Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
Trends Biotechnol ; 39(3): 298-310, 2021 03.
Article En | MEDLINE | ID: mdl-32807530

Intracellular delivery of therapeutic antibodies is highly desirable but remains a challenge for biomedical research and the pharmaceutical industry. Approximately two-thirds of disease-associated targets are found inside the cell. Difficulty blocking these targets with available drugs creates a need for technology to deliver highly specific therapeutic antibodies intracellularly. Historically, antibodies have not been believed to traverse the cell membrane and neutralize intracellular targets. Emerging evidence has revealed that anti-DNA autoantibodies found in systemic lupus erythematosus (SLE) patients can penetrate inside the cell. Harnessing this technology has the potential to accelerate the development of drugs against intracellular targets. Here, we dissect the mechanisms of the intracellular localization of SLE antibodies and discuss how to apply these insights to engineer successful cell-penetrating antibody drugs.


Antibodies, Antinuclear , Biological Products , Drug Delivery Systems , Lupus Erythematosus, Systemic , Antibodies, Antinuclear/administration & dosage , Antibodies, Antinuclear/chemistry , Antibodies, Antinuclear/metabolism , Biological Products/administration & dosage , Humans , Intracellular Space , Lupus Erythematosus, Systemic/drug therapy
2.
Mol Metab ; 10: 87-99, 2018 04.
Article En | MEDLINE | ID: mdl-29453154

OBJECTIVE: Insulin resistance is a key feature of Type 2 Diabetes (T2D), and improving insulin sensitivity is important for disease management. Allosteric modulation of the insulin receptor (IR) with monoclonal antibodies (mAbs) can enhance insulin sensitivity and restore glycemic control in animal models of T2D. METHODS: A novel human mAb, IRAB-A, was identified by phage screening using competition binding and surface plasmon resonance assays with the IR extracellular domain. Cell based assays demonstrated agonist and sensitizer effects of IRAB-A on IR and Akt phosphorylation, as well as glucose uptake. Lean and diet-induced obese mice were used to characterize single-dose in vivo pharmacological effects of IRAB-A; multiple-dose IRAB-A effects were tested in obese mice. RESULTS: In vitro studies indicate that IRAB-A exhibits sensitizer and agonist properties distinct from insulin on the IR and is translated to downstream signaling and function; IRAB-A bound specifically and allosterically to the IR and stabilized insulin binding. A single dose of IRAB-A given to lean mice rapidly reduced fed blood glucose for approximately 2 weeks, with concomitant reduced insulin levels suggesting improved insulin sensitivity. Phosphorylated IR (pIR) from skeletal muscle and liver were increased by IRAB-A; however, phosphorylated Akt (pAkt) levels were only elevated in skeletal muscle and not liver vs. control; immunochemistry analysis (IHC) confirmed the long-lived persistence of IRAB-A in skeletal muscle and liver. Studies in diet-induced obese (DIO) mice with IRAB-A reduced fed blood glucose and insulinemia yet impaired glucose tolerance and led to protracted insulinemia during a meal challenge. CONCLUSION: Collectively, the data suggest IRAB-A acts allosterically on the insulin receptor acting non-competitively with insulin to both activate the receptor and enhance insulin signaling. While IRAB-A produced a decrease in blood glucose in lean mice, the data in DIO mice indicated an exacerbation of insulin resistance; these data were unexpected and suggested the interplay of complex unknown pharmacology. Taken together, this work suggests that IRAB-A may be an important tool to explore insulin receptor signaling and pharmacology.


Allosteric Site , Antibodies, Monoclonal/pharmacology , Hypoglycemic Agents/pharmacology , Receptor, Insulin/agonists , 3T3 Cells , Allosteric Regulation , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Blood Glucose/metabolism , Cell Line, Tumor , Humans , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/immunology , Insulin/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle, Skeletal/metabolism , Receptor, Insulin/chemistry , Receptor, Insulin/immunology , Signal Transduction
3.
Diabetes ; 66(1): 206-217, 2017 Jan.
Article En | MEDLINE | ID: mdl-27797911

A hallmark of type 2 diabetes is impaired insulin receptor (IR) signaling that results in dysregulation of glucose homeostasis. Understanding the molecular origins and progression of diabetes and developing therapeutics depend on experimental models of hyperglycemia, hyperinsulinemia, and insulin resistance. We present a novel monoclonal antibody, IRAB-B, that is a specific, potent IR antagonist that creates rapid and long-lasting insulin resistance. IRAB-B binds to the IR with nanomolar affinity and in the presence of insulin efficiently blocks receptor phosphorylation within minutes and is sustained for at least 3 days in vitro. We further confirm that IRAB-B antagonizes downstream signaling and metabolic function. In mice, a single dose of IRAB-B induces rapid onset of hyperglycemia within 6 h, and severe hyperglycemia persists for 2 weeks. IRAB-B hyperglycemia is normalized in mice treated with exendin-4, suggesting that this model can be effectively treated with a GLP-1 receptor agonist. Finally, a comparison of IRAB-B with the IR antagonist S961 shows distinct antagonism in vitro and in vivo. IRAB-B appears to be a powerful tool to generate both acute and chronic insulin resistance in mammalian models to elucidate diabetic pathogenesis and evaluate therapeutics.


Antibodies, Monoclonal/therapeutic use , Insulin Resistance/physiology , Receptor, Insulin/metabolism , Animals , Blotting, Western , Cell Line , Diabetes Mellitus, Type 2 , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Mice , Mice, Inbred C57BL , Peptides/pharmacology , Phosphorylation , Protein Binding , Receptor, Insulin/antagonists & inhibitors , Signal Transduction/drug effects
4.
J Med Chem ; 59(24): 11039-11049, 2016 12 22.
Article En | MEDLINE | ID: mdl-28002958

The discovery of novel 4-hydroxy-2-(heterocyclic)pyrimidine-5-carboxamide inhibitors of hypoxia-inducible factor (HIF) prolyl hydroxylases (PHD) is described. These are potent, selective, orally bioavailable across several species, and active in stimulating erythropoiesis. Mouse and rat studies showed hematological changes with elevations of plasma EPO and circulating reticulocytes following single oral dose administration, while 4-week q.d. po administration in rat elevated hemoglobin levels. A major focus of the optimization process was to decrease the long half-life observed in higher species with early compounds. These efforts led to the identification of 28 (MK-8617), which has advanced to human clinical trials for anemia.


Anemia/drug therapy , Drug Discovery , Enzyme Inhibitors/pharmacology , Hypoxia-Inducible Factor-Proline Dioxygenases/antagonists & inhibitors , Pyridazines/pharmacology , Pyrimidines/pharmacology , Administration, Oral , Anemia/enzymology , Animals , Dose-Response Relationship, Drug , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/chemistry , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mice , Mice, Inbred C57BL , Models, Molecular , Molecular Structure , Pyridazines/administration & dosage , Pyridazines/chemistry , Pyrimidines/administration & dosage , Pyrimidines/chemistry , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship
5.
Cell Signal ; 28(8): 1037-47, 2016 Aug.
Article En | MEDLINE | ID: mdl-27155325

Insulin receptor signaling is a complex cascade leading to a multitude of intracellular functional responses. Three natural ligands, insulin, IGF1 and IGF2, are each capable of binding with different affinities to the insulin receptor, and result in variable biological responses. However, it is likely these affinity differences alone cannot completely explain the myriad of diverse cellular outcomes. Ligand binding initiates activation of a signaling cascade resulting in phosphorylation of the IR itself and other intracellular proteins. The direct catalytic activity along with the temporally coordinated assembly of signaling proteins is critical for insulin receptor signaling. We hypothesized that determining differential phosphorylation among individual tyrosine sites activated by ligand binding or dephosphorylation by phosphatases could provide valuable insight into insulin receptor signaling. Here, we present a sensitive, novel immunoassay adapted from Meso Scale Discovery technology to quantitatively measure changes in site-specific phosphorylation levels on endogenous insulin receptors from HuH7 cells. We identified insulin receptor phosphorylation patterns generated upon differential ligand activation and phosphatase-mediated deactivation. The data demonstrate that insulin, IGF1 and IGF2 elicit different insulin receptor phosphorylation kinetics and potencies that translate to downstream signaling. Furthermore, we show that insulin receptor deactivation, regulated by tyrosine phosphatases, occurs distinctively across specific tyrosine residues. In summary, we present a novel, quantitative and high-throughput assay that has uncovered differential ligand activation and site-specific deactivation of the insulin receptor. These results may help elucidate some of the insulin signaling mechanisms, discriminate ligand activity and contribute to a better understanding of insulin receptor signaling. We propose this methodology as a powerful approach to characterize agonists and antagonists of the insulin receptor and can be adapted to serve as a platform to evaluate ligands of alternate receptor systems.


Phosphoserine/metabolism , Protein Tyrosine Phosphatases/metabolism , Receptor, Insulin/metabolism , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Insulin/pharmacology , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/pharmacology , Kinetics , Ligands , Models, Biological , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Time Factors , Vanadates/pharmacology
6.
PLoS One ; 10(10): e0140694, 2015.
Article En | MEDLINE | ID: mdl-26492563

TIM-3 (T cell immunoglobulin and mucin-domain containing protein 3) is a member of the TIM family of proteins that is preferentially expressed on Th1 polarized CD4+ and CD8+ T cells. Recent studies indicate that TIM-3 serves as a negative regulator of T cell function (i.e. T cell dependent immune responses, proliferation, tolerance, and exhaustion). Despite having no recognizable inhibitory signaling motifs, the intracellular tail of TIM-3 is apparently indispensable for function. Specifically, the conserved residues Y265/Y272 and surrounding amino acids appear to be critical for function. Mechanistically, several studies suggest that TIM-3 can associate with interleukin inducible T cell kinase (ITK), the Src kinases Fyn and Lck, and the p85 phosphatidylinositol 3-kinase (PI3K) adaptor protein to positively or negatively regulate IL-2 production via NF-κB/NFAT signaling pathways. To begin to address this discrepancy, we examined the effect of TIM-3 in two model systems. First, we generated several Jurkat T cell lines stably expressing human TIM-3 or murine CD28-ECD/human TIM-3 intracellular tail chimeras and examined the effects that TIM-3 exerts on T cell Receptor (TCR)-mediated activation, cytokine secretion, promoter activity, and protein kinase association. In this model, our results demonstrate that TIM-3 inhibits several TCR-mediated phenotypes: i) NF-kB/NFAT activation, ii) CD69 expression, and iii) suppression of IL-2 secretion. To confirm our Jurkat cell observations we developed a primary human CD8+ cell system that expresses endogenous levels of TIM-3. Upon TCR ligation, we observed the loss of NFAT reporter activity and IL-2 secretion, and identified the association of Src kinase Lck, and PLC-γ with TIM-3. Taken together, our results support the conclusion that TIM-3 is a negative regulator of TCR-function by attenuating activation signals mediated by CD3/CD28 co-stimulation.


Antigens, CD/metabolism , Interleukin-2/metabolism , Membrane Proteins/metabolism , NFATC Transcription Factors/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Calcium/metabolism , Cell Differentiation , Cells, Cultured , Cytokines/metabolism , Genes, Reporter , Hepatitis A Virus Cellular Receptor 2 , Humans , Lymphocyte Activation/immunology , Membrane Proteins/chemistry , NF-kappa B/metabolism , Promoter Regions, Genetic/genetics , Protein Structure, Tertiary , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
7.
Int J Mol Sci ; 15(10): 17686-704, 2014 Sep 30.
Article En | MEDLINE | ID: mdl-25272226

Non-small cell lung cancer (NSCLC) affects millions of patients each year worldwide. Existing therapies include epidermal growth factor receptor (EGFR) inhibition using small molecules or antibodies with good efficacy. Unfortunately, intrinsic and acquired resistance to EGFR therapy remains a persistent complication for disease treatment. A greater understanding of the role of EGFR in NSCLC etiology is crucial to improving patient outcomes. In this study, the role of EGFR in tumor angiogenesis was examined in H292 NSCLC cells under the pretense that confluent cells would exhibit a more angiogenic and growth-centered phenotype. Indeed, confluent H292 cells potentiated endothelial cell angiogenesis in co-culture models in an EGFR-dependent manner. While confluent H292 cells did not exhibit any change in EGFR protein expression, EGFR localization to the extracellular membrane was increased. EGFR membrane localization coincided with a comparable potentiation of maximal EGFR phosphorylation and was followed by a 3-fold increase in vascular endothelial growth factor A (VEGF-A) production as compared to subconfluent cells. EGFR-mediated VEGF-A production was determined to be dependent on signal transducer and activator of transcription 3 (STAT3) activation and not phosphoinositide 3-kinase (PI3K) signaling. These results identify unique cell density dependent phenotypes within a monoclonal NSCLC cell line and provide a potential mechanism of resistance to anti-EGFR therapy in metastatic NSCLC.


Epidermal Growth Factor/pharmacology , Up-Regulation/drug effects , Vascular Endothelial Growth Factor A/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line , Cell Proliferation/drug effects , Cetuximab , Coculture Techniques , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Neovascularization, Physiologic/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-met/metabolism , RNA, Messenger/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/genetics
8.
J Med Chem ; 55(7): 2945-59, 2012 Apr 12.
Article En | MEDLINE | ID: mdl-22364528

The discovery of 1,3,8-triazaspiro[4.5]decane-2,4-diones (spirohydantoins) as a structural class of pan-inhibitors of the prolyl hydroxylase (PHD) family of enzymes for the treatment of anemia is described. The initial hit class, spirooxindoles, was identified through affinity selection mass spectrometry (AS-MS) and optimized for PHD2 inhibition and optimal PK/PD profile (short-acting PHDi inhibitors). 1,3,8-Triazaspiro[4.5]decane-2,4-diones (spirohydantoins) were optimized as an advanced lead class derived from the original spiroindole hit. A new set of general conditions for C-N coupling, developed using a high-throughput experimentation (HTE) technique, enabled a full SAR analysis of the spirohydantoins. This rapid and directed SAR exploration has resulted in the first reported examples of hydantoin derivatives with good PK in preclinical species. Potassium channel off-target activity (hERG) was successfully eliminated through the systematic introduction of acidic functionality to the molecular structure. Undesired upregulation of alanine aminotransferese (ALT) liver enzymes was mitigated and a robust on-/off-target margin was achieved. Spirohydantoins represent a class of highly efficacious, short-acting PHD1-3 inhibitors causing a robust erythropoietin (EPO) upregulation in vivo in multiple preclinical species. This profile deems spirohydantoins as attractive short-acting PHDi inhibitors with the potential for treatment of anemia.


Anemia/drug therapy , Aza Compounds/chemical synthesis , Hydantoins/chemical synthesis , Hypoxia-Inducible Factor 1/metabolism , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Spiro Compounds/chemical synthesis , Animals , Aza Compounds/pharmacokinetics , Aza Compounds/pharmacology , Dogs , ERG1 Potassium Channel , Erythropoietin/biosynthesis , Ether-A-Go-Go Potassium Channels/metabolism , High-Throughput Screening Assays , Humans , Hydantoins/pharmacokinetics , Hydantoins/pharmacology , Hypoxia-Inducible Factor-Proline Dioxygenases , Indoles/chemical synthesis , Indoles/pharmacokinetics , Indoles/pharmacology , Liver/drug effects , Liver/enzymology , Macaca mulatta , Mass Spectrometry , Mice , Mice, Inbred C57BL , Protein Binding , Rats , Spiro Compounds/pharmacokinetics , Spiro Compounds/pharmacology , Structure-Activity Relationship , Up-Regulation
9.
J Biotechnol ; 157(1): 198-206, 2012 Jan.
Article En | MEDLINE | ID: mdl-22100268

Pichia pastoris is a methylotropic yeast that has gained great importance as an organism for protein expression in recent years. Here, we report the expression of recombinant human erythropoietin (rhEPO) in glycoengineered P. pastoris. We show that glycosylation fidelity is maintained in fermentation volumes spanning six orders of magnitude and that the protein can be purified to high homogeneity. In order to increase the half-life of rhEPO, the purified protein was coupled to polyethylene glycol (PEG) and then compared to the currently marketed erythropoiesis stimulating agent, Aranesp(®) (darbepoetin). In in vitro cell proliferation assays the PEGylated protein was slightly, and the non-PEGylated protein was significantly more active than comparator. Pharmacodynamics as well as pharmacokinetic activity of PEGylated rhEPO in animals was comparable to that of Aranesp(®). Taken together, our results show that glycoengineered P. pastoris is a suitable production host for rhEPO, yielding an active biologic that is comparable to those produced in current mammalian host systems.


Erythropoietin/biosynthesis , Pichia/metabolism , Protein Engineering/methods , Animals , Cell Proliferation/drug effects , Darbepoetin alfa , Erythropoietin/analogs & derivatives , Erythropoietin/blood , Erythropoietin/genetics , Erythropoietin/pharmacokinetics , Erythropoietin/pharmacology , Female , Glycosylation , Humans , Male , Mice , Pichia/genetics , Polyethylene Glycols , Polysaccharides/chemistry , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics
10.
Bioorg Med Chem Lett ; 20(3): 1173-6, 2010 Feb 01.
Article En | MEDLINE | ID: mdl-20022493

A variety of N-linked tertiary amines and heteroarylamines were examined at the 4-position of sulfonylated proline dipeptides in order to improve VLA-4 receptor off-rates and overcome the issue of CYP3A4 time-dependent inhibition of ester prodrugs. A tight-binding inhibitor 5j with a long off-rate provided sustained receptor occupancy despite poor oral pharmacokinetics.


Dipeptides/chemistry , Dipeptides/metabolism , Integrin alpha4beta1/antagonists & inhibitors , Proline/chemistry , Proline/metabolism , Animals , Binding, Competitive/physiology , Dipeptides/pharmacology , Heterocyclic Compounds/chemistry , Heterocyclic Compounds/metabolism , Heterocyclic Compounds/pharmacology , Humans , Integrin alpha4beta1/metabolism , Proline/pharmacology , Protein Binding/physiology , Rats
11.
Bioorg Med Chem Lett ; 19(19): 5803-6, 2009 Oct 01.
Article En | MEDLINE | ID: mdl-19713111

A series of prolyl-N-isonicotinoyl-(L)-4-aminophenylalanine derivatives substituted at the proline 4-position with cyclic amines was evaluated as VLA-4 antagonists. The ring size and presence or absence of fluorine affected potency and receptor occupancy. The analog with 3,3-difluoropiperidine at the proline 4-position (13) was the most potent compound and had very good duration of receptor occupancy in vitro. The ethyl ester prodrug of 13 demonstrated excellent receptor occupancy after oral dosing in rats.


Dipeptides/chemistry , Integrin alpha4beta1/antagonists & inhibitors , Phenylalanine/analogs & derivatives , Prodrugs/chemistry , Administration, Oral , Animals , Dipeptides/administration & dosage , Dipeptides/chemical synthesis , Drug Discovery , Integrin alpha4beta1/metabolism , Phenylalanine/administration & dosage , Phenylalanine/chemical synthesis , Prodrugs/administration & dosage , Prodrugs/chemical synthesis , Rats
12.
Vet Immunol Immunopathol ; 130(1-2): 79-87, 2009 Jul 15.
Article En | MEDLINE | ID: mdl-19250687

The purpose of this study was to characterize the alpha(4)beta(1) receptor (CD49d/CD29, very late antigen-4, VLA-4) on circulating equine leukocytes and to evaluate the intrinsic potency of an alpha(4)beta(1) receptor antagonist (Compound B) in the horse. Ultimately, these studies would allow us to determine the suitability of treating recurrent airway obstruction (RAO; heaves) affected horses by blocking the cellular recruitment of lymphocytes and neutrophils into the lung. The data demonstrates the alpha(4)beta(1) integrin is present on horse lymphocytes and neutrophils (fluorescence-assisted cell sorter, FACS) and can bind low molecular weight alpha(4)beta(1) antagonists (Compounds A and B) with high affinity. K(D) values for the binding of Compound A to non-activated alpha(4)beta(1) on isolated horse PBMCs (peripheral blood mononuclear cells) and activated neutrophils were 17 pM and 27 pM, respectively. Compound B was identified as a suitable antagonist for performing a series of in vivo experiments. Compound B was found to possess excellent potency in horse whole blood, possessing IC(50) and IC(90) values of 39 pM and 172 pM, respectively. This represents a 3.9-fold molar excess of drug over the alpha(4)beta(1) concentration in blood. Following oral administration of Compound B (5 mg/kg) to beagle dogs and rhesus monkeys, rapid and sustained alpha(4)beta(1) receptor occupancy (>80%) was achieved and maintained for a period of 24 h. When Compound B was administered intravenously to the horse, by either a slow or rapid infusion at a dose of 0.3 mg/kg, receptor blockade of >80% was observed out to 24 h with a concomitant leukocytosis. We believe that Compound B possesses suitable intrinsic and pharmacological properties to be evaluated clinically in horses affected by RAO.


Airway Obstruction/veterinary , Horse Diseases/immunology , Integrin alpha4beta1/immunology , Leukocytes/immunology , Airway Obstruction/blood , Airway Obstruction/drug therapy , Airway Obstruction/immunology , Animals , Binding, Competitive , Dogs , Female , Flow Cytometry/veterinary , Horse Diseases/blood , Horse Diseases/drug therapy , Horses , Integrin alpha4beta1/antagonists & inhibitors , Macaca mulatta , Male , Rats , Rats, Sprague-Dawley
13.
Bioorg Med Chem Lett ; 15(18): 4053-6, 2005 Sep 15.
Article En | MEDLINE | ID: mdl-16039122

A series of VLA-4 antagonist were synthesized wherein carboxylic acid was replaced by various acid surrogates. The effect of these acid surrogates toward potency was evaluated in a binding assay. A number of acid surrogates were potent antagonist of VLA-4, albeit significantly less potent than the corresponding carboxylic acid. Heterocyclic acid surrogate, oxadiazolidinone 3, demonstrated an improved pharmacokinetic property when dosed intravenously.


Acids/chemistry , Integrin alpha4beta1/antagonists & inhibitors , Acids/metabolism , Acids/pharmacology , Animals , Inhibitory Concentration 50 , Injections, Intravenous , Integrin alpha4beta1/metabolism , Molecular Structure , Oxadiazoles/administration & dosage , Oxadiazoles/chemistry , Oxadiazoles/pharmacokinetics , Oxadiazoles/pharmacology , Rats , Structure-Activity Relationship
14.
Chem Biodivers ; 2(1): 112-22, 2005 Jan.
Article En | MEDLINE | ID: mdl-17191924

HIV-1 Tat is one of six regulatory proteins that are required for viral replication and is an attractive target for the development of new anti-HIV agents. Screening of microbial extracts using a whole cell Tat-dependent transactivation assay, which guided the separation of the active broths, led to the identification of five structurally diverse classes (M(R) range 232-1126) of natural products. These include i) three sesquiterpenoids, namely, sporogen-AO1, petasol, and 6-dehydropetasol, ii) two resorcylic 14-membered lactones, namely monorden and monocillin IV, iii) a ten-membered lactone, iv) a quinoline and quinoxiline bicyclic octadepsipeptides, namely echinomycin and UK-63598, and v) a cyclic heptapeptide, ternatin. These compounds displayed varying degrees of potencies with IC50 values ranging from 0.0002 to 100 microM. The most active compound was the quinoxiline bicyclic octadepsipeptides, UK-63598, which inhibited Tat-dependent transactivation with an IC50 value of 0.2 nM and exhibited a 100-fold therapeutic window with respect to toxicity. In a single-cycle antiviral assay, UK-6358 inhibited viral replication with an IC50 value of 0.5 nM; however, it appeared to be equally toxic at that concentration. Monocillin IV was significantly less active (Tat transactivation inhibitory IC50 of 5 microM) but was not toxic at 100 microM in an equivalent cytotoxicity assay. The compound exhibited antiviral activity with an IC50 value of 6.2 microM in the single-cycle antiviral assay and a sixfold therapeutic window. Details of the isolation, fermentation, and biological activities of these structurally diverse natural products are described.


Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacology , Bacteria/metabolism , Fungi/metabolism , Gene Expression Regulation, Viral/drug effects , Gene Products, tat/antagonists & inhibitors , Transcriptional Activation/drug effects , Bacteria/chemistry , Cell Line , Fungi/chemistry , Gene Products, tat/genetics , Gene Products, tat/metabolism , Humans , Molecular Structure
15.
Biochem Biophys Res Commun ; 324(1): 108-13, 2004 Nov 05.
Article En | MEDLINE | ID: mdl-15464989

HIV-1 protease is one of several key enzymes required for the replication and maturation of HIV-1 virus. An almost two-decade research effort by academic and pharmaceutical institutions resulted in the successful commercialization of seven drugs that are potent inhibitors of HIV-1 protease activity and which, if used correctly, are highly effective in managing viral load. However, identification of clinical viral isolates that are resistant to these drugs indicates that this is a significant problem and that new classes of inhibitors are continually needed. Screening of microbial extracts followed by bioassay-guided isolation led to the discovery of a natural hinnuliquinone, a C(2)-symmetric bis-indolyl quinone natural product that inhibited the wild-type and a clinically resistant (A44) strain of HIV-1 protease with K(i) values of 0.97 and 1.25microM, respectively. Crystallographic analysis of the inhibitor-bound HIV-1 protease helped explain the importance of the C(2)-symmetry of hinnuliquinone for activity. Details of the isolation, biological activity, and crystallographic analysis of the inhibitor-bound protease are herein described.


Benzoquinones/chemistry , Benzoquinones/metabolism , Fungal Proteins/chemistry , Fungal Proteins/metabolism , HIV Protease Inhibitors/chemistry , HIV Protease Inhibitors/metabolism , Indoles/chemistry , Indoles/metabolism , Quinones , Catalytic Domain , Dimerization , HIV Infections/drug therapy , HIV Protease/metabolism , HIV Protease Inhibitors/therapeutic use , HIV-1/enzymology , Humans , Models, Molecular , Molecular Conformation , Molecular Structure , Quercus/microbiology , Quinones/chemistry , Quinones/metabolism
16.
Bioorg Med Chem Lett ; 13(4): 713-7, 2003 Feb 24.
Article En | MEDLINE | ID: mdl-12639565

Integration of viral DNA into host cell DNA is an essential step in retroviral (HIV-1) replication and is catalyzed by HIV-1 integrase. HIV-1 integrase is a novel therapeutic target and is the focus of efforts to identify effective inhibitors that will prevent/or cure HIV infections. Four novel naphtho-gamma-pyrones, belonging to the chaetochromin and ustilaginoidin family, were discovered as inhibitors of HIV-1 integrase from the screening of fungal extracts using a recombinant in vitro assay. These compounds inhibit both the coupled and strand transfer activity of HIV-1 integrase with IC(50) values of 1-3 and 4-12 microM, respectively. The discovery, structure elucidation, chemical modification and the structure-activity relationship of these compounds are described.


Fusarium/chemistry , HIV Integrase Inhibitors/isolation & purification , Pyrones/isolation & purification , Anti-HIV Agents/chemistry , Anti-HIV Agents/isolation & purification , Anti-HIV Agents/pharmacology , HIV Integrase Inhibitors/chemistry , HIV Integrase Inhibitors/pharmacology , Inhibitory Concentration 50 , Mass Spectrometry , Molecular Structure , Nuclear Magnetic Resonance, Biomolecular , Pyrans , Pyrones/chemistry , Pyrones/pharmacology , Structure-Activity Relationship
17.
J Nat Prod ; 65(8): 1091-5, 2002 Aug.
Article En | MEDLINE | ID: mdl-12193009

Tat is a small HIV protein essential for both viral replication and the progression of HIV disease. In our efforts to discover Tat inhibitors from natural product screening of microbial fermentation extracts, we discovered durhamycin A (1) as a potent inhibitor (IC(50) = 4.8 nM) of Tat transactivation. Detailed NMR and MS/MS studies were utilized to elucidate the structure of 1 as a new member of the aureolic acid family of antibiotics. It consists of tetrasaccharide and disaccharide moieties attached to the aglycone, which is hitherto unknown in the aureolic acid family. Three other novel analogues, durhamycin B (2), compound (3), and the aglycone (4), were also discovered or chemically prepared that were less potent than durhamycin A.


Anti-HIV Agents/pharmacology , Gene Products, tat/antagonists & inhibitors , Gene Products, tat/physiology , HIV-1/physiology , Plicamycin/analogs & derivatives , Costa Rica , Drug Design , Gene Expression Regulation, Viral , Gene Products, tat/metabolism , Hydrolysis , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Structure , Plicamycin/pharmacology , Protein Binding/drug effects , Structure-Activity Relationship , Substrate Specificity , tat Gene Products, Human Immunodeficiency Virus
18.
Biochemistry ; 41(22): 7125-41, 2002 Jun 04.
Article En | MEDLINE | ID: mdl-12033947

Integrins alpha9beta1 and alpha4beta1 form a distinct structural class, but while alpha4beta1 has been subjected to extensive study, alpha9beta1 remains poorly characterized. We have used the small molecule N-(benzenesulfonyl)-(L)-prolyl-(L)-O-(1-pyrrolidinylcarbonyl)tyrosine (3) to investigate the biochemical properties of alpha9beta1 and directly compare these properties with those of alpha4beta1. Compound 3 has a high affinity for both integrins with K(D) values of < or =3 and 180 pM for alpha9beta1 in 1 mM Mn2+ (activating) and 1 mM Ca2+ and 1 mM Mg2+ (nonactivating) conditions and < or =5 and 730 pM for alpha4beta1 under the corresponding conditions. Ca2+ treatment promoted the binding of 3 to both integrins (EC50 = 30 microM Ca2+ in both cases). Compound 3 binding to both integrins was also stimulated by the addition of the activating monoclonal antibody TS2/16. These findings indicate that the mechanisms by which metal ions and TS2/16 regulate ligand binding to alpha9beta1 and alpha4beta1 are similar. The binding of 3 to both integrins induced the mAb 9EG7 LIBS epitope, a property consistent with occupancy of the receptor's ligand binding site by 3. But whereas EGTA treatment inhibited the binding of 9EG7 to alpha4beta1, it stimulated the binding of 9EG7 to alpha9beta1. The 9EG7 and TS2/16 effects point to contributions of the beta1-chains on binding. Cross-linking data revealed that the integrin alpha-chains are also involved in binding the small molecule, as stable linkages were observed on both the alpha9 chain of alpha9beta1 and the alpha4 chain of alpha4beta1. Extensive structure-activity analyses with natural and synthetic ligands indicate distinct features of the ligand binding pockets. Most notable was the estimated >1000-fold difference in the affinity of the integrins for VCAM-1, which binds alpha4beta1with an apparent K(D) of 10 nM and alpha9beta1 with an apparent K(D) of >10 microM. Differences were also seen in the binding of alpha9beta1 and alpha4beta1 to osteopontin. Compound 3 competed effectively for the binding of VCAM-1 and osteopontin to both integrins. While these studies show many similarities in the biochemical properties of alpha9beta1 and alpha4beta1, they identify important differences in their structure and function that can be exploited in the design of selective alpha9beta1 and alpha4beta1 inhibitors.


Dipeptides/metabolism , Integrins/metabolism , Metals/metabolism , Receptors, Lymphocyte Homing/metabolism , Sialoglycoproteins/metabolism , Sulfones/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Binding, Competitive/physiology , Calcium/metabolism , Dipeptides/chemical synthesis , Humans , Integrin alpha4beta1 , Integrins/antagonists & inhibitors , Jurkat Cells , K562 Cells , Ligands , Magnesium/metabolism , Manganese/metabolism , Oligopeptides/metabolism , Osteopontin , Protein Binding/physiology , Receptors, Lymphocyte Homing/antagonists & inhibitors , Sialoglycoproteins/pharmacology , Sulfones/chemical synthesis , Vascular Cell Adhesion Molecule-1/pharmacology
19.
Curr Opin Drug Discov Devel ; 5(2): 225-44, 2002 Mar.
Article En | MEDLINE | ID: mdl-11926129

The hypothesis that the inhibition of farnesyl protein transferase (FPTase) may be beneficial in the treatment of certain cancers is finally being clinically tested. The efforts of almost a decade of preclinical research have resulted in several new chemical entities that are currently undergoing phase I, II or III clinical trials. The number of compounds being tested is small and disproportionate given the amount of effort by a large number of organizations in targeting FPTase. The three compounds, BMS-214662 (Bristol-Myers Squibb), R-115777 (Janssen Pharmaceutica BV) and Sch-66336 (Schering-Plough Research Institute) are the most advanced. While initial results with these three candidate drugs are encouraging, it is disappointing that the clinical efficacy of many other candidates has been less successful. In this review, we summarize the clinical and preclinical data published in 2001, and discuss the natural product inhibitors reported between 1998 to 2001. In addition, a comprehensive history of the FPTase inhibitory discovery effort at Merck, from CAAX tetrapeptides to peptidomimetics to macrocyclic compounds, is summarized for the first time.


Alkyl and Aryl Transferases/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Animals , Humans
20.
Org Lett ; 4(7): 1123-6, 2002 Apr 04.
Article En | MEDLINE | ID: mdl-11922798

[structure: see text] HIV-1 integrase is a critical enzyme for viral replication, and its inhibition is an emerging target for potential antiviral chemotherapy. We have discovered a novel inhibitor, integramycin, from screening of fermentation extracts using an in vitro assay. Integramycin possesses a hexacyclic ring system and exhibited an IC50 value of 4 microM against HIV-1 integrase (strand transfer). The isolation, structure elucidation, stereochemistry, conformation, and biological activity has been described.


HIV Integrase Inhibitors/chemistry , HIV-1 , Micromonosporaceae/chemistry , Naphthalenes/chemistry , Spiro Compounds/chemistry , Fermentation , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Conformation , Naphthalenes/pharmacology , Spectrometry, Mass, Electrospray Ionization , Spiro Compounds/pharmacology
...