Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 13 de 13
Filtrer
1.
Mol Oncol ; 16(6): 1309-1328, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34669238

RÉSUMÉ

Small-cell lung cancer (SCLC) is a heterogeneous disease, consisting of intratumoral and intertumoral neuroendocrine (ASCL1 and/or NEUROD1), mesenchymal-like, and YAP-driven transcriptional states. Lysine-specific demethylase 1 (LSD1; also known as KDM1A) inhibitors have recently been progressed to clinical trials in SCLC based on a promising preclinical antitumor activity. A potential clinical limitation of LSD1 inhibitors is the heterogeneous drug responses that have been observed in SCLC cell lines and patient-derived models. Based on these observations, we studied molecular and transcriptional signatures that predict patient response to this class of drug. Employing SCLC patient-derived transcriptional signatures, we define that SCLC cell lines sensitive to LSD1 inhibitors are enriched in neuroendocrine transcriptional markers, whereas cell lines enriched in a mesenchymal-like transcriptional program demonstrate intrinsic resistance to LSD1 inhibitors. We have identified a reversible, adaptive resistance mechanism to LSD1 inhibitors through epigenetic reprogramming to a TEAD4-driven mesenchymal-like state. Our data suggest that only a segment of SCLC patients, with a defined neuroendocrine differentiation state, will likely benefit from LSD1 inhibitors. It provides novel evidence for the selection of a TEAD4-driven mesenchymal-like subpopulation resistant to LSD1 inhibitors in SCLC patients that may require effective drug combinations to sustain effective clinical responses.


Sujet(s)
Tumeurs du poumon , Carcinome pulmonaire à petites cellules , Protéines de liaison à l'ADN/génétique , Résistance aux substances , Histone Demethylases , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Protéines du muscle , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Carcinome pulmonaire à petites cellules/génétique , Carcinome pulmonaire à petites cellules/métabolisme , Facteurs de transcription à domaine TEA , Facteurs de transcription/génétique
2.
Future Oncol ; 16(1): 4289-4301, 2020 Jan.
Article de Anglais | MEDLINE | ID: mdl-31778074

RÉSUMÉ

Aim: This first-in-human, dose-finding study evaluated safety, pharmacokinetics and pharmacodynamics of crizotinib and established a recommended Phase II dose (RP2D) among patients with advanced solid malignancies. Patients & methods: Patients received oral crizotinib in a 3 + 3 dose escalation design. Results: Thirty-six patients received crizotinib (50 mg once daily-300 mg twice daily); maximum tolerated dose (and RP2D) was 250 mg twice daily. Most patients (89%) experienced ≥1 treatment-related adverse event. Three patients had grade 3 dose-limiting toxicities: alanine aminotransferase increased (n = 1) and fatigue (n = 2). Generally, an increase in soluble MET was found with increasing crizotinib concentrations. Conclusion: Crizotinib demonstrated a favorable safety profile. The observed pharmacodynamic effect on soluble MET provide evidence for targeted MET inhibition by crizotinib. Clinicaltrials. gov identifier: NCT00585195.


Sujet(s)
Kinase du lymphome anaplasique/antagonistes et inhibiteurs , Antinéoplasiques/usage thérapeutique , Crizotinib/usage thérapeutique , Tumeurs/traitement médicamenteux , Protein-tyrosine kinases/antagonistes et inhibiteurs , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Protéines proto-oncogènes/antagonistes et inhibiteurs , Antinéoplasiques/pharmacocinétique , Marqueurs biologiques tumoraux/métabolisme , Crizotinib/pharmacocinétique , Relation dose-effet des médicaments , Femelle , Études de suivi , Régulation de l'expression des gènes tumoraux , Humains , Mâle , Dose maximale tolérée , Adulte d'âge moyen , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Pronostic , Distribution tissulaire
3.
Int J Cancer ; 138(1): 195-205, 2016 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-26152787

RÉSUMÉ

Dysregulation of the Notch1 receptor has been shown to facilitate the development and progression of colorectal cancer (CRC) and has been identified as an independent predictor of disease progression and worse survival. Although mutations in the NOTCH1 receptor have not been described in CRC, we have previously discovered a NOTCH1 gene copy number gain in a portion of CRC tumor samples. Here, we demonstrated that a NOTCH1 gene copy number gain is significantly associated with worse survival and a high percentage of gene duplication in a cohort of patients with advanced CRC. In our CRC patient-derived tumor xenograft (PDTX) model, tumors harboring a NOTCH1 gain exhibited significant elevation of the Notch1 receptor, JAG1 ligand and cleaved Notch1 activity. In addition, a significant association was identified between a gain in NOTCH1 gene copy number and sensitivity to a Notch1-targeting antibody. These findings suggest that patients with metastatic CRC that harbor a gain in NOTCH1 gene copy number have worse survival and that targeting this patient population with a Notch1 antibody may yield improved outcomes.


