Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 23
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
In Vivo ; 36(4): 1615-1627, 2022.
Article de Anglais | MEDLINE | ID: mdl-35738590

RÉSUMÉ

BACKGROUND/AIM: The therapeutic potential of bromodomain and extra-terminal motif (BET) inhibitors in hematological cancers has been well established in preclinical and early-stage clinical trials, although as of yet, no BETtargeting agent has achieved approval. To add insight into potential response to mivebresib (ABBV-075), a broadspectrum BET inhibitor, co-clinical modeling of individual patient biopsies was conducted in the context of a Phase I trial in acute myeloid leukemia (AML). MATERIALS AND METHODS: Co-clinical modeling involves taking the patient's biopsy and implanting it in mice with limited passage so that it closely retains the original characteristics of the malignancy and allows comparisons of response between animal model and clinical data. Procedures were developed, initially with neonate NOD/Shi-scid-IL2rγnull (NOG) mice and then optimized with juvenile NOG-EXL as host mice, eventually resulting in a robust rate of engraftment (16 out of 26, 62%). RESULTS: Results from the co-clinical AML patient-derived xenograft (PDX) modeling (6 with >60% inhibition of bone marrow blasts) were consistent with the equivalent clinical data from patients receiving mivebresib in monotherapy, and in combination with venetoclax. The modeling system also demonstrated the activity of a novel BD2-selective BET inhibitor (ABBV-744) in the preclinical AML setting. Both agents were also highly effective in inhibiting blast counts in the spleen (10/10 and 5/6 models, respectively). CONCLUSION: These findings confirm the validity of the model system in the co-clinical setting, establish highly relevant in vivo models for the discovery of cancer therapy, and indicate the therapeutic value of BET inhibitors for AML and, potentially, myelofibrosis treatment.


Sujet(s)
Leucémie aigüe myéloïde , Pyridones , Animaux , Lignée cellulaire tumorale , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/anatomopathologie , Souris , Souris de lignée NOD , Souris SCID , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Sulfonamides
2.
Mol Cancer Ther ; 20(10): 1809-1819, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34253595

RÉSUMÉ

Dual bromodomain BET inhibitors that bind with similar affinities to the first and second bromodomains across BRD2, BRD3, BRD4, and BRDT have displayed modest activity as monotherapy in clinical trials. Thrombocytopenia, closely followed by symptoms characteristic of gastrointestinal toxicity, have presented as dose-limiting adverse events that may have prevented escalation to higher dose levels required for more robust efficacy. ABBV-744 is a highly selective inhibitor for the second bromodomain of the four BET family proteins. In contrast to the broad antiproliferative activities observed with dual bromodomain BET inhibitors, ABBV-744 displayed significant antiproliferative activities largely although not exclusively in cancer cell lines derived from acute myeloid leukemia and androgen receptor positive prostate cancer. Studies in acute myeloid leukemia xenograft models demonstrated antitumor efficacy for ABBV-744 that was comparable with the pan-BET inhibitor ABBV-075 but with an improved therapeutic index. Enhanced antitumor efficacy was also observed with the combination of ABBV-744 and the BCL-2 inhibitor, venetoclax compared with monotherapies of either agent alone. These results collectively support the clinical evaluation of ABBV-744 in AML (Clinical Trials.gov identifier: NCT03360006).


Sujet(s)
Composés hétérocycliques bicycliques/pharmacologie , Leucémie aigüe myéloïde/traitement médicamenteux , Protéines/antagonistes et inhibiteurs , Protéines proto-oncogènes c-bcl-2/antagonistes et inhibiteurs , Pyridines/pharmacologie , Pyrroles/pharmacologie , Sulfonamides/pharmacologie , Animaux , Antinéoplasiques/pharmacologie , Apoptose , Prolifération cellulaire , Association de médicaments , Femelle , Humains , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Souris , Souris de lignée NOD , Souris SCID , Cellules cancéreuses en culture , Tests d'activité antitumorale sur modèle de xénogreffe
3.
J Chem Inf Model ; 61(3): 1412-1426, 2021 03 22.
Article de Anglais | MEDLINE | ID: mdl-33661005

