Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 62
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Antimicrob Agents Chemother ; : e0103924, 2024 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-39240093

RÉSUMÉ

Oral broad-spectrum antivirals are urgently needed for the treatment of many emerging and contemporary RNA viruses. We previously synthesized 1-O-octadecyl-2-O-benzyl-sn-glyceryl-P-RVn (ODBG-P-RVn, V2043), a phospholipid prodrug of GS-441524 (remdesivir nucleoside, RVn), and demonstrated its in vivo efficacy in a SARS-CoV-2 mouse model. Structure-activity relationship studies focusing on the prodrug scaffold identified two modifications, 3-fluoro-4-methoxy-benzyl (V2053) and 4-cyano-benzyl (V2067), that significantly enhanced the in vitro broad-spectrum antiviral activity against multiple RNA viruses when compared to V2043. Here, we demonstrate that V2043, V2053, and V2067 are all orally bioavailable, well-tolerated, and achieve high sustained plasma levels after single oral daily dosing. All three phospholipid prodrugs are significantly more active than RVn in vitro and significantly reduce SARS-CoV-2 lung titers in prophylaxis and treatment mouse models of SARS-CoV-2 B.1.351 infection. On a molar basis, V2043 and V2067 are substantially more active than obeldesivir/GS-5245 and molnupiravir in vivo. Together, these data support the continued development of phospholipid RVn prodrugs for the treatment of SARS-CoV-2 and other RNA viruses of clinical concern.

2.
Antiviral Res ; 219: 105718, 2023 11.
Article de Anglais | MEDLINE | ID: mdl-37758067

RÉSUMÉ

Broad spectrum oral antivirals are urgently needed for the early treatment of many RNA viruses of clinical concern. We previously described the synthesis of 1-O-octadecyl-2-O-benzyl-glycero-3-phospho-RVn (V2043), an orally bioavailable lipid prodrug of remdesivir nucleoside (RVn, GS-441524) with broad spectrum antiviral activity against viruses with pandemic potential. Here we compared the relative activity of V2043 with new RVn lipid prodrugs containing sn-1 alkyl ether or sn-2 glycerol modifications. We found that 3-F-4-MeO-Bn, 3-CN-Bn, and 4-CN-Bn sn-2 glycerol modifications improved antiviral activity compared to V2043 when tested in vitro against clinically important RNA viruses from 5 virus families. These results support the continued development of V2043 and sn-2 glycerol modified RVn lipid prodrugs for the treatment of a broad range of RNA viruses for which there are limited therapies.


Sujet(s)
Antiviraux , Promédicaments , Antiviraux/pharmacologie , Promédicaments/pharmacologie , Nucléosides/pharmacologie , Glycérol , Lipides/pharmacologie
3.
J Med Chem ; 66(8): 5802-5819, 2023 04 27.
Article de Anglais | MEDLINE | ID: mdl-37040439

RÉSUMÉ

Early antiviral treatments, including intravenous remdesivir (RDV), reduce hospitalization and severe disease caused by COVID-19. An orally bioavailable RDV analog may facilitate earlier treatment of non-hospitalized COVID-19 patients. Here we describe the synthesis and evaluation of alkyl glyceryl ether phosphodiesters of GS-441524 (RVn), lysophospholipid analogs which allow for oral bioavailability and stability in plasma. Oral treatment of SARS-CoV-2-infected BALB/c mice with 1-O-octadecyl-2-O-benzyl-sn-glyceryl-3-phospho-RVn (60 mg/kg orally, once daily for 5 days starting 12h after infection) reduced lung viral load by 1.5 log10 units versus vehicle at day 2 and to below the limit of detection at day 5. Structure/activity evaluation of additional analogs that have hydrophobic ethers at the sn-2 of glycerol revealed improved in vitro antiviral activity by introduction of a 3-fluoro-4-methoxy-substituted benzyl or a 3- or 4-cyano-substituted benzyl. Collectively, our data support the development of RVn phospholipid prodrugs as oral antiviral agents for prevention and treatment of SARS-CoV-2 infections.


Sujet(s)
Antiviraux , COVID-19 , Animaux , Souris , SARS-CoV-2 , Phospholipides
4.
Microbiol Spectr ; 9(3): e0153721, 2021 12 22.
Article de Anglais | MEDLINE | ID: mdl-34817209

RÉSUMÉ

The necessity for intravenous administration of remdesivir confines its utility for treatment of coronavirus disease 2019 (COVID-19) to hospitalized patients. We evaluated the broad-spectrum antiviral activity of ODBG-P-RVn, an orally available, lipid-modified monophosphate prodrug of the remdesivir parent nucleoside (GS-441524), against viruses that cause diseases of human public health concern, including severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). ODBG-P-RVn showed 20-fold greater antiviral activity than GS-441524 and had activity nearly equivalent to that of remdesivir in primary-like human small airway epithelial cells. Our results warrant in vivo efficacy evaluation of ODBG-P-RVn. IMPORTANCE While remdesivir remains one of the few drugs approved by the FDA to treat coronavirus disease 2019 (COVID-19), its intravenous route of administration limits its use to hospital settings. Optimizing the stability and absorption of remdesivir may lead to a more accessible and clinically potent therapeutic. Here, we describe an orally available lipid-modified version of remdesivir with activity nearly equivalent to that of remdesivir against emerging viruses that cause significant disease, including Ebola and Nipah viruses. Our work highlights the importance of such modifications to optimize drug delivery to relevant and appropriate human tissues that are most affected by such diseases.


Sujet(s)
AMP/usage thérapeutique , Adénosine/usage thérapeutique , Alanine/usage thérapeutique , Antiviraux/usage thérapeutique , Traitements médicamenteux de la COVID-19 , Nucléosides/usage thérapeutique , Promédicaments/usage thérapeutique , Adénosine/analogues et dérivés , AMP/analogues et dérivés , Alanine/analogues et dérivés , Animaux , Éthers de glycéryle/usage thérapeutique , Humains , Lipides , SARS-CoV-2
5.
bioRxiv ; 2021 Aug 10.
Article de Anglais | MEDLINE | ID: mdl-34401879

RÉSUMÉ

The intravenous administration of remdesivir for COVID-19 confines its utility to hospitalized patients. We evaluated the broad-spectrum antiviral activity of ODBG-P-RVn, an orally available, lipid-modified monophosphate prodrug of the remdesivir parent nucleoside (GS-441524) against viruses that cause diseases of human public health concern, including SARS-CoV-2. ODBG-P-RVn showed 20-fold greater antiviral activity than GS-441524 and had near-equivalent activity to remdesivir in primary-like human small airway epithelial cells. Our results warrant investigation of ODBG-P-RVn efficacy in vivo.

6.
Antimicrob Agents Chemother ; 65(10): e0115521, 2021 09 17.
Article de Anglais | MEDLINE | ID: mdl-34310217

RÉSUMÉ

Remdesivir (RDV; GS-5734) is currently the only FDA-approved antiviral drug for the treatment of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The drug is approved for use in adults or children 12 years or older who are hospitalized for the treatment of COVID-19 on the basis of an acceleration of clinical recovery for inpatients with this disease. Unfortunately, the drug must be administered intravenously, restricting its use to those requiring hospitalization for relatively advanced disease. RDV is also unstable in plasma and has a complex activation pathway which may contribute to its highly variable antiviral efficacy in SARS-CoV-2-infected cells. Potent orally bioavailable antiviral drugs for early treatment of SARS-CoV-2 infection are urgently needed, and several, including molnupiravir and PF-07321332, are currently in clinical development. We focused on making simple, orally bioavailable lipid analogs of remdesivir nucleoside (RVn; GS-441524) that are processed to RVn monophosphate, the precursor of the active RVn triphosphate, by a single-step intracellular cleavage. In addition to high oral bioavailability, stability in plasma, and simpler metabolic activation, new oral lipid prodrugs of RVn had submicromolar anti-SARS-CoV-2 activity in a variety of cell types, including Vero E6, Calu-3, Caco-2, human pluripotent stem cell (PSC)-derived lung cells, and Huh7.5 cells. In Syrian hamsters, oral treatment with 1-O-octadecyl-2-O-benzyl-glycero-3-phosphate RVn (ODBG-P-RVn) was well tolerated and achieved therapeutic levels in plasma above the 90% effective concentration (EC90) for SARS-CoV-2. The results suggest further evaluation as an early oral treatment for SARS-CoV-2 infection to minimize severe disease and reduce hospitalizations.


Sujet(s)
Traitements médicamenteux de la COVID-19 , Promédicaments , Adénosine/analogues et dérivés , AMP/analogues et dérivés , Alanine/analogues et dérivés , Animaux , Antiviraux/pharmacologie , Cellules Caco-2 , Cricetinae , Humains , Lipides , SARS-CoV-2
7.
bioRxiv ; 2021 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-32869033

RÉSUMÉ

Remdesivir (RDV, GS-5734) is currently the only FDA-approved antiviral drug for the treatment of SARS CoV-2 infection. The drug is approved for use in adults or children 12-years or older who are hospitalized for the treatment of COVID-19 on the basis of an acceleration of clinical recovery for inpatients with this disease. Unfortunately, the drug must be administered intravenously, restricting its use to those requiring hospitalization for relatively advanced disease. RDV is also unstable in plasma and has a complex activation pathway which may contribute to its highly variable antiviral efficacy in SARS-CoV-2 infected cells. Potent orally bioavailable antiviral drugs for early treatment of SARS-CoV-2 infection are urgently needed and several including molnupiravir and PF-07321332 are currently in clinical development. We focused on making simple, orally bioavailable lipid analogs of Remdesivir nucleoside (RVn, GS-441524) that are processed to RVn-monophosphate, the precursor of the active RVn-triphosphate, by a single-step intracellular cleavage. In addition to high oral bioavailability, stability in plasma and simpler metabolic activation, new oral lipid prodrugs of RVn had submicromolar anti-SARS-CoV-2 activity in a variety of cell types including Vero E6, Calu-3, Caco-2, human pluripotent stem cell (PSC)-derived lung cells and Huh7.5 cells. In Syrian hamsters oral treatment with ODBG-P-RVn was well tolerated and achieved therapeutic levels in plasma above the EC90 for SARS-CoV-2. The results suggest further evaluation as an early oral treatment for SARS-CoV-2 infection to minimize severe disease and reduce hospitalizations.

8.
Antiviral Res ; 171: 104614, 2019 11.
Article de Anglais | MEDLINE | ID: mdl-31550449

RÉSUMÉ

Pre-exposure prophylaxis (PrEP) with topically or systemically administered antiretroviral agents can prevent acquisition of human immunodeficiency virus type 1 (HIV-1) infection. However, in clinical trials using tenofovir-containing agents, HIV-1 acquisition is reduced but not eliminated. Incomplete adherence remains the major contributor to failure. Sustained release or long-acting antiretroviral agents may provide better HIV-1 protection by reducing the clinical impact of incomplete adherence. To reduce dosing frequency, we synthesized a novel tenofovir prodrug, octadecyloxyethyl benzyl tenofovir (ODE-Bn-TFV), that is designed to release TFV slowly in tissues, and showed potent anti-HIV activity in vitro (EC50 = 1.7 nM). In cells exposed to 14C labeled TFV, ODE-Bn-TFV or the quickly activated monoester ODE-TFV, rapid cellular uptake for both lipophilic analogs was noted, achieving 50-fold higher levels than unmodified TFV after 48 h. Following exposure to ODE-[8-14C]TFV, the intracellular diphosphate levels were approximately four-fold higher than with ODE-Bn-TFV. However, intracellular TFVpp drug levels fell rapidly yielding a half-life of about two days. TFVpp levels in ODE-Bn-TFV treated cells decreased much more slowly and reached half-maximal levels in about seven days. These results suggest early accumulation of ODE-Bn-TFV followed by sustained intracellular release following cleavage of the ester bonds linking the ODE and benzyl moieties to the active molecular precursor, thereby potentially allowing for less frequent administration than with more rapidly activated forms of tenofovir.


Sujet(s)
Ténofovir/analogues et dérivés , Ténofovir/pharmacologie , Adénine/analogues et dérivés , Adénine/pharmacocinétique , Techniques de chimie synthétique , Esters , Humains , Structure moléculaire , Organophosphates/pharmacocinétique , Ténofovir/synthèse chimique , Distribution tissulaire
9.
Chemotherapy ; 63(4): 225-237, 2018.
Article de Anglais | MEDLINE | ID: mdl-30372692

RÉSUMÉ

Acute myeloid leukemia (AML) continues to be a deadly disease, with only 50-70% of patients achieving complete remission and less than 30% of adults having sustained long-term remissions. In order to address these unmet medical needs, we carried out a high-throughput screen of an in-house library of on- and off-patent drugs with the OCI/AML-2 cell line. Through this screen, we discovered adefovir dipi-voxil (adefovir-DP) as being active against human AML. In addition to adefovir-DP, there are second-generation formulations of adefovir, including octadecyloxyethyl adefovir (ODE-adefovir) and hexadecyloxypropyl adefovir (HDP-adefovir), which were designed to overcome the pharmacokinetic problems of the parent compound adefovir. Given the known clinical benefit of nucleoside analogs for the treatment of AML, we undertook studies to evaluate the potential benefit of adefovir-based molecules. In AML cell lines and patient samples, adefovir-DP and ODE-adefovir were highly potent, whereas HDP-adefovir was significantly less active. Interestingly, ODE-adefovir was remarkably less toxic than adefovir-DP towards normal hematopoietic cells. In addition, ODE-adefovir at a dose of 15 mg/kg/day showed potent activity against human AML in a NOD/SCID mouse model, with a reduction of human leukemia in mouse bone marrow of > 40% in all mice tested within 20 days of treatment. Based on its chemical structure, we hypothesized that the cytotoxicity of ODE-adefovir toward AML was through cell cycle arrest and DNA damage. Indeed, ODE-adefovir treatment induced cell cycle arrest in the S phase and increased levels of pH2Ax, indicating the induction of DNA damage. Furthermore, there was an increase in phospho-p53, transactivation of proapoptotic genes and activation of the intrinsic apoptotic pathway. Subsequent investigation unveiled strong synergism between ODE-adefovir and ara-C, making their coadministration of potential clinical benefit. Expression of MRP4, a nucleoside transporter, appeared to influence the response of AML cells to ODE-adefovir, as its inhibition potentiated ODE-adefovir killing. Taken together, our findings indicate that clinical development of ODE-adefovir or related compounds for the treatment of AML is warranted.


Sujet(s)
Adénine/analogues et dérivés , Apoptose/effets des médicaments et des substances chimiques , Cytarabine/pharmacologie , Phosphonates/pharmacologie , Adénine/composition chimique , Adénine/pharmacologie , Adénine/usage thérapeutique , Animaux , Caspases/métabolisme , Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Cytarabine/usage thérapeutique , Altération de l'ADN/effets des médicaments et des substances chimiques , Préparation de médicament , Résistance aux médicaments antinéoplasiques , Synergie des médicaments , Femelle , Histone/métabolisme , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Souris , Souris de lignée NOD , Souris SCID , Phosphonates/composition chimique , Phosphonates/usage thérapeutique , Poly(ADP-ribose) polymerases/métabolisme , Transplantation hétérologue , Cellules cancéreuses en culture
10.
Antiviral Res ; 158: 122-126, 2018 10.
Article de Anglais | MEDLINE | ID: mdl-30096340

RÉSUMÉ

On September 22, 2008, a physician on Prince of Wales Island, Alaska, notified the Alaska Department of Health and Social Services (ADHSS) of an unusually high number of adult patients with recently diagnosed pneumonia (n = 10), including three persons who required hospitalization and one who died. ADHSS and CDC conducted an investigation to determine the cause and distribution of the outbreak, identify risk factors for hospitalization, and implement control measures. This report summarizes the results of that investigation, which found that the outbreak was caused by adenovirus 14 (Ad14), an emerging adenovirus serotype in the United States that is associated with a higher rate of severe illness compared with other adenoviruses. Among the 46 cases identified in the outbreak from September 1 through October 27, 2008, the most frequently observed characteristics included the following: male (70%), Alaska Native (61%), underlying pulmonary disease (44%), aged > or = 65 years (26%), and current smoker (48%). Patients aged > or = 65 years had a fivefold increased risk for hospitalization. The most commonly reported symptoms were cough (100%), shortness of breath (87%), and fever (74%). Of the 11 hospitalized patients, three required intensive care, and one required mechanical ventilation. One death was reported. Ad14 isolates obtained during the outbreak were identical genetically to those in recent community-acquired outbreaks in the United States which suggests the emergence of a new, and possibly more virulent Ad14 variant. Clinicians should consider Ad14 infection in the differential diagnosis for patients with community-acquired pneumonia, particularly when unexplained clusters of severe respiratory infections are detected.


Sujet(s)
Adénovirus humains/effets des médicaments et des substances chimiques , Esters/pharmacologie , Nucléosides/pharmacologie , Sérogroupe , Cellules A549 , Infections humaines à adénovirus/épidémiologie , Infections humaines à adénovirus/virologie , Adénovirus humains/pathogénicité , Sujet âgé , Épidémies de maladies , Femelle , Fièvre , Humains , Mâle , Nucléosides/analogues et dérivés , Analyse de régression , Infections de l'appareil respiratoire/virologie , États-Unis
12.
Drug Deliv ; 24(1): 1703-1712, 2017 Nov.
Article de Anglais | MEDLINE | ID: mdl-29115885

RÉSUMÉ

Proliferative vitreoretinopathy (PVR) is a blinding eye disease and there is no effective pharmacological measure to prevent PVR development. The difficulty comes from lack of potent antiproliferative agent and lack of sustained delivery to cover high-risk time window for PVR to develop. Lipid prodrug of PMEG, hexadecyloxypropyl 9-[(2-phosphonomethoxy)ethyl]guanine (HDP-PMEG), was prepared and was evaluated as a pharmacological adjuvant to surgical management of PVR. A dose-escalation study determined that the highest nontoxic dose for intravitreal use in pigmented rabbits was 3 µg per eye. The genotoxicity of HDP-PMEG was harnessed as a perioperative preventative measure against PVR in a rabbit eye model while the sustained intravitreal pharmacological effect was evaluated on a laser-induced fibrovascular model in rat eye. After intravitreal 3 µg, HDP-PMEG particles in the rabbit vitreous was visible for at least 6 weeks. A single 50-min intravitreal infusion of HDP-PMEG demonstrated significant inhibition of PVR formation when compared with the eyes infused with only BSS (BSS vs. HDP-PMEG: estimate = 1.14, OR = 3.1, p = .027). A single intravitreal 104 ng (equivalent to 3 µg for rabbit eye) of HDP-PMEG significantly inhibit laser-induced fibrovascular proliferation in rat eye by 55% (least square mean pixel, BSS = 4763569.5 vs. HDP-PMEG = 2148129.7, p < .0001, generalized estimating equation [GEE]). Retinal fluorescein angiography showed the odds for BSS intervened eyes to have higher-rated FA leaking grades were 38.5 times compared with HDP-PMEG treated eyes (p < .0001, GEE). Our study results indicate that single intravitreal HDP-PMEG may be a promising ocular drug delivery as a perioperative intervention to prevent PVR reoccurrence following primary surgical management.


Sujet(s)
Prolifération cellulaire/effets des médicaments et des substances chimiques , Préparations à action retardée/pharmacologie , Guanine/analogues et dérivés , Lipides/pharmacologie , Composés organiques du phosphore/pharmacologie , Promédicaments/pharmacologie , Corps vitré/effets des médicaments et des substances chimiques , Animaux , Modèles animaux de maladie humaine , Systèmes de délivrance de médicaments/méthodes , Guanine/pharmacologie , Lapins , Rats , Rats de lignée BN , Vitréorétinopathie proliférante/traitement médicamenteux
13.
J Med Chem ; 59(23): 10470-10478, 2016 12 08.
Article de Anglais | MEDLINE | ID: mdl-27933957

RÉSUMÉ

Human papillomavirus (HPV) high-risk genotypes such as HPV-16 and HPV-18 cause the majority of anogenital tract carcinomas, including cervical cancer, the second most common malignancy in women worldwide. Currently there are no approved antiviral agents that reduce or eliminate HPV and reverse virus-associated pathology. We synthesized and evaluated several alkoxyalkyl acyclic nucleoside phosphonate diesters and identified octadecyloxyethyl benzyl 9-[(2-phosphonomethoxy)ethyl]guanine (ODE-Bn-PMEG) as an active compound which strongly inhibited transient amplification of HPV-11, -16, and -18 origin-containing plasmid DNA in transfected cells at concentrations well below its cytotoxic concentrations. ODE-Bn-PMEG demonstrated increased uptake in human foreskin fibroblast cells and was readily converted in vitro to the active antiviral metabolite, PMEG diphosphate. The P-chiral enantiomers of ODE-Bn-PMEG were obtained and appeared to have equivalent antiviral activities against HPV. ODE-Bn-PMEG is a promising candidate for the local treatment of HPV-16 and HPV-18 and other high-risk types, an important unmet medical need.


Sujet(s)
Antiviraux/pharmacologie , ADN viral/effets des médicaments et des substances chimiques , Guanine/analogues et dérivés , Techniques d'amplification d'acides nucléiques , Phosphonates/pharmacologie , Papillomaviridae/effets des médicaments et des substances chimiques , Antiviraux/synthèse chimique , Antiviraux/composition chimique , Lignée cellulaire tumorale , Relation dose-effet des médicaments , Guanine/synthèse chimique , Guanine/composition chimique , Guanine/pharmacologie , Cellules HEK293 , VIH (Virus de l'Immunodéficience Humaine)/effets des médicaments et des substances chimiques , Herpèsvirus humain de type 2/effets des médicaments et des substances chimiques , Humains , Agranulocytes/effets des médicaments et des substances chimiques , Agranulocytes/virologie , Structure moléculaire , Phosphonates/synthèse chimique , Phosphonates/composition chimique , Papillomaviridae/génétique , Relation structure-activité , Réplication virale/effets des médicaments et des substances chimiques
14.
Eur J Pharm Biopharm ; 89: 271-9, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25513956

RÉSUMÉ

There still is an unmet need for a safe and sustained intravitreal drug delivery system. In this study we are proposing and characterizing a micelle based, clear-media intravitreal drug delivery system using the lipid derivatized nucleoside analog, hexadecyloxypropyl-cidofovir (HDP-CDV, CMX 001). HDP-CDV forms micelles in water and in vitreous supernatant with the critical micelle concentration of 19 µg/mL and 9 µg/mL, respectively at 37 °C. The formed micelles had the average size of 274.7 nm and the Zeta potential of -47.1 mV. Drug release study in the excised rabbit vitreous showed a sustained release profile with a half-life of 2.7 days. The micelle formulation of HDP-CDV demonstrated a good safety profile in two animal species (rabbit and guinea pig) following intravitreal injection. The sustained efficacy was tested in a pretreatment study design and the drug potency was tested in an ongoing herpes simplex virus (HSV-1) retinitis model. The pretreatment studies using single intravitreal injection and later HSV-1 infection revealed at least 9 weeks of vitreous presence and therapeutic level of HDP-CDV, with 71% eyes protection from infection. The treatment study demonstrated that intravitreal administration halted active HSV-1 retinitis in 80% of the infected eyes while cidofovir (CDV) treatment failed to suppress active HSV-1 retinitis. In summary, lipid derivatized nucleoside analogs can be formulated as a micelle intravitreal injection and provides a sustained drug release in vitreous for chronic retinal diseases.


Sujet(s)
Cytosine/analogues et dérivés , Préparations à action retardée/administration et posologie , Préparations à action retardée/composition chimique , Phosphonates/administration et posologie , Phosphonates/composition chimique , Corps vitré/effets des médicaments et des substances chimiques , Animaux , Antiviraux/administration et posologie , Antiviraux/composition chimique , Chimie pharmaceutique/méthodes , Cytosine/administration et posologie , Cytosine/composition chimique , Systèmes de délivrance de médicaments/méthodes , Cochons d'Inde , Période , Herpès/traitement médicamenteux , Herpèsvirus humain de type 1/effets des médicaments et des substances chimiques , Injections intravitréennes/méthodes , Micelles , Promédicaments/administration et posologie , Promédicaments/composition chimique , Lapins , Rétine/effets des médicaments et des substances chimiques , Rétine/virologie , Corps vitré/virologie
15.
Mol Vis ; 18: 1907-17, 2012.
Article de Anglais | MEDLINE | ID: mdl-22876115

RÉSUMÉ

PURPOSE: The objectives of this study were to synthesize and characterize two types of cytarabine (Ara-C) lipid produgs and evaluate the prodrugs for sustained intraocular delivery after administration by intravitreal injection. METHODS: Hexadecyloxypropyl cytarabine 5'-monophosphate (HDP-P-Ara-C) and hexadecyloxypropyl cytarabine 3',5'-cyclic monophosphate (HDP-cP-Ara-C) were synthesized starting from cytarabine (1-ß-D-arabinofuranosylcytosine). Their vitreal clearance profile was simulated using a custom dissolution chamber, in vitro cytotoxicity was evaluated using cell proliferation assays, and in vivo ocular properties in rat and rabbit eyes were assessed using biomicroscopy, indirect ophthalmoscopy, tonometry, electroretinography, and histology. RESULTS: HDP-P-Ara-C was cleared from the dissolution chamber (flow rate 2 µL/min) within 7 days. In contrast, HDP-cP-Ara-C, a much more insoluble prodrug, was still detectable 36 days after the dissolution process was started. HDP-P-Ara-C had a 50% cytotoxicity concentration of 52±2.6 µM in human retinal pigment epithelium (ARPE-19) and 32±2.2 µM in a rat Müller cell line, rMC-1. The 50% cytotoxicity concentration values for HDP-cP-Ara-C in ARPE-19 and rMC-1 cells were 50 µM and 25 µM, respectively. HDP-P-Ara-C was not detectable 2 weeks after the highest intravitreal dose (228 µg/rat eye) was injected, and no ocular toxicity was found. With HDP-cP-Ara-C, the drug depot was visible for 26 weeks following a single intravitreal injection (800 µg/rabbit eye). For both compounds, the electroretinogram, intraocular pressure, and other toxicity studies were negative except for the highest dose of HDP-cP-Ara-C (800 µg/eye), which had focal toxicity from the direct touch of the retina and decreased dark adapted a-waves and decreased flicker electroretinogram amplitudes (generalized estimating equations, p=0.039 and 0.01). CONCLUSIONS: The cyclic monophosphate prodrug, HDP-cP-Ara-C, was found to have physiochemical properties better suited for sustained delivery of cytarabine to posterior segments of the eye. These properties included limited aqueous solubility, in vitro antiproliferative activity, and good tolerability after injection into rabbit eyes.


Sujet(s)
Antimétabolites antinéoplasiques/administration et posologie , Arabinonucléotides/administration et posologie , Cytidine monophosphate/analogues et dérivés , Préparations à action retardée/administration et posologie , Promédicaments/administration et posologie , Rétine/effets des médicaments et des substances chimiques , Vitréorétinopathie proliférante/traitement médicamenteux , Corps vitré/effets des médicaments et des substances chimiques , Animaux , Antimétabolites antinéoplasiques/synthèse chimique , Arabinonucléotides/synthèse chimique , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Cytidine monophosphate/administration et posologie , Cytidine monophosphate/synthèse chimique , Préparations à action retardée/synthèse chimique , Chambres de culture à diffusion , Électrorétinographie , Humains , Injections intravitréennes , Cinétique , Ophtalmoscopie , Promédicaments/synthèse chimique , Lapins , Rats , Rétine/métabolisme , Rétine/anatomopathologie , Épithélium pigmentaire de la rétine/effets des médicaments et des substances chimiques , Épithélium pigmentaire de la rétine/métabolisme , Épithélium pigmentaire de la rétine/anatomopathologie , Solubilité , Vitréorétinopathie proliférante/métabolisme , Vitréorétinopathie proliférante/anatomopathologie , Corps vitré/métabolisme , Corps vitré/anatomopathologie
16.
Invest Ophthalmol Vis Sci ; 52(13): 9391-6, 2011 Dec 09.
Article de Anglais | MEDLINE | ID: mdl-22058340

RÉSUMÉ

PURPOSE: To evaluate the intraocular safety and pharmacokinetics of hexadecyloxypropyl-cidofovir (HDP-CDV), the hydrolysis product of HDP-cyclic-CDV, a long-lasting intravitreal cidofovir prodrug for cytomegalovirus (CMV) retinitis. METHODS: HDP-cyclic-CDV was suspended in phosphate-buffered saline (PBS) at 37°C and formation of HDP-CDV was monitored by high-performance liquid chromatography (HPLC) analysis for 30 weeks. The safety and pharmacokinetics of HDP-CDV intravitreal injections were studied using New Zealand Red rabbits and (14)C labeled HDP-CDV. Ocular tissues from five time points (1, 3, 7, 14, and 35 days) were analyzed by scintillation counting and HPLC to characterize the pharmacokinetics. RESULTS: During the hydrolysis study, approximately 35% of the HDP-cyclic-CDV was converted to HDP-CDV. Evaluation of safety found no toxicity after intravitreal injection of HDP-CDV up to 28 µg/eye. Intravitreal pharmacokinetics of HDP-CDV in the retina, choroid, and vitreous followed a two-phase elimination process and elimination half-lives of 8.4 days (retina), 6.9 days (choroid), and 6.2 days (vitreous). In the retina, cidofovir and an unknown metabolite were detected in the first 2 weeks, and the maximum metabolite concentrations were present 48 hours after the maximum HDP-CDV concentration. CONCLUSIONS: HDP-cyclic CDV, under simulated physiologic conditions, slowly converts to HDP-CDV, another potent anti-CMV prodrug that may be taken up by retinal cells and metabolized further to the active antiviral metabolite, cidofovir diphosphate. Taken together, these observations help to explain the ability of a single intravitreal dose of HDP-cyclic-CDV to prevent viral retinitis for up to 68 days in a rabbit model.


Sujet(s)
Corps ciliaire/métabolisme , Rétinite à cytomégalovirus/traitement médicamenteux , Cytosine/analogues et dérivés , Phosphonates/pharmacocinétique , Rétine/métabolisme , Corps vitré/métabolisme , Animaux , Chromatographie en phase liquide à haute performance , Corps ciliaire/effets des médicaments et des substances chimiques , Rétinite à cytomégalovirus/métabolisme , Cytosine/administration et posologie , Cytosine/pharmacocinétique , Préparations à action retardée , Modèles animaux de maladie humaine , Relation dose-effet des médicaments , Études de suivi , Hydrolyse , Injections intravitréennes , Phosphonates/administration et posologie , Lapins , Rétine/effets des médicaments et des substances chimiques , Corps vitré/effets des médicaments et des substances chimiques
17.
Antimicrob Agents Chemother ; 55(11): 5063-72, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21896914

RÉSUMÉ

(S)-1-[3-hydroxy-2-(phosphonomethoxy)propyl]cytosine (HPMPC [cidofovir]) and (S)-9-[3-hydroxy-2-(phosphonomethoxy)propyl]adenine (HPMPA) are potent inhibitors of a variety of DNA viruses. These drugs possess a 3'-hydroxyl equivalent which could support chain extension from an incorporated drug molecule. HPMPC and HPMPA were initially reported to lack activity against human immunodeficiency virus type 1 (HIV-1); more recent results have shown that the octadecyloxyethyl (ODE) and hexadecyloxypropyl (HDP) esters of HPMPA are potent inhibitors of the virus. We have synthesized the ODE esters of a series of (S)-[3-hydroxy-2-(phosphonomethoxy)propyl] (HPMP) nucleosides, including HPMPC, HPMP-guanine (HPMPG), HPMP-thymine (HPMPT), and HPMP-diaminopurine (HPMPDAP), as well as the ODE ester of the obligate chain terminator (S)-9-[3-methoxy-2-(phosphonomethoxy)-propyl]adenine (MPMPA). All compounds except ODE-HPMPT were inhibitors of HIV-1 replication at low nanomolar concentrations. These compounds were also inhibitors of the replication of HIV-1 variants that are resistant to various nucleoside reverse transcriptase (RT) inhibitors at concentrations several times lower than would be expected to be achieved in vivo. To investigate the mechanism of the antiviral activity, the active metabolites of HPMPC and HPMPA were studied for their effects on reactions catalyzed by HIV-1 RT. Incorporation of HPMPC and HPMPA into a DNA primer strand resulted in multiple inhibitory effects exerted on the enzyme and showed that neither compound acts as an absolute chain terminator. Further, inhibition of HIV-1 RT also occurred when these drugs were located in the template strand. These results indicate that HPMPC and HPMPA inhibit HIV-1 by a complex mechanism and suggest that this class of drugs has a broader spectrum of activity than previously shown.


Sujet(s)
Agents antiVIH/pharmacologie , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Nucléosides/pharmacologie , Agents antiVIH/effets indésirables , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Humains , Structure moléculaire , Nucléosides/composition chimique
18.
Bioorg Med Chem ; 19(15): 4616-25, 2011 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-21719300

RÉSUMÉ

We reported previously that octadecyloxyethyl 9-(S)-[3-hydroxy-2-(phosphonomethoxy)-propyl]adenine (ODE-(S)-HPMPA) was active against genotype 1b and 2a hepatitis C virus (HCV) replicons. This is surprising because acyclic nucleoside phosphonates have been regarded as having antiviral activity only against double stranded DNA viruses, HIV and HBV. We synthesized octadecyloxyethyl 9-(S)-[3-methoxy-2-(phosphonomethoxy)propyl]-adenine and found it to be active in genotype 1b and 2a HCV replicons with EC50 values of 1-2 µM and a CC50 of > 150 µM. Analogs with substitutions at the 3'-hydroxyl larger than methyl or ethyl, or with other purine bases were less active but most compounds had significant antiviral activity against HIV-1 in vitro. The most active anti-HIV compound was octadecyloxyethyl 9-(R)-[3-methoxy-2-(phosphonomethoxy)propyl]guanine with an EC50 < 0.01 nanomolar and a selectivity index of > 4.4 million.


Sujet(s)
Adénine/analogues et dérivés , Antiviraux/pharmacologie , Infections à VIH/traitement médicamenteux , VIH-1 (Virus de l'Immunodéficience Humaine de type 1)/effets des médicaments et des substances chimiques , Hepacivirus/effets des médicaments et des substances chimiques , Hépatite C/traitement médicamenteux , Phosphonates/pharmacologie , Promédicaments/pharmacologie , Adénine/synthèse chimique , Adénine/composition chimique , Adénine/pharmacologie , Antiviraux/synthèse chimique , Antiviraux/composition chimique , Humains , Phosphonates/synthèse chimique , Phosphonates/composition chimique , Promédicaments/synthèse chimique , Promédicaments/composition chimique , Réplication virale/effets des médicaments et des substances chimiques
19.
Bioorg Med Chem ; 19(9): 2950-8, 2011 May 01.
Article de Anglais | MEDLINE | ID: mdl-21493074

RÉSUMÉ

Alkoxyalkyl esters of cidofovir (CDV) are orally active agents which inhibit the replication of a variety of double stranded DNA (dsDNA) viruses including variola, vaccinia, ectromelia, herpes simplex virus, cytomegalovirus, adenovirus and others. One of these compounds, hexadecyloxypropyl-CDV (HDP-CDV, CMX001) is in clinical development for prevention and treatment of poxvirus infection, vaccination complications, and for infections caused by cytomegalovirus, adenovirus, herpesviruses and other dsDNA viruses. This class of lipid analogs is potentially prone to undergo omega oxidation of the alkyl moiety which can lead to a short chain carboxylic acid lacking antiviral activity. To address this issue, we synthesized a series of alkoxyalkyl or alkyl glycerol esters of CDV and (S)-HPMPA having modifications in the structure of the alkyl residue. Antiviral activity was assessed in cells infected with vaccinia, cowpox or ectromelia viruses. Metabolic stability was determined in S9 membrane fractions from rat, guinea pig, monkey and human liver. All compounds had substantial antiviral activity in cells infected with vaccinia, cowpox or ectromelia. Metabolic stability was lowest in monkey liver S9 incubations where rapid disappearance of HDP-CDV and HDP-(S)-HPMPA was noted. Metabolic stability in monkey preparations increased substantially when a ω-1 methyl group (15-methyl-HDP-CDV) or a terminal cyclopropyl residue (14-cyclopropyl-tetradecyloxypropyl-CDV) was present in the alkyl chain. The most stable compound was 1-O-octadecyl-2-O-benzyl-sn-glycero-3-CDV (ODBG-CDV) which was not metabolized extensively by monkey liver S9. In rat, guinea pig or human liver S9 incubations, most of the modified antiviral compounds were considerably more stable.


Sujet(s)
Adénine/analogues et dérivés , Antiviraux/synthèse chimique , Cytosine/analogues et dérivés , Phosphonates/composition chimique , Adénine/synthèse chimique , Adénine/composition chimique , Adénine/pharmacologie , Animaux , Antiviraux/composition chimique , Antiviraux/pharmacologie , Cidofovir , Virus de la variole bovine/effets des médicaments et des substances chimiques , Cytosine/synthèse chimique , Cytosine/composition chimique , Cytosine/pharmacologie , Virus de l'ectromélie/effets des médicaments et des substances chimiques , Esters , Cochons d'Inde , Haplorhini , Humains , Foie/métabolisme , Phosphonates/synthèse chimique , Phosphonates/pharmacologie , Rats , Virus de la vaccine/effets des médicaments et des substances chimiques
20.
Mol Vis ; 17: 627-37, 2011 Mar 02.
Article de Anglais | MEDLINE | ID: mdl-21386925

RÉSUMÉ

PURPOSE: To investigate the safety and inhibitory effects of hexadecyloxypropyl 9-[2-(phosphonomethoxy) ethyl] guanine (HDP-PMEG) on ocular cell proliferation and collagen matrix contraction. METHODS: For the antiproliferation studies, various ocular cell monolayers were exposed to HDP-PMEG, PMEG, 5-fluorouracil (5-FU), and daunorubicin (DNB). For the collagen contraction studies, retinal pigment epithelium (RPE) cells seeded onto type I collagen lattices were exposed for a single 5- or 50-min period to various concentrations of HDP-PMEG or 5-FU. For the cytotoxicity study, trypan blue exclusion tests were performed using a human Müller cell line. Cytotoxicity was determined up to 4 days after treatment. RESULTS: The proliferation of RPE cells, scleral fibroblasts, vessel endothelial cells, and ocular melanoma cells can all be significantly inhibited by HDP-PMEG. Its inhibitory effects on those cells were uniformly stronger than that of 5-FU. Contraction of the collagen matrix containing RPE cells was significantly inhibited by HDP-PMEG and by 5-FU at concentrations of 20 µM and 2,000 µM, respectively, as compared with controls (p<0.05). The safety profile of HDP-PMEG was significantly better than 5-FU and daunorubicin. The ocular therapeutic index is 1,100 for HDP-PMEG, 17.2 for 5-FU, and 1.25 for daunorubicin. CONCLUSIONS: HDP-PMEG possesses a significant inhibitory effect on the proliferation of RPE, retinal glial cells, scleral fibroblasts, and ocular melanoma cells. HDP-PMEG is also genotoxic and may be used as a single short application for the modulation of unwanted ocular proliferation.


Sujet(s)
Oeil/cytologie , Guanine/analogues et dérivés , Composés organiques du phosphore/pharmacologie , Dosage biologique , Mort cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire , Prolifération cellulaire/effets des médicaments et des substances chimiques , Collagène/métabolisme , Matrice extracellulaire/effets des médicaments et des substances chimiques , Matrice extracellulaire/métabolisme , Fluorouracil/pharmacologie , Guanine/composition chimique , Guanine/pharmacologie , Humains , Composés organiques du phosphore/composition chimique , Facteurs temps
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE