Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 15 de 15
Filtrer
1.
Arthritis Rheumatol ; 70(11): 1778-1789, 2018 11.
Article de Anglais | MEDLINE | ID: mdl-29855175

RÉSUMÉ

OBJECTIVE: To investigate the safety and efficacy of ABT-122, a tumor necrosis factor (TNF)- and interleukin-17A (IL-17A)-targeted dual variable domain immunoglobulin, in patients with active psoriatic arthritis (PsA) who have experienced an inadequate response to methotrexate. METHODS: Patients (n = 240) were randomized to receive ABT-122 (120 or 240 mg every week), adalimumab (40 mg every other week), or placebo in a 12-week double-blind, parallel-group study. The primary efficacy end point was the proportion of patients achieving ≥20% improvement in disease activity according to the American College of Rheumatology response criteria (ACR20) at week 12. Secondary and exploratory 12-week end points included 50% improvement (ACR50) and 70% improvement (ACR70) response rates, and proportion of patients meeting the Psoriasis Area and Severity Index (PASI) response criteria for ≥75% (PASI75) and ≥90% (PASI90) improvement in skin scores among those with ≥3% of their body surface area affected by psoriasis. RESULTS: In both ABT-122 dose groups, ACR20 response rates at week 12 (64.8-75.3%) were superior to that in patients receiving placebo (25.0%) (P < 0.001) but similar to that in patients receiving adalimumab (68.1%). ACR50 and ACR70 response rates were also superior in both ABT-122 dose groups (36.6-53.4% and 22.5-31.5%, respectively) compared to the placebo group (12.5% and 4.2%, respectively) (P < 0.05). Among eligible patients in the placebo, adalimumab, ABT-122 120 mg every week, and ABT-122 240 mg every week treatment groups, PASI75 responses were achieved in 27.3%, 57.6%, 74.4%, and 77.6% of patients, respectively, whereas PASI90 responses were achieved in 18.2%, 45.5%, 48.8%, and 46.9% of patients, respectively. Frequencies of treatment-emergent adverse events, including infections, were similar across all treatment groups, causing no discontinuations. No serious infections or systemic hypersensitivity reactions were reported with ABT-122. CONCLUSION: Dual neutralization of TNF and IL-17A with ABT-122 had efficacy and safety that was similar to, and not broadly differentiated from, that of adalimumab over a 12-week treatment course in patients with PsA.


Sujet(s)
Antirhumatismaux/usage thérapeutique , Arthrite psoriasique/traitement médicamenteux , Immunoglobulines/usage thérapeutique , Interleukine-17/antagonistes et inhibiteurs , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs , Adulte , Méthode en double aveugle , Femelle , Humains , Mâle , Méthotrexate/usage thérapeutique , Adulte d'âge moyen , Résultat thérapeutique
2.
BMC Rheumatol ; 2: 23, 2018.
Article de Anglais | MEDLINE | ID: mdl-30886973

RÉSUMÉ

BACKGROUND: Anti-cytokine therapies such as adalimumab, tocilizumab, and the small molecule JAK inhibitor tofacitinib have proven that cytokines and their subsequent downstream signaling processes are important in the pathogenesis of rheumatoid arthritis. Tofacitinib, a pan-JAK inhibitor, is the first approved JAK inhibitor for the treatment of RA and has been shown to be effective in managing disease. However, in phase 2 dose-ranging studies tofacitinib was associated with dose-limiting tolerability and safety issues such as anemia. Upadacitinib (ABT-494) is a selective JAK1 inhibitor that was engineered to address the hypothesis that greater JAK1 selectivity over other JAK family members will translate into a more favorable benefit:risk profile. Upadacitinib selectively targets JAK1 dependent disease drivers such as IL-6 and IFNγ, while reducing effects on reticulocytes and natural killer (NK) cells, which potentially contributed to the tolerability issues of tofacitinib. METHODS: Structure-based hypotheses were used to design the JAK1 selective inhibitor upadacitinib. JAK family selectivity was defined with in vitro assays including biochemical assessments, engineered cell lines, and cytokine stimulation. In vivo selectivity was defined by the efficacy of upadacitinib and tofacitinib in a rat adjuvant induced arthritis model, activity on reticulocyte deployment, and effect on circulating NK cells. The translation of the preclinical JAK1 selectivity was assessed in healthy volunteers using ex vivo stimulation with JAK-dependent cytokines. RESULTS: Here, we show the structural basis for the JAK1 selectivity of upadacitinib, along with the in vitro JAK family selectivity profile and subsequent in vivo physiological consequences. Upadacitinib is ~ 60 fold selective for JAK1 over JAK2, and > 100 fold selective over JAK3 in cellular assays. While both upadacitinib and tofacitinib demonstrated efficacy in a rat model of arthritis, the increased selectivity of upadacitinib for JAK1 resulted in a reduced effect on reticulocyte deployment and NK cell depletion relative to efficacy. Ex vivo pharmacodynamic data obtained from Phase I healthy volunteers confirmed the JAK1 selectivity of upadactinib in a clinical setting. CONCLUSIONS: The data presented here highlight the JAK1 selectivity of upadacinitinib and supports its use as an effective therapy for the treatment of RA with the potential for an improved benefit:risk profile.

