Development of ST-246® for Treatment of Poxvirus Infections

Robert Jordan, Janet M Leeds, Shanthakumar Tyavanagimatt, Dennis E Hruby, Robert Jordan, Janet M Leeds, Shanthakumar Tyavanagimatt, Dennis E Hruby

Abstract

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.

Keywords: ST-246; Smallpox; Tecovirimat; antiviral drug; egress inhibitor; orthopoxvirus; p37.

Figures

Figure 1.
Figure 1.
Structure activity relationships and chemical information for ST-246. A summary of antiviral activity of chemical analogs of ST-246 (adapted from Bailey et al. [18]). EWG—Electron withdrawing groups; EDG—Electron donating groups. R refers to modifications of the phenyl ring.
Figure 2.
Figure 2.
ST-246 inhibits production of extracellular virus and systemic virus spread in vitro and in vivo. (A) Diagram showing the four infectious forms of vaccinia virus and viral genes required for each step in the morphogenesis process. Vaccinia virus genes involved in each step in the pathway are shown (adapted from Smith et al. [23]). (B) ST-246 inhibits extracellular virus (CEV and EEV) formation. BSC-40 cell monolayers were infected with vaccinia virus at 5 pfu/cell in the presence and absence of 10 μM ST-246. The cell monolayers were radiolabeled with 35-S methionine and vaccinia virus particles, either cell-associated (upper graph) or released into the culture medium (lower graph) were fractionated by equilibrium centrifugation on cesium chloride gradients. The radiolabeled material in each fraction was quantified by liquid scintillation. The assignment of each type of virus particle was based upon their reported density (adapted from Chen et al.) [48] (C) ST-246 inhibits plaque formation. BSC-40 cell monolayers (1 × 106 cells/well) were infected with 10-fold serial dilutions of vaccinia virus in the presence and absence of 5 μM ST-246. At 3 days post-infection, the cultures were fixed in 5% glutaraldehyde and stained with crystal violet to visualize plaques. (D) ST-246 protects mice from systemic disease. Mice were inoculated with a lethal dose of vaccinia virus (WR) via the intranasal route. ST-246 was administered at 100 mg/kg as a liquid suspension by oral gavage once per day for 14 days. Shown are mice treated with ST-246 or placebo at day 8 post-infection.
Figure 3.
Figure 3.
Dose normalized exposure of ST-246 in BALB/c mice from Day 1 of multiple studies. Dose normalized area-under-the curve values are plotted versus dose for mouse toxicology studies. The results suggest that absorption limits increased exposure as the dose is increased. AUCinf = Area under concentration time curve for plasma drug levels after oral dosing from time 0 extrapolated to infinite time.
Figure 4.
Figure 4.
Correlation between maximum monkeypox viral DNA levels in the blood versus maximum lesion number in non-human primates infected with monkeypox virus. Viral DNA levels were determined by quantitative PCR assay. Maximum viral DNA levels and lesion number were determined over a period of 21 days starting at the time of infection (Day 0). All doses were administered by oral gavage. Dose levels ranged from 0.3 mg/kg/day to 300 mg/kg/day for 14 days. Treatment was initiated on Day 1 (E3), Day 5 (E6), Days 3 or 4 (21G) or lesion onset on Day 3 or 4 (26G). Studies 21G and 26G were randomized, double blind, placebo-controlled studies conducted in compliance with GLP guidelines. Pearson’s correlation coefficient was calculated to show the degree of correlation.
Figure 5.
Figure 5.
Efficacy of ST-246 in a non-human primate model of monkeypox virus infection. Cynomolgus macaques were infected with 5 × 107 pfu of monkeypox virus via intravenous inoculation. At 3 days post-inoculation, ST-246 was administered by oral gavage once per day for 14 consecutive days. Viral DNA in the blood was determined by quantitative PCR. Viral DNA and lesion counts were measured every day for 21 days (adapted from Jordan et al.) [63]. (A) The maximum viral DNA levels in the blood and lesion number for individual animals over the course of the experiment were plotted per treatment group. The average value (bar) for each treatment group is shown. (B) Daily lesion numbers and viral DNA levels in the blood were shown for vehicle-treated animals (blue circles) and ST-246-treated animals at 3 mg /k g (red squares) with the average values plotted as a single black line. Cross symbols indicate day of death. The dashed horizontal line shows the limit of quantification of viral DNA.
Figure 6.
Figure 6.
A comparison of ST-246 exposure (area under the concentration time curve, AUC) in monkeys and humans. Monkey doses were converted to human equivalent doses by multiplying first by 0.32 in accordance with FDA Guidance for Industry (entitled Estimating the Maximum Safe Starting Dose in Initial Clinical Trials for Therapeutics in Adult Healthy Volunteers, published July 2005) and then multiplying by the average weight of human (78.6 kg) from the clinical study. The maximum, minimum and mean AUC for monkeys is shown in the green triangles, brown squares, and blue diamonds, respectively. The range in AUC values in humans is indicated by the symbols. The red dashed line indicates the dose of ST-246 that protects non-human primates from lethal infection.

References

    1. Fenner F, Henderson DA, Arita I, Jazek Z, Ladnyi ID. Smallpox and Its Eradication. World Health Organization; Geneva, Switzerland: 1988. pp. 1–1421.
    1. Parker S, Nuara A, Buller RM, Schultz DA. Human monkeypox: An emerging zoonotic disease. Future Microbiol. 2007;2:17–34.
    1. Mukinda VB, Mwema G, Kilundu M, Heymann DL, Khan AS, Esposito JJ. Re-emergence of human monkeypox in Zaire in 1996. Monkeypox Epidemiologic Working Group. Lancet. 1997;349:1449–1450.
    1. Monath TP, Caldwell JR, Mundt W, Fusco J, Johnson CS, Buller M, Liu J, Gardner B, Downing G, Blum PS, Kemp T, Nichols R, Weltzin R. ACAM2000 clonal Vero cell culture vaccinia virus (New York City Board of Health strain)—A second-generation smallpox vaccine for biological defense. Int J Infect Dis. 2004;8:S31–S44.
    1. Baxby D, Bennett M, Getty B. Human cowpox 1969–93: A review based on 54 cases. Br J Dermatol. 1994;131:598–607.
    1. Vorou RM, Papavassiliou VG, Pierroutsakos IN. Cowpox virus infection: An emerging health threat. Curr Opin Infect Dis. 2008;21:153–156.
    1. Essbauer S, Pfeffer M, Meyer H. Zoonotic poxviruses. Vet Microbiol. 2010;140:229–236.
    1. Reynolds MG, Carroll DS, Olson VA, Hughes C, Galley J, Likos A, Montgomery JM, Suu-Ire R, Kwasi MO, Root JJ, Braden Z, Abel J, Clemmons C, Regnery R, Karem K, Damon IK. A silent enzootic of an orthopoxvirus in Ghana, West Africa: evidence for multi-species involvement in the absence of widespread human disease. Am J Trop Med Hyg. 2010;82:746–754.
    1. Vora S, Damon I, Fulginiti V, Weber SG, Kahana M, Stein SL, Gerber SI, Garcia-Houchins S, Lederman E, Hruby D, Collins L, Scott D, Thompson K, Barson JV, Regnery R, Hughes C, Daum RS, Li Y, Zhao H, Smith S, Braden Z, Karem K, Olson V, Davidson W, Trindade G, Bolken T, Jordan R, Tien D, Marcinak J. Severe eczema vaccinatum in a household contact of a smallpox vaccinee. Clin Infect Dis. 2008;46:1555–1561.
    1. Fulginiti VA, Papier A, Lane JM, Neff JM, Henderson DA. Smallpox vaccination: A review, part II. Adverse events. Clin Infect Dis. 2003;37:251–271.
    1. Bray M. Pathogenesis and potential antiviral therapy of complications of smallpox vaccination. Antivir Res. 2003;58:101–114.
    1. Lane JM, Ruben FL, Neff JM, Millar JD. Complications of smallpox vaccination, 1968: Results of ten statewide surveys. J Infect Dis. 1970;122:303–309.
    1. Gubser C, Hue S, Kellam P, Smith GL. Poxvirus genomes: A phylogenetic analysis. J Gen Virol. 2004;85:105–117.
    1. Johnston JB, McFadden G. Poxvirus immunomodulatory strategies: Current perspectives. J Virol. 2003;77:6093–6100.
    1. McLysaght A, Baldi PF, Gaut BS. Extensive gene gain associated with adaptive evolution of poxviruses. Proc Natl Acad Sci U S A. 2003;100:15655–15660.
    1. Matz-Rensing K, Ellerbrok H, Ehlers B, Pauli G, Floto A, Alex M, Czerny CP, Kaup FJ. Fatal poxvirus outbreak in a colony of New World monkeys. Vet Pathol. 2006;43:212–218.
    1. Yang G, Pevear DC, Davies MH, Collett MS, Bailey T, Rippen S, Barone L, Burns C, Rhodes G, Tohan S, Huggins JW, Baker RO, Buller RL, Touchette E, Waller K, Schriewer J, Neyts J, DeClercq E, Jones K, Hruby D, Jordan R. An orally bioavailable antipoxvirus compound (ST-246) inhibits extracellular virus formation and protects mice from lethal orthopoxvirus Challenge. J Virol. 2005;79:13139–13149.
    1. Bailey TR, Rippin SR, Opsitnick E, Burns CJ, Pevear DC, Collett MS, Rhodes G, Tohan S, Huggins JW, Baker RO, Kern ER, Keith KA, Dai D, Yang G, Hruby D, Jordan R. N-(3,3a,4,4a,5,5a,6,6a-Octahydro-1,3-dioxo-4,6–ethenocycloprop[f]isoindol-2-(1H)-yl)carboxamides: Identification of novel orthopoxvirus egress inhibitors. J Med Chem. 2007;50:1442–1444.
    1. Duraffour S, Snoeck R, de Vos R, van Den Oord JJ, Crance JM, Garin D, Hruby DE, Jordan R, De, Clercq E, Andrei G. Activity of the anti-orthopoxvirus compound ST-246 against vaccinia, cowpox and camelpox viruses in cell monolayers and organotypic raft cultures. Antivir Ther. 2007;12:1205–1216.
    1. Smith SK, Olson VA, Karem KL, Jordan R, Hruby DE, Damon IK. In vitro efficacy of ST246 against smallpox and monkeypox. Antimicrob Agents Chemother. 2009;53:1007–1012.
    1. Jordan R. 2010. Siga Technologies, Inc., Corvallis, OR, USA. Unpublished work.
    1. Moss B. Poxviridae and Their Replication. 4th ed. Lippincott-Raven; Philadelphia, PA, USA: 2001. pp. 2849–2884.
    1. Smith GL, Vanderplasschen A, Law M. The formation and function of extracellular enveloped vaccinia virus. J Gen Virol. 2002;83:2915–2931.
    1. Byrd CM, Bolken TC, Hruby DE. The vaccinia virus I7L gene product is the core protein proteinase. J Virol. 2002;76:8973–8976.
    1. Blasco R, Moss B. Extracellular vaccinia virus formation and cell-to-cell virus transmission are prevented by deletion of the gene encoding the 37,000-Dalton outer envelope protein. J Virol. 1991;65:5910–5920.
    1. Roper RL, Wolffe EJ, Weisberg A, Moss B. The envelope protein encoded by the A33R gene is required for formation of actin-containing microvilli and efficient cell-to-cell spread of vaccinia virus. J Virol. 1998;72:4192–4204.
    1. Cudmore S, Cossart P, Griffiths G, Way M. Actin-based motility of vaccinia virus. Nature. 1995;378:636–638.
    1. Payne LG. Significance of extracellular enveloped virus in the in vitro and in vivo dissemination of vaccinia. J Gen Virol. 1980;50:89–100.
    1. Payne LG, Kristensson K. Extracellular release of enveloped vaccinia virus from mouse nasal epithelial cells in vivo. J Gen Virol. 1985;66:643–646.
    1. McIntosh AA, Smith GL. Vaccinia virus glycoprotein A34R is required for infectivity of extracellular enveloped virus. J Virol. 1996;70:272–281.
    1. Stern RJ, Thompson JP, Moyer RW. Attenuation of B5R mutants of rabbitpox virus in vivo is related to impaired growth and not an enhanced host inflammatory response. Virology. 1997;233:118–129.
    1. Zhang WH, Wilcock D, Smith GL. Vaccinia Virus F12L Protein Is Required for Actin Tail Formation, Normal Plaque Size, and Virulence. J Virol. 2000;74:11654–11662.
    1. Boulter EA, Zwartouw HT, Titmuss DH, Maber HB. The nature of the immune state produced by inactivated vaccinia virus in rabbits. Am J Epidemiol. 1971;94:612–620.
    1. Reeves PM, Bommarius B, Lebeis S, McNulty S, Christensen J, Swimm A, Chahroudi A, Chavan R, Feinberg MB, Veach D, Bornmann W, Sherman M, Kalman D. Disabling poxvirus pathogenesis by inhibition of Abl-family tyrosine kinases. Nat Med. 2005;11:731–739.
    1. Yang H, Kim SK, Kim M, Reche PA, Morehead TJ, Damon IK, Welsh RM, Reinherz EL. Antiviral chemotherapy facilitates control of poxvirus infections through inhibition of cellular signal transduction. J Clin Invest. 2005;115:379–387.
    1. Vanderplasschen A, Mathew E, Hollinshead M, Sim RB, Smith GL. Extracellular enveloped vaccinia virus is resistant to complement because of incorporation of host complement control proteins into its envelope. Proc Natl Acad Sci U S A. 1998;95:7544–7549.
    1. Vanderplasschen A, Hollinshead M, Smith GL. Antibodies against vaccinia virus do not neutralize extracellular enveloped virus but prevent virus release from infected cells and comet formation. J Gen Virol. 1997;78(Pt. 8):2041–2048.
    1. Husain M, Weisberg A, Moss B. Topology of epitope-tagged F13L protein, a major membrane component of extracellular vaccinia virions. Virology. 2003;308:233–242.
    1. Hiller G, Weber K. Golgi-derived membranes that contain an acylated viral polypeptide are used for vaccinia virus envelopment. J Virol. 1985;55:651–659.
    1. Husain M, Moss B. Similarities in the induction of post-Golgi vesicles by the vaccinia virus F13L protein and phospholipase D. J Virol. 2002;76:7777–7789.
    1. Husain M, Moss B. Vaccinia virus F13L protein with a conserved phospholipase catalytic motif induces colocalization of the B5R envelope glycoprotein in post-Golgi vesicles. J Virol. 2001;75:7528–7542.
    1. Schweizer A, Kornfeld S, Rohrer J. Proper sorting of the cation-dependent mannose 6-phosphate receptor in endosomes depends on a pair of aromatic amino acids in its cytoplasmic tail. Proc Natl Acad Sci U S A. 1997;94:14471–14476.
    1. Orsel JG, Sincock PM, Krise JP, Pfeffer SR. Recognition of the 300-kDa mannose 6-phosphate receptor cytoplasmic domain by 47-kDa tail-interacting protein. Proc Natl Acad Sci U S A. 2000;97:9047–9051.
    1. Carroll KS, Hanna J, Simon I, Krise J, Barbero P, Pfeffer SR. Role of Rab9 GTPase in facilitating receptor recruitment by TIP47. Science. 2001;292:1373–1376.
    1. Diaz E, Pfeffer SR. TIP47: A cargo selection device for mannose 6-phosphate receptor trafficking. Cell. 1998;93:433–443.
    1. Blot G, Janvier K, Le Panse S, Benarous R, Berlioz-Torrent C. Targeting of the human immunodeficiency virus type 1 envelope to the trans-Golgi network through binding to TIP47 is required for env incorporation into virions and infectivity. J Virol. 2003;77:6931–6945.
    1. Lopez-Verges S, Camus G, Blot G, Beauvoir R, Benarous R, Berlioz-Torrent C. Tail-interacting protein TIP47 is a connector between Gag and Env and is required for Env incorporation into HIV-1 virions. Proc Natl Acad Sci U S A. 2006;103:14947–14952.
    1. Murray JL, Mavrakis M, McDonald NJ, Yilla M, Sheng J, Bellini WJ, Zhao L, Le Doux JM, Shaw MW, Luo CC, Lippincott-Schwartz J, Sanchez A, Rubin DH, Hodge TW. Rab9 GTPase is required for replication of human immunodeficiency virus type 1, filoviruses, and measles virus. J Virol. 2005;79:11742–11751.
    1. Chen Y, Honeychurch KM, Yang G, Byrd CM, Harver C, Hruby DE, Jordan R. Vaccinia virus p37 interacts with host proteins associated with LE-derived transport vesicle biogenesis. Virol J. 2009;6:44. doi: 10.1186/1743-422X-6-44..
    1. Davies B, Morris T. Physiological parameters in laboratory animals and humans. Pharm Res. 1993;10:1093–1095.
    1. Breman JG, Henderson DA. Diagnosis and management of smallpox. N Engl J Med. 2002;346:1300–1308.
    1. Sofi Ibrahim M, Kulesh DA, Saleh SS, Damon IK, Esposito JJ, Schmaljohn AL, Jahrling PB. Real-Time PCR Assay To Detect Smallpox Virus. J Clin Microbiol. 2003;41:3835–3839.
    1. Hooper JW, Thompson E, Wilhelmsen C, Zimmerman M, Ichou MA, Steffen SE, Schmaljohn CS, Schmaljohn AL, Jahrling PB. Smallpox DNA vaccine protects nonhuman primates against lethal monkeypox. J Virol. 2004;78:4433–4443.
    1. Kim M, Yang H, Kim SK, Reche PA, Tirabassi RS, Hussey RE, Chishti Y, Rheinwald JG, Morehead TJ, Zech T, Damon IK, Welsh RM, Reinherz EL. Biochemical and Functional Analysis of Smallpox Growth Factor (SPGF) and Anti-SPGF Monoclonal Antibodies. J Biol Chem. 2004;279:25838–25848.
    1. Bras G. The morbid anatomy of smallpox. Doc Med Geogr Trop. 1952;4:303–351.
    1. Smallpox Case Definitions. As described in “Guide A” of the Smallpox Response Plan and Guidelines, Coordinating Center for Infectious Diseases (CCID) National Center for Preparedness, Detection, and Control of Infectious Diseases (NCPDCID) Division of Bioterrorism Preparedness and Response (DBPR) 2008.
    1. Zaucha GM, Jahrling PB, Geisbert TW, Swearengen JR, Hensley L. The pathology of experimental aerosolized monkeypox virus infection in cynomolgus monkeys (Macaca fascicularis) Lab Invest. 2001;81:1581–1600.
    1. Stagles MJ, Watson AA, Boyd JF, More IA, McSeveney D. The histopathology and electron microscopy of a human monkeypox lesion. Trans R Soc Trop Med Hyg. 1985;79:192–202.
    1. Fenner F, Buller RML. Mousepox. In: Nathanson N, editor. Viral Pathogenesis. Lippincott Raven; Philadelphia, PA, USA: 1997. pp. 535–553.
    1. Westwood JC, Boulter EA, Bowen ET, Maber HB. Experimental respiratory infection with poxviruses. I. Clinical virological and epidemiological studies. Br J Exp Pathol. 1966;47:453–465.
    1. Stittelaar KJ, Neyts J, Naesens L, van Amerongen G, van Lavieren RF, Holy A, De Clercq E, Niesters HG, Fries E, Maas C, Mulder PG, van der Zeijst BA, Osterhaus AD. Antiviral treatment is more effective than smallpox vaccination upon lethal monkeypox virus infection. Nature. 2006;439:745–748.
    1. Stittelaar KJ, van Amerongen G, Kondova I, Kuiken T, van Lavieren RF, Pistoor FH, Niesters HG, van Doornum G, van der Zeijst BA, Mateo L, Chaplin PJ, Osterhaus AD. Modified vaccinia virus Ankara protects macaques against respiratory challenge with monkeypox virus. J Virol. 2005;79:7845–7851.
    1. Huggins J, Goff A, Hensley L, Mucker E, Shamblin J, Wlazlowski C, Johnson W, Chapman J, Larsen T, Twenhafel N, Karem K, Damon IK, Byrd CM, Bolken TC, Jordan R, Hruby D. Nonhuman primates are protected from smallpox virus or monkeypox virus challenges by the antiviral drug ST-246. Antimicrob Agents Chemother. 2009;53:2620–2625.
    1. Jordan R, Goff A, Frimm A, Corrado ML, Hensley LE, Byrd CM, Mucker E, Shamblin J, Bolken TC, Wlazlowski C, Johnson W, Chapman J, Twenhafel N, Tyavanagimatt S, Amantana A, Chinsangaram J, Hruby DE, Huggins J. ST-246 antiviral efficacy in a nonhuman primate monkeypox model: Determination of the minimal effective dose and human dose justification. Antimicrob Agents Chemother. 2009;53:1817–1822.
    1. Huggins J. United States Army Institute of Infectious Disease (USAMRIID), Frederick, MD, USA. 2010. Personal communication.
    1. Esteban DJ, Buller RM. Ectromelia virus: The causative agent of mousepox. J Gen Virol. 2005;86:2645–2659.
    1. Martinez MJ, Bray MP, Huggins JW. A mouse model of aerosol-transmitted orthopoxviral disease: Morphology of experimental aerosol-transmitted orthopoxviral disease in a cowpox virus-BALB/c mouse system. Arch Pathol Lab Med. 2000;124:362–377.
    1. Smee DF. Progress in the discovery of compounds inhibiting orthopoxviruses in animal models. Antivir Chem Chemother. 2008;19:115–124.
    1. Quenelle DC, Buller RM, Parker S, Keith KA, Hruby DE, Jordan R, Kern ER. Efficacy of delayed treatment with ST-246 given orally against systemic orthopoxvirus infections in mice. Antimicrob Agents Chemother. 2007;51:689–695.
    1. Nalca A, Hatkin JM, Garza NL, Nichols DK, Norris SW, Hruby DE, Jordan R. Evaluation of orally delivered ST-246 as postexposure prophylactic and antiviral therapeutic in an aerosolized rabbitpox rabbit model. Antivir Res. 2008;79:121–127.
    1. Sbrana E, Jordan R, Hruby DE, Mateo RI, Xiao SY, Siirin M, Newman PC, Da Rosa AP, Tesh RB. Efficacy of the antipoxvirus compound ST-246 for treatment of severe orthopoxvirus infection. Am J Trop Med Hyg. 2007;76:768–773.
    1. Berhanu A, King DS, Mosier S, Jordan R, Jones KF, Hruby DE, Grosenbach DW. ST-246 inhibits in vivo poxvirus dissemination, virus shedding, and systemic disease manifestation. Antimicrob Agents Chemother. 2009;53:4999–5009.
    1. Grosenbach DW, Berhanu A, King DS, Mosier S, Jones KF, Jordan RA, Bolken TC, Hruby DE. Efficacy of ST-246 versus lethal poxvirus challenge in immunodeficient mice. Proc Natl Acad Sci U S A. 2010;107:838–843.
    1. Grosenbach DW, Jordan R, King DS, Berhanu A, Warren TK, Kirkwood-Watts DL, Tyavanagimatt S, Tan Y, Wilson RL, Jones KF, Hruby DE. Immune responses to the smallpox vaccine given in combination with ST-246, a small-molecule inhibitor of poxvirus dissemination. Vaccine. 2008;26:933–946.
    1. Jahrling PB, Hensley LE, Martinez MJ, LeDuc JW, Rubins KH, Relman DA, Huggins JW. Exploring the potential of variola virus infection of cynomolgus macaques as a model for human smallpox. Proc Natl Acad Sci U S A. 2004;101:15196–15200.
    1. Jordan R. 2010. Siga Technologies, Inc., Corvallis, OR, USA. Double blind, randomized, placebo-controlled, repeat-dose efficacy study of the therapeutic window of the proposed primate equivalence of the human dose (400 mg/day) of oral ST-246 polyform I in cynomolgus monkeys infected with monkeypox virus. Unpublished work.
    1. Jordan R. 2010. Siga Technologies, Inc., Corvallis, OR, USA. Double blind, randomized, placebo-controlled, repeat-dose efficacy study of the therapeutic window of the proposed primate equivalence of the humand dose (400 mg/day) of oral ST-246 polyform I in cynomolgus monkeys infected with variola virus. Unpublished work.
    1. Jordan R, Tien D, Bolken TC, Jones KF, Tyavanagimatt SR, Strasser J, Frimm A, Corrado ML, Strome PG, Hruby DE. Single-dose safety and pharmacokinetics of ST-246, a novel orthopoxvirus egress inhibitor. Antimicrob Agents Chemother. 2008;52:1721–1727.
    1. Jordan R, Chinsangaram J, Bolken TC, Tyavanagimatt SR, Tien D, Jones KF, Frimm A, Corrado ML, Pickens M, Landis P, Clarke J, Marbury TC, Hruby DE. Safety and pharmacokinetics of the antiorthopoxvirus compound ST-246 following repeat oral dosing in healthy adult subjects. Antimicrob Agents Chemother. 2010;54:2560–2566.
    1. Centers for Disease Control and Prevention (CDC) Progressive vaccinia in a military smallpox vaccinee—United States, 2009. MMWR Morb Mortal Wkly Rep. 2009;58:532–536.

Source: PubMed

3
Subscribe