Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 11 de 11
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Sci Rep ; 7(1): 15270, 2017 11 10.
Article de Anglais | MEDLINE | ID: mdl-29127379

RÉSUMÉ

Activating mutations in the Wnt pathway are a characteristic feature of colorectal cancer (CRC). The R-spondin (RSPO) family is a group of secreted proteins that enhance Wnt signaling and RSPO2 and RSPO3 gene fusions have been reported in CRC. We have previously shown that Wnt pathway blockers exhibit potent combinatorial activity with taxanes to inhibit tumor growth. Here we show that RSPO3 antagonism synergizes with paclitaxel based chemotherapies in patient-derived xenograft models (PDX) with RSPO3 fusions and in tumors with common CRC mutations such as APC, ß-catenin, or RNF43. In these latter types of tumors that represent over 90% of CRC, RSPO3 is produced by stromal cells in the tumor microenvironment and the activating mutations appear to sensitize the tumors to Wnt-Rspo synergy. The combination of RSPO3 inhibition and taxane treatment provides an approach to effectively target oncogenic WNT signaling in a significant number of patients with colorectal and other intestinal cancers.


Sujet(s)
Composés pontés/pharmacologie , Tumeurs colorectales , Mutation , Protéines tumorales , Paclitaxel/pharmacologie , Taxoïdes/pharmacologie , Thrombospondines , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Voie de signalisation Wnt , Animaux , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/génétique , Souris , Souris de lignée NOD , Souris SCID , Protéines tumorales/antagonistes et inhibiteurs , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Thrombospondines/antagonistes et inhibiteurs , Thrombospondines/génétique , Thrombospondines/métabolisme , Microenvironnement tumoral/génétique , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , Voie de signalisation Wnt/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
2.
Sci Adv ; 3(6): e1700090, 2017 06.
Article de Anglais | MEDLINE | ID: mdl-28691093

RÉSUMÉ

The WNT pathway mediates intercellular signaling that regulates cell fate in both normal development and cancer. It is widely appreciated that the WNT pathway is frequently dysregulated in human cancers through a variety of genetic and epigenetic mechanisms. Targets in the WNT pathway are being extensively pursued for the development of new anticancer therapies, and we have advanced two WNT antagonists for clinical development: vantictumab (anti-FZD) and ipafricept (FZD8-Fc). We examined the antitumor efficacy of these WNT antagonists in combination with various chemotherapies in a large set of patient-derived xenograft models. In responsive models, WNT blockade led to profound synergy with taxanes such as paclitaxel, and the combination activity with taxanes was consistently more effective than with other classes of chemotherapy. Taxane monotherapy increased the frequency of cells with active WNT signaling. This selection of WNT-active chemotherapy-resistant tumorigenic cells was prevented by WNT-antagonizing biologics and required sequential dosing of the WNT antagonist followed by the taxane. The WNT antagonists potentiated paclitaxel-mediated mitotic blockade and promoted widespread mitotic cell death. By blocking WNT/ß-catenin signaling before mitotic blockade by paclitaxel, we found that this treatment effectively sensitizes cancer stem cells to taxanes. This combination strategy and treatment regimen has been incorporated into ongoing clinical testing for vantictumab and ipafricept.


Sujet(s)
Antinéoplasiques/pharmacologie , Mitose/effets des médicaments et des substances chimiques , Taxoïdes/pharmacologie , Protéines de type Wingless/antagonistes et inhibiteurs , Mort cellulaire/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Humains , Paclitaxel/pharmacologie , Voie de signalisation Wnt/effets des médicaments et des substances chimiques , bêta-Caténine/antagonistes et inhibiteurs
3.
Nature ; 545(7654): 360-364, 2017 05 18.
Article de Anglais | MEDLINE | ID: mdl-28489825