RÉSUMÉ

Drug design with patient centricity for ease of administration and pill burden requires robust understanding of the impact of chemical modifications on relevant physicochemical properties early in lead optimization. To this end, we have developed a physics-based ensemble approach to predict aqueous thermodynamic crystalline solubility, with a 2D chemical structure as the input. Predictions for the bromodomain and extraterminal domain (BET) inhibitor series show very close match (0.5 log unit) with measured thermodynamic solubility for cases with low crystal anisotropy and good match (1 log unit) for high anisotropy structures. The importance of thermodynamic solubility is clearly demonstrated by up to a 4 log unit drop in solubility compared to kinetic (amorphous) solubility in some cases and implications thereof, for instance on human dose. We have also demonstrated that incorporating predicted crystal structures in thermodynamic solubility prediction is necessary to differentiate (up to 4 log unit) between solubility of molecules within the series. Finally, our physics-based ensemble approach provides valuable structural insights into the origins of 3-D conformational landscapes, crystal polymorphism, and anisotropy that can be leveraged for both drug design and development.


Sujet(s)
Physique , Eau , Humains , Conformation moléculaire , Solubilité , Thermodynamique
4.
J Med Chem ; 63(10): 5585-5623, 2020 05 28.
Article de Anglais | MEDLINE | ID: mdl-32324999

RÉSUMÉ

The BET family of proteins consists of BRD2, BRD3, BRD4, and BRDt. Each protein contains two distinct bromodomains (BD1 and BD2). BET family bromodomain inhibitors under clinical development for oncology bind to each of the eight bromodomains with similar affinities. We hypothesized that it may be possible to achieve an improved therapeutic index by selectively targeting subsets of the BET bromodomains. Both BD1 and BD2 are highly conserved across family members (>70% identity), whereas BD1 and BD2 from the same protein exhibit a larger degree of divergence (∼40% identity), suggesting selectivity between BD1 and BD2 of all family members would be more straightforward to achieve. Exploiting the Asp144/His437 and Ile146/Val439 sequence differences (BRD4 BD1/BD2 numbering) allowed the identification of compound 27 demonstrating greater than 100-fold selectivity for BRD4 BD2 over BRD4 BD1. Further optimization to improve BD2 selectivity and oral bioavailability resulted in the clinical development compound 46 (ABBV-744).


Sujet(s)
Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/métabolisme , Découverte de médicament/méthodes , Pyridines/composition chimique , Pyridines/métabolisme , Pyrroles/composition chimique , Pyrroles/métabolisme , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/métabolisme , Animaux , Femelle , Cellules HeLa , Humains , Souris , Souris SCID , Domaines protéiques/effets des médicaments et des substances chimiques , Domaines protéiques/physiologie , Structure secondaire des protéines , Structure tertiaire des protéines , Pyridines/pharmacologie , Pyrroles/pharmacologie , Tests d'activité antitumorale sur modèle de xénogreffe/méthodes
5.
Nature ; 578(7794): 306-310, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31969702

RÉSUMÉ

Proteins of the bromodomain and extra-terminal (BET) domain family are epigenetic readers that bind acetylated histones through their bromodomains to regulate gene transcription. Dual-bromodomain BET inhibitors (DbBi) that bind with similar affinities to the first (BD1) and second (BD2) bromodomains of BRD2, BRD3, BRD4 and BRDt have displayed modest clinical activity in monotherapy cancer trials. A reduced number of thrombocytes in the blood (thrombocytopenia) as well as symptoms of gastrointestinal toxicity are dose-limiting adverse events for some types of DbBi1-5. Given that similar haematological and gastrointestinal defects were observed after genetic silencing of Brd4 in mice6, the platelet and gastrointestinal toxicities may represent on-target activities associated with BET inhibition. The two individual bromodomains in BET family proteins may have distinct functions7-9 and different cellular phenotypes after pharmacological inhibition of one or both bromodomains have been reported10,11, suggesting that selectively targeting one of the bromodomains may result in a different efficacy and tolerability profile compared with DbBi. Available compounds that are selective to individual domains lack sufficient potency and the pharmacokinetics properties that are required for in vivo efficacy and tolerability assessment10-13. Here we carried out a medicinal chemistry campaign that led to the discovery of ABBV-744, a highly potent and selective inhibitor of the BD2 domain of BET family proteins with drug-like properties. In contrast to the broad range of cell growth inhibition induced by DbBi, the antiproliferative activity of ABBV-744 was largely, but not exclusively, restricted to cell lines of acute myeloid leukaemia and prostate cancer that expressed the full-length androgen receptor (AR). ABBV-744 retained robust activity in prostate cancer xenografts, and showed fewer platelet and gastrointestinal toxicities than the DbBi ABBV-07514. Analyses of RNA expression and chromatin immunoprecipitation followed by sequencing revealed that ABBV-744 displaced BRD4 from AR-containing super-enhancers and inhibited AR-dependent transcription, with less impact on global transcription compared with ABBV-075. These results underscore the potential value of selectively targeting the BD2 domain of BET family proteins for cancer therapy.


Sujet(s)
Protéines du cycle cellulaire/antagonistes et inhibiteurs , Protéines du cycle cellulaire/composition chimique , Tumeurs de la prostate/traitement médicamenteux , Tumeurs de la prostate/métabolisme , Domaines protéiques/effets des médicaments et des substances chimiques , Pyridines/pharmacologie , Pyrroles/pharmacologie , Facteurs de transcription/antagonistes et inhibiteurs , Facteurs de transcription/composition chimique , Animaux , Protéines du cycle cellulaire/génétique , Protéines du cycle cellulaire/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Éléments activateurs (génétique)/génétique , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Mâle , Souris , Pyridines/effets indésirables , Pyridines/toxicité , Pyrroles/effets indésirables , Pyrroles/toxicité , Rats , Récepteurs aux androgènes/métabolisme , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme , Transcription génétique/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe
6.
J Med Chem ; 61(12): 5138-5153, 2018 06 28.
Article de Anglais | MEDLINE | ID: mdl-29852069

RÉSUMÉ

The emergence of drug-resistant HIV from a widespread antiviral chemotherapy targeting HIV protease in the past decades is unavoidable and provides a challenge to develop alternative inhibitors. We synthesized a series of allophenylnorstatine-based peptidomimetics with various P3, P2, and P2́ moieties. The derivatives with P2 tetrahydrofuranylglycine (Thfg) were found to be potent against wild type HIV-1 protease and the virus, leading to a highly potent compound 21f (KNI-1657) against lopinavir/ritonavir- or darunavir-resistant strains. Co-crystal structures of 21f and the wild-type protease revealed numerous key hydrogen bonding interactions with Thfg. These results suggest that the strategy to design allophenylnorstatine-based peptidomimetics combined with Thfg residue would be promising for generating candidates to overcome multidrug resistance.


Sujet(s)
Résistance virale aux médicaments/effets des médicaments et des substances chimiques , Inhibiteurs de protéase du VIH/composition chimique , Inhibiteurs de protéase du VIH/pharmacologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Peptidomimétiques/pharmacologie , Cristallographie aux rayons X , Darunavir/pharmacologie , Glycine/composition chimique , Protéase du VIH/composition chimique , Protéase du VIH/métabolisme , Humains , Lopinavir/pharmacologie , Peptidomimétiques/composition chimique , Phénylbutyrates/composition chimique , Sérum/métabolisme , Relation structure-activité
7.
Bioorg Med Chem Lett ; 28(10): 1804-1810, 2018 06 01.
Article de Anglais | MEDLINE | ID: mdl-29678460

RÉSUMÉ

Novel conformationally constrained BET bromodomain inhibitors have been developed. These inhibitors were optimized in two similar, yet distinct chemical series, the 6-methyl-1H-pyrrolo[2,3-c]pyridin-7(6H)-ones (A) and the 1-methyl-1H-pyrrolo[2,3-c]pyridin-7(6H)-ones (B). Each series demonstrated excellent activity in binding and cellular assays, and lead compounds from each series demonstrated significant efficacy in in vivo tumor xenograft models.


Sujet(s)
Protéines nucléaires/antagonistes et inhibiteurs , Pyridones/composition chimique , Facteurs de transcription/antagonistes et inhibiteurs , Animaux , Sites de fixation , Protéines du cycle cellulaire , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cristallographie aux rayons X , Évaluation préclinique de médicament , Période , Humains , Souris , Microsomes/métabolisme , Simulation de dynamique moléculaire , Myélome multiple/traitement médicamenteux , Protéines nucléaires/métabolisme , Structure tertiaire des protéines , Pyridones/pharmacocinétique , Pyridones/pharmacologie , Pyridones/usage thérapeutique , Relation structure-activité , Facteurs de transcription/métabolisme , Transplantation hétérologue
8.
Article de Anglais | MEDLINE | ID: mdl-29084747

RÉSUMÉ

Glecaprevir (formerly ABT-493) is a novel hepatitis C virus (HCV) NS3/4A protease inhibitor (PI) with pangenotypic activity. It inhibited the enzymatic activity of purified NS3/4A proteases from HCV genotypes 1 to 6 in vitro (half-maximal [50%] inhibitory concentration = 3.5 to 11.3 nM) and the replication of stable HCV subgenomic replicons containing proteases from genotypes 1 to 6 (50% effective concentration [EC50] = 0.21 to 4.6 nM). Glecaprevir had a median EC50 of 0.30 nM (range, 0.05 to 3.8 nM) for HCV replicons containing proteases from 40 samples from patients infected with HCV genotypes 1 to 5. Importantly, glecaprevir was active against the protease from genotype 3, the most-difficult-to-treat HCV genotype, in both enzymatic and replicon assays demonstrating comparable activity against the other HCV genotypes. In drug-resistant colony selection studies, glecaprevir generally selected substitutions at NS3 amino acid position A156 in replicons containing proteases from genotypes 1a, 1b, 2a, 2b, 3a, and 4a and substitutions at position D/Q168 in replicons containing proteases from genotypes 3a, 5a, and 6a. Although the substitutions A156T and A156V in NS3 of genotype 1 reduced susceptibility to glecaprevir, replicons with these substitutions demonstrated a low replication efficiency in vitro Glecaprevir is active against HCV with most of the common NS3 amino acid substitutions that are associated with reduced susceptibility to other currently approved HCV PIs, including those at positions 155 and 168. Combination of glecaprevir with HCV inhibitors with other mechanisms of action resulted in additive or synergistic antiviral activity. In summary, glecaprevir is a next-generation HCV PI with potent pangenotypic activity and a high barrier to the development of resistance.


Sujet(s)
Antiviraux/pharmacologie , Résistance virale aux médicaments/effets des médicaments et des substances chimiques , Hepacivirus/effets des médicaments et des substances chimiques , Inhibiteurs de protéases/pharmacologie , Quinoxalines/pharmacologie , Sulfonamides/pharmacologie , Protéines virales non structurales/antagonistes et inhibiteurs , Substitution d'acide aminé , Acides amino-isobutyriques , Agents antiVIH/pharmacologie , Cyclopropanes , Synergie des médicaments , Génotype , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Hepacivirus/génétique , Humains , Lactames macrocycliques , Leucine/analogues et dérivés , Proline/analogues et dérivés , Réplicon/effets des médicaments et des substances chimiques , Protéines virales non structurales/génétique , Réplication virale/effets des médicaments et des substances chimiques
9.
J Med Chem ; 60(20): 8369-8384, 2017 10 26.
Article de Anglais | MEDLINE | ID: mdl-28949521

RÉSUMÉ

The development of bromodomain and extraterminal domain (BET) bromodomain inhibitors and their examination in clinical studies, particularly in oncology settings, has garnered substantial recent interest. An effort to generate novel BET bromodomain inhibitors with excellent potency and drug metabolism and pharmacokinetics (DMPK) properties was initiated based upon elaboration of a simple pyridone core. Efforts to develop a bidentate interaction with a critical asparagine residue resulted in the incorporation of a pyrrolopyridone core, which improved potency by 9-19-fold. Additional structure-activity relationship (SAR) efforts aimed both at increasing potency and improving pharmacokinetic properties led to the discovery of the clinical candidate 63 (ABBV-075/mivebresib), which demonstrates excellent potency in biochemical and cellular assays, advantageous exposures and half-life both in animal models and in humans, and in vivo efficacy in mouse models of cancer progression and inflammation.


Sujet(s)
Découverte de médicament , Protéines/antagonistes et inhibiteurs , Pyridones/pharmacologie , Sulfonamides/pharmacologie , Animaux , Lignée cellulaire tumorale , Chromatographie en phase liquide à haute performance , Transfert d'énergie par résonance de fluorescence , Période , Humains , Spectrométrie de masse , Souris , Spectroscopie par résonance magnétique du proton , Pyridones/composition chimique , Pyridones/pharmacocinétique , Relation structure-activité , Sulfonamides/composition chimique , Sulfonamides/pharmacocinétique
10.
J Med Chem ; 60(9): 3828-3850, 2017 05 11.
Article de Anglais | MEDLINE | ID: mdl-28368119

RÉSUMÉ

Members of the BET family of bromodomain containing proteins have been identified as potential targets for blocking proliferation in a variety of cancer cell lines. A two-dimensional NMR fragment screen for binders to the bromodomains of BRD4 identified a phenylpyridazinone fragment with a weak binding affinity (1, Ki = 160 µM). SAR investigation of fragment 1, aided by X-ray structure-based design, enabled the synthesis of potent pyridone and macrocyclic pyridone inhibitors exhibiting single digit nanomolar potency in both biochemical and cell based assays. Advanced analogs in these series exhibited high oral exposures in rodent PK studies and demonstrated significant tumor growth inhibition efficacy in mouse flank xenograft models.