Sujet(s)
Tumeurs colorectales/génétique , Tumeurs colorectales/mortalité , Variations de nombre de copies de segment d'ADN , Dosage génique , Récepteur Notch1/génétique , Animaux , Anticorps monoclonaux/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Apoptose/génétique , Marqueurs biologiques tumoraux , Protéines de liaison au calcium/métabolisme , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques/génétique , Femelle , Duplication de gène , Humains , Protéines et peptides de signalisation intercellulaire/métabolisme , Protéine jagged-1 , Mâle , Protéines membranaires/métabolisme , Souris , Métastase tumorale , Pronostic , Récepteur Notch1/antagonistes et inhibiteurs , Récepteur Notch1/métabolisme , Protéines serrate-jagged , Transduction du signal/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
4.
Clin Cancer Res ; 21(6): 1487-96, 2015 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-25564152

RÉSUMÉ

PURPOSE: To identify and characterize novel, activating mutations in Notch receptors in breast cancer and to determine response to the gamma secretase inhibitor (GSI) PF-03084014. EXPERIMENTAL DESIGN: We used several computational approaches, including novel algorithms, to analyze next-generation sequencing data and related omic datasets from The Cancer Genome Atlas (TCGA) breast cancer cohort. Patient-derived xenograft (PDX) models were sequenced, and Notch-mutant models were treated with PF-03084014. Gene-expression and functional analyses were performed to study the mechanism of activation through mutation and inhibition by PF-03084014. RESULTS: We identified mutations within and upstream of the PEST domains of NOTCH1, NOTCH2, and NOTCH3 in the TCGA dataset. Mutations occurred via several genetic mechanisms and compromised the function of the PEST domain, a negative regulatory domain commonly mutated in other cancers. Focal amplifications of NOTCH2 and NOTCH3 were also observed, as were heterodimerization or extracellular domain mutations at lower incidence. Mutations and amplifications often activated the Notch pathway as evidenced by increased expression of canonical Notch target genes, and functional mutations were significantly enriched in the triple-negative breast cancer subtype (TNBC). PDX models were also identified that harbored PEST domain mutations, and these models were highly sensitive to PF-03084014. CONCLUSIONS: This work suggests that Notch-altered breast cancer constitutes a bona fide oncogenic driver segment with the most common alteration being PEST domain mutations present in multiple Notch receptors. Importantly, functional studies suggest that this newly identified class can be targeted with Notch inhibitors, including GSIs.


Sujet(s)
Amyloid precursor protein secretases/antagonistes et inhibiteurs , Récepteur Notch1/génétique , Récepteur Notch2/génétique , Récepteurs Notch/génétique , Tumeurs du sein triple-négatives/traitement médicamenteux , Algorithmes , Animaux , Séquence nucléotidique , Lignée cellulaire tumorale , Prolifération cellulaire , Biologie informatique/méthodes , Variations de nombre de copies de segment d'ADN/génétique , Femelle , Dosage génique/génétique , Humains , Souris , Souris SCID , Structure tertiaire des protéines/génétique , Récepteur Notch3 , Analyse de séquence d'ADN , Transduction du signal/génétique , 1,2,3,4-Tétrahydro-naphtalènes/pharmacologie , Tumeurs du sein triple-négatives/génétique , Valine/analogues et dérivés , Valine/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
5.
J Thorac Oncol ; 9(3): 285-94, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24496003

RÉSUMÉ

INTRODUCTION: The aim of this study was to identify anaplastic lymphoma kinase (ALK) rearrangements in lung cancer patient-derived xenograft (PDX) models and to explore their responses to crizotinib. METHODS: Screening of 99 lung cancer PDX models by the NanoString ALK fusion assay identified two ALK-rearranged non-small-cell lung cancer (NSCLC) tumors, including one harboring a previously known echinoderm microtubule-associated protein-like 4 (EML4)-ALK fusion and another containing an unknown ALK fusion variant. Expression array, RNA-Seq, reverse transcription polymerase chain reaction, and direct sequencing were then conducted to confirm the rearrangements and to identify the novel fusion partner in the xenograft and/or the primary patient tumor. Finally, pharmacological studies were performed in PDX models to evaluate their responses to ALK inhibitor crizotinib. RESULTS: Two ALK-rearranged NSCLC PDX models were identified: one carried a well-known EML4-ALK variant 3a/b and the other harbored a novel huntingtin interacting protein 1 (HIP1)-ALK fusion gene. Exon 28 of the HIP1 gene located on chromosome 7 was fused to exon 20 of the ALK gene located on chromosome 2. Both cases were clinically diagnosed as squamous cell carcinoma. Compared with the other lung cancer PDX models, both ALK-rearranged models displayed elevated ALK mRNA expression. Furthermore, in vivo efficacy studies demonstrated that, similar to the EML4-ALK-positive model, the HIP1-ALK-containing PDX model was sensitive to treatment with crizotinib. CONCLUSIONS: Discovery of HIP1 as a fusion partner of ALK in NSCLC is a novel finding. In addition, the HIP1-ALK-rearranged tumor is sensitive to treatment with crizotinib in vivo, implicating HIP1-ALKas an oncogenic driver of lung tumorigenesis. Collectively, our results indicate that HIP1-ALK-positive NSCLC may benefit from clinical applications of crizotinib.


Sujet(s)
Carcinome pulmonaire non à petites cellules/génétique , Protéines de liaison à l'ADN/génétique , Tumeurs du poumon/génétique , Protéines de fusion oncogènes/génétique , Inhibiteurs de protéines kinases/pharmacologie , Pyrazoles/pharmacologie , Pyridines/pharmacologie , Récepteurs à activité tyrosine kinase/génétique , Kinase du lymphome anaplasique , Animaux , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Carcinome pulmonaire non à petites cellules/anatomopathologie , Crizotinib , Protéines de liaison à l'ADN/antagonistes et inhibiteurs , Femelle , Humains , Hybridation fluorescente in situ , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/anatomopathologie , Souris , Souris de lignée BALB C , Protéines de fusion oncogènes/antagonistes et inhibiteurs , ARN messager/génétique , Réaction de polymérisation en chaine en temps réel , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , RT-PCR , Cellules cancéreuses en culture
6.
J Mol Diagn ; 16(2): 229-43, 2014 Mar.
Article de Anglais | MEDLINE | ID: mdl-24418728

RÉSUMÉ

Approximately 7% of non-small cell lung carcinomas (NSCLCs) harbor oncogenic fusions involving ALK, ROS1, and RET. Although tumors harboring ALK fusions are highly sensitive to crizotinib, emerging preclinical and clinical data demonstrate that patients with ROS1 or RET fusions may also benefit from inhibitors targeting these kinases. Using a transcript-based method, we designed a combination of 3' overexpression and fusion-specific detection strategies to detect ALK, ROS1 and RET fusion transcripts in NSCLC tumors. We validated the assay in 295 NSCLC specimens and showed that the assay is highly sensitive and specific. ALK results were 100% concordant with fluorescence in situ hybridization (FISH) (n = 52) and 97.8% concordant with IHC (n = 179) [sensitivity, 96.8% (95% CI 91.0%-98.9%); specificity, 98.8% (95% CI 93.6%-99.8%)]. For ROS1 and RET, we also observed 100% concordance with FISH (n = 46 and n = 15, respectively). We identified seven ROS1 and 14 RET fusion-positive tumors and confirmed the fusion status by RT-PCR and FISH. One RET fusion involved a novel partner, cutlike homeobox 1 gene (CUX1), yielding an in-frame CUX1-RET fusion. ROS1 and RET fusions were significantly enriched in tumors without KRAS/EGFR/ALK alterations. ALK/ROS1/RET/EGFR/KRAS alterations were mutually exclusive. As a single-tube assay, this test shows promise as a more practical and cost-effective screening modality for detecting rare but targetable fusions in NSCLC.