RÉSUMÉ

The Notch signalling pathway mediates cell fate decisions and is tumour suppressive or oncogenic depending on the context. During lung development, Notch pathway activation inhibits the differentiation of precursor cells to a neuroendocrine fate. In small-cell lung cancer, an aggressive neuroendocrine lung cancer, loss-of-function mutations in NOTCH genes and the inhibitory effects of ectopic Notch activation indicate that Notch signalling is tumour suppressive. Here we show that Notch signalling can be both tumour suppressive and pro-tumorigenic in small-cell lung cancer. Endogenous activation of the Notch pathway results in a neuroendocrine to non-neuroendocrine fate switch in 10-50% of tumour cells in a mouse model of small-cell lung cancer and in human tumours. This switch is mediated in part by Rest (also known as Nrsf), a transcriptional repressor that inhibits neuroendocrine gene expression. Non-neuroendocrine Notch-active small-cell lung cancer cells are slow growing, consistent with a tumour-suppressive role for Notch, but these cells are also relatively chemoresistant and provide trophic support to neuroendocrine tumour cells, consistent with a pro-tumorigenic role. Importantly, Notch blockade in combination with chemotherapy suppresses tumour growth and delays relapse in pre-clinical models. Thus, small-cell lung cancer tumours generate their own microenvironment via activation of Notch signalling in a subset of tumour cells, and the presence of these cells may serve as a biomarker for the use of Notch pathway inhibitors in combination with chemotherapy in select patients with small-cell lung cancer.


Sujet(s)
Tumeurs du poumon/métabolisme , Tumeurs du poumon/anatomopathologie , Récepteurs Notch/métabolisme , Transduction du signal , Carcinome pulmonaire à petites cellules/métabolisme , Carcinome pulmonaire à petites cellules/anatomopathologie , Microenvironnement tumoral , Animaux , Différenciation cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Modèles animaux de maladie humaine , Femelle , Humains , Tumeurs du poumon/traitement médicamenteux , Mâle , Souris , Récidive tumorale locale/prévention et contrôle , Récepteurs Notch/agonistes , Récepteurs Notch/antagonistes et inhibiteurs , Récepteurs Notch/déficit , Protéines de répression/métabolisme , Carcinome pulmonaire à petites cellules/traitement médicamenteux
4.
Cancer Res ; 76(3): 713-23, 2016 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-26719531

RÉSUMÉ

Deregulation of the ß-catenin signaling has long been associated with cancer. Intracellular components of this pathway, including axin, APC, and ß-catenin, are frequently mutated in a range of human tumors, but the contribution of specific extracellular ligands that promote cancer development through this signaling axis remains unclear. We conducted a reporter-based screen in a panel of human tumors to identify secreted factors that stimulate ß-catenin signaling. Through this screen and further molecular characterization, we found that R-spondin (RSPO) proteins collaborate with Wnt proteins to activate ß-catenin. RSPO family members were expressed in several human tumors representing multiple malignancies, including ovarian, pancreatic, colon, breast, and lung cancer. We generated specific monoclonal antibody antagonists of RSPO family members and found that anti-RSPO treatment markedly inhibited tumor growth in human patient-derived tumor xenograft models, either as single agents or in combination with chemotherapy. Furthermore, blocking RSPO signaling reduced the tumorigenicity of cancer cells based on serial transplantation studies. Moreover, gene-expression analyses revealed that anti-RSPO treatment in responsive tumors strongly inhibited ß-catenin target genes known to be associated with cancer and normal stem cells. Collectively, our results suggest that the RSPO family is an important stimulator of ß-catenin activity in many human tumors and highlight a new effective approach for therapeutically modulating this fundamental signaling axis.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Thrombospondines/métabolisme , bêta-Caténine/métabolisme , Animaux , Carcinogenèse , Lignée cellulaire tumorale , Cellules HEK293 , Humains , Souris , Souris de lignée NOD , Souris SCID , Thérapie moléculaire ciblée , Tumeurs/génétique , Tumeurs/anatomopathologie , Transduction du signal , Thrombospondines/biosynthèse , Thrombospondines/génétique , Thrombospondines/immunologie , Protéines de type Wingless/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
5.
Clin Cancer Res ; 21(9): 2084-95, 2015 May 01.
Article de Anglais | MEDLINE | ID: mdl-25934888

RÉSUMÉ

PURPOSE: The Notch pathway plays an important role in both stem cell biology and cancer. Dysregulation of Notch signaling has been reported in several human tumor types. In this report, we describe the development of an antibody, OMP-59R5 (tarextumab), which blocks both Notch2 and Notch3 signaling. EXPERIMENTAL DESIGN: We utilized patient-derived xenograft tumors to evaluate antitumor effect of OMP-59R5. Immunohistochemistry, RNA microarray, real-time PCR, and in vivo serial transplantation assays were employed to investigate the mechanisms of action and pharmacodynamic readouts. RESULTS: We found that anti-Notch2/3, either as a single agent or in combination with chemotherapeutic agents was efficacious in a broad spectrum of epithelial tumors, including breast, lung, ovarian, and pancreatic cancers. Notably, the sensitivity of anti-Notch2/3 in combination with gemcitabine in pancreatic tumors was associated with higher levels of Notch3 gene expression. The antitumor effect of anti-Notch2/3 in combination with gemcitabine plus nab-paclitaxel was greater than the combination effect with gemcitabine alone. OMP-59R5 inhibits both human and mouse Notch2 and Notch3 function and its antitumor activity was characterized by a dual mechanism of action in both tumor and stromal/vascular cells in xenograft experiments. In tumor cells, anti-Notch2/3 inhibited expression of Notch target genes and reduced tumor-initiating cell frequency. In the tumor stroma, OMP-59R5 consistently inhibited the expression of Notch3, HeyL, and Rgs5, characteristic of affecting pericyte function in tumor vasculature. CONCLUSIONS: These findings indicate that blockade of Notch2/3 signaling with this cross-reactive antagonist antibody may be an effective strategy for treatment of a variety of tumor types.


Sujet(s)
Anticorps monoclonaux/pharmacologie , Antinéoplasiques/pharmacologie , Tumeurs expérimentales/traitement médicamenteux , Cellules souches tumorales/effets des médicaments et des substances chimiques , Récepteur Notch2/antagonistes et inhibiteurs , Récepteurs Notch/antagonistes et inhibiteurs , Animaux , Humains , Immunohistochimie , Souris , Séquençage par oligonucléotides en batterie , Réaction de polymérisation en chaine en temps réel , Récepteur Notch3 , Tests d'activité antitumorale sur modèle de xénogreffe
6.
Thyroid ; 23(12): 1569-77, 2013 Dec.
Article de Anglais | MEDLINE | ID: mdl-23705946

RÉSUMÉ

BACKGROUND: A limited number of approved therapeutic options are available to metastatic medullary thyroid cancer (MTC) patients, and the response to conventional chemotherapy and/or radiotherapy strategies is inadequate. Sporadic and inherited mutations in the tyrosine kinase RET result in oncogenic activation that is associated with the pathogenesis of MTC. Cabozantinib is a potent inhibitor of MET, RET, and vascular endothelial factor receptor 2 (VEGFR2), as well as other tyrosine kinases that have been implicated in tumor development and progression. The object of this study was to determine the in vitro biochemical and cellular inhibitory profile of cabozantinib against RET, and in vivo antitumor efficacy using a xenograft model of MTC. METHODS: Cabozantinib was evaluated in biochemical and cell-based assays that determined the potency of the compound against wild type and activating mutant forms of RET. Additionally, the pharmacodynamic modulation of RET and MET and in vivo antitumor activity of cabozantinib was examined in a MTC tumor model following subchronic oral administration. RESULTS: In biochemical assays, cabozantinib inhibited multiple forms of oncogenic RET kinase activity, including M918T and Y791F mutants. Additionally, it inhibited proliferation of TT tumor cells that harbor a C634W activating mutation of RET that is most often associated with MEN2A and familial MTC. In these same cells grown as xenograft tumors in nude mice, oral administration of cabozantinib resulted in dose-dependent tumor growth inhibition that correlated with a reduction in circulating plasma calcitonin levels. Moreover, immunohistochemical analyses of tumors revealed that cabozantinib reduced levels of phosphorylated MET and RET, and decreased tumor cellularity, proliferation, and vascularization. CONCLUSIONS: Cabozantinib is a potent inhibitor of RET and prevalent mutationally activated forms of RET known to be associated with MTC, and effectively inhibits the growth of a MTC tumor cell model in vitro and in vivo.


Sujet(s)
Anilides/pharmacologie , Carcinome médullaire/traitement médicamenteux , Pyridines/pharmacologie , Récepteurs à activité tyrosine kinase/antagonistes et inhibiteurs , Tumeurs de la thyroïde/traitement médicamenteux , Anilides/usage thérapeutique , Animaux , Carcinome médullaire/métabolisme , Carcinome médullaire/anatomopathologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Souris , Souris nude , Phosphorylation/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Protéines proto-oncogènes c-ret/antagonistes et inhibiteurs , Pyridines/usage thérapeutique , Tumeurs de la thyroïde/métabolisme , Tumeurs de la thyroïde/anatomopathologie , Résultat thérapeutique , Récepteur-2 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs
7.
J Med Chem ; 55(9): 4322-35, 2012 May 10.
Article de Anglais | MEDLINE | ID: mdl-22497444

RÉSUMÉ

Targeting glycosphingolipid synthesis has emerged as a novel approach for treating metabolic diseases. 32 (EXEL-0346) represents a new class of glucosylceramide synthase (GCS) inhibitors. This report details the elaboration of hit 8 with the goal of achieving and maintaining maximum GCS inhibition in vivo. 32 inhibited GCS with an IC(50) of 2 nM and achieved maximum hepatic GCS inhibition after four or five daily doses in rodents. Robust improvements in glucose tolerance in DIO mice and ZDF rats were observed after 2 weeks of q.d. dosing. Four weeks of dosing resulted in decreased plasma triglycerides and reduced hepatic fat deposition. Thus, 32 provides insight into the amount of metabolic regulation that can be restored following achievement of maximal target knockdown.


Sujet(s)
Antienzymes/synthèse chimique , Glucosyltransferases/antagonistes et inhibiteurs , Phénylalanine/analogues et dérivés , Tissu adipeux blanc/effets des médicaments et des substances chimiques , Tissu adipeux blanc/métabolisme , Animaux , Diabète de type 2/traitement médicamenteux , Diabète de type 2/enzymologie , Antienzymes/composition chimique , Antienzymes/pharmacologie , Femelle , Gangliosides/métabolisme , Hyperglycémie provoquée , Glucosyltransferases/métabolisme , Humains , Foie/effets des médicaments et des substances chimiques , Foie/enzymologie , Spectroscopie par résonance magnétique , Spectrométrie de masse , Souris , Souris de lignée C57BL , Souris nude , Phénylalanine/pharmacologie , Rats , Rat Sprague-Dawley , Rat Zucker , Relation structure-activité , Triglycéride/sang
8.
Mol Cancer Ther ; 10(12): 2298-308, 2011 Dec.
Article de Anglais | MEDLINE | ID: mdl-21926191

RÉSUMÉ

The signaling pathway of the receptor tyrosine kinase MET and its ligand hepatocyte growth factor (HGF) is important for cell growth, survival, and motility and is functionally linked to the signaling pathway of VEGF, which is widely recognized as a key effector in angiogenesis and cancer progression. Dysregulation of the MET/VEGF axis is found in a number of human malignancies and has been associated with tumorigenesis. Cabozantinib (XL184) is a small-molecule kinase inhibitor with potent activity toward MET and VEGF receptor 2 (VEGFR2), as well as a number of other receptor tyrosine kinases that have also been implicated in tumor pathobiology, including RET, KIT, AXL, and FLT3. Treatment with cabozantinib inhibited MET and VEGFR2 phosphorylation in vitro and in tumor models in vivo and led to significant reductions in cell invasion in vitro. In mouse models, cabozantinib dramatically altered tumor pathology, resulting in decreased tumor and endothelial cell proliferation coupled with increased apoptosis and dose-dependent inhibition of tumor growth in breast, lung, and glioma tumor models. Importantly, treatment with cabozantinib did not increase lung tumor burden in an experimental model of metastasis, which has been observed with inhibitors of VEGF signaling that do not target MET. Collectively, these data suggest that cabozantinib is a promising agent for inhibiting tumor angiogenesis and metastasis in cancers with dysregulated MET and VEGFR signaling.


Sujet(s)
Anilides/usage thérapeutique , Processus de croissance cellulaire/effets des médicaments et des substances chimiques , Métastase tumorale/prévention et contrôle , Tumeurs/traitement médicamenteux , Néovascularisation pathologique/prévention et contrôle , Pyridines/usage thérapeutique , Inhibiteurs de l'angiogenèse/pharmacologie , Inhibiteurs de l'angiogenèse/usage thérapeutique , Anilides/pharmacologie , Animaux , Lignée cellulaire tumorale , Évaluation préclinique de médicament , Femelle , Humains , Souris , Souris nude , Modèles biologiques , Tumeurs/anatomopathologie , Néovascularisation pathologique/traitement médicamenteux , Inhibiteurs de protéines kinases/pharmacologie , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Pyridines/pharmacologie , Récepteur-2 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs
9.
Clin Cancer Res ; 16(13): 3507-16, 2010 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-20472683

RÉSUMÉ

PURPOSE: Foretinib is an oral multikinase inhibitor targeting Met, RON, Axl, and vascular endothelial growth factor receptor. We conducted a phase I, first-time-in-human, clinical trial using escalating doses of oral foretinib. The primary objectives are to identify a maximum tolerated dose and determine the safety profile of foretinib. Secondary objectives included evaluation of plasma pharmacokinetics, long-term safety after repeated administration, preliminary antitumor activity, and pharmacodynamic activity. EXPERIMENTAL DESIGN: Patients had histologically confirmed metastatic or unresectable solid tumors for which no standard measures exist. All patients received foretinib orally for 5 consecutive days every 14 days. Dose escalation followed a conventional "3+3" design. RESULTS: Forty patients were treated in eight dose cohorts. The maximum tolerated dose was defined as 3.6 mg/kg, with a maximum administered dose of 4.5 mg/kg. Dose-limiting toxicities included grade 3 elevations in aspartate aminotransferase and lipase. Additional non-dose-limiting adverse events included hypertension, fatigue, diarrhea, vomiting, proteinuria, and hematuria. Responses were observed in two patients with papillary renal cell cancer and one patient with medullary thyroid cancer. Stable disease was identified in 22 patients. Foretinib pharmacokinetics increased linearly with dose. Pharmacodynamic evaluation indicated inhibition of MET phosphorylation and decreased proliferation in select tumor biopsies at submaximal doses. CONCLUSIONS: The recommended dose of foretinib was determined to be 240 mg, given on the first 5 days of a 14-day cycle. This dose and schedule were identified as having acceptable safety and pharmacokinetics, and will be the dose used in subsequent phase II trials.


Sujet(s)
Anilides/usage thérapeutique , Antinéoplasiques/usage thérapeutique , Tumeurs/traitement médicamenteux , Inhibiteurs de protéines kinases/usage thérapeutique , Protéines proto-oncogènes c-met/antagonistes et inhibiteurs , Quinoléines/usage thérapeutique , Récepteur-2 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs , Administration par voie orale , Adulte , Sujet âgé , Antinéoplasiques/effets indésirables , Calendrier d'administration des médicaments , Femelle , Humains , Mâle , Dose maximale tolérée , Adulte d'âge moyen , Métastase tumorale , Tumeurs/anatomopathologie , Inhibiteurs de protéines kinases/effets indésirables
10.
Clin Cancer Res ; 13(12): 3713-23, 2007 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-17575237

RÉSUMÉ

PURPOSE: Agents inhibiting the epidermal growth factor receptor (EGFR) have shown clinical benefit in a subset of non-small cell lung cancer patients expressing amplified or mutationally activated EGFR. However, responsive patients can relapse as a result of selection for EGFR gene mutations that confer resistance to ATP competitive EGFR inhibitors, such as erlotinib and gefitinib. We describe here the activity of EXEL-7647 (XL647), a novel spectrum-selective kinase inhibitor with potent activity against the EGF and vascular endothelial growth factor receptor tyrosine kinase families, against both wild-type (WT) and mutant EGFR in vitro and in vivo. EXPERIMENTAL DESIGN: The activity of EGFR inhibitors against WT and mutant EGFRs and their effect on downstream signal transduction was examined in cellular assays and in vivo using A431 and MDA-MB-231 (WT EGFR) and H1975 (L858R and T790M mutant EGFR) xenograft tumors. RESULTS: EXEL-7647 shows potent and long-lived inhibition of the WT EGFR in vivo. In addition, EXEL-7647 inhibits cellular proliferation and EGFR pathway activation in the erlotinib-resistant H1975 cell line that harbors a double mutation (L858R and T790M) in the EGFR gene. In vivo efficacy studies show that EXEL-7647 substantially inhibited the growth of H1975 xenograft tumors and reduced both tumor EGFR signaling and tumor vessel density. Additionally, EXEL-7647, in contrast to erlotinib, substantially inhibited the growth and vascularization of MDA-MB-231 xenografts, a model which is more reliant on signaling through vascular endothelial growth factor receptors. CONCLUSIONS: These studies provide a preclinical basis for clinical trials of XL647 in solid tumors and in patients bearing tumors that are resistant to existing EGFR-targeted therapies.


Sujet(s)
Antinéoplasiques/pharmacologie , Composés azabicycliques/pharmacologie , Récepteurs ErbB/effets des médicaments et des substances chimiques , Récepteurs ErbB/génétique , Inhibiteurs de protéines kinases/pharmacologie , Quinazolines/pharmacologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Relation dose-effet des médicaments , Résistance aux médicaments antinéoplasiques/génétique , Chlorhydrate d'erlotinib , Femelle , Géfitinib , Humains , Immunohistochimie , Méthode TUNEL , Souris , Souris nude , Souris SCID , Mutation , Phosphorylation/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
11.
Curr Biol ; 16(12): 1244-8, 2006 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-16782017

RÉSUMÉ

Lymphangiogenesis results in the formation of a vascular network distinct from arteries and veins that serves to drain interstitial fluid from surrounding tissues and plays a pivotal role in the immune defense of vertebrates as well as in the progression of cancer and other diseases . In mammals, lymph vessels are lined by endothelial cells possibly sprouting from embryonic veins, and their development appears to be critically dependent on the function of PROX1 and VEGFC signaling . The existence of a lymphatic system in teleosts has been a matter of debate for decades. Here we show on the morphological, molecular, and functional levels that zebrafish embryos develop a lymphatic vasculature that serves to retrieve components of the interstitium to the lymph system. We demonstrate the existence of vessels that are molecularly and functionally distinct from blood vessels and show that the development of these vessels depends on Vegfc and VEGFR-3/Flt4 signaling. These findings imply that the molecular components controlling lymphangiogenesis in zebrafish and mammals are conserved and that the zebrafish lymphatic system develops early enough to allow in vivo observations, lineage tracing, and genetic as well as pharmacological screens.


Sujet(s)
Vaisseaux lymphatiques/embryologie , Transduction du signal , Facteur de croissance endothéliale vasculaire de type C/métabolisme , Protéines de poisson-zèbre/métabolisme , Danio zébré/embryologie , Angiopoïétine-2/métabolisme , Animaux , Animal génétiquement modifié/anatomie et histologie , Animal génétiquement modifié/métabolisme , Transport biologique , Embryon non mammalien/cytologie , Embryon non mammalien/métabolisme , Endothélium lymphatique/cytologie , Endothélium lymphatique/métabolisme , Liquide extracellulaire/métabolisme , Protéines à fluorescence verte/analyse , Vaisseaux lymphatiques/métabolisme , Récepteur-3 au facteur croissance endothéliale vasculaire/métabolisme , Danio zébré/génétique , Danio zébré/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...