Sujet(s)
Composés macrocycliques/composition chimique , Composés macrocycliques/pharmacologie , Pyridones/composition chimique , Pyridones/pharmacologie , Animaux , Cristallographie aux rayons X , Découverte de médicament , Composés macrocycliques/pharmacocinétique , Structure moléculaire , Pyridones/pharmacocinétique , Rats , Relation structure-activité
11.
Cancer Res ; 77(11): 2976-2989, 2017 06 01.
Article de Anglais | MEDLINE | ID: mdl-28416490

RÉSUMÉ

ABBV-075 is a potent and selective BET family bromodomain inhibitor that recently entered phase I clinical trials. Comprehensive preclinical characterization of ABBV-075 demonstrated broad activity across cell lines and tumor models, representing a variety of hematologic malignancies and solid tumor indications. In most cancer cell lines derived from solid tumors, ABBV-075 triggers prominent G1 cell-cycle arrest without extensive apoptosis. In this study, we show that ABBV-075 efficiently triggers apoptosis in acute myeloid leukemia (AML), non-Hodgkin lymphoma, and multiple myeloma cells. Apoptosis induced by ABBV-075 was mediated in part by modulation of the intrinsic apoptotic pathway, exhibiting synergy with the BCL-2 inhibitor venetoclax in preclinical models of AML. In germinal center diffuse large B-cell lymphoma, BCL-2 levels or venetoclax sensitivity predicted the apoptotic response to ABBV-075 treatment. In vivo combination studies uncovered surprising benefits of low doses of ABBV-075 coupled with bortezomib and azacitidine treatment, despite the lack of in vitro synergy between ABBV-075 and these agents. The in vitro/in vivo activities of ABBV-075 described here may serve as a useful reference to guide the development of ABBV-075 and other BET family inhibitors for cancer therapy. Cancer Res; 77(11); 2976-89. ©2017 AACR.


Sujet(s)
Antagonistes des androgènes/usage thérapeutique , Pyridones/usage thérapeutique , Sulfonamides/usage thérapeutique , Antagonistes des androgènes/pharmacologie , Apoptose , Lignée cellulaire tumorale , Synergie des médicaments , Humains , Pyridones/pharmacologie , Sulfonamides/pharmacologie , Transfection
12.
Bioorg Med Chem Lett ; 27(10): 2225-2233, 2017 05 15.
Article de Anglais | MEDLINE | ID: mdl-28268136
13.
Mol Cancer Ther ; 16(2): 388-396, 2017 02.
Article de Anglais | MEDLINE | ID: mdl-27903752

RÉSUMÉ

An increasing number of BET family protein inhibitors have recently entered clinical trials. It has been reported that attempts of monitoring target engagement of the BET bromodomain inhibitor OTX015 using literature-described putative pharmacodynamic markers, such as c-Myc, BRD2, etc., failed to detect pharmacodynamic marker responses in AML patients treated at active dose and those with clinical responses. Here, we report the identification and characterization of HEXIM1 and other genes as robust pharmacodynamic markers for BET inhibitors. Global gene expression profiling studies were carried out using cancer cells and surrogate tissues, such as whole blood and skin, to identify genes that are modulated by BET family proteins. Candidate markers were further characterized for concentration- and time-dependent responses to the BET inhibitor ABBV-075 in vitro and in vivo HEXIM1 was found to be the only gene that exhibited robust and consistent modulation by BET inhibitors across multiple cancer indications and surrogate tissues. Markers such as SERPINI1, ZCCHC24, and ZMYND8 were modulated by ABBV-075 and other BET inhibitors across cancer cell lines and xenograft tumors but not in blood and skin. Significant downregulation of c-Myc, a well-publicized target of BET inhibitors, was largely restricted to hematologic cancer cell lines. Incorporating well-characterized pharmacodynamic markers, such as HEXIM1 and other genes described here, can provide a better understanding of potential efficacy and toxicity associated with inhibiting BET family proteins and informs early clinical decisions on BET inhibitor development programs. Mol Cancer Ther; 16(2); 388-96. ©2016 AACR.


