Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
2.
Bioorg Med Chem ; 73: 117035, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36208543

ABSTRACT

Hereditary angioedema (HAE) is a rare and potentially life-threatening disease that affects an estimated 1 in 50,000 individuals worldwide. Berotralstat (BCX7353) is the only small molecule approved by the US Food and Drug Administration (FDA) for the prophylactic treatment of HAE attacks in patients 12 years and older. During the discovery of BCX7353, we also identified a novel series of small molecules containing a quaternary carbon as potent and orally bioavailable Plasma Kallikrein (PKal) inhibitors. Lead compound was identified as a potent inhibitor following a detailed lead optimization process that balanced the lipophilic efficiency (LipE) and pharmacokinetic (PK) profile.


Subject(s)
Angioedemas, Hereditary , Plasma Kallikrein , Angioedemas, Hereditary/drug therapy , Angioedemas, Hereditary/prevention & control , Antiviral Agents/therapeutic use , Carbon , Humans , United States
3.
J Med Chem ; 64(17): 12453-12468, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34436898

ABSTRACT

Hereditary angioedema (HAE) is a rare and potentially life-threatening disease that affects an estimated 1 in 50 000 individuals worldwide. Until recently, prophylactic HAE treatment options were limited to injectables, a burdensome administration route that has driven the need for an oral treatment. A substantial body of evidence has shown that potent and selective plasma kallikrein inhibitors that block the generation of bradykinin represent a promising approach for the treatment of HAE. Berotralstat (BCX7353, discovered by BioCryst Pharmaceuticals using a structure-guided drug design strategy) is a synthetic plasma kallikrein inhibitor that is potent and highly selective over other structurally related serine proteases. This once-daily, small-molecule drug is the first orally bioavailable prophylactic treatment for HAE attacks, having successfully completed a Phase III clinical trial (meeting its primary end point) and recently receiving the U.S. Food and Drug Administration's approval for the prophylactic treatment of HAE attacks in patients 12 years and older.


Subject(s)
Angioedemas, Hereditary/drug therapy , Kallikreins/antagonists & inhibitors , Pyrazoles/chemistry , Pyrazoles/pharmacology , Administration, Oral , Catalytic Domain , Drug Design , Humans , Models, Molecular , Molecular Structure , Protein Binding , Protein Conformation , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacology , Structure-Activity Relationship
4.
Blood ; 118(8): 2184-90, 2011 Aug 25.
Article in English | MEDLINE | ID: mdl-21730354

ABSTRACT

Forodesine and nelarabine (the pro-drug of ara-G) are 2 nucleoside analogues with promising anti-leukemic activity. To better understand which pediatric patients might benefit from forodesine or nelarabine (ara-G) therapy, we investigated the in vitro sensitivity to these drugs in 96 diagnostic pediatric leukemia patient samples and the mRNA expression levels of different enzymes involved in nucleoside metabolism. Forodesine and ara-G cytotoxicities were higher in T-cell acute lymphoblastic leukemia (T-ALL) samples than in B-cell precursor (BCP)-ALL and acute myeloid leukemia (AML) samples. Resistance to forodesine did not preclude ara-G sensitivity and vice versa, indicating that both drugs rely on different resistance mechanisms. Differences in sensitivity could be partly explained by significantly higher accumulation of intracellular dGTP in forodesine-sensitive samples compared with resistant samples, and higher mRNA levels of dGK but not dCK. The mRNA levels of the transporters ENT1 and ENT2 were higher in ara-G-sensitive than -resistant samples. We conclude that especially T-ALL, but also BCP-ALL, pediatric patients may benefit from forodesine or nelarabine (ara-G) treatment.


Subject(s)
Antineoplastic Agents/therapeutic use , Arabinonucleosides/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Prolymphocytic, B-Cell/drug therapy , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Prodrugs/therapeutic use , Purine Nucleosides/therapeutic use , Pyrimidinones/therapeutic use , Cell Line, Tumor , Child , Deoxycytidine Kinase/genetics , Deoxyguanine Nucleotides/metabolism , Drug Resistance, Neoplasm , Equilibrative Nucleoside Transporter 1/genetics , Equilibrative-Nucleoside Transporter 2/genetics , Gene Expression , Humans , In Vitro Techniques , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Prolymphocytic, B-Cell/genetics , Leukemia, Prolymphocytic, B-Cell/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Purines/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
5.
Antiviral Res ; 90(1): 17-21, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21316393

ABSTRACT

New and emerging influenza virus strains, such as the pandemic influenza A (H1N1) virus require constant vigilance for antiviral drug sensitivity and resistance. Efficacy of intramuscularly (IM) administered neuraminidase (NA) inhibitor, peramivir, was evaluated in mice infected with recently isolated pandemic A/California/04/2009 (H1N1, swine origin, mouse adapted) influenza virus. A single IM injection of peramivir (four dose groups), given 1h prior to inoculation, significantly reduced weight loss (p < 0.001) and mortality (p < 0.05) in mice infected with LD90 dose of pandemic A/California/04/2009 (H1N1) influenza virus compared to vehicle group. There was 20% survival in the vehicle-treated group, whereas in the peramivir-treated groups, survival increased in a dose-dependent manner with 60, 60, 90 and 100% survivors for the 1, 3, 10, and 30 mg/kg doses, respectively. Weight loss on day 4 in the vehicle-treated group was 3.4 gm, and in the peramivir-treated groups was 2.1, 1.5, 1.8 and 1.8 g for the 1, 3, 10 and 30 mg/kg dose groups, respectively. In the treatment model, peramivir given 24h after infection as a single IM injection at 50mg/kg dose, showed significant protection against lethality and weight loss. There was 13% survival in the vehicle-treated group while in the peramivir-treated group at 24, 48, and 72 h post infection, survival was 100, 40, and 50%, respectively. Survival in the oseltamivir groups (10 mg/kg/d twice a day, orally for 5 days) was 90, 30 and 20% at 24, 48 and 72 h, respectively. These data demonstrate efficacy of parenterally administered peramivir against the recently isolated pandemic influenza virus in murine infection models.


Subject(s)
Antiviral Agents/administration & dosage , Cyclopentanes/administration & dosage , Guanidines/administration & dosage , Influenza A Virus, H1N1 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Acids, Carbocyclic , Animals , Body Weight , Disease Models, Animal , Female , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/mortality , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Rodent Diseases/drug therapy , Rodent Diseases/mortality , Rodent Diseases/pathology , Rodent Diseases/virology , Survival Analysis
6.
Antiviral Res ; 88(3): 276-80, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20943201

ABSTRACT

Efficacy of combination of the intramuscularly administered neuraminidase (NA) inhibitor, peramivir, and the orally administered M2 ion channel blocker, rimantadine was evaluated in mouse influenza A/Victoria/3/75 (H3N2) model. Mice were challenged with a sub-lethal virus dose (0-40% mortality in placebo group) and changes in body weights were analyzed by three-dimensional effect analysis to assess mode of drug interactions. Compounds were administered in a 5-day treatment course starting 1h before viral inoculation. The peramivir and rimantadine doses ranged from 0.3-3 mg/kg/d and 5-30 mg/kg/d, respectively. The maximum mean weight loss of 5.19 g was observed in the vehicle-infected group on day 10. In the 1 and 3 mg/kg/d peramivir monotherapy groups, the weight losses were 4.3 and 3.55 g, respectively. In the rimantadine monotherapy group, the weight losses were 3.43, 2.1, and 1.64 g for the 5, 10, and 30 mg/kg/d groups, respectively. Combination of 1mg/kg/d peramivir with 5 and 10 mg/kg/d rimantadine produced weight losses of 1.69 and 0.69 (p<0.05 vs. vehicle and individual agent), respectively, whereas the combination of 3.0 mg/kg/d peramivir with 10 and 30 mg/kg/d rimantadine did not show any weight loss (p<0.05 vs. vehicle and individual agent). The three-dimensional analysis of the weight loss for the majority of the drug combinations of peramivir and rimantadine tested demonstrated synergistic antiviral effects.


Subject(s)
Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Cyclopentanes , Guanidines , Influenza A Virus, H3N2 Subtype/drug effects , Orthomyxoviridae Infections/drug therapy , Rimantadine , Weight Loss/drug effects , Acids, Carbocyclic , Animals , Cyclopentanes/pharmacology , Cyclopentanes/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Administration Routes , Drug Administration Schedule , Drug Synergism , Female , Guanidines/pharmacology , Guanidines/therapeutic use , Mice , Mice, Inbred BALB C , Orthomyxoviridae Infections/mortality , Rimantadine/pharmacology , Rimantadine/therapeutic use , Survival Rate
7.
Int Immunopharmacol ; 10(7): 784-90, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20399911

ABSTRACT

The profound suppression of T-cell immunity seen in purine nucleoside phosphorylase (PNP; EC 2.4.2.1) deficient patients supports potential application of inhibitors of PNP in the therapy of T-cell mediated diseases. BCX-4208 is a novel potent transition state analog inhibitor of human PNP with an IC(50) of 0.5 nM. PNP inhibition leads to elevation of dGuo which is converted to dGTP mainly in lymphocytes causing imbalance in deoxynucleotide (dNTP) pools and cell apoptosis. In in vitro studies, neither BCX-4208 nor dGuo alone inhibits proliferation of lymphocytes. BCX-4208 in the presence of 10 microM deoxyguanosine (dGuo) inhibits lymphocyte proliferation induced by MLR, IL-2 or Con A with IC(50)s of 0.159, 0.26 and 0.73 microM, respectively. The IC(50) for dGuo in the presence of 1 microM BCX-4208 for the IL-2 stimulated lymphocytes was 3.12 microM. dGTP in human lymphocytes is elevated and a 3-5 fold increase in dGTP results in 50% inhibition after in vitro exposure to BCX-4208 and dGuo. Flow cytometric analyses of human lymphocytes using annexin V staining reveal that BCX-4208 in the presence of dGuo induces cellular apoptosis in T-cells (CD3+), B-cells (CD20+, CD19+) and NK (CD56+) cells. BCX-4208 is orally bioavailable in mice and elevates plasma dGuo levels to 3.7 microM (predose levels<0.004 microM), similar to levels seen in PNP-deficient patients and levels needed to cause apoptosis in T and B-cells. These data support the evaluation of BCX-4208 in the treatment of T-cell and B-cell mediated diseases. BCX-4208 is currently undergoing early clinical investigation in psoriasis and gout.


Subject(s)
Apoptosis/drug effects , B-Lymphocytes/drug effects , Enzyme Inhibitors/therapeutic use , Psoriasis/drug therapy , T-Lymphocytes/drug effects , Administration, Oral , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Proliferation/drug effects , Cells, Cultured , Deoxyguanosine/genetics , Deoxyguanosine/metabolism , Enzyme Inhibitors/pharmacology , Graft Rejection/drug therapy , Graft Rejection/immunology , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/immunology , Humans , Lymphocyte Culture Test, Mixed , Organ Transplantation , Psoriasis/immunology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
8.
Thromb Res ; 117(3): 343-9, 2006.
Article in English | MEDLINE | ID: mdl-16378835

ABSTRACT

Tissue factor (TF) is a transmembrane glycoprotein that binds its zymogen cofactor, Factor VIIa (FVIIa) on the cell surface. Together (TF/FVIIa) they activate Factor X (FX) and Factor IX (FIX) and start the extrinsic pathway of blood coagulation. As such, the TF/FVIIa complex plays an important role in normal physiology as well as in thrombotic diseases such as unstable angina (UA), disseminated intravascular coagulation (DIC), and deep vein thrombosis (DVT). In addition to its function as an initiator of coagulation, TF/FVIIa plays an important role in inflammation. Expression of TF on the cell surface and its appearance as a soluble molecule are characteristic features of acute and chronic inflammation in conditions such as sepsis and atherosclerosis. Here we demonstrate that BCX-3607, a small molecule potent inhibitor of TF/FVIIa, reduces thrombus weight in an animal model of DVT. BCX-3607 also decreases the level of interleukin-6 (IL-6) in a LPS-stimulated mouse model of endotoxemia. Additionally, in vitro studies indicate that BCX-3607 blocks the generation of TF/FVIIa-induced IL-8 mRNA in human keratinocytes and reduces the TF/FVIIa-mediated generation of IL-6 and IL-8 in human umbilical vein endothelial cells (HUVEC). Therefore, BCX-3607 might block the TF/FVIIa-mediated coagulation and inflammation associated with pathological conditions.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Factor VIIa/antagonists & inhibitors , Fibrinolytic Agents/pharmacology , Pyridines/pharmacology , Thromboplastin/antagonists & inhibitors , Animals , Atherosclerosis/drug therapy , Blotting, Northern , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelium, Vascular/metabolism , Endotoxemia/pathology , Humans , Inflammation , Interleukin-6/metabolism , Interleukin-8/metabolism , Keratinocytes/metabolism , Lipopolysaccharides/metabolism , Male , Mice , Models, Biological , Models, Chemical , Prothrombin Time , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sepsis , Time Factors
9.
Antiviral Res ; 69(1): 39-45, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16325932