3.
Toxicol Pathol ; 44(7): 998-1012, 2016 10.
Article de Anglais | MEDLINE | ID: mdl-27324990

RÉSUMÉ

Spleen tyrosine kinase (Syk) is a nonreceptor tyrosine kinase that is an important signaling enzyme downstream of immunoreceptors containing an intracellular immunoreceptor tyrosine activating motif (ITAM). These receptors encompass a wide variety of biological functions involved in autoimmune disease pathogenesis. There has been considerable interest in the development of inhibitors of the Syk pathway for the treatment of rheumatoid arthritis and systemic lupus erythematosus. We report that Syk inhibition mechanistically caused peri-islet hemorrhages and fibrin deposition in the rat pancreas and that this finding is due to a homeostatic functional defect in platelets. In more limited studies, similar lesions could not be induced in mice, dogs, and cynomolgus monkeys at similar or higher plasma drug concentrations. Irradiation-induced thrombocytopenia caused a phenotypically similar peri-islet pancreas lesion and the formation of this lesion could be prevented by platelet transfusion. In addition, Syk inhibitor-induced lesions were prevented by the coadministration of prednisone. A relatively greater sensitivity of rat platelets to Syk inhibition was supported by functional analyses demonstrating rat-specific differences in response to convulxin, a glycoprotein VI agonist that signals through Syk. These data demonstrate that the Syk pathway is critical in platelet-endothelial cell homeostasis in the peri-islet pancreatic microvasculature in rats.


Sujet(s)
Plaquettes/métabolisme , Antienzymes/toxicité , Hémorragie/induit chimiquement , Ilots pancréatiques/effets des médicaments et des substances chimiques , Syk kinase/antagonistes et inhibiteurs , Animaux , Plaquettes/effets des médicaments et des substances chimiques , Chiens , Ilots pancréatiques/anatomopathologie , Macaca fascicularis , Souris , Rats , Rat Sprague-Dawley , Spécificité d'espèce
4.
Bioorg Med Chem Lett ; 25(20): 4399-404, 2015 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-26372653

RÉSUMÉ

Previous work investigating tricyclic pyrrolopyrazines as kinase cores led to the discovery that 1-cyclohexyl-6H-pyrrolo[2,3-e][1,2,4]triazolo[4,3-a]pyrazine (12) had Jak inhibitory activity. Herein we describe our initial efforts to develop orally bioavailable analogs of 12 with improved selectivity of Jak1 over Jak2.


Sujet(s)
Janus kinase 1/antagonistes et inhibiteurs , Inhibiteurs de protéines kinases/pharmacologie , Pyrazines/pharmacologie , Triazoles/pharmacologie , Animaux , Lignée cellulaire , Relation dose-effet des médicaments , Humains , Janus kinase 1/métabolisme , Mâle , Modèles moléculaires , Structure moléculaire , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Pyrazines/synthèse chimique , Pyrazines/composition chimique , Rats , Rat Sprague-Dawley , Relation structure-activité , Triazoles/synthèse chimique , Triazoles/composition chimique
5.
Bioorg Med Chem Lett ; 23(3): 693-8, 2013 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-23265875

RÉSUMÉ

Interest in therapeutic kinase inhibitors continues to grow beyond success in oncology. To date, ATP-mimetic kinase inhibitors have focused primarily on monocyclic and bicyclic heterocyclic cores. We sought to expand on the repertoire of potential cores for kinase inhibition by exploring tricyclic variants of classical bicyclic hinge binding motifs such as pyrrolopyridine and pyrrolopyrazine. Herein we describe the syntheses of eight alternative tricyclic cores as well as in vitro screening results for representative kinases of potential therapeutic interest.


