Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
ACS Pharmacol Transl Sci ; 7(5): 1302-1309, 2024 May 10.
Article En | MEDLINE | ID: mdl-38751629

The B-cell lymphoma-2 (Bcl-2) family of proteins plays a vital role in tumorigenesis. Cancer cells utilize the expression of Bcl-2 to evade therapy and develop resistance. Bcl-2 overexpression also causes cancer cells to be more invasive and metastatic. About 80% of cancer deaths are due to metastases, and yet targeted therapies for metastatic cancers are scarce. We discovered a small molecule, BFC1103, which changes the conformation of Bcl-2 to convert the antiapoptotic protein to a proapoptotic protein. BFC1103-induced apoptosis is dependent on the expression levels of Bcl-2, with higher levels causing more apoptosis. BFC1103 suppressed the growth of breast cancer lung metastasis. BFC1103 has the potential for further optimization and development for clinical testing in metastatic cancers that express Bcl-2. This study demonstrates a new approach to target Bcl-2 using a small molecule, BFC1103, to suppress metastatic disease.

2.
Cancer Res Commun ; 4(3): 634-644, 2024 03 04.
Article En | MEDLINE | ID: mdl-38329389

Cancer cells exploit the expression of anti-apoptotic protein Bcl-2 to evade apoptosis and develop resistance to therapeutics. High levels of Bcl-2 leads to sequestration of pro-apoptotic proteins causing the apoptotic machinery to halt. In this study, we report discovery of a small molecule, BFC1108 (5-chloro-N-(2-ethoxyphenyl)-2-[(4-methoxybenzyol)amino]benzamide), which targets Bcl-2 and converts it into a pro-apoptotic protein. The apoptotic effect of BFC1108 is not inhibited, but rather potentiated, by Bcl-2 overexpression. BFC1108 induces a conformational change in Bcl-2, resulting in the exposure of its BH3 domain both in vitro and in vivo. BFC1108 suppresses the growth of triple-negative breast cancer xenografts with high Bcl-2 expression and inhibits breast cancer lung metastasis. This study demonstrates a novel approach to targeting Bcl-2 using BFC1108, a small molecule Bcl-2 functional converter that effectively induces apoptosis in Bcl-2-expressing cancers. SIGNIFICANCE: We report the identification of a small molecule that exposes the Bcl-2 killer conformation and induces death in Bcl-2-expressing cancer cells. Selective targeting of Bcl-2 and elimination of cancer cells expressing Bcl-2 opens up new therapeutic avenues.


Apoptosis Regulatory Proteins , Apoptosis , Humans , Apoptosis Regulatory Proteins/metabolism , Protein Binding
3.
Blood Adv ; 7(19): 5835-5842, 2023 10 10.
Article En | MEDLINE | ID: mdl-37552106

In patients with cytopenic myelofibrosis, treatment with the JAK2/IRAK1 inhibitor pacritinib was associated with anemia benefit in the phase 3 PERSIST-2 study. The impact of pacritinib on transfusion independence (TI) has not been previously described, nor has the mechanism by which pacritinib improves anemia been elucidated. Because it has been previously postulated that inhibition of activin A receptor, type 1 (ACVR1)/activin receptor-like kinase-2 improves anemia in patients with myelofibrosis via suppression of hepcidin production, we assessed the relative inhibitory potency of pacritinib compared with other JAK2 inhibitors against ACVR1. Pacritinib inhibited ACVR1 with greater potency (half-maximal inhibitory concentration [IC50] = 16.7 nM; Cmax:IC50 = 12.7) than momelotinib (IC50 = 52.5 nM; Cmax:IC50 = 3.2), fedratinib (IC50 = 273 nM; Cmax:IC50 = 1.0), or ruxolitinib (IC50 > 1000; Cmax:IC50 < 0.01). Pacritinib's inhibitory activity against ACVR1 was corroborated via inhibition of downstream SMAD signaling in conjunction with marked suppression of hepcidin production. Among patients on PERSIST-2 who were not transfusion independent at baseline based on Gale criteria, a significantly greater proportion achieved TI on pacritinib compared with those treated on best available therapy (37% vs 7%, P = .001), and significantly more had a ≥50% reduction in transfusion burden (49% vs 9%, P < .0001). These data indicate that the anemia benefit of the JAK2/IRAK1 inhibitor pacritinib may be a function of potent ACVR1 inhibition.


Anemia , Janus Kinase Inhibitors , Primary Myelofibrosis , Humans , Primary Myelofibrosis/complications , Primary Myelofibrosis/drug therapy , Hepcidins , Janus Kinase 2 , Anemia/etiology , Anemia/complications , Activin Receptors, Type I
4.
Blood Adv ; 4(22): 5825-5835, 2020 11 24.
Article En | MEDLINE | ID: mdl-33232476

PAC203 is a randomized dose-finding study of pacritinib, an oral JAK2/IRAK1 inhibitor, in patients with advanced myelofibrosis who are intolerant of or resistant to ruxolitinib. Patients were randomized 1:1:1 to pacritinib 100 mg once per day, 100 mg twice per day, or 200 mg twice per day. Enhanced eligibility criteria, monitoring, and dose modifications were implemented to mitigate risk of cardiac and hemorrhagic events. Efficacy was based on ≥35% spleen volume response (SVR) and ≥50% reduction in the 7-component total symptom score (TSS) through week 24. Of 161 patients, 73% were intolerant of and 76% had become resistant to ruxolitinib; 50% met criteria for both. Severe thrombocytopenia (platelet count <50 × 103/µL) was present in 44%. SVR rates were highest with 200 mg twice per day (100 mg once per day, 0%; 100 mg twice per day, 1.8%; 200 mg twice per day, 9.3%), particularly among patients with baseline platelet counts <50 × 103/µL (17%; 4 of 24). Although TSS response rate was similar across doses (100 mg once per day, 7.7%; 100 mg twice per day, 7.3%; 200 mg twice per day, 7.4%), median percent reduction in TSS suggested a dose-response relationship (-3%, -16%, and -27%, respectively). Pharmacokinetic and pharmacodynamic modeling based on all available data showed greatest SVR and TSS reduction at 200 mg twice per day compared with lower doses. Common adverse events were gastrointestinal events, thrombocytopenia, and anemia. There was no excess of grade ≥3 hemorrhagic or cardiac events at 200 mg twice per day. Pacritinib 200 mg twice per day demonstrated clinical activity and an acceptable safety profile and was selected as the recommended dose for a pivotal phase 3 study in patients with myelofibrosis and severe thrombocytopenia. This trial was registered at www.clinicaltrials.gov as #NCT03165734.


Primary Myelofibrosis , Bridged-Ring Compounds , Humans , Primary Myelofibrosis/drug therapy , Pyrimidines/adverse effects , Treatment Outcome
5.
PLoS Pathog ; 14(12): e1007439, 2018 12.
Article En | MEDLINE | ID: mdl-30576397

Arenaviruses are a significant cause of hemorrhagic fever, an often-fatal disease for which there is no approved antiviral therapy. Lassa fever in particular generates high morbidity and mortality in West Africa, where the disease is endemic, and a recent outbreak in Nigeria was larger and more geographically diverse than usual. We are developing LHF-535, a small-molecule viral entry inhibitor that targets the arenavirus envelope glycoprotein, as a therapeutic candidate for Lassa fever and other hemorrhagic fevers of arenavirus origin. Using a lentiviral pseudotype infectivity assay, we determined that LHF-535 had sub-nanomolar potency against the viral envelope glycoproteins from all Lassa virus lineages, with the exception of the glycoprotein from the LP strain from lineage I, which was 100-fold less sensitive than that of other strains. This reduced sensitivity was mediated by a unique amino acid substitution, V434I, in the transmembrane domain of the envelope glycoprotein GP2 subunit. This position corresponds to the attenuation determinant of Candid#1, a live-attenuated Junín virus vaccine strain used to prevent Argentine hemorrhagic fever. Using a virus-yield reduction assay, we determined that LHF-535 potently inhibited Junín virus, but not Candid#1, and the Candid#1 attenuation determinant, F427I, regulated this difference in sensitivity. We also demonstrated that a daily oral dose of LHF-535 at 10 mg/kg protected mice from a lethal dose of Tacaribe virus. Serial passage of Tacaribe virus in LHF-535-treated Vero cells yielded viruses that were resistant to LHF-535, and the majority of drug-resistant viruses exhibited attenuated pathogenesis. These findings provide a framework for the clinical development of LHF-535 as a broad-spectrum inhibitor of arenavirus entry and provide an important context for monitoring the emergence of drug-resistant viruses.