Sujet(s)
Antinéoplasiques/pharmacologie , Marqueurs biologiques , Tumeurs/génétique , Protéines nucléaires/antagonistes et inhibiteurs , Protéines de liaison à l'ARN/génétique , Animaux , Antinéoplasiques/pharmacocinétique , Biopsie , Lignée cellulaire tumorale , Analyse de regroupements , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Femelle , Analyse de profil d'expression de gènes , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Humains , Souris , Tumeurs/traitement médicamenteux , Tumeurs/métabolisme , Tumeurs/anatomopathologie , Spécificité d'organe/génétique , Protéines de liaison à l'ARN/métabolisme , Facteurs de transcription , Tests d'activité antitumorale sur modèle de xénogreffe
15.
J Med Chem ; 52(23): 7604-17, 2009 Dec 10.
Article de Anglais | MEDLINE | ID: mdl-19954246

RÉSUMÉ

A series of HIV protease inhibitor based on the allophenylnorstatine structure with various P(2)' moieties were synthesized. Among these analogues, we discovered that a small allyl group would maintain potent enzyme inhibitory activity compared to the o-methylbenzyl moiety in clinical candidate 1 (KNI-764, also known as JE-2147, AG-1776, or SM-319777). Introduction of an anilinic amino group to 2 (KNI-727) improved water-solubility and anti-HIV-1 activity. X-ray crystallographic analysis of 13k (KNI-1689) with a beta-methallyl group at P(2)' position revealed hydrophobic interactions with Ala28, Ile84, and Ile50' similar to that of 1. The presence of an additional methyl group on the allyl group in compound 13k significantly increased anti-HIV activity over 1 while providing a rational drug design for structural minimization and improving membrane permeability.


Sujet(s)
Inhibiteurs de protéase du VIH/composition chimique , Inhibiteurs de protéase du VIH/pharmacologie , Protéase du VIH/composition chimique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/enzymologie , Phénylbutyrates/composition chimique , Phénylbutyrates/pharmacologie , Lignée cellulaire , Protéase du VIH/métabolisme , Inhibiteurs de protéase du VIH/synthèse chimique , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Humains , Modèles moléculaires , Conformation moléculaire , Phénylbutyrates/synthèse chimique , Relation structure-activité , Spécificité du substrat
16.
J Med Chem ; 52(9): 2964-70, 2009 May 14.
Article de Anglais | MEDLINE | ID: mdl-19348416

RÉSUMÉ

We studied the synthesis, cleavage rates, and oral administration of prodrugs of the HIV protease inhibitors (PIs) lopinavir and ritonavir. Phosphate esters attached directly to the central hydroxyl groups of these PIs did not demonstrate enzyme-mediated cleavage in vitro and did not provide measurable plasma levels of the parent drugs in vivo. However, oxymethylphosphate (OMP) and oxyethylphosphate (OEP) prodrugs provided improved rates of cleavage, high levels of aqueous solubility, and high plasma levels of the parent drugs when dosed orally in rats and dogs. Dosing unformulated capsules containing the solid prodrugs led to plasma levels equivalent to those observed for dosing formulated solutions of the parent drugs. A direct synthetic process for the preparation of OMP and OEP prodrugs was developed, and the improved synthetic method may be applicable to the preparation of analogous soluble prodrugs of other drug classes with limited solubility.


Sujet(s)
Promédicaments/composition chimique , Promédicaments/pharmacocinétique , Pyrimidinones/composition chimique , Pyrimidinones/pharmacocinétique , Ritonavir/composition chimique , Ritonavir/pharmacocinétique , Eau/composition chimique , Administration par voie orale , Animaux , Chiens , Femelle , Protéase du VIH/métabolisme , Inhibiteurs de protéase du VIH/administration et posologie , Inhibiteurs de protéase du VIH/composition chimique , Inhibiteurs de protéase du VIH/pharmacocinétique , Concentration en ions d'hydrogène , Lopinavir , Mâle , Promédicaments/administration et posologie , Pyrimidinones/administration et posologie , Rats , Rat Sprague-Dawley , Ritonavir/administration et posologie , Solubilité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...