Sujet(s)
Dépistage génétique/méthodes , Tumeurs du poumon/diagnostic , Tumeurs du poumon/génétique , Protéines de fusion oncogènes/génétique , Protein-tyrosine kinases/génétique , Protéines proto-oncogènes c-ret/génétique , Protéines proto-oncogènes/génétique , Récepteurs à activité tyrosine kinase/génétique , Kinase du lymphome anaplasique , Carcinome pulmonaire non à petites cellules/diagnostic , Carcinome pulmonaire non à petites cellules/génétique , Femelle , Ordre des gènes , Humains , Hybridation fluorescente in situ , Mâle , Reproductibilité des résultats , RT-PCR , Sensibilité et spécificité , Analyse de séquence d'ADN , Transcription génétique , Translocation génétique
7.
Mol Cancer Ther ; 12(12): 2929-39, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-24107449

RÉSUMÉ

Figitumumab (CP-751,871), a potent and fully human monoclonal anti-insulin-like growth factor 1 receptor (IGF1R) antibody, has been investigated in clinical trials of several solid tumors. To identify biomarkers of sensitivity and resistance to figitumumab, its in vitro antiproliferative activity was analyzed in a panel of 93 cancer cell lines by combining in vitro screens with extensive molecular profiling of genomic aberrations. Overall response was bimodal and the majority of cell lines were resistant to figitumumab. Nine of 15 sensitive cell lines were derived from colon cancers. Correlations between genomic characteristics of cancer cell lines with figitumumab antiproliferative activity revealed that components of the IGF pathway, including IRS2 (insulin receptor substrate 2) and IGFBP5 (IGF-binding protein 5), played a pivotal role in determining the sensitivity of tumors to single-agent figitumumab. Tissue-specific differences among the top predictive genes highlight the need for tumor-specific patient selection strategies. For the first time, we report that alteration or expression of the MYB oncogene is associated with sensitivity to IGF1R inhibitors. MYB is dysregulated in hematologic and epithelial tumors, and IGF1R inhibition may represent a novel therapeutic opportunity. Although growth inhibitory activity with single-agent figitumumab was relatively rare, nine combinations comprising figitumumab plus chemotherapeutic agents or other targeted agents exhibited properties of synergy. Inhibitors of the ERBB family were frequently synergistic and potential biomarkers of drug synergy were identified. Several biomarkers of antiproliferative activity of figitumumab both alone and in combination with other therapies may inform the design of clinical trials evaluating IGF1R inhibitors.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Antinéoplasiques/pharmacologie , Résistance aux médicaments antinéoplasiques/génétique , Récepteur IGF de type 1/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Analyse de regroupements , Variations de nombre de copies de segment d'ADN , Synergie des médicaments , Analyse de profil d'expression de gènes , Humains , Concentration inhibitrice 50 , Mutation , Locus de caractère quantitatif , Récepteur IGF de type 1/métabolisme , Transduction du signal
8.
Clin Cancer Res ; 18(18): 5008-19, 2012 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-22806875

RÉSUMÉ

PURPOSE: We aimed to assess the biologic activity of PF-03084014 in breast xenograft models. The biomarkers for mechanism and patient stratification were also explored. EXPERIMENTAL DESIGN: The in vitro and in vivo properties of PF-03084014 were investigated. The mRNA expressions of 40 key Notch pathway genes at baseline or after treatment were analyzed to link with the antitumor efficacy of PF-03084014 in a panel of breast cancer xenograft models. RESULTS: In vitro, PF-03084014 exhibited activity against tumor cell migration, endothelial cell tube formation, and mammosphere formation. In vivo, we observed apoptosis, antiproliferation, reduced tumor cell self-renewal ability, impaired tumor vasculature, and decreased metastasis activity after the treatment of PF-03084014. PF-03084014 treatment displayed significant antitumor activity in 10 of the 18 breast xenograft models. However, the antitumor efficacy in most models did not correlate with the in vitro antiproliferation results in the corresponding cell lines, suggesting the critical involvement of tumor microenvironment during Notch activation. In the tested breast xenograft models, the baseline expressions of the Notch receptors, ligands, and the cleaved Notch1 failed to predict the antitumor response to PF-03084014, whereas several Notch pathway target genes, including HEY2, HES4, and HES3, strongly corresponded with the response with a P value less than 0.01. Many of the best molecular predictors of response were also significantly modulated following PF-03084014 treatment. CONCLUSIONS: PF-03084014 showed antitumor and antimetastatic properties via pleiotropic mechanisms. The Notch pathway downstream genes may be used to predict the antitumor activity of PF-03084014 and enrich for responders among breast cancer patients.


Sujet(s)
Amyloid precursor protein secretases/antagonistes et inhibiteurs , Antinéoplasiques/pharmacologie , Marqueurs biologiques tumoraux/métabolisme , Tumeurs du sein/métabolisme , 1,2,3,4-Tétrahydro-naphtalènes/pharmacologie , Valine/analogues et dérivés , Amyloid precursor protein secretases/métabolisme , Antinéoplasiques/administration et posologie , Marqueurs biologiques tumoraux/génétique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/génétique , Lignée cellulaire tumorale , Analyse de regroupements , Femelle , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/effets des médicaments et des substances chimiques , Cellules endothéliales de la veine ombilicale humaine/métabolisme , Humains , Métastase tumorale/traitement médicamenteux , Récepteurs Notch/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , 1,2,3,4-Tétrahydro-naphtalènes/administration et posologie , Valine/administration et posologie , Valine/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe
9.
Mol Cancer Ther ; 11(3): 710-9, 2012 Mar.
Article de Anglais | MEDLINE | ID: mdl-22222631

RÉSUMÉ

PF-03814735 is a novel, reversible inhibitor of Aurora kinases A and B that finished a phase I clinical trial for the treatment of advanced solid tumors. To find predictive biomarkers of drug sensitivity, we screened a diverse panel of 87 cancer cell lines for growth inhibition upon PF-03814735 treatment. Small cell lung cancer (SCLC) and, to a lesser extent, colon cancer lines were very sensitive to PF-03814735. The status of the Myc gene family and retinoblastoma pathway members significantly correlated with the efficacy of PF-03814735. Whereas RB1 inactivation, intact CDKN2A/p16, and normal CCND1/Cyclin D1 status are hallmarks of SCLC, activation or amplification of any of the three Myc genes (MYC, MYCL1, and MYCN) clearly differentiated cell line sensitivity within the SCLC panel. By contrast, we found that expression of Aurora A and B were weak predictors of response. We observed a decrease in histone H3 phosphorylation and polyploidization of sensitive lines, consistent with the phenotype of Aurora B inhibition. In vivo experiments with two SCLC xenograft models confirmed the sensitivity of Myc gene-driven models to PF-03814735 and a possible schedule dependence of MYC/c-Myc-driven tumors. Altogether our results suggest that SCLC and other malignancies driven by the Myc family genes may be suitable indications for treatment by Aurora B kinase inhibitors.


Sujet(s)
Marqueurs biologiques tumoraux/génétique , Composés hétérocycliques 3 noyaux/pharmacologie , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Pyrimidines/pharmacologie , Animaux , Antinéoplasiques/pharmacologie , Aurora kinase A , Aurora kinase B , Aurora kinases , Marqueurs biologiques tumoraux/métabolisme , Technique de Western , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Analyse de profil d'expression de gènes , Génomique/méthodes , Histone/métabolisme , Humains , Tumeurs du poumon/traitement médicamenteux , Tumeurs du poumon/génétique , Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Souris , Souris nude , Phosphorylation/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme , Interférence par ARN , RT-PCR , Carcinome pulmonaire à petites cellules/traitement médicamenteux , Carcinome pulmonaire à petites cellules/génétique , Carcinome pulmonaire à petites cellules/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
10.
Eur J Pharmacol ; 489(3): 139-49, 2004 Apr 12.
Article de Anglais | MEDLINE | ID: mdl-15087236

RÉSUMÉ

Taste receptors are thought to couple to the G protein Galpha-gustducin to initiate signal transduction cascades leading to taste perception. To further characterize the G protein-coupling selectivity of these receptors, we expressed them in HEK293 cells and monitored the modulation of different signaling pathways upon stimulation. We found that the bitter compound cycloheximide induces phosphorylation of extracellular signal-regulated kinases1 and 2 (ERK 1/2) and inhibits cAMP accumulation in HEK293 cells expressing the mouse bitter T2R(5) receptor. These effects are totally abolished upon treatment with pertussis toxin. On the other hand, sweeteners and monosodium glutamate induce phosphorylation of ERK1/2 and inhibit cAMP accumulation in HEK293 cells expressing the human sweet T1R(2)/T1R(3) receptor and the human umami T1R(1)/T1R(3) receptor, respectively. The effects of these taste modalities are also prevented by treatment with pertussis toxin. Collectively, our results show that taste receptors can functionally couple to Galpha(i/o) proteins to transmit intracellular signals.


Sujet(s)
Sous-unités alpha Gi-Go des protéines G/physiologie , Système de signalisation des MAP kinases/physiologie , Récepteurs couplés aux protéines G/métabolisme , Calicules gustatifs/physiologie , Goût/effets des médicaments et des substances chimiques , Inhibiteurs des adénylate cyclases , Adenylate Cyclase/métabolisme , Animaux , Aspartame/pharmacologie , Lignée cellulaire , Colforsine/antagonistes et inhibiteurs , Colforsine/pharmacologie , Cyclamates/pharmacologie , AMP cyclique/antagonistes et inhibiteurs , AMP cyclique/composition chimique , AMP cyclique/métabolisme , Cycloheximide/métabolisme , Cycloheximide/pharmacologie , Relation dose-effet des médicaments , Fructose/pharmacologie , Humains , Souris , Mitogen-Activated Protein Kinase 1/métabolisme , Mitogen-Activated Protein Kinase 3/métabolisme , Toxine pertussique/pharmacologie , Phosphorylation/effets des médicaments et des substances chimiques , Récepteurs couplés aux protéines G/effets des médicaments et des substances chimiques , Récepteurs couplés aux protéines G/génétique , Saccharine/métabolisme , Saccharine/pharmacologie , Glutamate de sodium/métabolisme , Glutamate de sodium/pharmacologie , Saccharose/métabolisme , Saccharose/pharmacologie , Goût/génétique , Calicules gustatifs/effets des médicaments et des substances chimiques , Transducine/physiologie , Tryptophane/métabolisme , Tryptophane/pharmacologie
11.
Bioorg Med Chem Lett ; 14(2): 535-9, 2004 Jan 19.
Article de Anglais | MEDLINE | ID: mdl-14698198

RÉSUMÉ

The SAR for the affinity to the A(1) adenosine receptor and relative intrinsic efficacy (IE, [(35)S]-GTPgammaS binding) of a series of 5'-carbamate and 5'-thionocarbamate derivatives of tecadenoson is described. Based on this SAR, selected compounds were evaluated in guinea pig isolated hearts to determine whether they were partial or full agonists with respect to their negative dromotropism, an A(1) AdoR mediated effect. Progress towards obtaining a partial A(1) AdoR agonist to potentially control ventricular rate during atrial fibrillation has been made with the discovery of several potent partial A(1) AdoR agonists (compounds 13, 14, and 17).


Sujet(s)
Adénosine/analogues et dérivés , Adénosine/pharmacologie , Fibrillation auriculaire/traitement médicamenteux , Fibrillation auriculaire/métabolisme , Rythme cardiaque/effets des médicaments et des substances chimiques , Récepteur A1 à l'adénosine/métabolisme , Adénosine/métabolisme , Adénosine/usage thérapeutique , Animaux , Cochons d'Inde , Rythme cardiaque/physiologie , Humains , Techniques in vitro , Foie/métabolisme , Liaison aux protéines/physiologie , Rats , Relation structure-activité
12.
J Pharmacol Exp Ther ; 305(1): 225-31, 2003 Apr.
Article de Anglais | MEDLINE | ID: mdl-12649373

RÉSUMÉ

Elevated serum nonesterified free fatty acid (NEFA) concentrations are detrimental to both the mechanical and electrical function of the heart. A(1) adenosine receptor agonists are potent and efficacious inhibitors of lipolysis; however, their cardiovascular effects have limited their use to lower serum NEFA. Our objective was to determine whether the antilipolytic effect of N-[3-(R)-tetrahydrofuranyl]-6-aminopurine riboside (CVT-510), an A(1) agonist, could be distinguished from its bradycardia effect and demonstrated in rats with normal or elevated serum NEFA. Rats were instrumented with telemetry transmitters for continuous recording of heart rate, and catheterized, for delivery of drugs and blood sampling. CVT-510 caused a rapid and sustained dose-dependent decrease in NEFA at doses that did not cause bradycardia (2, 5, and 20 micro g/kg). Significant bradycardia was observed at 50 micro g/kg. Norepinephrine (NE) increased NEFA from 0.5 +/- 0.01 to 0.9 +/- 0.2 mM and this effect lasted for 2 h. CVT-510 (10 micro g/kg) given at 40 min postinjection of NE reversed the rise in NEFA (69% reduction). When CVT-510 (20 micro g/kg) was given 15 min before a 30-min long infusion of NE, the lipolytic response to NE was prevented. To mimic the antilipolytic effect of CVT-510 in awake rats, hearts were perfused with palmitate at concentrations similar to those observed in the in vivo studies (0.8 and 0.2 mM), which decreased myocardial oxygen consumption (MVO(2)) by 11%. Thus, CVT-510 at doses > or =5-fold lower than those that slow heart rate caused a marked and sustained lowering of normal or elevated NEFA, that when mimicked in vitro decreased MVO(2) and would be expected to improve cardiac efficiency.


Sujet(s)
Adénosine/analogues et dérivés , Adénosine/pharmacologie , Système cardiovasculaire/effets des médicaments et des substances chimiques , Furanes/pharmacologie , Lipolyse/effets des médicaments et des substances chimiques , Agonistes des récepteurs purinergiques P1 , Adipocytes/effets des médicaments et des substances chimiques , Adipocytes/métabolisme , Animaux , Catécholamines/physiologie , Interactions médicamenteuses , Acides gras/métabolisme , Coeur/effets des médicaments et des substances chimiques , Lipolyse/physiologie , Mâle , Myocarde/métabolisme , Norépinéphrine/pharmacologie , Consommation d'oxygène , Rats , Rat Sprague-Dawley
13.
Br J Pharmacol ; 135(6): 1457-66, 2002 Mar.
Article de Anglais | MEDLINE | ID: mdl-11906959

RÉSUMÉ

1. Adipocyte A(1)-adenosine receptors (A(1) AdoR) tonically inhibit adenylyl cyclase and lipolysis. Three potential explanations for tonic activity of A(1)AdoR of rat epididymal adipocytes were investigated: high affinity of adenosine for the receptor, efficient coupling of receptor activation to response, and spontaneous activity of the receptor in the absence of agonist. 2. The affinity of adenosine for the adipocyte A(1)AdoR was determined as 4.6 microM by analysis of effects of an irreversible receptor antagonist on agonist concentration-response relationships. In contrast, the potency of adenosine to decrease cyclic AMP in isolated adipocytes was 1.4 nM. 3. Occupancy by agonist of the A(1)AdoR was efficiently coupled to functional response (decrease of adipocyte cyclic AMP content). Activation by adenosine of less than 1% of A(1)AdoRs caused a near-maximal decrease of cyclic AMP in adipocytes. Thus the receptor reserve for adenosine to decrease cyclic AMP content of adipocytes was greater than 99%. 4. Affinities and receptor reserves for other A(1)AdoR agonists were determined. Agonists appeared to differ more in their affinity for the receptor than in their intrinsic efficacy to activate it. 5. A(1)AdoRs were inactive in the absence of agonist. 6. It is concluded that adipocyte A(1)AdoR are tonically activated by endogenous adenosine at nanomolar concentrations. The expression of a high density of A(1)AdoR that are efficiently coupled to a functional response enables the adipocyte to respond with high sensitivity to the low-affinity agonist, adenosine. Adipocytes may be a model for cells whose functions are tonically modulated by adenosine present in the interstitium of well-oxygenated tissues.


Sujet(s)
Adipocytes/métabolisme , Agonistes des récepteurs purinergiques P1 , Récepteurs purinergiques P1/métabolisme , Adénosine/métabolisme , Adénosine/pharmacologie , Adipocytes/effets des médicaments et des substances chimiques , Animaux , Séparation cellulaire , AMP cyclique/métabolisme , Relation dose-effet des médicaments , Mâle , Rats , Rat Sprague-Dawley , Récepteurs purinergiques P1/physiologie , Vasodilatateurs/métabolisme , Vasodilatateurs/pharmacologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE