Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Front Oncol ; 12: 953013, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36185300

RESUMEN

During the last two decades, kinase inhibitors have become the major drug class for targeted cancer therapy. Although the number of approved kinase inhibitors increases rapidly, comprehensive in vitro profiling and comparison of inhibitor activities is often lacking in the public domain. Here we report the extensive profiling and comparison of 21 kinase inhibitors approved by the FDA for oncology indications since June 2018 and 13 previously approved comparators on panels of 255 biochemical kinase assays and 134 cancer cell line viability assays. Comparison of the cellular inhibition profiles of the EGFR inhibitors gefitinib, dacomitinib, and osimertinib identified the uncommon EGFR p.G719S mutation as a common response marker for EGFR inhibitors. Additionally, the FGFR inhibitors erdafitinib, infigratinib, and pemigatinib potently inhibited the viability of cell lines which harbored oncogenic alterations in FGFR1-3, irrespective of the specific clinical indications of the FGFR inhibitors. These results underscore the utility of in vitro kinase inhibitor profiling in cells for identifying new potential stratification markers for patient selection. Furthermore, comparison of the in vitro inhibition profiles of the RET inhibitors pralsetinib and selpercatinib revealed they had very similar biochemical and cellular selectivity. As an exception, an NTRK3 fusion-positive cell line was potently inhibited by pralsetinib but not by selpercatinib, which could be explained by the targeting of TRK kinases in biochemical assays by pralsetinib but not selpercatinib. This illustrates that unexpected differences in cellular activities between inhibitors that act through the same primary target can be explained by subtle differences in biochemical targeting. Lastly, FLT3-mutant cell lines were responsive to both FLT3 inhibitors gilteritinib and midostaurin, and the PI3K inhibitor duvelisib. Biochemical profiling revealed that the FLT3 and PI3K inhibitors targeted distinct kinases, indicating that unique dependencies can be identified by combined biochemical and cellular profiling of kinase inhibitors. This study provides the first large scale kinase assay or cell panel profiling study for newly approved kinase inhibitors, and shows that comprehensive in vitro profiling of kinase inhibitors can provide rationales for therapy selection and indication expansion of approved kinase inhibitors.

2.
Mol Cancer Ther ; 18(2): 470-481, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30381447

RESUMEN

Kinase inhibitors form the largest class of precision medicine. From 2013 to 2017, 17 have been approved, with 8 different mechanisms. We present a comprehensive profiling study of all 17 inhibitors on a biochemical assay panel of 280 kinases and proliferation assays of 108 cancer cell lines. Drug responses of the cell lines were related to the presence of frequently recurring point mutations, insertions, deletions, and amplifications in 15 well-known oncogenes and tumor-suppressor genes. In addition, drug responses were correlated with basal gene expression levels with a focus on 383 clinically actionable genes. Cell lines harboring actionable mutations defined in the FDA labels, such as mutant BRAF(V600E) for cobimetinib, or ALK gene translocation for ALK inhibitors, are generally 10 times more sensitive compared with wild-type cell lines. This sensitivity window is more narrow for markers that failed to meet endpoints in clinical trials, for instance CDKN2A loss for CDK4/6 inhibitors (2.7-fold) and KRAS mutation for cobimetinib (2.3-fold). Our data underscore the rationale of a number of recently opened clinical trials, such as ibrutinib in ERBB2- or ERBB4-expressing cancers. We propose and validate new response biomarkers, such as mutation in FBXW7 or SMAD4 for EGFR and HER2 inhibitors, ETV4 and ETV5 expression for MEK inhibitors, and JAK3 expression for ALK inhibitors. Potentially, these new markers could be combined to improve response rates. This comprehensive overview of biochemical and cellular selectivities of approved kinase inhibitor drugs provides a rich resource for drug repurposing, basket trial design, and basic cancer research.


Asunto(s)
Neoplasias/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Adenina/análogos & derivados , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Aprobación de Drogas , Reposicionamiento de Medicamentos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias/tratamiento farmacológico , Piperidinas , Mutación Puntual , Mapas de Interacción de Proteínas , Pirazoles/farmacología , Pirimidinas/farmacología
3.
Drug Discov Today ; 12(13-14): 521-6, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17631245

RESUMEN

Advances in detection technologies have enabled an increased use of cell-based functional assays in early drug discovery, in particular for G protein-coupled receptors. Screening assays that use live cells are less prone to generate false positives than assays using lysed cell samples. The use of cryopreserved cells instead of cells that are continuously maintained in culture decreases day-to-day variation, removes passage effects and improves the consistency of cell-based assay results. Cryopreservation techniques uncouple cell culturing from drug-screening activities and allow the use of cells as reagents, just like enzymes in biochemical assays.


Asunto(s)
Bioensayo , Criopreservación/métodos , Evaluación Preclínica de Medicamentos/métodos , Línea Celular , Diseño de Fármacos , Humanos , Transporte de Proteínas , Receptores Acoplados a Proteínas G/metabolismo
4.
Mol Cancer Ther ; 16(11): 2609-2617, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28751540

RESUMEN

The spindle assembly checkpoint kinase TTK (Mps1) is a key regulator of chromosome segregation and is the subject of novel targeted therapy approaches by small-molecule inhibitors. Although the first TTK inhibitors have entered phase I dose escalating studies in combination with taxane chemotherapy, a patient stratification strategy is still missing. With the aim to identify a genomic biomarker to predict the response of tumor cells to TTK inhibitor therapy, we profiled a set of preclinical and clinical TTK inhibitors from different chemical series on a panel of 66 genetically characterized cell lines derived from different tumors (Oncolines). Cell lines harboring activating mutations in the CTNNB1 gene, encoding the Wnt pathway signaling regulator ß-catenin, were on average up to five times more sensitive to TTK inhibitors than cell lines wild-type for CTNNB1 The association of CTNNB1-mutant status and increased cancer cell line sensitivity to TTK inhibition was confirmed with isogenic cell line pairs harboring either mutant or wild-type CTNNB1 Treatment of a xenograft model of a CTNNB1-mutant cell line with the TTK inhibitor NTRC 0066-0 resulted in complete inhibition of tumor growth. Mutations in CTNNB1 occur at relatively high frequency in endometrial cancer and hepatocellular carcinoma, which are known to express high TTK levels. We propose mutant CTNNB1 as a prognostic drug response biomarker, enabling the selection of patients most likely to respond to TTK inhibitor therapy in proof-of-concept clinical trials. Mol Cancer Ther; 16(11); 2609-17. ©2017 AACR.


Asunto(s)
Carcinoma Hepatocelular/tratamiento farmacológico , Proteínas de Ciclo Celular/genética , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Neoplasias Hepáticas/tratamiento farmacológico , Terapia Molecular Dirigida , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , beta Catenina/genética , Animales , Biomarcadores Farmacológicos , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Ratones , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Vía de Señalización Wnt/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Oncotarget ; 8(24): 38309-38325, 2017 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-28415765

RESUMEN

Inhibition of the spindle assembly checkpoint kinase TTK causes chromosome mis-segregation and tumor cell death. However, high levels of TTK correlate with chromosomal instability (CIN), which can lead to aneuploidy. We show that treatment of tumor cells with the selective small molecule TTK inhibitor NTRC 0066-0 overrides the mitotic checkpoint, irrespective of cell line sensitivity. In stable aneuploid cells NTRC 0066-0 induced acute CIN, whereas in cells with high levels of pre-existing CIN there was only a small additional fraction of cells mis-segregating their chromosomes. In proliferation assays stable aneuploid cells were more sensitive than cell lines with pre-existing CIN. Tetraploids are thought to be an intermediate between diploid and unstable aneuploid cells. TTK inhibitors had the same potency on post-tetraploid and parental diploid cells, which is remarkable because the post-tetraploids are more resistant to mitotic drugs. Finally, we confirm that the reference compound reversine is a TTK inhibitor and like NTRC 0066-0, inhibits the proliferation of patient-derived colorectal cancer organoids. In contrast, treatment with TTK inhibitor did not reduce the viability of non-proliferating T cell acute lymphoblastic leukemia cells samples. Consequently, TTK inhibitor therapy is expected to spare non-dividing cells, and may be used to target stable aneuploid tumors.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Inestabilidad Cromosómica/efectos de los fármacos , Neoplasias/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Aneuploidia , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Humanos , Puntos de Control de la Fase M del Ciclo Celular/efectos de los fármacos , Neoplasias/enzimología
6.
J Mol Biol ; 429(14): 2211-2230, 2017 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-28539250

RESUMEN

The protein kinase threonine tyrosine kinase (TTK; also known as Mps1) is a critical component of the spindle assembly checkpoint and a promising drug target for the treatment of aggressive cancers, such as triple negative breast cancer. While the first TTK inhibitors have entered clinical trials, little is known about how the inhibition of TTK with small-molecule compounds affects cellular activity. We studied the selective TTK inhibitor NTRC 0066-0, which was developed in our own laboratory, together with 11 TTK inhibitors developed by other companies, including Mps-BAY2b, BAY 1161909, BAY 1217389 (Bayer), TC-Mps1-12 (Shionogi), and MPI-0479605 (Myrexis). Parallel testing shows that the cellular activity of these TTK inhibitors correlates with their binding affinity to TTK and, more strongly, with target residence time. TTK inhibitors are therefore an example where target residence time determines activity in in vitro cellular assays. X-ray structures and thermal stability experiments reveal that the most potent compounds induce a shift of the glycine-rich loop as a result of binding to the catalytic lysine at position 553. This "lysine trap" disrupts the catalytic machinery. Based on these insights, we developed TTK inhibitors, based on a (5,6-dihydro)pyrimido[4,5-e]indolizine scaffold, with longer target residence times, which further exploit an allosteric pocket surrounding Lys553. Their binding mode is new for kinase inhibitors and can be classified as hybrid Type I/Type III. These inhibitors have very potent anti-proliferative activity that rivals classic cytotoxic therapy. Our findings will open up new avenues for more applications for TTK inhibitors in cancer treatment.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proliferación Celular/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas de Ciclo Celular/química , Línea Celular Tumoral , Cristalografía por Rayos X , Humanos , Cinética , Modelos Moleculares , Unión Proteica , Conformación Proteica/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/química , Proteínas Tirosina Quinasas/química
7.
Mol Cancer Ther ; 15(12): 3097-3109, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27587489

RESUMEN

Cancer cell line panels are important tools to characterize the in vitro activity of new investigational drugs. Here, we present the inhibition profiles of 122 anticancer agents in proliferation assays with 44 or 66 genetically characterized cancer cell lines from diverse tumor tissues (Oncolines). The library includes 29 cytotoxics, 68 kinase inhibitors, and 11 epigenetic modulators. For 38 compounds this is the first comparative profiling in a cell line panel. By strictly maintaining optimized assay protocols, biological variation was kept to a minimum. Replicate profiles of 16 agents over three years show a high average Pearson correlation of 0.8 using IC50 values and 0.9 using GI50 values. Good correlations were observed with other panels. Curve fitting appears a large source of variation. Hierarchical clustering revealed 44 basic clusters, of which 26 contain compounds with common mechanisms of action, of which 9 were not reported before, including TTK, BET and two clusters of EZH2 inhibitors. To investigate unexpected clusterings, sets of BTK, Aurora and PI3K inhibitors were profiled in biochemical enzyme activity assays and surface plasmon resonance binding assays. The BTK inhibitor ibrutinib clusters with EGFR inhibitors, because it cross-reacts with EGFR. Aurora kinase inhibitors separate into two clusters, related to Aurora A or pan-Aurora selectivity. Similarly, 12 inhibitors in the PI3K/AKT/mTOR pathway separated into different clusters, reflecting biochemical selectivity (pan-PI3K, PI3Kßγδ-isoform selective or mTOR-selective). Of these, only allosteric mTOR inhibitors preferentially targeted PTEN-mutated cell lines. This shows that cell line profiling is an excellent tool for the unbiased classification of antiproliferative compounds. Mol Cancer Ther; 15(12); 3097-109. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Aurora Quinasas/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Serina-Treonina Quinasas TOR/metabolismo , Aurora Quinasas/antagonistas & inhibidores , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Análisis por Conglomerados , Variaciones en el Número de Copia de ADN , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Perfilación de la Expresión Génica/métodos , Humanos , Mutación , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteómica/métodos , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
8.
Toxicol In Vitro ; 19(4): 491-503, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15826807

RESUMEN

In this study the focus is on the comparison of fluorometric assays, using Alamar Blue (AB) and Hoechst 33342 coloration, and luminometric assays, using Cyto-Lite and ATP-Lite, for toxicity measurements. With AB, ATP-Lite and Cyto-Lite the energy status of the cell is measured and with Hoechst 33342 the amount of DNA. These assays were carried out with different dosages of several toxic compounds with the following permanent cell lines: human liver (Hep G2), human endometrium (ECC-1), human cervix (HeLa) and Chinese hamster ovary (CHO) cells. In these assays toxicity of 110 compounds was assessed in Hep G2 cells. With 60 of those, toxicity was assessed in Hela, ECC-1 and CHO cells. These compounds were non-narcotic antitussives, nasal decongestants, narcotic analgesics, hypnotics, vasodilators, specific cellular energy blockers, cellular proliferation inhibitors, ion channel blockers, estrogens, antiestrogens, androgens, progestagens and others. The outcome of this study is that all four cell lines were responsive to the same set of 60 drugs with a comparable indication of toxicity. Hep G2 cells appear slightly more sensitive, as compared to the other three cell lines. Evaluation up to dosages of 3.2 x 10(-4) or even 3.2 x 10(-3)M for some of the compounds for these four assays in Hep G2 cells demonstrated toxicity for 45 of the 60 (75%) reference compounds with known toxicity in these assays. With a new set of 50 compounds, among which there were estrogens, androgens, progestagens and antiestrogens, 18 (36%) were identified as toxic up to a concentration of 3.2 x 10(-5)M. In conclusion, many of the 60 tested reference compounds gave similar dose and toxicity effects on these permanent cell lines. Therefore, all these cell lines can be used for toxicity screening with AB, ATP-Lite, Cyto-Lite and Hoechst 33342. However, species specific cell lines may reveal species specific effects, as shown with digoxin.


Asunto(s)
Adenosina Trifosfato/metabolismo , Supervivencia Celular/efectos de los fármacos , ADN/metabolismo , NADP/metabolismo , Animales , Bencimidazoles , Células CHO , Línea Celular , Colorantes , Cricetinae , Técnicas Citológicas , Endometrio/citología , Endometrio/metabolismo , Femenino , Fluorometría , Células HeLa , Humanos , Hígado/citología , Hígado/metabolismo , Oxazinas , Xantenos
9.
Toxicol In Vitro ; 19(4): 505-16, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15826808

RESUMEN

In this study fluorometric assays have been used for medium throughput screening on toxicity. Dichlorofluorescein diacetate, monochlorobimane and calcein-AM were fluorophores for the measurement of the formation of reactive oxygen species (ROS), the quantification of glutathione and the membrane stability, respectively. These assays have been carried out in the presence or absence of toxic compounds and with four different cell lines, i.e. human liver (Hep G2), human endometrium (ECC-1), human cervix (HeLa) and Chinese hamster ovary cells (CHO). In these assays the toxic dose of 60 reference compounds was assessed for Hep G2, HeLa, ECC-1 and CHO cells and of 40 pharmaceutical compounds for Hep G2 (ROS, glutathione) or HeLa (calcein) cells, only. These compounds were narcotic analgesics, hypnotics, vasodilators, specific cellular energy blockers, cellular proliferation inhibitors, ion channel blockers, estrogens, antiestrogens, androgens, progestagens and others. The outcome of this study revealed that all four cell lines were responsive to the same set of drugs. Only for some drugs Hep G2 cells appear slightly more sensitive, as compared to the other three cell lines. In general the HeLa cell line was the most sensitive cell line for the calcein uptake, while the Hep G2 cell line shows slightly more sensitivity for dichorofluorescein and monochlorobimane assays than the other three cell lines. Further evaluation at higher toxic dosages with Hep G2 cells for ROS and glutathione depletion and HeLa cells for calcein uptake, demonstrated toxic effects for 56 of the 100 reference compounds in these assays, among which there were estrogens, androgens, progestagens and antiestrogens. In conclusion, almost all tested compounds gave similar dose and toxicity effects on the permanent cell lines used in this study. Only three compounds showed more tissue specific cell responses. This shows that in principle all four cell lines can be used for toxicity screening.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Fluoresceínas/metabolismo , Glutatión/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Células CHO , Línea Celular , Cricetinae , Endometrio/citología , Endometrio/metabolismo , Femenino , Fluorometría , Células HeLa , Humanos , Hígado/citología , Hígado/metabolismo
10.
Cell Cycle ; 14(17): 2810-20, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26151317

RESUMEN

Up to 80% of human cancers, in particular solid tumors, contain cells with abnormal chromosomal numbers, or aneuploidy, which is often linked with marked chromosomal instability. Whereas in some tumors the aneuploidy occurs by missegregation of one or a few chromosomes, aneuploidy can also arise during proliferation of inherently unstable tetraploid cells generated by whole genome doubling from diploid cells. Recent findings from cancer genome sequencing projects suggest that nearly 40% of tumors underwent whole genome doubling at some point of tumorigenesis, yet its contribution to cancer phenotypes and benefits for malignant growth remain unclear. Here, we investigated the consequences of a whole genome doubling in both cancerous and non-transformed p53 positive human cells. SNP array analysis and multicolor karyotyping revealed that induced whole-genome doubling led to variable aneuploidy. We found that chromosomal instability (CIN) is a frequent, but not a default outcome of whole genome doubling. The CIN phenotypes were accompanied by increased tolerance to mitotic errors that was mediated by suppression of the p53 signaling. Additionally, the expression of pro-apoptotic factors, such as iASPP and cIAP2, was downregulated. Furthermore, we found that whole genome doubling promotes resistance to a broad spectrum of chemotherapeutic drugs and stimulates anchorage-independent growth even in non-transformed p53-positive human cells. Taken together, whole genome doubling provides multifaceted benefits for malignant growth. Our findings provide new insight why genome-doubling promotes tumorigenesis and correlates with poor survival in cancer.


Asunto(s)
Antineoplásicos/farmacología , Inestabilidad Cromosómica/fisiología , Resistencia a Múltiples Medicamentos/fisiología , Mitosis/fisiología , Tetraploidía , Línea Celular Tumoral , Inestabilidad Cromosómica/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Resistencia a Múltiples Medicamentos/efectos de los fármacos , Células HCT116 , Humanos , Mitosis/efectos de los fármacos
11.
PLoS One ; 10(5): e0125021, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26018524

RESUMEN

The aim of combination drug treatment in cancer therapy is to improve response rate and to decrease the probability of the development of drug resistance. Preferably, drug combinations are synergistic rather than additive, and, ideally, drug combinations work synergistically only in cancer cells and not in non-malignant cells. We have developed a workflow to identify such targeted synergies, and applied this approach to selectively inhibit the proliferation of cell lines with mutations in genes that are difficult to modulate with small molecules. The approach is based on curve shift analysis, which we demonstrate is a more robust method of determining synergy than combination matrix screening with Bliss-scoring. We show that the MEK inhibitor trametinib is more synergistic in combination with the BRAF inhibitor dabrafenib than with vemurafenib, another BRAF inhibitor. In addition, we show that the combination of MEK and BRAF inhibitors is synergistic in BRAF-mutant melanoma cells, and additive or antagonistic in, respectively, BRAF-wild type melanoma cells and non-malignant fibroblasts. This combination exemplifies that synergistic action of drugs can depend on cancer genotype. Next, we used curve shift analysis to identify new drug combinations that specifically inhibit cancer cell proliferation driven by difficult-to-drug cancer genes. Combination studies were performed with compounds that as single agents showed preference for inhibition of cancer cells with mutations in either the CTNNB1 gene (coding for ß-catenin), KRAS, or cancer cells expressing increased copy numbers of MYC. We demonstrate that the Wnt-pathway inhibitor ICG-001 and trametinib acted synergistically in Wnt-pathway-mutant cell lines. The ERBB2 inhibitor TAK-165 was synergistic with trametinib in KRAS-mutant cell lines. The EGFR/ERBB2 inhibitor neratinib acted synergistically with the spindle poison docetaxel and with the Aurora kinase inhibitor GSK-1070916 in cell lines with MYC amplification. Our approach can therefore efficiently discover novel drug combinations that selectively target cancer genes.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Terapia Molecular Dirigida/métodos , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , beta Catenina/genética , Compuestos Aza/administración & dosificación , Compuestos Aza/farmacología , Bencimidazoles/administración & dosificación , Bencimidazoles/farmacología , Línea Celular Tumoral/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Imidazoles/administración & dosificación , Imidazoles/farmacología , Indazoles/administración & dosificación , Indazoles/farmacología , Indoles/administración & dosificación , Indoles/farmacología , Melanoma/tratamiento farmacológico , Melanoma/genética , Melanoma/patología , Mutación , Oxazoles/administración & dosificación , Oxazoles/farmacología , Oximas/administración & dosificación , Oximas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Piridonas/administración & dosificación , Piridonas/farmacología , Pirimidinonas/administración & dosificación , Pirimidinonas/farmacología , Quinolinas/administración & dosificación , Quinolinas/farmacología , Sulfonamidas/administración & dosificación , Sulfonamidas/farmacología , Triazoles/administración & dosificación , Triazoles/farmacología , Vemurafenib , beta Catenina/metabolismo
12.
PLoS One ; 9(3): e92146, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24651269

RESUMEN

The anti-proliferative activities of all twenty-five targeted kinase inhibitor drugs that are in clinical use were measured in two large assay panels: (1) a panel of proliferation assays of forty-four human cancer cell lines from diverse tumour tissue origins; and (2) a panel of more than 300 kinase enzyme activity assays. This study provides a head-on comparison of all kinase inhibitor drugs in use (status Nov. 2013), and for six of these drugs, the first kinome profiling data in the public domain. Correlation of drug activities with cancer gene mutations revealed novel drug sensitivity markers, suggesting that cancers dependent on mutant CTNNB1 will respond to trametinib and other MEK inhibitors, and cancers dependent on SMAD4 to small molecule EGFR inhibitor drugs. Comparison of cellular targeting efficacies reveals the most targeted inhibitors for EGFR, ABL1 and BRAF(V600E)-driven cell growth, and demonstrates that the best targeted agents combine high biochemical potency with good selectivity. For ABL1 inhibitors, we computationally deduce optimized kinase profiles for use in a next generation of drugs. Our study shows the power of combining biochemical and cellular profiling data in the evaluation of kinase inhibitor drug action.


Asunto(s)
Marcación de Gen , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Inhibidores de Proteínas Quinasas/uso terapéutico , Análisis de Varianza , Marcadores Genéticos , Humanos , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Proteoma/metabolismo , Reproducibilidad de los Resultados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA