Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
Diabetes Obes Metab ; 12(4): 348-55, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20380656

ABSTRACT

AIM: To determine efficacy and tolerability of dutogliptin, a dipeptidyl peptidase 4 (DPP4) inhibitor, in patients with type 2 diabetes mellitus. METHODS: This was a 12-week, multicentre, randomized, double-blind, placebo-controlled trial in 423 patients with type 2 diabetes with suboptimal metabolic control. Following a 2-week single-blind placebo run-in, patients aged 18-75 years with a body mass index of 25-48 kg/m(2) and baseline HbA1c of 7.3-11.0% were randomized 2:2:1 to receive once-daily oral therapy with either dutogliptin (400 or 200 mg) or placebo on a background medication of either metformin alone, a thiazolidinedione (TZD) alone or a combination of metformin plus a TZD. RESULTS: Average HbA1c at baseline was 8.4%. Administration of dutogliptin 400 and 200 mg for 12 weeks decreased HbA1c by -0.52% (p < 0.001) and -0.35% (p = 0.006), respectively (placebo-corrected values), with absolute changes in HbA1c for the 400 mg, 200 mg and placebo groups of -0.82, -0.64 and -0.3%, respectively. The proportion of patients achieving an HbA1c < 7% was 27, 21 and 12% at dutogliptin doses of 400 and 200 mg or placebo, respectively (p = 0.008 for comparison of 400 mg vs. placebo). Fasting plasma glucose (FPG) levels were significantly reduced in both active treatment groups compared to placebo: the placebo-corrected difference was -1.00 mmol/l (p < 0.001) for the 400 mg group and -0.88 mmol/l (p = 0.003) for the 200 mg group. Dutogliptin caused significantly greater reductions in postprandial glucose AUC (0-2h) in both the 400 and 200 mg groups (placebo corrected values -2.58 mmol/l/h, p < 0.001 and -1.63 mmol/l/h, p = 0.032, respectively). In general, patients tolerated the study drug well. There were minor, not clinically meaningful differences in adverse events (AEs) between dutogliptin-treated patients and placebo controls, and 60% of all reported AEs were mild. Vital signs and body weight were stable, and routine safety laboratory parameters did not change compared with placebo. Trough ex vivo DPP4 inhibition at the end of the 12-week treatment period was 80 and 70%, at the 400 and 200 mg doses of dutogliptin, respectively. CONCLUSIONS: Dutogliptin treatment for 12 weeks improved glycaemic control in patients with type 2 diabetes who were on a background medication of metformin, a TZD or metformin plus a TZD. Tolerability was favourable for both doses tested. The 400 mg dose of dutogliptin resulted in larger changes of HbA1c and FPG and more subjects reached an HbA1c target of < 7% than the 200 mg dose.


Subject(s)
Boronic Acids/administration & dosage , Diabetes Mellitus, Type 2/drug therapy , Glycated Hemoglobin/metabolism , Hypoglycemic Agents/administration & dosage , Adolescent , Adult , Aged , Diabetes Mellitus, Type 2/metabolism , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Female , Humans , Male , Middle Aged , Placebos , Treatment Outcome , Young Adult
2.
Diabetes Obes Metab ; 10(4): 293-300, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18333888

ABSTRACT

AIM: To determine the efficacy and tolerability of PHX1149, a novel dipeptidyl peptidase-4 (DPP4) inhibitor, in patients with type 2 diabetes. METHODS: This is a multicentre, randomized, double-blind, placebo-controlled, 4-week study in patients with type 2 diabetes with suboptimal metabolic control. Patients with a baseline haemoglobin A(1c) (HbA(1c)) of 7.3 to 11.0% were randomized 1 : 1 : 1 : 1 to receive once-daily oral therapy with either PHX1149 (100, 200 or 400 mg) or placebo; patients were on a constant background therapy of either metformin alone or metformin plus a glitazone. RESULTS: Treatment with 100, 200 or 400 mg of PHX1149 significantly decreased postprandial glucose area under the curve AUC(0-2 h) by approximately 20% (+0.11 +/- 0.50, -2.08 +/- 0.51, -1.73 +/- 0.49 and -1.88 +/- 0.48 mmol/l x h, respectively, for placebo and 100, 200 and 400 mg (p = 0.002, 0.008 and 0.004 vs. placebo). Postprandial AUC(0-2 h) of intact glucagon-like peptide-1, the principal mediator of the biological effects of DPP4 inhibitors, was increased by 3.90 +/- 2.83, 11.63 +/- 2.86, 16.42 +/- 2.72 and 15.75 +/- 2.71 pmol/l x h, respectively, for placebo and 100, 200 and 400 mg (p = 0.053, 0.001 and 0.002 vs. placebo). Mean HbA(1c) was lower in all dose groups; the placebo-corrected change in the groups receiving 400 mg PHX1149 was -0.28% (p = 0.02). DPP4 inhibition on day 28 was 53, 73 and 78% at 24 h postdose in the groups receiving 100, 200 and 400 mg PHX1149, respectively. There were no differences in adverse events between PHX1149-treated and placebo subjects. CONCLUSIONS: Addition of the DPP4 inhibitor PHX1149 to a stable regimen of metformin or metformin plus a glitazone in patients with type 2 diabetes was well tolerated and improved blood glucose control.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Thiazolidinediones/therapeutic use , Administration, Oral , Adult , Aged , Area Under Curve , Biomarkers/blood , Blood Glucose/analysis , Diabetes Mellitus, Type 2/blood , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Dose-Response Relationship, Drug , Double-Blind Method , Drug Administration Schedule , Drug Therapy, Combination , Female , Glycated Hemoglobin/analysis , Humans , Male , Middle Aged , Postprandial Period , Treatment Outcome
3.
Leukemia ; 20(11): 2008-14, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16990784

ABSTRACT

Somatic mutations of FLT3 resulting in constitutive kinase activation are the most common acquired genomic abnormality found in acute myeloid leukemia (AML). The majority of these mutations are internal tandem duplications (ITD) of the juxtamembrane region (JM). In addition, a minority of cases of AML are associated with mutation of the FLT3 activation loop (AL), typically involving codons D835 and/or I836. We hypothesized that other novel mutations of FLT3 could also contribute to leukemogenesis. We genotyped 109 cases of AML and identified two novel gain-of-function mutations. The first mutation, N841 H, is similar to previously described mutations involving amino-acid substitutions of codon 841. The other novel mutation, FLT3 K663Q, is the first AML-associated gain-of-function mutation located outside the JM and AL domains. Of note, this mutation was potently inhibited by Sunitinib (SU11248), a previously described FLT3 kinase inhibitor. Sunitinib reduced the proliferation and induced apoptosis of transformed Ba/F3 cells expressing FLT3 K663Q. The potency of Sunitinib against FLT3 K663Q was similar to its potency against FLT3 ITD mutations. We conclude that FLT3 mutations in AML can involve novel regions of the TK1. Future studies are needed to define the incidence and prognostic significance of FLT3 mutations outside the well-established JM and AL regions.


Subject(s)
Antineoplastic Agents/pharmacology , Indoles/pharmacology , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/genetics , Pyrroles/pharmacology , fms-Like Tyrosine Kinase 3/genetics , Acute Disease , Amino Acid Sequence , Amino Acid Substitution , Animals , Apoptosis/drug effects , Base Sequence , Cell Division/drug effects , Cell Line, Tumor , Humans , Leukemia, Myeloid/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mutagenesis, Site-Directed , Neoplasm Transplantation , Point Mutation , Sunitinib , fms-Like Tyrosine Kinase 3/metabolism
4.
Clin Cancer Res ; 7(12): 4230-8, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11751524

ABSTRACT

The epidermal growth factor receptor (EGFR) and HER-2 tyrosine kinases have been implicated in the development, progression, and severity of several human cancers and are attractive targets for therapeutic intervention. SU11925 was developed as a small molecule inhibitor of the tyrosine kinase activity of both EGFR and HER-2. In cellular assays, SU11925 exhibited similar potency against EGFR and HER-2, inhibiting EGF-stimulated EGFR autophosphorylation in A431 (human epidermoid carcinoma) cells with an IC(50) of 30 nM and HER-2 phosphorylation in SK-OV-3TP5 (human ovarian carcinoma) cells with an IC(50) of 38 nM. In contrast to its similar activity against the two targets in cellular assays, approximately 10-fold higher plasma concentrations of SU11925 were required to inhibit HER-2 phosphorylation in HER-2-overexpressing tumors compared with EGFR phosphorylation in EGFR-overexpressing tumors in vivo. Consistent with the proposed mechanism of action of this inhibitor, SU11925 inhibited the s.c. growth of EGFR- and HER-2-dependent tumors in athymic mice at doses that produced substantial inhibition of target receptor phosphorylation in vivo. An unexpected finding from these studies was that higher plasma concentrations of SU11925 were required to inhibit EGFR phosphorylation in vivo in tumors that also express high levels of HER-2 than in tumors that express EGFR alone. This observation, which suggests that it is more difficult to inhibit EGFR phosphorylation in vivo in cells that express high levels of HER-2, was confirmed with ZD1839 (Iressa), a selective EGFR inhibitor that also targets the tyrosine kinase catalytic site. The potential clinical implications of this observation are discussed.


Subject(s)
ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/physiology , Genes, erbB-2 , Piperazines/pharmacology , Piperazines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Quinazolines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Squamous Cell , ErbB Receptors/antagonists & inhibitors , Female , Gefitinib , Humans , Mice , Mice, Nude , Ovarian Neoplasms , Phosphorylation , Receptor, ErbB-2 , Transplantation, Heterologous , Tumor Cells, Cultured
5.
J Acquir Immune Defic Syndr ; 27(5): 450-8, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11511821

ABSTRACT

OBJECTIVE: To determine whether genotypic changes in HIV-1 (HIV) reverse transcriptase (RT) occur during adefovir dipivoxil (ADV) therapy that may alter the susceptibility of HIV to adefovir or the related nucleotide inhibitor, tenofovir. DESIGN AND METHODS: GS-96-408 was a 1:1 randomized, double-blind, phase III clinical trial assessing the safety and efficacy of 120-mg daily ADV compared with placebo for the treatment of HIV when added to stable background antiretroviral therapy (ART). Of 442 patients enrolled, 142 were prospectively selected for a virology substudy. Baseline and posttreatment (weeks 24-48) plasma samples were genotypically analyzed in HIV RT. HIV from ADV-treated patients who developed RT mutations at week 24 were also phenotypically analyzed. RESULTS: Nucleoside-associated RT mutations arose with similar frequency among the ADV-and placebo-treated patients, 32% (n = 23) and 28% (n = 20), respectively, during the 24-week blinded treatment phase. RT mutations previously selected by adefovir in vitro (K70E or K65R) did not develop in any patient. Most mutations were typical zidovudine (ZDV)-resistance mutations (e.g., M41L, D67N, K70R, T215Y) in patients taking ZDV or stavudine (d4T) concomitantly, demonstrating directly in the placebo arm that d4T is able to select for these mutations. There appeared to be more patients developing D67N and K70R mutations in the ADV arm versus more T215Y mutations in the placebo arm. Between weeks 24 and 48, 19 of 50 patients (38%) in the ADV arm developed similar RT mutations. The mean HIV RNA responses at weeks 24 and 48 among the ADV-treated patients developing RT mutations were -0.68 log(10) copies/ml (n = 23) and -0.52 log(10) copies/ml (n = 19), respectively, similar to the overall week-24 and week-48 responses (-0.53 and 0.48 log(10) copies/ml, respectively). Patient-derived HIV expressing the observed RT mutations showed insignificant decreases in adefovir susceptibility compared with wild-type in 12 of 16 cases (< threefold). HIV from 1 patient showed significantly reduced susceptibility to tenofovir, which was in association with a double insertion mutation after codon 69 that was also present at baseline. CONCLUSIONS: HIV RT changes that arose during ADV therapy appear attributable to the patient's background ART. ADV therapy may have influenced the pattern of ZDV-associated resistance mutations that developed, but this did not result in an observed loss of viral load suppression. There was a trend toward decreased phenotypic susceptibility to adefovir in ADV-treated patients, with 4 of 16 analyzed patients showing mild, but significantly decreased susceptibility associated with the additional ZDV-associated mutations. Decreased susceptibility to the related nucleotide analog, tenofovir, was not observed to develop in ADV-treated patients.


Subject(s)
Adenine/analogs & derivatives , Adenine/pharmacology , HIV Infections/drug therapy , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , Organophosphonates , Organophosphorus Compounds/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Adenine/therapeutic use , Anti-HIV Agents/pharmacology , Anti-HIV Agents/therapeutic use , Double-Blind Method , Genes, Viral , Genotype , HIV Infections/virology , HIV Reverse Transcriptase/metabolism , HIV-1/enzymology , HIV-1/genetics , HIV-1/physiology , Humans , Microbial Sensitivity Tests , Mutation , Phenotype , Reverse Transcriptase Inhibitors/therapeutic use , Tenofovir
6.
Blood ; 97(5): 1413-21, 2001 Mar 01.
Article in English | MEDLINE | ID: mdl-11222388

ABSTRACT

SU5416 and SU6668 are potent antiangiogenic small-molecule inhibitors of receptor tyrosine kinases, including those of the vascular endothelial growth factor and platelet-derived growth factor receptor families. The stem cell factor (SCF) receptor, c-kit, is structurally related to these receptors and, although not expressed on mature peripheral blood cells, is expressed in leukemic blasts derived from 60% to 80% of acute myeloid leukemia (AML) patients. The c-kit kinase inhibitory activity of SU5416 and SU6668 was evaluated in MO7E cells, a human myeloid leukemia cell line. Tyrosine autophosphorylation of the receptor, induced by SCF, was inhibited in these cells by SU5416 and SU6668 in a dose-dependent manner (inhibitory concentration of 50% [IC(50)] 0.1-1 microM). Inhibition of extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation, a signaling event downstream of c-kit activation, was also inhibited in a dose-dependent manner. Both compounds also inhibited SCF-induced proliferation of MO7E cells (IC(50) 0.1 microM for SU5416; 0.29 microM for SU6668). Furthermore, both SU5416 and SU6668 induced apoptosis in a dose- and time-dependent manner as measured by the increase in activated caspase-3 and the enhanced cleavage of its substrate poly(ADP-ribose) polymerase. These findings with MO7E cells were extended to leukemic blasts from c-kit(+) patients. In patient blasts, both SU5416 and SU6668 inhibited SCF-induced phosphorylation of c-kit and ERK1/2 and induced apoptosis. These studies indicate that SU5416 and SU6668 inhibit biologic functions of c-kit in addition to exhibiting antiangiogenic properties and suggest that the combination of these activities may provide a novel therapeutic approach for the treatment of AML.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Leukemia, Myeloid/pathology , Proto-Oncogene Proteins c-kit/drug effects , Apoptosis/drug effects , Caspase 3 , Caspases/drug effects , Caspases/metabolism , Cell Division/drug effects , Dose-Response Relationship, Drug , Humans , Indoles/pharmacology , Leukemia, Myeloid/drug therapy , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Oxindoles , Phosphorylation/drug effects , Propionates , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrroles/pharmacology , Stem Cell Factor/pharmacology , Tumor Cells, Cultured/drug effects
7.
Clin Cancer Res ; 6(7): 2931-40, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10914743

ABSTRACT

SU101 (leflunomide, N-[4-(trifluoromethyl)-phenyl] 5-methylisoxazole-4-carboxamide), an inhibitor of platelet-derived growth factor receptor signaling, has shown promising clinical activity in Phase I and II studies. Currently, SU101 in combination with cytotoxic agents is in late-stage clinical development for the treatment of cancers. In previous reports, efficacy in vivo versus varied tumor xenografts was observed. As part of the preclinical development of SU101 as a cancer therapy, the combination of SU101 with cytotoxic agents was studied in athymic mice bearing small, established, s.c. human tumor cell xenografts of glioblastoma (SF763T cells), lung (Calu-6 cells), or head and neck (KB cells) origin. In the SF763T model, the combination of SU101 with carmustine resulted in a statistically significant growth inhibition of 74% compared with the vehicle control; this combination was more effective than either agent alone. In the Calu-6 model, the combination of SU101, cisplatin, and etoposide resulted in a growth inhibition of 75% that was statistically greater than that of the vehicle-treated control group and groups treated with one or two agents. In the KB model, the combination of SU101, 5-fluorouracil, and cisplatin resulted in a statistically significant growth inhibition of 69% compared with the vehicle control. Treatment with one or two agents did not significantly inhibit growth in this model. Importantly, in addition to enhanced efficacy resulting from combination therapies, the combination treatments tested were well tolerated, as evidenced by lack of mortality. These data suggest that SU101 in combination with cytotoxic agents may provide clinical benefit and warrant further clinical investigation.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Glioblastoma/drug therapy , Head and Neck Neoplasms/drug therapy , Isoxazoles/therapeutic use , Lung Neoplasms/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/toxicity , Antineoplastic Combined Chemotherapy Protocols/toxicity , Carmustine/administration & dosage , Carmustine/toxicity , Cell Division/drug effects , Cell Survival/drug effects , Cisplatin/administration & dosage , Cisplatin/toxicity , Etoposide/administration & dosage , Etoposide/toxicity , Humans , Isoxazoles/administration & dosage , Isoxazoles/toxicity , Leflunomide , Mice , Mice, Nude , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Anticancer Drug Des ; 15(1): 29-41, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10888034

ABSTRACT

Angiogenesis, or the sprouting of new blood vessels, is a central process in the growth of solid tumors. For many cancers, the extent of vascularization of a tumor is a negative prognostic indicator signifying aggressive disease and increased potential for metastasis. Recent efforts to understand the molecular basis of tumor-associated angiogenesis have identified several potential therapeutic targets, including the receptor tyrosine kinases for the angiogenic factor vascular endothelial growth factor (VEGF). Here we review the approach taken at SUGEN, Inc. to discover and develop small molecule inhibitors of receptor tyrosine kinases as anti-angiogenic agents. We focus on SU5416, a selective inhibitor of VEGF receptors that is currently in clinical development for the treatment of advanced malignancies. Its biochemical, biological and pharmacological properties are reviewed and clinical implications discussed.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Indoles/pharmacology , Pyrroles/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Growth Factor/antagonists & inhibitors , Animals , Humans , Receptors, Vascular Endothelial Growth Factor
9.
Adv Cancer Res ; 79: 1-38, 2000.
Article in English | MEDLINE | ID: mdl-10818676

ABSTRACT

Angiogenesis, the sprouting of new blood vessels, plays a role in diverse disease states including cancer, diabetic retinopathy, age-related macular degeneration, rheumatoid arthritis, psoriasis, atherosclerosis, and restenosis. With regard to cancer, the clinical association of tumor vascularity with tumor aggressiveness has been clearly demonstrated in numerous tumor types. The observation of increased microvessel density in tumors not only serves as an independent prognostic indicator, but also suggests that anti-angiogenic therapy may be an important component of treatment regimens for cancer patients. The complexity of the angiogenic process, which involves both positive and negative regulators, provides a number of targets for therapy. Many positive regulators, including growth factor receptors, matrix metalloproteinases, and integrins, have been correlated with increased vascularity of tumors and poor prognosis for patient survival. Thus, these serve as ideal targets for anti-angiogenesis therapy. Many inhibitors of these targets are currently undergoing clinical evaluation as potential anti-cancer agents. In this article, we discuss the role of positive regulators in angiogenesis and tumor growth and describe the anti-angiogenic agents under development.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/therapy , Neovascularization, Pathologic/drug therapy , Angiogenesis Inhibitors/pharmacology , Animals , Carrier Proteins/physiology , Clinical Trials as Topic , Cytokines/physiology , Drug Design , Extracellular Matrix Proteins/metabolism , Fibrinolysin/physiology , Growth Substances/physiology , Humans , Integrins/antagonists & inhibitors , Integrins/physiology , Metalloendopeptidases/physiology , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Proteins/physiology , Neoplasms/blood supply , Plasminogen Activators/physiology , Protease Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/physiology , Receptors, Growth Factor/drug effects , Receptors, Growth Factor/physiology , Thymidine Phosphorylase/physiology
10.
J Virol ; 74(4): 1767-74, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10644348

ABSTRACT

Simian immunodeficiency virus (SIV) infection of newborn macaques is a useful animal model of human pediatric AIDS to study pathogenesis and to develop intervention strategies aimed at preventing infection or delaying disease progression. In previous studies, we demonstrated that 9-¿2-(R)-(phosphonomethoxy)propyladenine (PMPA; tenofovir) was highly effective in protecting newborn macaques against infection with virulent wild-type (i.e., drug-susceptible) SIVmac251. In the present study, we determined how reduced drug susceptibility of the virus inoculum affects the chemoprophylactic success. SIVmac055 is a virulent isolate that has a fivefold-reduced in vitro susceptibility to PMPA, associated with a K65R mutation and additional amino acid changes (N69T, R82K, A158S, S211N) in reverse transcriptase (RT). Eight newborn macaques were inoculated orally with SIVmac055. The three untreated control animals became SIVmac055 infected; these animals had persistently high viremia and developed fatal immunodeficiency within 3 months. Five animals were treated once daily with PMPA (at 30 mg/kg of body weight) for 4 weeks, starting 24 h prior to oral SIVmac055 inoculation. Two of the five PMPA-treated animals had no evidence of infection. The other three PMPA-treated infant macaques became infected but had a delayed viremia, enhanced antiviral antibody responses, and a slower disease course (AIDS in 5 to 15 months). No reversion to wild-type susceptibility or loss of the K65R mutation was detected in virus isolates from any of the PMPA-treated or untreated SIVmac055-infected animals. Several additional amino acid changes developed in RT, but they were not exclusively associated with PMPA therapy. The results of this study suggest that prophylactic administration of PMPA to human newborns and to adults following exposure to human immunodeficiency virus will still be beneficial even in the presence of viral variants with reduced susceptibility to PMPA.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/pharmacology , Organophosphonates , Organophosphorus Compounds/pharmacology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Adenine/administration & dosage , Adenine/pharmacology , Administration, Oral , Animals , Animals, Newborn , Antiviral Agents/administration & dosage , Macaca mulatta , Organophosphorus Compounds/administration & dosage , Research Design , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/drug effects , Tenofovir
11.
Clin Cancer Res ; 6(12): 4848-58, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11156244

ABSTRACT

SU5416, a selective inhibitor of the tyrosine kinase activity of the vascular endothelial growth factor (VEGF) receptor Flk-1/KDR, is currently in Phase III clinical trials for the treatment of advanced malignancies. In cellular assays, SU5416 inhibits the VEGF-dependent mitogenic/proliferative response of human umbilical vein endothelial cells (HUVECs). In tumor xenograft models, SU5416 inhibits the growth of tumors from a variety of origins by inhibiting tumor angiogenesis. In three different human tumor xenograft models, infrequent (once or twice a week) administration of SU5416 is efficacious despite the fact that it has a short plasma half-life (30 min), which suggests that SU5416 has long-lasting inhibitory activity in vivo. The goal of the present study was to determine the basis for the prolonged activity of SU5416. The results indicate that a short (3 h) exposure to 5 microM SU5416 (to mimic plasma levels of the compound as measured in patients who were receiving SU5416 therapy) produced long-lasting (at least 72 h) inhibition of the VEGF-dependent proliferation of HUVECs in culture, which indicate that SU5416 has long-lasting inhibitory activity in vitro as well as in vivo. SU5416 treatment of HUVECs did not affect surface expression of Flk-1/KDR or the affinity of the receptor for VEGF. Instead, the durability of the in vitro activity of SU5416 was shown to be attributable to its long-lasting ability to specifically inhibit VEGF-dependent phosphorylation of Flk-1/KDR and subsequent downstream signaling, although SU5416 is not an irreversible inhibitor of Flk-1/KDR tyrosine kinase activity. The long-lasting inhibition of cellular responses to VEGF was attributable to the accumulation of SU5416 in cells, as shown using radiolabeled compound, such that inhibitory cellular concentrations of SU5416 are maintained long after the removal of the compound from the medium. The long-lasting inhibitory activity of SU5416 in vitro is consistent with the finding that SU5416 has demonstrated evidence of biological activity in clinical studies when administered twice a week despite a short plasma half-life.


Subject(s)
Angiogenesis Inhibitors/pharmacokinetics , Indoles/pharmacokinetics , Neovascularization, Pathologic , Pyrroles/pharmacokinetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/metabolism , 3T3 Cells , Animals , Cell Division/drug effects , Cell Membrane/metabolism , Cell Separation , Cells, Cultured , Culture Media, Serum-Free/metabolism , Dose-Response Relationship, Drug , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Epitopes , Female , Flow Cytometry , Humans , Kinetics , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Neoplasm Transplantation , Phosphorylation/drug effects , Protein Binding/drug effects , Protein-Tyrosine Kinases/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/biosynthesis , Receptors, Vascular Endothelial Growth Factor , Signal Transduction/drug effects , Time Factors , Tumor Cells, Cultured , Umbilical Cord/cytology , Umbilical Cord/drug effects
12.
JAMA ; 282(24): 2305-12, 1999.
Article in English | MEDLINE | ID: mdl-10612317

ABSTRACT

CONTEXT: Adefovir dipivoxil is a nucleotide analog that has demonstrated effective antiretroviral activity against human immunodeficiency virus (HIV) with once-daily administration. OBJECTIVE: To determine if adefovir confers antiretroviral or immunologic benefit when added to stable antiretroviral therapy. DESIGN: Multicenter, 24-week, randomized, double-blind, placebo-controlled study. Enrollment was conducted from June 3, 1996, through May 6, 1997. SETTING: Thirty-three US HIV treatment centers. PARTICIPANTS: Of 1171 patients screened, 442 patients infected with HIV receiving stable antiretroviral therapy for at least 8 weeks with plasma HIV RNA greater than 2500 copies/mL and CD4+ cell count above 0.20 x 10(9)/L were randomized. INTERVENTION: Patients were randomized to receive either a single 120-mg/d dose of adefovir dipivoxil (n = 219) or an indistinguishable placebo (n = 223). All patients received L-carnitine, 500 mg/d. Open-label adefovir was offered after 24 weeks and was continued until the end of the study. MAIN OUTCOME MEASURES: Changes in HIV RNA from baseline, based on area under the curve and CD4+ cell levels, adverse events, and effect of baseline genotypic resistance on response to adefovir. RESULTS: Patients assigned to adefovir demonstrated a 0.4-log10 decline from baseline in HIV RNA compared with no change in the placebo group (P<.001), which continued through 48 weeks. CD4+ cell counts did not change. During the initial 24 weeks, elevated hepatic enzyme levels (P<.001), gastrointestinal tract complaints (P<.001), and weight loss (P<.001) were associated with use of adefovir. Between 24 weeks and 48 weeks elevations in serum creatinine occurred in 60% of patients, usually returning to baseline after discontinuation of adefovir. Patients with lamivudine or lamivudine and zidovudine resistance mutations demonstrated anti-HIV effects with adefovir (P< or =.01 vs placebo group). CONCLUSIONS: This study suggests that once-daily adefovir therapy reduces HIV RNA and is active against isolates resistant to lamivudine or lamivudine and zidovudine. Nephrotoxicity occurred when treatment extended beyond 24 weeks but was reversible.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/therapeutic use , HIV Infections/drug therapy , Organophosphonates , Reverse Transcriptase Inhibitors/therapeutic use , Adenine/therapeutic use , Adult , Anti-HIV Agents/pharmacology , CD4 Lymphocyte Count , Double-Blind Method , Drug Resistance, Microbial , Drug Therapy, Combination , Female , Genotype , HIV Infections/immunology , HIV Infections/virology , HIV-1/drug effects , HIV-1/genetics , Humans , Lamivudine/pharmacology , Male , RNA, Viral , Viral Load , Zidovudine/pharmacology
14.
J Med Virol ; 58(4): 402-7, 1999 Aug.
Article in English | MEDLINE | ID: mdl-10421408

ABSTRACT

Five AIDS patients with cytomegalovirus (CMV) retinitis who had received ganciclovir (GCV) therapy were followed with serial blood sampling to detectchanges both in CMV load and in the genetic composition of genes UL97 and UL54 whilst receiving cidofovir (CDV) therapy. CDV neither reduced CMV load in blood nor prevented its quantitative resurgence during therapy. These effects were not explained by the initial presence or development of CDV-associated drug resistance mutations in UL54. In two patients, UL97 genotypic resistance to GCV involving either a L595S mutation or a deletion of amino acids 590-603 were present at the initiation of CDV and, in both patients, repopulation of CMV strains with wild-type UL97 sequences occurred during CDV therapy. These data are consistent with GCV-resistant strains containing UL97 mutations being less fit than their wild-type counterparts and so being able to persist only with the selective pressure of GCV.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Antiviral Agents/therapeutic use , Cytomegalovirus/genetics , DNA, Viral/blood , DNA-Directed DNA Polymerase/genetics , Organophosphonates , Phosphotransferases (Alcohol Group Acceptor)/genetics , Viral Proteins , Acquired Immunodeficiency Syndrome/complications , Cidofovir , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/virology , Cytosine/analogs & derivatives , Cytosine/therapeutic use , DNA, Viral/genetics , Ganciclovir/therapeutic use , Genotype , Humans , Mutation , Organophosphorus Compounds/therapeutic use , Prospective Studies , Retinitis/complications , Retinitis/pathology , Retinitis/virology , Viral Load
15.
Antimicrob Agents Chemother ; 43(4): 802-12, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10103184

ABSTRACT

Simian immunodeficiency virus (SIV) infection of newborn rhesus macaques is a useful animal model of human immunodeficiency virus infection for the study of the emergence and clinical implications of drug-resistant viral mutants. We previously demonstrated that SIV-infected infant macaques receiving prolonged treatment with 9-[2-(phosphonomethoxy)propyl]adenine (PMPA) developed viral mutants with fivefold reduced susceptibility to PMPA in vitro and that the development of these mutants was associated with the development of a K65R mutation and additional compensatory mutations in reverse transcriptase (RT). To study directly the virulence and clinical implications of these SIV mutants, two uncloned SIVmac isolates with similar fivefold reduced in vitro susceptibilities to PMPA but distinct RT genotypes, SIVmac055 (K65R, N69T, R82K A158S,S211N) and SIVmac385 (K65R, N69S, I118V), were each inoculated intravenously into six newborn rhesus macaques; 3 weeks later, three animals of each group were started on PMPA treatment. All six untreated animals developed persistently high levels of viremia and fatal immunodeficiency within 4 months. In contrast, the six PMPA-treated animals, despite having persistently high virus levels, survived significantly longer: 5 to 9 months for the three SIVmac055-infected infants and > or = 21 months for the three SIVmac385-infected infants. Virus from only one untreated animal demonstrated reversion to wild-type susceptibility and loss of the K65R mutation. In several other animals, additional RT mutations, including K64R and Y115F, were detected, but the biological role of these mutations is unclear since they did not affect the in vitro susceptibility of the virus to PMPA. In conclusion, this study demonstrates that although SIVmac mutants with the PMPA-selected K65R mutation in RT were highly virulent, PMPA treatment still offered strong therapeutic benefits. These results suggest that the potential emergence of HIV mutants with reduced susceptibility to PMPA in patients during prolonged PMPA therapy may not eliminate its therapeutic benefits.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/therapeutic use , Organophosphonates , Organophosphorus Compounds/therapeutic use , Simian Acquired Immunodeficiency Syndrome/drug therapy , Adenine/administration & dosage , Adenine/therapeutic use , Animals , Animals, Newborn , Antiviral Agents/administration & dosage , Macaca , Mutation , Organophosphorus Compounds/administration & dosage , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/pathogenicity , Survival Analysis , Tenofovir
16.
Clin Infect Dis ; 28(3): 528-33, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10194072

ABSTRACT

Ganciclovir and cidofovir, two antiviral agents used in the treatment of cytomegalovirus (CMV) retinitis, have a synergistic effect inhibiting CMV replication in vitro. In a phase I study, seven patients with AIDS-related CMV retinitis were treated with cidofovir (5 mg/kg intravenously every 2 weeks) combined with ganciclovir (1 g orally three times a day). During a median of 5.5 months (range, 1-12 months) of combined therapy, only one patient had retinitis progression, and only two of 28 blood cultures (specimens of which were obtained on a monthly basis) yielded CMV. Dose-limiting adverse ocular effects (anterior uveitis [two patients] and hypotony [two patients]) occurred in three of seven patients. The results suggest that combination therapy with intravenous cidofovir and oral ganciclovir (a regimen that does not require indwelling central venous catheter access) might enhance clinical efficacy. Less frequent administration of cidofovir in combination with oral ganciclovir should be prospectively studied to determine if the incidence of treatment-associated toxicity might be reduced.


Subject(s)
AIDS-Related Opportunistic Infections/drug therapy , Antiviral Agents/therapeutic use , Cytomegalovirus Retinitis/drug therapy , Cytosine/analogs & derivatives , Ganciclovir/therapeutic use , Organophosphonates , Organophosphorus Compounds/therapeutic use , Adult , Antiviral Agents/adverse effects , Cidofovir , Cytosine/adverse effects , Cytosine/therapeutic use , Drug Therapy, Combination , Female , Ganciclovir/adverse effects , Humans , Male , Middle Aged , Organophosphorus Compounds/adverse effects , Treatment Outcome
17.
J Virol ; 73(4): 2947-55, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10074144

ABSTRACT

Simian immunodeficiency virus (SIV) infection of newborn macaques is a useful animal model of human pediatric AIDS to study disease pathogenesis and to develop intervention strategies aimed at delaying disease. In the present study, we demonstrate that very early events of infection greatly determine the ultimate disease course, as short-term antiviral drug administration during the initial viremia stage significantly delayed the onset of AIDS. Fourteen newborn macaques were inoculated orally with uncloned, highly virulent SIVmac251. The four untreated control animals showed persistently high virus levels and poor antiviral immune responses; they developed fatal immunodeficiency within 15 weeks. In contrast, SIV-infected newborn macaques which were started on 9-[2-(R)-(phosphonomethoxy)propyl]adenine (PMPA) treatment at 5 days of age and continued for either 14 or 60 days showed reduced virus levels and enhanced antiviral immune responses. This short-term PMPA treatment did not induce detectable emergence of SIV mutants with reduced in vitro susceptibility to PMPA. Although viremia increased in most animals after PMPA treatment was withdrawn, all animals remained disease-free for at least 6 months. Our data suggest that short-term treatment with a potent antiviral drug regimen during the initial viremia will significantly prolong AIDS-free survival for HIV-infected infants and adults.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/administration & dosage , Organophosphonates , Organophosphorus Compounds/administration & dosage , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Immunodeficiency Virus/isolation & purification , Adenine/administration & dosage , Animals , Animals, Newborn , Humans , Injections, Subcutaneous , Macaca mulatta , Tenofovir , Time Factors
18.
J Infect Dis ; 179(1): 92-100, 1999 Jan.
Article in English | MEDLINE | ID: mdl-9841827

ABSTRACT

In a phase II study of 6-12 months of adefovir dipivoxil treatment in human immunodeficiency virus (HIV)-infected patients, HIV from 8 of 29 patients developed mutations in reverse transcriptase (RT) potentially attributable to adefovir dipivoxil therapy. Recombinant HIV from pre- and posttreatment plasma samples from these 8 patients showed no change or minor decreases in adefovir susceptibility, consistent with the durable antiviral effect observed. Additionally, HIV from 8 patients developed the M184V RT mutation because of concomitant lamivudine use. Recombinant HIV pairs from all 4 patients with zidovudine-resistant HIV showed statistically significant increases in adefovir susceptibility of 3- to 4-fold (to near wild type IC50), and HIV pairs from 2 of 4 patients with zidovudine-sensitive HIV showed a 2- to 3-fold increase in susceptibility. In growth kinetics studies, expression of the M184V RT mutation resulted in attenuated viral growth in peripheral blood mononuclear cell cultures. These studies suggest that patients possessing HIV with zidovudine and lamivudine resistance mutations may benefit from adefovir dipivoxil therapy.


Subject(s)
Adenine/analogs & derivatives , HIV Infections/drug therapy , HIV Infections/virology , HIV Reverse Transcriptase/genetics , HIV-1/drug effects , HIV-1/genetics , Lamivudine/therapeutic use , Organophosphonates , Point Mutation , Reverse Transcriptase Inhibitors/therapeutic use , Adenine/administration & dosage , Adenine/therapeutic use , Drug Resistance, Microbial/genetics , Drug Therapy, Combination , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/physiology , Humans , In Vitro Techniques , Lamivudine/administration & dosage , Reverse Transcriptase Inhibitors/administration & dosage , Virus Replication/drug effects , Virus Replication/genetics , Zidovudine/therapeutic use
19.
Antivir Ther ; 4(2): 87-94, 1999.
Article in English | MEDLINE | ID: mdl-10682153

ABSTRACT

9-(2-phosphonomethoxypropyl)adenine (PMPA) has demonstrated remarkable anti-simian immunodeficiency virus (SIV) activity in macaque models of SIV infection and transmission prevention. Recently, PMPA and its oral prodrug, bis-POC PMPA, have also shown potent anti-human immunodeficiency virus type 1 (HIV-1) activity in Phase I clinical studies. In vitro experiments were performed to address the resistance properties of PMPA. After eight passages in increasing concentrations of PMPA, HIV-1IIIB was able to grow in the presence of 2 microM PMPA, fivefold above the IC50 of PMPA for wild-type parental virus. Sequence analysis of the reverse transcriptase (RT) genes from four of 15 RT clones demonstrated the presence of a K65R substitution in RT and recombinant HIV expressing the K65R RT mutation showed a threefold to fourfold increase in IC50 value for PMPA as compared to wild-type. Additional experiments demonstrated that viruses expressing other nucleoside-associated RT resistance mutations all showed wild-type or < threefold reduced susceptibility to PMPA in vitro. Interestingly, lamivudine-resistant viruses expressing the M184V RT mutation showed wild-type to slightly increased susceptibility to PMPA in vitro and addition of the M184V mutation to HIV with the K65R mutation resulted in reversion to wild-type susceptibility for PMPA. In agreement with the cell culture findings, Escherichia coli-expressed K65R RT showed fivefold reduced susceptibility to PMPA diphosphate, the active moiety of PMPA. Furthermore, in combination experiments, PMPA with hydroxyurea showed synergistic inhibition of HIV replication in vitro. The potent antiretroviral activity and favourable resistance profile of PMPA and bis-POC PMPA are being further investigated in ongoing clinical trials.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/pharmacology , HIV-1/drug effects , Organophosphonates , Organophosphorus Compounds/pharmacology , Adenine/pharmacology , Drug Resistance , HIV Reverse Transcriptase/genetics , Hydroxyurea/pharmacology , Mutation , Tenofovir
20.
J Infect Dis ; 178(6): 1821-5, 1998 Dec.
Article in English | MEDLINE | ID: mdl-9815243

ABSTRACT

Blood culture isolates from patients receiving first- (peripheral retinitis) or second-line (relapsing retinitis) therapy with intravenous cidofovir were obtained from three clinical trials for in vitro antiviral susceptibility analyses. Isolates from 6 patients obtained after 14.3 weeks (mean) of first-line cidofovir therapy showed complete susceptibility to cidofovir, ganciclovir, and foscarnet. Isolates from 20 patients were obtained after 17.3 weeks (mean) of second-line cidofovir therapy. Ten showed complete susceptibility to all inhibitors, 3 showed low-level ganciclovir resistance (<6-fold) but were sensitive to cidofovir and foscarnet, and 7 showed moderately reduced susceptibility (<8-fold) to cidofovir and high-level resistance (8- to 23-fold) to ganciclovir in vitro. Four of these 7 isolates showed reduced susceptibility (4-fold) to foscarnet. Notably, there was no difference in time to retinitis progression in patients that were on cidofovir therapy when sensitive isolates were compared with those showing reduced susceptibility to cidofovir in vitro.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cytomegalovirus Retinitis/drug therapy , Cytomegalovirus/drug effects , Cytosine/analogs & derivatives , Organophosphonates , Organophosphorus Compounds/therapeutic use , Cidofovir , Cytomegalovirus/genetics , Cytomegalovirus/isolation & purification , Cytomegalovirus Retinitis/virology , Cytosine/therapeutic use , Drug Resistance, Microbial , Foscarnet/pharmacology , Ganciclovir/pharmacology , Genotype , Humans , Microbial Sensitivity Tests , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL
...