ABSTRACT

In the event of an influenza outbreak, antivirals including the neuraminidase (NA) inhibitors, peramivir, oseltamivir, and zanamivir may provide valuable benefit when vaccine production is delayed, limited, or cannot be used. Here we demonstrate the efficacy of a single intramuscular injection of peramivir in the mouse influenza model. Peramivir potently inhibits the neuraminidase enzyme N9 from H1N9 virus in vitro with a 50% inhibitory concentration (IC(50)) of 1.3+/-0.4 nM. On-site dissociation studies indicate that peramivir remains tightly bound to N9 NA (t(1/2)>24h), whereas, zanamivir and oseltamivir carboxylate dissociated rapidly from the enzyme (t(1/2)=1.25 h). A single intramuscular injection of peramivir (10mg/kg) significantly reduces weight loss and mortality in mice infected with influenza A/H1N1, while oseltamivir demonstrates no efficacy by the same treatment regimen. This may be due to tight binding of peramivir to the N1 NA enzymes similar to that observed for N9 enzyme. Additional efficacy studies indicate that a single injection of peramivir (2-20mg/kg) was comparable to a q.d.x 5 day course of orally administered oseltamivir (2-20mg/kg/day) in preventing lethality in H3N2 and H1N1 influenza models. A single intramuscular injection of peramivir may successfully treat influenza infections and provide an alternate option to oseltamivir during an influenza outbreak.


Subject(s)
Antiviral Agents/therapeutic use , Cyclopentanes/therapeutic use , Guanidines/therapeutic use , Influenza A Virus, H1N1 Subtype , Neuraminidase/antagonists & inhibitors , Neuraminidase/therapeutic use , Orthomyxoviridae Infections/drug therapy , Acids, Carbocyclic , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/chemistry , Cyclopentanes/administration & dosage , Cyclopentanes/chemistry , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Guanidines/administration & dosage , Guanidines/chemistry , Injections, Intramuscular , Mice , Neuraminidase/administration & dosage
10.
Int Immunopharmacol ; 3(6): 879-87, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12781704

ABSTRACT

Purine nucleoside phosphorylase (PNP) deficiency in humans produces a relatively selective depletion of T cells. BCX-1777 is a potent inhibitor of PNP. BCX-1777 in the presence of deoxyguanosine (dGuo) inhibits the proliferation of CEM-SS [T-acute lymphoblastic leukemia (T-ALL)] cells with an IC(50)=0.015 microM. This inhibition by BCX-1777 and dGuo is accompanied by elevation of dGTP (154-fold) and dATP (8-fold). Deoxycytidine (dCyt) completely and lamivudine (3TC) partially reverse this inhibition caused by BCX-1777 and dGuo. dNTP analysis of these samples indicates that, in the presence of dCyt, where complete reversal of inhibition is observed, dGTP and dATP pools revert back to the control levels. In samples containing 3TC, where partial reversal of inhibition was observed, dGTP decreased from 154-fold to 38-fold and dATP levels further increased from 8-fold to 30-fold compared to the control sample. In CEM-SS cells, inhibition of proliferation by BCX-1777 and dGuo is not due to accumulation of dATP because in the presence of 3TC, where reversal of inhibition is observed, dATP levels are further increased. These studies clearly indicate that inhibition of T cells is due to accumulation of dGTP resulting in cell death with characteristics of apoptosis. The half-life of dGTP in CEM-SS cells is 18 h, which is longer than that observed in human lymphocytes (4 h), suggesting that the nucleotidase level in CEM-SS cells is lower than in human lymphocytes. A 154-fold accumulation of dGTP in CEM-SS cells in the presence of BCX-1777 and dGuo compared to a 15-fold accumulation of dGTP in human lymphocytes suggests that kinase level is higher in CEM-SS cells compared to human lymphocytes. High kinase and low nucleotidase levels make CEM-SS cells more sensitive to inhibition by BCX-1777 and dGuo than human lymphocytes. Currently, BCX-1777 is in phase I/II clinical trial for the treatment of T cell malignancies.


Subject(s)
Leukemia-Lymphoma, Adult T-Cell/drug therapy , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Pyrimidinones/pharmacology , Pyrroles/pharmacology , Apoptosis/drug effects , Cell Division/drug effects , Cell Line , Culture Media , HIV Reverse Transcriptase/antagonists & inhibitors , Half-Life , Humans , Lamivudine/pharmacology , Lymphocytes/drug effects , Lymphocytes/metabolism , Purine Nucleosides , Reverse Transcriptase Inhibitors/pharmacology , Stavudine/pharmacology
11.
Int Immunopharmacol ; 2(7): 913-23, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12188033

ABSTRACT

Purine nucleoside phosphorylase (PNP) deficiency in humans produces a relatively selective depletion of T-cells. Inhibitors of PNP are therefore of interest as potential T-cell selective immunosuppressive agents. BCX-1777 is a potent inhibitor of PNP and in vitro T-cell proliferation. Inhibition of human T-cells by BCX-1777 and deoxyguanosine (dGuo) is accompanied by deoxyguanosine triphosphate (dGTP) accumulation. Unlike human T-cells, mouse, rat, dog and monkey T-cells are neither inhibited (IC50>100 microM) nor accumulate dGTP in the presence of BCX-1777 and dGuo. Cells pretreated with BCX-1777 and dGuo for 24 h (to elevate dGTP levels) prior to stimulation demonstrated 80% inhibition similar to the inhibition observed with cells treated with BCX-1777 and dGuo during the stimulation and proliferation process. This further confirms that inhibition of T-cells is due to the accumulation of dGTP in these cells. Deoxynucleotide (dNTP) analysis of the cells treated with BCX-1777 and dGuo for 48 h showed no significant change in deoxycytidine triphosphate (dCTP) and deoxyadenosine triphosphate (dATP) pools. However, a decrease (2-fold) in thymidine triphosphate (dTTP) pools, and a large increase in dGTP pools (15-fold) were observed. Results from various groups have shown that alteration in the dNTP supply results in DNA fragmentation and cell death with characteristics of apoptosis. Indeed, apoptosis is observed in human T-lymphocytes treated with BCX-1777 and dGuo. To compare the in vivo efficacy of BCX-1777 with another potent T-cell inhibitor, cyclosporin, these drugs were tested in a xenogeneic graft-vs.-host disease model (XGVHD). In this model, human lymphocytes are engrafted into severe combined immunodeficient mice (SCID) mice inducing severe XGVHD. The efficacy of BCX-1777 in the XGVHD model was comparable to cyclosporin and a combination of BCX-1777 and cyclosporin treatment showed a trend towards increased efficacy compared to cyclosporin alone. These results suggest that BCX-1777 may be useful for the treatment of disease characterized by activated T-cell responses.


Subject(s)
Cyclosporine/therapeutic use , Deoxyguanine Nucleotides/metabolism , Graft vs Host Disease/drug therapy , Growth Inhibitors/therapeutic use , Immunosuppressive Agents/therapeutic use , Pyrimidinones/therapeutic use , Pyrroles/therapeutic use , Transplantation, Heterologous/immunology , Animals , Cyclosporine/pharmacology , Dogs , Drug Therapy, Combination , Graft vs Host Disease/chemically induced , Graft vs Host Disease/metabolism , Growth Inhibitors/pharmacology , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Lymphocytes/cytology , Lymphocytes/drug effects , Lymphocytes/metabolism , Macaca fascicularis , Mice , Mice, Inbred BALB C , Mice, SCID , Purine Nucleosides , Pyrimidinones/pharmacology , Pyrroles/pharmacology , Rats , Rats, Sprague-Dawley , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...