Antiviral Agents/pharmacology , Lassa Fever , Lassa virus/genetics , Virulence/drug effects , Virulence/genetics , Animals , Chlorocebus aethiops , Drug Resistance, Viral/drug effects , Drug Resistance, Viral/genetics , HEK293 Cells , Humans , Lassa virus/drug effects , Mice , Mutation , Vero Cells , Viral Envelope Proteins/genetics
6.
PLoS One ; 8(4): e61514, 2013.
Article En | MEDLINE | ID: mdl-23637845

Plasma pharmacokinetics of ST-246, smallpox therapeutic, was evaluated in mice, rabbits, monkeys and dogs following repeat oral administrations by gavage. The dog showed the lowest Tmax of 0.83 h and the monkey, the highest value of 3.25 h. A 2- to 4-fold greater dose-normalized Cmax was observed for the dog compared to the other species. The mouse showed the highest dose-normalized AUC, which was 2-fold greater than that for the rabbit and monkey both of which by approximation, recorded the lowest value. The Cl/F increased across species from 0.05 L/h for mouse to 42.52 L/h for dog. The mouse showed the lowest VD/F of 0.41 L and the monkey, the highest VD/F of 392.95 L. The calculated extraction ratios were 0.104, 0.363, 0.231 and 0.591 for mouse, rabbit, monkey and dog, respectively. The dog showed the lowest terminal half-life of 3.10 h and the monkey, the highest value of 9.94 h. The simple allometric human VD/F and MLP-corrected Cl/F were 2311.51 L and 51.35 L/h, respectively, with calculated human extraction ratio of 0.153 and terminal half-life of 31.20 h. Overall, a species-specific difference was observed for Cl/F with this parameter increasing across species from mouse to dog. The human MLP-corrected Cl/F, terminal half-life, extraction ratios were in close proximity to the observed estimates. In addition, the first-in-humans (FIH) dose of 485 mg, determined from the MLP-corrected allometry Cl/F, was well within the dose range of 400 mg and 600 mg administered in healthy adult human volunteers.


Antiviral Agents/pharmacokinetics , Benzamides/pharmacokinetics , Isoindoles/pharmacokinetics , Poxviridae Infections/drug therapy , Administration, Oral , Adult , Animals , Antiviral Agents/administration & dosage , Area Under Curve , Benzamides/administration & dosage , Benzamides/blood , Body Weight , Dogs , Female , Half-Life , Humans , Isoindoles/administration & dosage , Isoindoles/blood , Macaca fascicularis , Male , Mice , Orthopoxvirus , Rabbits
7.
Antimicrob Agents Chemother ; 57(4): 1902-12, 2013 Apr.
Article En | MEDLINE | ID: mdl-23403421

Dengue virus (DENV) is the predominant mosquito-borne viral pathogen that infects humans with an estimated 50 to 100 million infections per year worldwide. Over the past 50 years, the incidence of dengue disease has increased dramatically and the virus is now endemic in more than 100 countries. Moreover, multiple serotypes of DENV are now found in the same geographic region, increasing the likelihood of more severe forms of disease. Despite extensive research, there are still no approved vaccines or therapeutics commercially available to treat DENV infection. Here we report the results of a high-throughput screen of a chemical compound library using a whole-virus assay that identified a novel small-molecule inhibitor of DENV, ST-610, that potently and selectively inhibits all four serotypes of DENV replication in vitro. Sequence analysis of drug-resistant virus isolates has identified a single point mutation, A263T, in the NS3 helicase domain that confers resistance to this compound. ST-610 inhibits DENV NS3 helicase RNA unwinding activity in a molecular-beacon-based helicase assay but does not inhibit nucleoside triphosphatase activity based on a malachite green ATPase assay. ST-610 is nonmutagenic, is well tolerated (nontoxic) in mice, and has shown efficacy in a sublethal murine model of DENV infection with the ability to significantly reduce viremia and viral load compared to vehicle controls.


Benzoxazoles/therapeutic use , Dengue Virus/drug effects , Dengue Virus/enzymology , Dengue/drug therapy , RNA Helicases/antagonists & inhibitors , Viral Nonstructural Proteins/antagonists & inhibitors , Virus Replication/drug effects , Animals , Cell Line , Dengue Virus/genetics , Dengue Virus/pathogenicity , Dogs , Drug Resistance, Viral/genetics , HeLa Cells , Humans , Mice , RNA Helicases/genetics , Vero Cells , Viral Nonstructural Proteins/genetics
8.
Antimicrob Agents Chemother ; 57(1): 15-25, 2013 Jan.
Article En | MEDLINE | ID: mdl-23070172

Dengue viruses (DENV) infect 50 to 100 million people worldwide per year, of which 500,000 develop severe life-threatening disease. This mosquito-borne illness is endemic in most tropical and subtropical countries and has spread significantly over the last decade. While there are several promising vaccine candidates in clinical trials, there are currently no approved vaccines or therapeutics available for treatment of dengue infection. Here, we describe a novel small-molecule compound, ST-148, that is a potent inhibitor of all four serotypes of DENV in vitro. ST-148 significantly reduced viremia and viral load in vital organs and tended to lower cytokine levels in the plasma in a nonlethal model of DENV infection in AG129 mice. Compound resistance mapped to the DENV capsid (C) gene, and a direct interaction of ST-148 with C protein is suggested by alterations of the intrinsic fluorescence of the protein in the presence of compound. Thus, ST-148 appears to interact with the DENV C protein and inhibits a distinct step(s) of the viral replication cycle.


Antiviral Agents/pharmacology , Capsid Proteins/antagonists & inhibitors , Capsid Proteins/genetics , Dengue Virus/drug effects , Dengue/drug therapy , Heterocyclic Compounds, 3-Ring/pharmacology , Thiadiazoles/pharmacology , Viremia/drug therapy , Amino Acid Sequence , Animals , Antiviral Agents/chemical synthesis , Capsid Proteins/chemistry , Cell Line , Chlorocebus aethiops , Cricetinae , Dengue/virology , Dengue Virus/classification , Dengue Virus/genetics , Disease Models, Animal , Dogs , Escherichia coli/genetics , Heterocyclic Compounds, 3-Ring/chemical synthesis , Humans , Inhibitory Concentration 50 , Mice , Models, Molecular , Molecular Sequence Data , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Thiadiazoles/chemical synthesis , Viral Load/drug effects , Viral Plaque Assay , Viremia/virology , Virus Replication/drug effects
9.
Antimicrob Agents Chemother ; 57(3): 1136-43, 2013 Mar.
Article En | MEDLINE | ID: mdl-23254433

Although smallpox has been eradicated, the United States government considers it a "material threat" and has funded the discovery and development of potential therapeutic compounds. As reported here, the human efficacious dose for one of these compounds, ST-246, was determined using efficacy studies in nonhuman primates (NHPs), together with pharmacokinetic and pharmacodynamic analysis that predicted the appropriate dose and exposure levels to provide therapeutic benefit in humans. The efficacy analysis combined the data from studies conducted at three separate facilities that evaluated treatment following infection with a closely related virus, monkeypox virus (MPXV), in a total of 96 NHPs. The effect of infection on ST-246 pharmacokinetics in NHPs was applied to humans using population pharmacokinetic models. Exposure at the selected human dose of 600 mg is more than 4-fold higher than the lowest efficacious dose in NHPs and is predicted to provide protection to more than 95% of the population.


Antiviral Agents/therapeutic use , Benzamides/therapeutic use , Isoindoles/therapeutic use , Macaca fascicularis/virology , Monkeypox virus/drug effects , Mpox (monkeypox)/drug therapy , Smallpox/drug therapy , Animals , Antiviral Agents/pharmacokinetics , Antiviral Agents/pharmacology , Benzamides/pharmacokinetics , Benzamides/pharmacology , Drug Dosage Calculations , Female , Humans , Isoindoles/pharmacokinetics , Isoindoles/pharmacology , Male , Models, Statistical , Mpox (monkeypox)/mortality , Mpox (monkeypox)/virology , Monkeypox virus/growth & development , Smallpox/virology , Survival Analysis , Treatment Outcome , Variola virus/drug effects , Variola virus/growth & development
10.
Antimicrob Agents Chemother ; 56(9): 4900-5, 2012 Sep.
Article En | MEDLINE | ID: mdl-22777041

ST-246 is being evaluated as a treatment for pathogenic orthopoxvirus infections in humans. To this end, a phase 2, double-blind, randomized, placebo-controlled, multicenter trial was conducted to assess the safety, tolerability, and pharmacokinetics (PK) of ST-246 when administered as a single daily oral dose (400 mg or 600 mg) for 14 days in fed adult volunteers. ST-246 was safe and well tolerated, with no deaths or serious adverse events reported during the study. There was a low incidence of treatment-emergent adverse events (TEAEs), the most common of which were mild nausea and headache. There were no clinically significant results from laboratory assessments, vital sign measurements, physical examinations, or electrocardiograms. The PK and dose proportionality of ST-246 were determined. The PK analysis showed that steady state was achieved by day 5 for the ST-246 400-mg treatment group and by day 6 for the 600-mg group. The dose proportionality analysis showed that the 400- and 600-mg ratio of dose-normalized peak drug concentration in plasma (C(max)) and relative exposure for each dosing interval (AUC(τ)) ranged from 80% to 85%. However, the 90% confidence intervals did not include 1.0, so dose proportionality could not be concluded. Overall, ST-246 was shown to be safe, and the PK was predictable. These results support further testing of ST-246 in a multicenter pivotal clinical safety study for licensure application.


Antiviral Agents/pharmacokinetics , Benzamides/pharmacokinetics , Isoindoles/pharmacokinetics , Administration, Oral , Adolescent , Adult , Aged , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Antiviral Agents/blood , Area Under Curve , Benzamides/administration & dosage , Benzamides/adverse effects , Benzamides/blood , Biological Availability , Double-Blind Method , Drug Administration Schedule , Female , Half-Life , Humans , Isoindoles/administration & dosage , Isoindoles/adverse effects , Isoindoles/blood , Male , Middle Aged , Placebos
11.
Bioorg Med Chem Lett ; 22(13): 4263-72, 2012 Jul 01.
Article En | MEDLINE | ID: mdl-22664128

A series of acylthiourea derivatives were designed, synthesized, and evaluated for broad-spectrum antiviral activity with selected viruses from Poxviridae (vaccinia virus) and two different genera of the family Bunyaviridae (Rift Valley fever and La Crosse viruses). A compound selected from a library screen, compound 1, displayed submicromolar antiviral activity against both vaccinia virus (EC(50)=0.25 µM) and La Crosse virus (EC(50)=0.27 µM) in cytopathic effect (CPE) assays. SAR analysis was performed to further improve antiviral potency and to optimize drug-like properties of the initial hits. During our analysis, we identified 26, which was found to be nearly fourfold more potent than 1 against both vaccinia and La Crosse viruses. Selected compounds were further tested to more fully characterize the spectrum of antiviral activity. Many of these possessed single digit micromolar and sub-micromolar antiviral activity against a diverse array of targets, including influenza virus (Orthomyxoviridae), Tacaribe virus (Arenaviridae), and dengue virus (Flaviviridae).


Antiviral Agents/chemistry , Thiourea/chemistry , Antiviral Agents/chemical synthesis , Antiviral Agents/pharmacology , Arenavirus/drug effects , Dengue Virus/drug effects , La Crosse virus/drug effects , Orthomyxoviridae/drug effects , Structure-Activity Relationship , Thiourea/chemical synthesis , Thiourea/pharmacology , Vaccinia virus/drug effects
12.
Antimicrob Agents Chemother ; 56(7): 3582-6, 2012 Jul.
Article En | MEDLINE | ID: mdl-22526314

ST-246, a novel compound that inhibits egress of orthopoxvirus from mammalian cells, is being tested as a treatment for pathogenic orthopoxvirus infections in humans. This phase I, double-blind, randomized, crossover, exploratory study was conducted to compare the pharmacokinetics (PK) of a single daily 400-mg oral dose of ST-246 polymorph form I versus polymorph form V administered to fed, healthy human volunteers. Both forms appeared to be well tolerated, with no serious adverse events. The order of administration of the two forms had no effect on the results of the PK analyses. Form I and form V both exhibited comparable plasma concentration versus time profiles, but complete bioequivalence between the two forms was not found. Maximum drug concentration (C(max)) met the bioequivalence criteria, as the 90% confidence interval (CI) was 80.6 to 96.9%. However, the area under the concentration-time curve from time zero to time t (AUC(0-t)) and AUC(0-∞) did not meet the bioequivalence criteria (CIs of 67.8 to 91.0% and 73.9 to 104.7%, respectively). The extent of absorption of form I, as defined by AUC(0-∞), was 11.7% lower than that of form V. Since ST-246 form I is more thermostable than form V, form I was selected for further development and use in all future studies.


Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Benzamides/administration & dosage , Benzamides/pharmacokinetics , Capsules/administration & dosage , Isoindoles/administration & dosage , Isoindoles/pharmacokinetics , Orthopoxvirus/drug effects , Administration, Oral , Adolescent , Adult , Double-Blind Method , Female , Humans , Male , Middle Aged , Young Adult
13.
PLoS One ; 6(8): e23237, 2011.
Article En | MEDLINE | ID: mdl-21858040

BACKGROUND: ST-246® is an antiviral, orally bioavailable small molecule in clinical development for treatment of orthopoxvirus infections. An intravenous (i.v.) formulation may be required for some hospitalized patients who are unable to take oral medication. An i.v. formulation has been evaluated in three species previously used in evaluation of both efficacy and toxicology of the oral formulation. METHODOLOGY/PRINCIPAL FINDINGS: The pharmacokinetics of ST-246 after i.v. infusions in mice, rabbits and nonhuman primates (NHP) were compared to those obtained after oral administration. Ten minute i.v. infusions of ST-246 at doses of 3, 10, 30, and 75 mg/kg in mice produced peak plasma concentrations ranging from 16.9 to 238 µg/mL. Elimination appeared predominately first-order and exposure dose-proportional up to 30 mg/kg. Short i.v. infusions (5 to 15 minutes) in rabbits resulted in rapid distribution followed by slower elimination. Intravenous infusions in NHP were conducted at doses of 1 to 30 mg/kg. The length of single infusions in NHP ranged from 4 to 6 hours. The pharmacokinetics and tolerability for the two highest doses were evaluated when administered as two equivalent 4 hour infusions initiated 12 hours apart. Terminal elimination half-lives in all species for oral and i.v. infusions were similar. Dose-limiting central nervous system effects were identified in all three species and appeared related to high C(max) plasma concentrations. These effects were eliminated using slower i.v. infusions. CONCLUSIONS/SIGNIFICANCE: Pharmacokinetic profiles after i.v. infusion compared to those observed after oral administration demonstrated the necessity of longer i.v. infusions to (1) mimic the plasma exposure observed after oral administration and (2) avoid C(max) associated toxicity. Shorter infusions at higher doses in NHP resulted in decreased clearance, suggesting saturated distribution or elimination. Elimination half-lives in all species were similar between oral and i.v. administration. The administration of ST-246 was well tolerated as a slow i.v. infusion.


Benzamides/pharmacokinetics , Drug Evaluation, Preclinical/methods , Isoindoles/pharmacokinetics , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Antiviral Agents/pharmacokinetics , Area Under Curve , Benzamides/administration & dosage , Benzamides/adverse effects , Biological Availability , Dose-Response Relationship, Drug , Female , Humans , Infusions, Intravenous , Isoindoles/administration & dosage , Isoindoles/adverse effects , Macaca fascicularis , Male , Metabolic Clearance Rate , Mice , Mice, Inbred BALB C , Rabbits , Time Factors , Tissue Distribution , Tremor/chemically induced
14.
Antimicrob Agents Chemother ; 54(6): 2560-6, 2010 Jun.
Article En | MEDLINE | ID: mdl-20385870

ST-246, a novel compound that inhibits egress of orthopoxvirus from infected cells, is being evaluated as a treatment for pathogenic orthopoxvirus infections in humans. This phase I, double-blind, randomized, placebo-controlled, escalating multiple-dose study was conducted to determine the safety, tolerability, and pharmacokinetics of ST-246 administered as a single daily oral dose of 250, 400, or 800 mg for 21 days to nonfasting healthy human volunteers. ST-246 appeared to be well tolerated, with no serious adverse events (AEs). Headache, for which one subject in the 800-mg group discontinued the study, was the most commonly reported AE in all treatment groups. The multiple-dose pharmacokinetics of ST-246 was well characterized. The day 21 mean elimination half-lives were calculated at 18.8, 19.8, and 20.7 h for each of the 250-, 400-, and 800-mg/day dose groups, respectively. Steady state was reached by day 6 (within 3 to 5 half-lives), saturable absorption was observed at the 800-mg dose level, and the fraction of parent drug excreted in the urine was very low. Based on these results, administration of 400 mg/day ST-246 can be expected to provide plasma concentrations above the efficacious concentration demonstrated in nonhuman primate models in earlier studies.


Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Benzamides/administration & dosage , Benzamides/pharmacokinetics , Isoindoles/administration & dosage , Isoindoles/pharmacokinetics , Orthopoxvirus/drug effects , Administration, Oral , Adolescent , Adult , Antiviral Agents/adverse effects , Benzamides/adverse effects , Double-Blind Method , Female , Half-Life , Humans , Isoindoles/adverse effects , Male , Middle Aged , Poxviridae Infections/drug therapy , Young Adult
15.
Viruses ; 2(11): 2409-2435, 2010 Nov.
Article En | MEDLINE | ID: mdl-21994624

ST-246 (Tecovirimat) is a small synthetic antiviral compound being developed to treat pathogenic orthopoxvirus infections of humans. The compound was discovered as part of a high throughput screen designed to identify inhibitors of vaccinia virus-induced cytopathic effects. The antiviral activity is specific for orthopoxviruses and the compound does not inhibit the replication of other RNA- and DNA-containing viruses or inhibit cell proliferation at concentrations of compound that are antiviral. ST-246 targets vaccinia virus p37, a viral protein required for envelopment and secretion of extracellular forms of virus. The compound is orally bioavailable and protects multiple animal species from lethal orthopoxvirus challenge. Preclinical safety pharmacology studies in mice and non-human primates indicate that ST-246 is readily absorbed by the oral route and well tolerated with the no observable adverse effect level (NOAEL) in mice measured at 2000 mg/kg and the no observable effect level (NOEL) in non-human primates measured at 300 mg/kg. Drug substance and drug product processes have been developed and commercial scale batches have been produced using Good Manufacturing Processes (GMP). Human phase I clinical trials have shown that ST-246 is safe and well tolerated in healthy human volunteers. Based on the results of the clinical evaluation, once a day dosing should provide plasma drug exposure in the range predicted to be antiviral based on data from efficacy studies in animal models of orthopoxvirus disease. These data support the use of ST-246 as a therapeutic to treat pathogenic orthopoxvirus infections of humans.

16.
Antimicrob Agents Chemother ; 53(5): 1817-22, 2009 May.
Article En | MEDLINE | ID: mdl-19223621

Therapeutics for the treatment of pathogenic orthopoxvirus infections are being sought. In the absence of patients with disease, animal models of orthopoxvirus disease are essential for evaluation of the efficacies of antiviral drugs and establishment of the appropriate dose and duration of human therapy. Infection of nonhuman primates (NHP) by the intravenous injection of monkeypox virus has been used to evaluate a promising therapeutic drug candidate, ST-246. ST-246 administered at 3 days postinfection (which corresponds to the secondary viremia stage of disease) at four different doses (from 100 mg/kg of body weight down to 3 mg/kg) once a day for 14 days was able to offer NHP 100% protection from a lethal infection with monkeypox virus and reduce the viral load and lesion formation. In NHP, the administration of ST-246 at a dose of 10 mg/kg/day for 14 days resulted in levels of blood exposure comparable to the levels attained in humans administered 400 mg in the fed state. These results suggest that administration of an oral dosage of 400 mg once daily for 14 days will be effective for the prevention or treatment of smallpox or monkeypox infections in humans.


Antiviral Agents , Benzamides , Isoindoles , Monkeypox virus/drug effects , Mpox (monkeypox)/drug therapy , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Benzamides/administration & dosage , Benzamides/pharmacokinetics , Benzamides/therapeutic use , Disease Models, Animal , Dose-Response Relationship, Drug , Humans , Isoindoles/administration & dosage , Isoindoles/pharmacokinetics , Isoindoles/therapeutic use , Macaca fascicularis , Mpox (monkeypox)/mortality , Mpox (monkeypox)/virology , Treatment Outcome
17.
Antimicrob Agents Chemother ; 52(5): 1721-7, 2008 May.
Article En | MEDLINE | ID: mdl-18316519

ST-246 is a novel, potent orthopoxvirus egress inhibitor that is being developed to treat pathogenic orthopoxvirus infections of humans. This phase I, double-blind, randomized, placebo-controlled single ascending dose study (first time with humans) was conducted to determine the safety, tolerability, and pharmacokinetics of ST-246 in healthy human volunteers. ST-246 was administered in single oral doses of 500, 1,000, and 2,000 mg to fasting healthy volunteers and 1,000 mg to nonfasting healthy volunteers. ST-246 was generally well tolerated with no serious adverse events, and no subject was withdrawn from the study due to ST-246. The most commonly reported drug-related adverse event was neutropenia, which was found, upon further analysis, not to be treatment related. ST-246 was readily absorbed following oral administration with mean times to maximum concentration from 2 h to 3 h. Absorption was greater in nonfasting volunteers than in fasting volunteers. Administration of ST-246 resulted in exposure levels predicted to be sufficient for inhibiting orthopoxvirus replication compared to exposure levels in nonhuman primates in which ST-246 protected animals from lethal orthopoxvirus infection.


Benzamides/pharmacokinetics , Isoindoles/pharmacokinetics , Orthopoxvirus/drug effects , Administration, Oral , Adult , Antiviral Agents/administration & dosage , Antiviral Agents/pharmacokinetics , Area Under Curve , Benzamides/adverse effects , Benzamides/blood , Dose-Response Relationship, Drug , Double-Blind Method , Fasting , Humans , Isoindoles/adverse effects , Isoindoles/blood , Metabolic Clearance Rate , Neutropenia/chemically induced , Orthopoxvirus/genetics
18.
Vaccine ; 26(7): 933-46, 2008 Feb 13.
Article En | MEDLINE | ID: mdl-18226434

The re-emerging threat of smallpox and the emerging threat of monkeypox highlight the need for effective poxvirus countermeasures. Currently approved smallpox vaccines have unacceptable safety profiles and, consequently, the general populace is no longer vaccinated, leading to an increasingly susceptible population. ST-246, a small-molecule inhibitor of poxvirus dissemination, has been demonstrated in various animal models to be safe and effective in preventing poxviral disease. This suggests that it may also be used to improve the safety of the traditional smallpox vaccine provided that it does not inhibit vaccine-induced protective immunity. In this study, we compared the immune responses elicited by the smallpox vaccine alone or in combination with ST-246 in mice. Normal lesion formation following dermal scarification with the attenuated New York City Board of Health strain (Dryvax), commonly referred to as a vaccine "take", was not inhibited although severe lesions and systemic disease due to vaccination with the virulent Western Reserve (VV-WR) strain were prevented. The vaccine given with ST-246 did not affect cellular immune responses or neutralizing antibody titers although anti-vaccinia ELISA titers were slightly reduced. Vaccination in combination with ST-246 provided equivalent short- and long-term protection against lethal intranasal challenge with VV-WR when compared to vaccine alone. These results suggest that ST-246 does not compromise protective immunity elicited by the vaccine and provide the basis for future studies examining the efficacy of ST-246 in preventing or treating adverse events due to vaccination.


Benzamides/immunology , Isoindoles/immunology , Smallpox Vaccine/immunology , Vaccinia virus/immunology , Vaccinia/immunology , Animals , Antibodies, Viral/blood , Benzamides/administration & dosage , Benzamides/pharmacology , Cytokines/metabolism , Drug Therapy, Combination , Female , Immunoglobulin G/blood , Isoindoles/administration & dosage , Isoindoles/pharmacology , Lymphocyte Activation , Mice , Mice, Inbred BALB C , Neutralization Tests , Smallpox Vaccine/administration & dosage , Time Factors , Vaccination , Vaccinia/prevention & control , Virus Replication/drug effects
...