Sujet(s)
Conception de médicament , Inhibiteurs de protéines kinases , Cellules cultivées , Cyclisation , Activation enzymatique/effets des médicaments et des substances chimiques , Concentration inhibitrice 50 , Structure moléculaire , Inhibiteurs de protéines kinases/synthèse chimique , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Pyrazines/synthèse chimique , Pyrazines/composition chimique , Pyrazines/pharmacologie , Pyridines/synthèse chimique , Pyridines/composition chimique , Pyridines/pharmacologie , Pyrroles/synthèse chimique , Pyrroles/composition chimique , Pyrroles/pharmacologie
6.
BMC Struct Biol ; 12: 22, 2012 Sep 20.
Article de Anglais | MEDLINE | ID: mdl-22995073

RÉSUMÉ

BACKGROUND: Structure-based drug design (SBDD) can accelerate inhibitor lead design and optimization, and efficient methods including protein purification, characterization, crystallization, and high-resolution diffraction are all needed for rapid, iterative structure determination. Janus kinases are important targets that are amenable to structure-based drug design. Here we present the first mouse Tyk2 crystal structures, which are complexed to 3-aminoindazole compounds. RESULTS: A comprehensive construct design effort included N- and C-terminal variations, kinase-inactive mutations, and multiple species orthologs. High-throughput cloning and expression methods were coupled with an abbreviated purification protocol to optimize protein solubility and stability. In total, 50 Tyk2 constructs were generated. Many displayed poor expression, inadequate solubility, or incomplete affinity tag processing. One kinase-inactive murine Tyk2 construct, complexed with an ATP-competitive 3-aminoindazole inhibitor, provided crystals that diffracted to 2.5-2.6 Å resolution. This structure revealed initial "hot-spot" regions for SBDD, and provided a robust platform for ligand soaking experiments. Compared to previously reported human Tyk2 inhibitor crystal structures (Chrencik et al. (2010) J Mol Biol 400:413), our structures revealed a key difference in the glycine-rich loop conformation that is induced by the inhibitor. Ligand binding also conferred resistance to proteolytic degradation by thermolysin. As crystals could not be obtained with the unliganded enzyme, this enhanced stability is likely important for successful crystallization and inhibitor soaking methods. CONCLUSIONS: Practical criteria for construct performance and prioritization, the optimization of purification protocols to enhance protein yields and stability, and use of high-throughput construct exploration enable structure determination methods early in the drug discovery process. Additionally, specific ligands stabilize Tyk2 protein and may thereby enable crystallization.


Sujet(s)
Conception de médicament , Indazoles/composition chimique , Indazoles/pharmacologie , TYK2 Kinase/antagonistes et inhibiteurs , TYK2 Kinase/composition chimique , Séquence d'acides aminés , Animaux , Cristallisation , Cristallographie aux rayons X , Stabilité enzymatique/effets des médicaments et des substances chimiques , Humains , Janus kinases/antagonistes et inhibiteurs , Janus kinases/métabolisme , Souris , Données de séquences moléculaires , Inhibiteurs de protéines kinases/composition chimique , Inhibiteurs de protéines kinases/pharmacologie , Structure secondaire des protéines , Protéolyse/effets des médicaments et des substances chimiques , Relation structure-activité , TYK2 Kinase/isolement et purification
7.
J Biomol Screen ; 17(7): 857-67, 2012 Aug.
Article de Anglais | MEDLINE | ID: mdl-22584786

RÉSUMÉ

Development of inhibitor compounds selective against undesirable targets is critical in drug discovery. Selectivity ratios for candidate compounds are evaluated by dividing potencies from two assays assessing the off-target and target. Because all potency measurements have underlying uncertainty, understanding error propagation is essential to interpreting selectivity data. Assay noise introduces ambiguity in the statistical significance of selectivity ratios, particularly at low replicate numbers when compounds are often prioritized for subsequent testing. The ability to differentiate potency results for any pair of compounds in one assay is evaluated using a metric called minimum significant ratio (MSR). Potency results of one compound tested in a pair of assays can be differentiated by the minimum significant selectivity ratio (MSSR). To differentiate selectivity ratios for any pair of compounds, we extend this concept by proposing two new parameters called the minimum significant ratio of selectivity ratios (MSRSR) and confidence in ratio of selectivity ratios (CRSR). Importantly, these tools can be used after a single selectivity measurement. We describe these methods and illustrate their usefulness using structure-activity relationship data from a Janus kinase inhibitor project, in which these tools informed a cogent retesting strategy and enabled rapid and objective decision making.


Sujet(s)
Découverte de médicament , Évaluation préclinique de médicament , Antienzymes/pharmacologie , Janus kinases/antagonistes et inhibiteurs , Préparations pharmaceutiques/analyse , Phénomènes physiologiques cellulaires , Interprétation statistique de données , Antienzymes/composition chimique , Janus kinases/métabolisme , Relation structure-activité
8.
Bioorg Med Chem Lett ; 20(1): 334-7, 2010 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-19926477
9.
Bioorg Med Chem Lett ; 19(6): 1722-5, 2009 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-19217782

RÉSUMÉ

COT (Tpl2 in mice) is a serine/threonine MAP3 kinase that regulates production of TNF-alpha and other pro-inflammatory cytokines such as IL-1beta via the ERK/MAP kinase pathway. As TNF-alpha and IL-1beta are clinically validated targets for therapeutic intervention in rheumatoid arthritis (RA), blocking COT provides a potential avenue for amelioration of disease. Herein we describe identification of a cellular active selective small molecule inhibitor of COT kinase.


Sujet(s)
Antienzymes/synthèse chimique , Antienzymes/pharmacologie , MAP Kinase Kinase Kinases/antagonistes et inhibiteurs , Protéines proto-oncogènes/antagonistes et inhibiteurs , Pyridines/synthèse chimique , Facteur de nécrose tumorale alpha/antagonistes et inhibiteurs , Animaux , Polyarthrite rhumatoïde/traitement médicamenteux , Chimie pharmaceutique/méthodes , Conception de médicament , Extracellular Signal-Regulated MAP Kinases/métabolisme , Humains , Liaison hydrogène , Concentration inhibitrice 50 , Interleukine-1 bêta/métabolisme , Ligands , MAP Kinase Kinase Kinases/composition chimique , Souris , Structure moléculaire , Protéines proto-oncogènes/composition chimique , Pyridines/pharmacologie , Facteur de nécrose tumorale alpha/composition chimique , Facteur de nécrose tumorale alpha/métabolisme
10.
J Med Chem ; 51(20): 6571-80, 2008 Oct 23.
Article de Anglais | MEDLINE | ID: mdl-18811133

RÉSUMÉ

A series of 2-aminopyrimidines was synthesized as ligands of the histamine H4 receptor (H4R). Working in part from a pyrimidine hit that was identified in an HTS campaign, SAR studies were carried out to optimize the potency, which led to compound 3, 4- tert-butyl-6-(4-methylpiperazin-1-yl)pyrimidin-2-ylamine. We further studied this compound by systematically modifying the core pyrimidine moiety, the methylpiperazine at position 4, the NH2 at position 2, and positions 5 and 6 of the pyrimidine ring. The pyrimidine 6 position benefited the most from this optimization, especially in analogs in which the 6- tert-butyl was replaced with aromatic and secondary amine moieties. The highlight of the optimization campaign was compound 4, 4-[2-amino-6-(4-methylpiperazin-1-yl)pyrimidin-4-yl]benzonitrile, which was potent in vitro and was active as an anti-inflammatory agent in an animal model and had antinociceptive activity in a pain model, which supports the potential of H 4R antagonists in pain.


Sujet(s)
Antihistaminiques/synthèse chimique , Antihistaminiques/pharmacologie , Pyrimidines/synthèse chimique , Pyrimidines/pharmacologie , Récepteurs histaminergiques/métabolisme , Animaux , Marqueurs biologiques , Antihistaminiques/composition chimique , Humains , Hyperplasie/induit chimiquement , Hyperplasie/prévention et contrôle , Ligands , Locomotion/effets des médicaments et des substances chimiques , Souris , Structure moléculaire , Pyrimidines/composition chimique , Rats , Relation structure-activité , Spécificité du substrat
11.
J Med Chem ; 51(20): 6547-57, 2008 Oct 23.
Article de Anglais | MEDLINE | ID: mdl-18817367

RÉSUMÉ

A new structural class of histamine H 4 receptor antagonists (6-14) was designed based on rotationally restricted 2,4-diaminopyrimidines. Series compounds showed potent and selective in vitro H 4 antagonism across multiple species, good CNS penetration, improved PK properties compared to reference H 4 antagonists, functional H 4 antagonism in cellular and in vivo pharmacological assays, and in vivo anti-inflammatory and antinociceptive efficacy. One compound, 10 (A-943931), combined the best features of the series in a single molecule and is an excellent tool compound to probe H 4 pharmacology. It is a potent H 4 antagonist in functional assays across species (FLIPR Ca (2+) flux, K b < 5.7 nM), has high (>190x) selectivity for H 4, and combines good PK in rats and mice (t 1/2 of 2.6 and 1.6 h, oral bioavailability of 37% and 90%) with anti-inflammatory activity (ED 50 = 37 micromol/kg, mouse) and efficacy in pain models (thermal hyperalgesia, ED 50 = 72 micromol/kg, rat).


Sujet(s)
Amines/composition chimique , Anti-inflammatoires/synthèse chimique , Antihistaminiques/synthèse chimique , Antihistaminiques/usage thérapeutique , Douleur/traitement médicamenteux , Pyrimidines/synthèse chimique , Récepteurs histaminergiques/métabolisme , Animaux , Anti-inflammatoires/composition chimique , Anti-inflammatoires/classification , Anti-inflammatoires/usage thérapeutique , Modèles animaux de maladie humaine , Antihistaminiques/composition chimique , Antihistaminiques/classification , Ligands , Souris , Structure moléculaire , Pyrimidines/composition chimique , Pyrimidines/classification , Pyrimidines/usage thérapeutique , Rats
13.
Anal Biochem ; 350(2): 268-76, 2006 Mar 15.
Article de Anglais | MEDLINE | ID: mdl-16356459

RÉSUMÉ

Cancer osaka thyroid (COT) is a member of the mitogen-activated protein kinase kinase kinase family of enzymes and plays a pivotal role in tumor necrosis factor-alpha production in macrophages. Consequently, COT is considered to be a promising target for antiinflammatory drug discovery. We describe here the development of in vitro COT assays in several formats and the advantages and disadvantages of each. A cascade assay requires very small amounts of enzyme and can provide a useful tool for high-throughput screening, but it is not desirable for compound mechanistic studies due to complicated kinetics. Direct assays are superior to cascade assays and are suitable for both compound screening and mechanistic studies. Among the direct assays, the homogeneous time-resolved fluorescence (HTRF) format is preferred over the radiometric format due to the robustness, throughput, and ease of use of the HTRF format. When the physiological protein substrate MEK1 (MAP/Erk kinase 1) was used to determine inhibitor potencies, false positives were observed due to compound interference by binding to MEK1. Using a MEK1 peptide substrate, these false positives were eliminated. In addition, we describe a simple method to study the ATP competitiveness of compounds. The knowledge gained through our studies with COT, and the methods described for our assays and compound mechanistic studies, can be readily applied to other kinase targets.


Sujet(s)
MAP Kinase Kinase Kinases/antagonistes et inhibiteurs , MAP Kinase Kinase Kinases/analyse , Protéines proto-oncogènes/antagonistes et inhibiteurs , Protéines proto-oncogènes/analyse , Adénosine triphosphate/métabolisme , Fluorescence , Humains , Cinétique , MAP Kinase Kinase 1/métabolisme , Système de signalisation des MAP kinases , Radio-isotopes du phosphore
14.
J Med Chem ; 48(19): 6066-83, 2005 Sep 22.
Article de Anglais | MEDLINE | ID: mdl-16162008

RÉSUMÉ

A series of novel thienopyrimidine-based receptor tyrosine kinase inhibitors has been discovered. Investigation of structure-activity relationships at the 5- and 6-positions of the thienopyrimidine nucleus led to a series of N,N'-diaryl ureas that potently inhibit all of the vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) receptor tyrosine kinases. A kinase insert domain-containing receptor (KDR) homology model suggests that these compounds bind to the "inactive conformation" of the enzyme with the urea portion extending into the back hydrophobic pocket adjacent to the adenosine 5'-triphosphate (ATP) binding site. A number of compounds have been identified as displaying excellent in vivo potency. In particular, compounds 28 and 76 possess favorable pharmacokinetic (PK) profiles and demonstrate potent antitumor efficacy against the HT1080 human fibrosarcoma xenograft tumor growth model (tumor growth inhibition (TGI) = 75% at 25 mg/kg.day, per os (po)).


Sujet(s)
Antinéoplasiques/synthèse chimique , Pyrimidines/synthèse chimique , Récepteurs aux facteurs de croissance dérivés des plaquettes/antagonistes et inhibiteurs , Urée/analogues et dérivés , Urée/synthèse chimique , Récepteur-2 au facteur croissance endothéliale vasculaire/antagonistes et inhibiteurs , Adénosine triphosphate/composition chimique , Animaux , Antinéoplasiques/composition chimique , Antinéoplasiques/pharmacologie , Oedème/induit chimiquement , Oedème/anatomopathologie , Oestradiol , Femelle , Humains , Souris , Souris de lignée BALB C , Souris SCID , Modèles moléculaires , Cellules NIH 3T3 , Phosphorylation , Pyrimidines/composition chimique , Pyrimidines/pharmacologie , Relation structure-activité , Urée/composition chimique , Urée/pharmacologie , Utérus/effets des médicaments et des substances chimiques , Utérus/anatomopathologie , Récepteur-2 au facteur croissance endothéliale vasculaire/composition chimique , Récepteur-2 au facteur croissance endothéliale vasculaire/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
15.
Arch Biochem Biophys ; 441(1): 64-74, 2005 Sep 01.
Article de Anglais | MEDLINE | ID: mdl-16087150

RÉSUMÉ

Cancer osaka thyroid (COT), a human MAP 3 K, is essential for lipopolysaccharide activation of the Erk MAPK cascade in macrophages. COT 30--467 is insoluble, whereas low levels of COT 30--397 can be expressed, but this protein is unstable. However, both COT 30--467 and COT 30--397 are expressed in a soluble and stable form when produced in complex with the C-terminal half of p105. The k(cat) of COT 30--397 is reduced approximately 47--fold in the COT 30--467/p105 Delta N complex. COT prefers Mn(2+) to Mg(2+) as the ATP metal cofactor, exhibiting an unusually high ATP K(m) in the presence of Mg(2+). When using Mn(2+) as the cofactor, the ATP K(m) is reduced to a level typical of most kinases. In contrast, the binding affinity of COT for its other substrate MEK is cofactor independent. Our results using purified proteins indicate that p105 binding improves COT solubility and stability while down-regulating kinase activity, consistent with cellular data showing that p105 functions as an inhibitor of COT.


Sujet(s)
MAP Kinase Kinase Kinases/composition chimique , MAP Kinase Kinase Kinases/isolement et purification , Facteur de transcription NF-kappa B/composition chimique , Précurseurs de protéines/composition chimique , Protéines proto-oncogènes/composition chimique , Protéines proto-oncogènes/isolement et purification , Sites de fixation , Activation enzymatique , Stabilité enzymatique , Humains , Cellules Jurkat , Cinétique , MAP Kinase Kinase Kinases/génétique , Sous-unité p50 de NF-kappa B , Liaison aux protéines , Ingénierie des protéines/méthodes , Protéines proto-oncogènes/génétique , Protéines recombinantes/composition chimique , Protéines recombinantes/isolement et purification , Relation structure-activité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE