4
|
Toffalorio F, Santarpia M, Radice D, Jaramillo CA, Spitaleri G, Manzotti M, Catania C, Jordheim LP, Pelosi G, Peters GJ, Tibaldi C, Funel N, Spaggiari L, de Braud F, De Pas T, Giovannetti E. 5'-nucleotidase cN-II emerges as a new predictive biomarker of response to gemcitabine/platinum combination chemotherapy in non-small cell lung cancer. Oncotarget 2018; 9:16437-16450. [PMID: 29662657 PMCID: PMC5893252 DOI: 10.18632/oncotarget.24505] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 02/02/2018] [Indexed: 02/04/2023] Open
Abstract
A number of pharmacogenetic studies have been carried out in non-small-cell lung cancer (NSCLC) to identify and characterize genes involved in chemotherapy activity. However, the results obtained so far are controversial and no reliable biomarker is currently used to predict clinical benefit from platinum-based chemotherapy, which represents the cornerstone of treatment of advanced NSCLC. This study investigated the expression levels of ERCC1 and of six genes (RRM1, RRM2, hENT1, dCK, cN-II and CDA) involved in gemcitabine metabolism in locally/advanced NSCLC patients treated with gemcitabine/platinum combination. Gene expression was assessed by quantitative-PCR in laser-microdissected specimens and correlated with tumor response. Frequency distribution of responses above and below the median expression level of biomarkers was compared using a two-sided Fisher’s test. 5′-nucleotidase (cN-II) was the only gene differently expressed (p = 0.016) in the responders (complete/partial-response) compared to non-responders (stable/progressive disease). In the multivariate analysis, overexpression of this catabolic enzyme of gemcitabine remained a significant negative predictive factor. Patients with low cN-II had a modest trend toward increased survival, while both survival and progression-free survival were significantly longer in a more homogenous validation cohort of 40 advanced NSCLC (8.0 vs. 5.1 months, p = 0.026). Moreover, in vitro studies showed that silencing or pharmacological inhibition of cN-II increased the cytotoxicity of gemcitabine. This is the first study demonstrating the role of cN-II as a predictor of response to gemcitabine/platinum combinations in NSCLC. Its validation in prospective studies may improve clinical outcome of selected patients.
Collapse
Affiliation(s)
- Francesca Toffalorio
- Medical Oncology Unit of Respiratory Tract and Sarcomas, New Drugs Development Division, European Institute of Oncology, Milan, Italy.,Medical Affairs, Roche Spa, Monza, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit of Respiratory Tract and Sarcomas, New Drugs Development Division, European Institute of Oncology, Milan, Italy.,Medical Oncology Unit, Department of Human Pathology, University of Messina, Messina, Italy
| | - Davide Radice
- Epidemiology and Biostatistics Division, European Institute of Oncology, Milan, Italy
| | | | - Gianluca Spitaleri
- Medical Oncology Unit of Respiratory Tract and Sarcomas, New Drugs Development Division, European Institute of Oncology, Milan, Italy.,Thoracic Oncology Division, European Institute of Oncology, Milan, Italy
| | - Michela Manzotti
- Division of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Chiara Catania
- Medical Oncology Unit of Respiratory Tract and Sarcomas, New Drugs Development Division, European Institute of Oncology, Milan, Italy.,Thoracic Oncology Division, European Institute of Oncology, Milan, Italy
| | - Lars Petter Jordheim
- Centre de Recherche en Cancérologie de Lyon, INSERM 1052/CNRS UMR 5286, Lyon, France
| | - Giuseppe Pelosi
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Inter-Hospital Pathology Division, Science and Technology Park, IRCCS MultiMedica, Milan, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - Carmelo Tibaldi
- Division of Oncology, Department of Oncology, S. Luca Hospital, Lucca, Italy
| | - Niccola Funel
- CNR-Nano, Institute of Nanoscience and Nanotechnology, Pisa, Italy.,Cancer Pharmacology Laboratory, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Lorenzo Spaggiari
- Thoracic Surgery Division, European Institute of Oncology, Milan, Italy
| | - Filippo de Braud
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.,Inter-Hospital Pathology Division, Science and Technology Park, IRCCS MultiMedica, Milan, Italy
| | - Tommaso De Pas
- Medical Oncology Unit of Respiratory Tract and Sarcomas, New Drugs Development Division, European Institute of Oncology, Milan, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands.,CNR-Nano, Institute of Nanoscience and Nanotechnology, Pisa, Italy.,Cancer Pharmacology Laboratory, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| |
Collapse
|
5
|
Grixti JM, O'Hagan S, Day PJ, Kell DB. Enhancing Drug Efficacy and Therapeutic Index through Cheminformatics-Based Selection of Small Molecule Binary Weapons That Improve Transporter-Mediated Targeting: A Cytotoxicity System Based on Gemcitabine. Front Pharmacol 2017; 8:155. [PMID: 28396636 PMCID: PMC5366350 DOI: 10.3389/fphar.2017.00155] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 03/10/2017] [Indexed: 12/23/2022] Open
Abstract
The transport of drug molecules is mainly determined by the distribution of influx and efflux transporters for which they are substrates. To enable tissue targeting, we sought to develop the idea that we might affect the transporter-mediated disposition of small-molecule drugs via the addition of a second small molecule that of itself had no inhibitory pharmacological effect but that influenced the expression of transporters for the primary drug. We refer to this as a “binary weapon” strategy. The experimental system tested the ability of a molecule that on its own had no cytotoxic effect to increase the toxicity of the nucleoside analog gemcitabine to Panc1 pancreatic cancer cells. An initial phenotypic screen of a 500-member polar drug (fragment) library yielded three “hits.” The structures of 20 of the other 2,000 members of this library suite had a Tanimoto similarity greater than 0.7 to those of the initial hits, and each was itself a hit (the cheminformatics thus providing for a massive enrichment). We chose the top six representatives for further study. They fell into three clusters whose members bore reasonable structural similarities to each other (two were in fact isomers), lending strength to the self-consistency of both our conceptual and experimental strategies. Existing literature had suggested that indole-3-carbinol might play a similar role to that of our fragments, but in our hands it was without effect; nor was it structurally similar to any of our hits. As there was no evidence that the fragments could affect toxicity directly, we looked for effects on transporter transcript levels. In our hands, only the ENT1-3 uptake and ABCC2,3,4,5, and 10 efflux transporters displayed measurable transcripts in Panc1 cultures, along with a ribonucleoside reductase RRM1 known to affect gemcitabine toxicity. Very strikingly, the addition of gemcitabine alone increased the expression of the transcript for ABCC2 (MRP2) by more than 12-fold, and that of RRM1 by more than fourfold, and each of the fragment “hits” served to reverse this. However, an inhibitor of ABCC2 was without significant effect, implying that RRM1 was possibly the more significant player. These effects were somewhat selective for Panc cells. It seems, therefore, that while the effects we measured were here mediated more by efflux than influx transporters, and potentially by other means, the binary weapon idea is hereby fully confirmed: it is indeed possible to find molecules that manipulate the expression of transporters that are involved in the bioactivity of a pharmaceutical drug. This opens up an entirely new area, that of chemical genomics-based drug targeting.
Collapse
Affiliation(s)
- Justine M Grixti
- Faculty of Biology, Medicine and Health, University of ManchesterManchester, UK; Manchester Institute of Biotechnology, University of ManchesterManchester, UK
| | - Steve O'Hagan
- Manchester Institute of Biotechnology, University of ManchesterManchester, UK; School of Chemistry, University of ManchesterManchester, UK; Centre for Synthetic Biology of Fine and Speciality Chemicals, University of ManchesterManchester, UK
| | - Philip J Day
- Faculty of Biology, Medicine and Health, University of ManchesterManchester, UK; Manchester Institute of Biotechnology, University of ManchesterManchester, UK
| | - Douglas B Kell
- Manchester Institute of Biotechnology, University of ManchesterManchester, UK; School of Chemistry, University of ManchesterManchester, UK; Centre for Synthetic Biology of Fine and Speciality Chemicals, University of ManchesterManchester, UK
| |
Collapse
|
6
|
Genotoxicity kinetics in murine normoblasts as an approach for the in vivo action of difluorodeoxycytidine. Cancer Chemother Pharmacol 2017; 79:843-853. [DOI: 10.1007/s00280-017-3290-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/14/2017] [Indexed: 12/30/2022]
|
7
|
Ni S, Qiu L, Zhang G, Zhou H, Han Y. Lymph cancer chemotherapy: delivery of doxorubicin-gemcitabine prodrug and vincristine by nanostructured lipid carriers. Int J Nanomedicine 2017; 12:1565-1576. [PMID: 28280326 PMCID: PMC5338998 DOI: 10.2147/ijn.s120685] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
PURPOSE Radiation and chemotherapy are the most common course of treatment for B-cell lymphoma. Doxorubicin (DOX), gemcitabine (GEM), and vincristine (VCR) are the commonly used antilymphoma chemotherapeutic drugs. The aim of this study is to construct a novel drug delivery system for the combination delivery of the three drugs on lymphoma. MATERIALS AND METHODS DOX-GEM prodrug was synthesized. Novel nanostructured lipid carriers (NLCs) containing DOX-GEM prodrug and VCR were prepared and used to treat B-cell lymphoma through in vivo treatment to a lymph cancer animal model. The systemic toxicity of the nanomedicine was also evaluated during the treatment. RESULTS DOX-GEM prodrug and VCR-loaded NLCs (DOX-GEM VCR NLCs) exhibited the highest antitumor effect in B-cell lymphoma cells and lymphoma animal xenografts when compared with the single drug-loaded NLCs and the drug solutions. CONCLUSION It could be concluded that the highest antitumor effect can be achieved by the system due to the stable drug-loading capacity, attractive anticancer therapeutic effects, and reduced toxicities in human Burkitt's lymphoma cell line and mice-bearing cancer model. The resulting DOX-GEM VCR NLCs could be an efficient antilymph cancer agent and could be developed further for the treatment of other tumors.
Collapse
Affiliation(s)
- Shuqin Ni
- Department of Internal Medicine Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Ji'nan, Shandong, People's Republic of China
| | - Lei Qiu
- Department of Internal Medicine Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Ji'nan, Shandong, People's Republic of China
| | - Guodong Zhang
- Department of Internal Medicine Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Ji'nan, Shandong, People's Republic of China
| | - Haiyan Zhou
- Department of Internal Medicine Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Ji'nan, Shandong, People's Republic of China
| | - Yong Han
- Department of Internal Medicine Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Science, Ji'nan, Shandong, People's Republic of China
| |
Collapse
|
8
|
Sripornsawan P, Okamoto Y, Nishikawa T, Kodama Y, Yamaki Y, Kurauchi K, Tanabe T, Nakagawa S, Shinkoda Y, Imuta N, Kawano Y. Gene expression ratio as a predictive determinant of nelarabine chemosensitivity in T-lymphoblastic leukemia/lymphoma. Pediatr Blood Cancer 2017; 64:250-253. [PMID: 27576612 DOI: 10.1002/pbc.26214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 07/13/2016] [Accepted: 07/21/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Nelarabine has been used for the treatment of T-cell malignancies including T-acute lymphoblastic leukemia (T-ALL)/T-lymphoblastic lymphoma. However, the mechanisms that underlie the susceptibility or resistance to nelarabine have not been fully elucidated. The aim of this study was to determine the significance of nelarabine transport and metabolism in the context of nelarabine cytotoxicity. PROCEDURE The expression profiles of six genes in the nelarabine pathway were analyzed in blast cells from six patients with T-ALL as well as in three T-ALL cell lines. In vitro cytotoxicity (LC50 of 9-β-d-arabinofuranosylguanine [ara-G]) was evaluated. RESULTS The mRNA expression of ENT1, DCK, CDA, NT5C2, RRM1, and RRM2 in patients showed inter-individual variability and was not correlated with the LC50 of ara-G. However, the ratio of (ENT1 × DCK)/(CDA × RRM1) expression was significantly correlated with LC50 (r = -0.831, P = 0.0405). CONCLUSIONS Chemosensitivity to nelarabine is influenced by the balance of the expression of these four genes, and the ratio of their expression predicts the response of T-cell malignancies to nelarabine.
Collapse
Affiliation(s)
- Pornpun Sripornsawan
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan.,Department of Pediatrics, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Yasuhiro Okamoto
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takuro Nishikawa
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yuichi Kodama
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yuni Yamaki
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Koichiro Kurauchi
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takayuki Tanabe
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Shunsuke Nakagawa
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yuichi Shinkoda
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Naoko Imuta
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yoshifumi Kawano
- Department of Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
9
|
Ohmine K, Kawaguchi K, Ohtsuki S, Motoi F, Ohtsuka H, Kamiie J, Abe T, Unno M, Terasaki T. Quantitative Targeted Proteomics of Pancreatic Cancer: Deoxycytidine Kinase Protein Level Correlates to Progression-Free Survival of Patients Receiving Gemcitabine Treatment. Mol Pharm 2015; 12:3282-91. [PMID: 26280109 DOI: 10.1021/acs.molpharmaceut.5b00282] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The purpose of the present study is to identify the determinant(s) of gemcitabine (dFdC)-sensitivity in pancreatic cancer tissues of patients treated with dFdC alone and in pancreatic cancer cell lines exposed to dFdC in vitro. Protein expression levels of 12 enzymes and 13 transporters potentially involved in transport and metabolism of dFdC in pancreatic cancer cell lines and tissues were quantified by means of our LC-MS/MS-based quantitative targeted proteomics technology. Protein expression levels of deoxycytidine kinase (dCK), uridine monophosphate-cytidine monophosphate (UMP-CMP) kinase, cytosolic nucleotidase III (cN-III), and equilibrative nucleoside transporter 1 (ENT1) were significantly correlated with IC50 or 1/IC50 in five cell lines with different sensitivities to dFdC (p < 0.05). Expression levels of the selected proteins in pancreatic cancer tissues of 10 patients with different progression-free survival (PFS) (49-955 days) were quantified, and their relationship with PFS was examined. Only the protein expression level of dCK was significantly correlated with PFS (p < 0.05). Multiple regression analysis was also performed, and combinations of ENT1, UMP-CMP kinase, CTPS1, and dCK were highly correlated with PFS. Our results indicate that the protein expression level of dCK in pancreatic cancer tissue is a good predictor of PFS, and thus dCK may be the best biomarker of dFdC sensitivity in pancreatic cancer patients treated with dFdC, although other proteins would also contribute to dFdC-sensitivity at the cellular level in vivo and in vitro.
Collapse
Affiliation(s)
- Ken Ohmine
- Membrane Transport and Drug Targeting Laboratory, Tohoku University Graduate School of Pharmaceutical Sciences , Sendai, Japan
| | - Kei Kawaguchi
- Division of Hepato-Biliary-Pancreatic Surgery, Tohoku University Graduate School of Medicine , Sendai, Japan
| | - Sumio Ohtsuki
- Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University , Kumamoto, Japan
| | - Fuyuhiko Motoi
- Division of Hepato-Biliary-Pancreatic Surgery, Tohoku University Graduate School of Medicine , Sendai, Japan
| | - Hideo Ohtsuka
- Division of Hepato-Biliary-Pancreatic Surgery, Tohoku University Graduate School of Medicine , Sendai, Japan
| | - Junichi Kamiie
- Laboratory of Veterinary Pathology, Azabu University School of Veterinary Medicine , Sagamihara, Japan
| | - Takaaki Abe
- Department of Clinical Biology and Hormonal Regulation, Tohoku University Graduate School of Medicine , Sendai, Japan
| | - Michiaki Unno
- Division of Hepato-Biliary-Pancreatic Surgery, Tohoku University Graduate School of Medicine , Sendai, Japan
| | - Tetsuya Terasaki
- Membrane Transport and Drug Targeting Laboratory, Tohoku University Graduate School of Pharmaceutical Sciences , Sendai, Japan
| |
Collapse
|
10
|
Cividini F, Filoni DN, Pesi R, Allegrini S, Camici M, Tozzi MG. IMP-GMP specific cytosolic 5'-nucleotidase regulates nucleotide pool and prodrug metabolism. Biochim Biophys Acta Gen Subj 2015; 1850:1354-61. [PMID: 25857773 DOI: 10.1016/j.bbagen.2015.03.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 03/26/2015] [Accepted: 03/31/2015] [Indexed: 12/20/2022]
Abstract
BACKGROUND Type II cytosolic 5'-nucleotidase (cN-II) catalyzes the hydrolysis of purine and, to some extent, of pyrimidine monophosphates. Recently, a number of papers demonstrated the involvement of cN-II in the mechanisms of resistance to antitumor drugs such as cytarabine, gemcitabine and fludarabine. Furthermore, cN-II is involved in drug resistance in patients affected by hematological malignancies influencing the clinical outcome. Although the implication of cN-II expression and/or activity appears to be correlated with drug resistance and poor prognosis, the molecular mechanism by which cN-II mediates drug resistance is still unknown. METHODS HEK 293 cells carrying an expression vector coding for cN-II linked to green fluorescent protein (GFP) and a control vector without cN-II were utilized. A highly sensitive capillary electrophoresis method was applied for nucleotide pool determination and cytotoxicity exerted by drugs was determined with 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT) assay. RESULTS Over-expression of cN-II causes a drop of nucleoside triphosphate concentration and a general disturbance of nucleotide pool. Over-expressing cells were resistant to fludarabine, gemcitabine and cytarabine independently of cN-II ability to hydrolyze their monophosphates. CONCLUSIONS An increase of cN-II expression is sufficient to cause both a general disturbance of nucleotide pool and an increase of half maximal inhibitory concentration (IC50) of the drugs. Since the monophosphates of cytarabine and gemcitabine are not substrates of cN-II, the protection observed cannot be directly ascribed to drug inactivation. GENERAL SIGNIFICANCE Our results indicate that cN-II exerts a relevant role in nucleotide and drug metabolism through not only enzyme activity but also a mechanism involving a protein-protein interaction, thus playing a general regulatory role in cell survival. SENTENCE Resistance to fludarabine, gemcitabine and cytarabine can be determined by an increase of cN-II both through dephosphorylation of active drugs and perturbation of nucleotide pool.
Collapse
Affiliation(s)
- Federico Cividini
- Dipartimento di Biologia, Unità di Biochimica, Università di Pisa, Via San Zeno 51, 56127, Pisa, Italy
| | - Daniela Nicole Filoni
- Dipartimento di Biologia, Unità di Biochimica, Università di Pisa, Via San Zeno 51, 56127, Pisa, Italy; Dipartimento di Chimica e Farmacia, Università di Sassari, Via Muroni 23A, 07100, Sassari, Italy
| | - Rossana Pesi
- Dipartimento di Biologia, Unità di Biochimica, Università di Pisa, Via San Zeno 51, 56127, Pisa, Italy
| | - Simone Allegrini
- Dipartimento di Chimica e Farmacia, Università di Sassari, Via Muroni 23A, 07100, Sassari, Italy.
| | - Marcella Camici
- Dipartimento di Biologia, Unità di Biochimica, Università di Pisa, Via San Zeno 51, 56127, Pisa, Italy
| | - Maria Grazia Tozzi
- Dipartimento di Biologia, Unità di Biochimica, Università di Pisa, Via San Zeno 51, 56127, Pisa, Italy
| |
Collapse
|
11
|
Cividini F, Pesi R, Chaloin L, Allegrini S, Camici M, Cros-Perrial E, Dumontet C, Jordheim L, Tozzi M. The purine analog fludarabine acts as a cytosolic 5′-nucleotidase II inhibitor. Biochem Pharmacol 2015; 94:63-8. [DOI: 10.1016/j.bcp.2015.01.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/26/2015] [Accepted: 01/26/2015] [Indexed: 11/29/2022]
|
12
|
Preclinical combination therapy of the investigational drug NAMI-A(+) with doxorubicin for mammary cancer. Invest New Drugs 2014; 33:53-63. [PMID: 25338748 DOI: 10.1007/s10637-014-0175-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 10/09/2014] [Indexed: 12/11/2022]
Abstract
AIM OF THE STUDY The tumor metastases targeting ruthenium complex NAMI-A synergistically improves the activity of gemcitabine in combination therapies. High-throughput screening was used to identify other potential drug combinations from a library of FDA approved drugs. Doxorubicin was identified as a hit compound and was therefore evaluated in combination with NAMI-A in vitro and in a preclinical in vivo model. RESULTS High-throughput screening identified eight structurally diverse compounds that synergize with NAMI-A including doxorubicin. The combination index on MCF-7 cells showed synergism as the concentration of NAMI-A increases independent of the doxorubicin concentration. In MCa mammary carcinoma of CBA mice, NAMI-A (35 mg/kg/day i.p. on days 7-12) followed by doxorubicin (10 mg/kg i.p. on day 16), significantly increased the effects of the individual drugs on metastases with 70 % animals resulting free of macroscopically detectable tumor nodules in the lungs at sacrifice. NAMI-A, unlike doxorubicin, cured 60 % of the treated mice but the combination therapy was toxic to the animals. CONCLUSIONS The combined therapy of NAMI-A with doxorubicin synergizes on lung metastasis in a preclinical mouse model. The combination therapy at the maximum tolerated doses of the two drugs is toxic. Hence, this combination is not suitable for clinical studies using maximum tolerated doses.
Collapse
|
13
|
Differential modulation of nicotine-induced gemcitabine resistance by GABA receptor agonists in pancreatic cancer cell xenografts and in vitro. BMC Cancer 2014; 14:725. [PMID: 25260978 PMCID: PMC4190427 DOI: 10.1186/1471-2407-14-725] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Accepted: 09/23/2014] [Indexed: 12/20/2022] Open
Abstract
Background Pancreatic cancer is frequently resistant to cancer therapeutics. Smoking and alcoholism are risk factors and pancreatic cancer patients often undergo nicotine replacement therapy (NRT) and treatment for alcohol dependence. Based on our report that low dose nicotine within the range of NRT causes gemcitabine resistance in pancreatic cancer, our current study has tested the hypothesis that GABA or the selective GABA-B-R agonist baclofen used to treat alcohol dependence reverse nicotine-induced gemcitabine resistance in pancreatic cancer. Methods Using mouse xenografts from the gemcitabine--sensitive pancreatic cancer cell line BXPC-3, we tested the effects of GABA and baclofen on nicotine-induced gemcitabine resistance. The levels of cAMP, p-SRC, p-ERK, p-AKT, p-CREB and cleaved caspase-3 in xenograft tissues were determined by ELISA assays. Expression of the two GABA-B receptors, metalloproteinase-2 and 9 and EGR-1 in xenograft tissues was monitored by Western blotting. Mechanistic studies were conducted in vitro, using cell lines BXPC-3 and PANC-1 and included analyses of cAMP production by ELISA assay and Western blots to determine protein expression of GABA-B receptors, metalloproteinase-2 and 9 and EGR-1. Results Our data show that GABA was as effective as gemcitabine and significantly reversed gemcitabine resistance induced by low dose nicotine in xenografts whereas baclofen did not. These effects of GABA were accompanied by decreases in cAMP, p-CREB, p-AKT, p-Src, p-ERK metalloproteinases-9 and -2 and EGR-1 and increases in cleaved caspase-3 in xenografts whereas baclofen had the opposite effects. In vitro exposure of cells to single doses or seven days of nicotine induced the protein expression of MMP-2, MMP-9 and EGR-1 and these responses were blocked by GABA. Baclofen downregulated the protein expression of GABA-B-Rs in xenograft tissues and in cells exposed to baclofen for seven days in vitro. This response was accompanied by inversed baclofen effects from inhibition of cAMP formation after single dose exposures to stimulation of cAMP formation in cells pretreated for seven days. Conclusions These findings suggest GABA as a promising single agent for the therapy of pancreatic cancer and to overcome nicotine-induced gemcitabine resistance whereas treatment with baclofen may increase gemcitabine resistance.
Collapse
|
14
|
DiNardo CD, O'Brien S, Gandhi VV, Ravandi F. Elacytarabine (CP-4055) in the treatment of acute myeloid leukemia. Future Oncol 2013; 9:1073-82. [PMID: 23902239 DOI: 10.2217/fon.13.130] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Elacytarabine (formerly CP-4055) is a lipid-conjugated derivative of the nucleoside analog cytarabine. Elacytarabine was rationally designed to circumvent cytarabine resistance related to decreased cellular uptake, due to the ability of the lipophilic drug moiety to enter the cell without the requirement of specialized nuclear transport proteins, including the hENT1. In preclinical and clinical studies, elacytarabine has demonstrated both safety and efficacy in acute myeloid leukemia (AML), with noteworthy activity among the cytarabine-refractory AML population. Elacytarabine was granted orphan drug designation status from the European Commission in 2007 and from the US FDA in 2008, with a fast-track approval designation from the FDA in 2010. Results of a recent randomized Phase III clinical trial, however, failed to show superiority of elacytarabine over the investigator's choice of therapy for relapsed or refractory AML.
Collapse
Affiliation(s)
- Courtney D DiNardo
- Department of Leukemia, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | |
Collapse
|
15
|
Avan A, Pacetti P, Reni M, Milella M, Vasile E, Mambrini A, Vaccaro V, Caponi S, Cereda S, Peters GJ, Cantore M, Giovannetti E. Prognostic factors in gemcitabine-cisplatin polychemotherapy regimens in pancreatic cancer: XPD-Lys751Gln polymorphism strikes back. Int J Cancer 2013; 133:1016-22. [PMID: 23390054 DOI: 10.1002/ijc.28078] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Revised: 01/08/2013] [Accepted: 01/09/2013] [Indexed: 01/18/2023]
Abstract
The use of platinated agents in combination chemotherapy regimens for advanced pancreatic cancer is controversial owing to the lack of an outstanding impact on the outcome and a substantial increase in hematologic and extra-hematologic toxicities. Pharmacogenetic studies to identify patients who could benefit most from such therapies are urgently needed. The Xeroderma-Pigmentosum group-D polymorphism at codon-751 (XPD-Lys751Gln) emerged as the most significant independent predictor for death- and progression-risk in our previous study on functional polymorphisms in 122 advanced pancreatic cancer patients treated with cisplatin-docetaxel-capecitabine-gemcitabine and cisplatin-epirubicin-capecitabine-gemcitabine (or EC-GemCap). To confirm the prognostic role of this variable, we further evaluated the correlation of XPD-Lys751Gln with outcome in another 125 patients treated with the same regimens, and 90 treated with gemcitabine monotherapy. Genotyping was successfully carried out in the vast majority of DNA samples. Genotype frequencies followed Hardy-Weinberg equilibrium, and XPD-Lys751Gln was associated with differential progression-free and overall-survival. Multivariate analysis confirmed its prognostic significance in platinum-based regimens. In particular, XPD-Gln751Gln was significantly associated with risk of death (hazard ratio, HR = 1.7, 95% confidence interval [CI], 1.1-2.6, p = 0.011) and risk of progression (HR = 1.7, 95% CI, 1.1-2.5, p = 0.013). No correlation was observed in gemcitabine monotherapy-treated patients. The analysis of DNA damage using extra-long-PCR in lymphocytes supported the association of XPD-Gln751Gln with greater resistance to cisplatin-induced damage. The increasing evidence of XPD-Lys751Gln impact on the outcome of gemcitabine-cisplatin-based polychemotherapy leads to plan prospective studies to validate the role of this polymorphism as a new tool for optimization of the currently available treatments in pancreatic cancer.
Collapse
Affiliation(s)
- Amir Avan
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
O'Brien S, Rizzieri DA, Vey N, Ravandi F, Krug UO, Sekeres MA, Dennis M, Venditti A, Berry DA, Jacobsen TF, Staudacher K, Bergeland T, Giles FJ. Elacytarabine has single-agent activity in patients with advanced acute myeloid leukaemia. Br J Haematol 2012; 158:581-8. [PMID: 22702906 DOI: 10.1111/j.1365-2141.2012.09186.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 05/03/2012] [Indexed: 01/14/2023]
Abstract
Elacytarabine is a novel cytotoxic nucleoside analogue, independent of nucleoside transporters (e.g. human Equilibrative Nucleoside Transporter 1 [hENT1]) for cell uptake, and mechanisms of action similar to those of cytarabine. This Phase II study assessed the efficacy and safety of elacytarabine in patients with advanced stage acute myeloid leukaemia (AML). Patients received 2000 mg/m(2) per d continuously i.v. during days 1-5 every 3 weeks. Patients were matched by six risk factors with historical controls; remission rate (assessed after 1 or 2 cycles) and 6-month survival were compared. Sixty-one patients, median age 58 years, were enrolled; 52% had five or six risk factors. The remission rate was 18% (95% confidence interval: 9-30%) vs. 4% in controls (P < 0·0001), 6-month survival rate was 43%, median overall survival was 5·3 months (vs. 1·5 months); 10 patients (16%) were referred for stem cell transplantation after treatment. Side effects were predictable and manageable. The most common grade 3/4 non-haematological adverse events were febrile neutropenia, hypokalemia, fatigue, hyponatraemia, dyspnoea and pyrexia. Thirty-day all-cause mortality, after start of treatment, was 13% vs. 25% in controls. Elacytarabine has monotherapy activity in patients with advanced AML. This study provides proof-of-concept that lipid esterification of nucleoside analogues is clinically relevant.
Collapse
Affiliation(s)
- Susan O'Brien
- Department of Leukemia, University of Texas, MD Anderson Cancer Center, Houston, TX 77230, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Funamizu N, Lacy CR, Fujita K, Furukawa K, Misawa T, Yanaga K, Manome Y. Tetrahydrouridine inhibits cell proliferation through cell cycle regulation regardless of cytidine deaminase expression levels. PLoS One 2012; 7:e37424. [PMID: 22616006 PMCID: PMC3353937 DOI: 10.1371/journal.pone.0037424] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 04/21/2012] [Indexed: 12/24/2022] Open
Abstract
Tetrahydrouridine (THU) is a well characterized and potent inhibitor of cytidine deaminase (CDA). Highly expressed CDA catalyzes and inactivates cytidine analogues, ultimately contributing to increased gemcitabine resistance. Therefore, a combination therapy of THU and gemcitabine is considered to be a potential and promising treatment for tumors with highly expressed CDA. In this study, we found that THU has an alternative mechanism for inhibiting cell growth which is independent of CDA expression. Three different carcinoma cell lines (MIAPaCa-2, H441, and H1299) exhibited decreased cell proliferation after sole administration of THU, while being unaffected by knocking down CDA. To investigate the mechanism of THU-induced cell growth inhibition, cell cycle analysis using flow cytometry was performed. This analysis revealed that THU caused an increased rate of G1-phase occurrence while S-phase occurrence was diminished. Similarly, Ki-67 staining further supported that THU reduces cell proliferation. We also found that THU regulates cell cycle progression at the G1/S checkpoint by suppressing E2F1. As a result, a combination regimen of THU and gemcitabine might be a more effective therapy than previously believed for pancreatic carcinoma since THU works as a CDA inhibitor, as well as an inhibitor of cell growth in some types of pancreatic carcinoma cells.
Collapse
Affiliation(s)
- Naotake Funamizu
- Department of Molecular Cell Biology, Institute of DNA Medicine, The Jikei University School of Medicine, Tokyo, Japan.
| | | | | | | | | | | | | |
Collapse
|
18
|
Crea F, Fornaro L, Paolicchi E, Masi G, Frumento P, Loupakis F, Salvatore L, Cremolini C, Schirripa M, Graziano F, Ronzoni M, Ricci V, Farrar WL, Falcone A, Danesi R. An EZH2 polymorphism is associated with clinical outcome in metastatic colorectal cancer patients. Ann Oncol 2012; 23:1207-1213. [PMID: 21926398 DOI: 10.1093/annonc/mdr387] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Despite therapeutic innovations, metastatic colorectal cancer (mCRC) is still characterized by poor prognosis and few molecular markers predict the risk of progression. Polycomb group genes (PcGs) are epigenetic modifiers involved in tumor suppressor gene silencing. PcG member EZH2 mediates gene silencing through histone-H3 lysine-27 methylation. In colorectal cancer (CRC), EZH2 overexpression predicts shorter survival. Recently, four EZH2 single-nucleotide polymorphisms (SNPs) have been described. The present study was aimed at evaluating the correlation between EZH2 SNPs and outcome parameters in mCRC patients. PATIENTS AND METHODS DNA was extracted from blood samples of 110 mCRC patients treated with first-line 5-fluorouracil, folinic acid, irinotecan (FOLFIRI) and bevacizumab. Genotyping was carried out by real-time PCR. Genotype was used to predict objective response, progression-free survival (PFS) and overall survival (OS). EZH2 messenger RNA levels were evaluated on lymphocytes of a parallel cohort of 50 CRC patients. RESULTS One allelic variant (rs3757441 C/C versus C/T or T/T) was significantly associated with shorter PFS and OS (P < 0.01 and P < 0.05, respectively). At multivariate analysis, the same variant resulted an independent predictor of PFS and OS (P < 0.05). The C/C variant was associated with significantly higher EZH2 expression (P < 0.05). CONCLUSION An EZH2 SNP may be useful to predict clinical outcome in mCRC patients.
Collapse
Affiliation(s)
- F Crea
- Division of Pharmacology, Department of Internal Medicine, University of Pisa, Pisa.
| | - L Fornaro
- Division of Medical Oncology, Department of Oncology, Transplants and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - E Paolicchi
- Division of Pharmacology, Department of Internal Medicine, University of Pisa, Pisa
| | - G Masi
- Division of Medical Oncology, Department of Oncology, Transplants and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - P Frumento
- Unit of Biostatistics, Institute of Environmental Health, Karolinska Institutet, Stockholm, Sweden
| | - F Loupakis
- Division of Medical Oncology, Department of Oncology, Transplants and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - L Salvatore
- Division of Medical Oncology, Department of Oncology, Transplants and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - C Cremolini
- Division of Medical Oncology, Department of Oncology, Transplants and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - M Schirripa
- Division of Medical Oncology, Department of Oncology, Transplants and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - F Graziano
- Medical Oncology Unit, Department of Onco-Hematology, Azienda Ospedaliera S. Salvatore, Pesaro
| | - M Ronzoni
- Division of Medical Oncology, S. Raffaele Scientific Institute, Milano, Italy
| | - V Ricci
- Division of Medical Oncology, S. Raffaele Scientific Institute, Milano, Italy
| | - W L Farrar
- Cancer Stem Cell Section, Laboratory of Cancer Prevention, National Institute of Cancer-Frederick, Frederick, USA
| | - A Falcone
- Division of Medical Oncology, Department of Oncology, Transplants and New Technologies in Medicine, University of Pisa, Pisa, Italy
| | - R Danesi
- Division of Pharmacology, Department of Internal Medicine, University of Pisa, Pisa
| |
Collapse
|
19
|
Abstract
The mainstay of acute myeloid leukemia chemotherapy is the nucleoside analog cytarabine (ara-C). Numerous studies suggest that the intracellular concentrations of the ara-C active metabolite, ara-CTP, vary widely among patients and, in turn, are associated with variability in clinical response to acute myeloid leukemia treatment. Thus, genetic variation in key genes in the ara-C metabolic pathway--specifically, deoxycytidine kinase (a rate-limiting activating enzyme), 5 nucleotidase, cytidine deaminase and deoxycytidylate deaminase (all three are inactivating enzymes), human equilibrative nucleoside transporter (ara-C uptake transporter) and ribonucleotide reductase (RRM1 and RRM2--enzymes regulating intracellular deoxycytidine triphosphate pools)--form the molecular basis of the interpatient variability observed in intracellular ara-CTP concentrations and response to ara-C. Understanding genetic variants in the key candidate genes involved in the metabolic activation of ara-C, as well as the pharmacodynamic targets of ara-C, will provide an opportunity to identify patients at an increased risk of adverse reactions or decreased likelihood of response, based upon their genetic profile, which in future could help in dose optimization to reduce drug toxicity without compromising efficacy. The pharmacogenetic studies on ara-C would also be equally applicable to other nucleoside analogs, such as gemcitabine, decitabine, clofarabine and so on, which are metabolized by the same pathway.
Collapse
Affiliation(s)
- Jatinder K Lamba
- Department of Experimental and Clinical Pharmacology, University of Minnesota, Minneapolis, MN 55455, USA.
| |
Collapse
|
20
|
Ferrandina G, Mey V, Nannizzi S, Ricciardi S, Petrillo M, Ferlini C, Danesi R, Scambia G, Del Tacca M. Expression of nucleoside transporters, deoxycitidine kinase, ribonucleotide reductase regulatory subunits, and gemcitabine catabolic enzymes in primary ovarian cancer. Cancer Chemother Pharmacol 2009; 65:679-86. [DOI: 10.1007/s00280-009-1073-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2009] [Accepted: 07/07/2009] [Indexed: 12/12/2022]
|
21
|
Lack ofin vitro interactions using human liver micro-somes between rabeprazole and anticancer drugs. Eur J Drug Metab Pharmacokinet 2009; 34:19-26. [DOI: 10.1007/bf03191379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
22
|
Affiliation(s)
- Mark Kirschbaum
- Division of Hematology, City of Hope National Cancer Center, Duarte, USA.
| |
Collapse
|
23
|
Leisewitz AV, Zimmerman EI, Jones SZ, Yang J, Graves LM. Imatinib-resistant CML cells have low ENT activity but maintain sensitivity to gemcitabine. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2008; 27:779-86. [PMID: 18600540 DOI: 10.1080/15257770802145892] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Philadelphia chromosome-positive chronic myelogenus leukemia (CML) is widely treated with imatinib mesylate (imatinib), a potent inhibitor of the Bcr-Abl tyrosine kinase. However, resistance to this compound remains a concern. Current treatment approaches include combinations of imatinib with nucleoside analogs such as gemcitabine, which requires equilibrative nucleoside transporters (ENTs) for uptake, to overcome this resistance. Here we report that imatinib treatment decreased ENT1-dependent activity and mRNA expression. Although, imatinib-resistant cells showed decreased levels of both ENT1 and ENT2 activity and expression, these cells remained sensitive to gemcitabine, suggesting that nucleoside analogs can be used as adjunctive therapy.
Collapse
Affiliation(s)
- Andrea V Leisewitz
- Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, North Carolina 27599, USA
| | | | | | | | | |
Collapse
|