Single- and Multiple-Ascending-Dose Study of the Safety, Tolerability, and Pharmacokinetics of the Polymyxin Derivative SPR206

Jon Bruss, Troy Lister, Vipul K Gupta, Emily Stone, Lisa Morelli, Yang Lei, David Melnick, Jon Bruss, Troy Lister, Vipul K Gupta, Emily Stone, Lisa Morelli, Yang Lei, David Melnick

Abstract

Carbapenem-resistant Acinetobacter baumannii and Enterobacterales are identified as urgent threats, and multidrug-resistant (MDR) Pseudomonas aeruginosa and extended-spectrum beta-lactamase (ESBL)-producing pathogens are identified as serious threats by the Centers for Disease Control and Prevention (CDC). SPR206 is a novel polymyxin derivative with potent in vitro and in vivo activity against A. baumannii, P. aeruginosa, and multiple clinically important species of Enterobacterales, including multidrug- and extensively drug-resistant strains. This was a first-in-human (FIH) double-blind, placebo-controlled, single-, and multiple-ascending-dose study of the safety, tolerability, and pharmacokinetics (PK) of SPR206 in 94 healthy subjects. Following intravenous (i.v.) administration (1-h infusion) at single doses of 10 mg to 400 mg and multiple doses of 25 mg to 150 mg every 8 h (q8h) for 7 days and 100 mg q8h for 14 days, SPR206 was generally safe and generally well tolerated. While the incidence of adverse events increased with dose, most were of mild severity. Systemic exposure (maximum concentration of drug in serum [Cmax] and area under the concentration-time curve [AUC]) to SPR206 was approximately dose proportional, time to peak concentrations ranged from 1.1 to 1.3 h, and half-life ranged from 2.4 to 4.1 h. No appreciable accumulation occurred with repeated dosing of SPR206, and trough concentrations suggest that steady state was achieved by day 2. Urinary excretion of unchanged SPR206 was dose dependent across single- (SAD) and multiple-ascending-dose (MAD) cohorts, and the percentage of dose excreted as SPR206 was up to >50%. Importantly, no evidence of nephrotoxicity was observed over 14 days of 100 mg q8h dosing of SPR206; a dosing regimen anticipated to exceed requirements for clinical efficacy. (This study has been registered at ClinicalTrials.gov under identifier NCT03792308.).

Keywords: antimicrobial safety; pharmacokinetics; polymyxins.

Figures

FIG 1
FIG 1
Chemical structure of SPR206.
FIG 2
FIG 2
Study design.
FIG 3
FIG 3
Mean (standard deviation) serum creatinine values over time with multiple ascending doses of SPR206 (safety population).
FIG 4
FIG 4
Mean (standard deviation) plasma SPR206 concentrations after single ascending doses (PK population).
FIG 5
FIG 5
Mean cumulative amount of SPR206 excreted in urine after single ascending doses (PK population).
FIG 6
FIG 6
Mean (standard deviation) plasma SPR206 plasma concentrations following multiple ascending doses on day 1, day 7, and day 14 (PK population).
FIG 7
FIG 7
Mean (standard deviation) SPR206 trough concentrations following 7-day and 14-day administration.

References

    1. World Health Organization. 2017. Global priority list of antibiotic-resistant bacteria to guide research, discovery, and development of new antibiotics. World Health Organization, Geneva, Switzerland. .
    1. Centers for Disease Control and Prevention. 2019. Antibiotic resistance threats in the United States, 2019. Department of Health and Human Services, Centers for Disease Control and Prevention, Atlanta, GA.
    1. Cai B, Echols R, Magee G, Arjona Ferreira JC, Morgan G, Ariyasu M, Sawada T, Nagata TD. 2017. Prevalence of carbapenem-resistant Gram-negative infections in the United States predominated by Acinetobacter baumannii and Pseudomonas aeruginosa. Open Forum Infect Dis 4:ofx176. 10.1093/ofid/ofx176.
    1. Sievert DM, Ricks P, Edwards JR, Schneider A, Patel J, Srinivasan A, Kallen A, Limbago B, Fridkin S, National Healthcare Safety Network (NHSN) Team and Participating NHSN Facilities. 2013. Antimicrobial-resistant pathogens associated with healthcare-associated infections: summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2009–2010. Infect Control Hosp Epidemiol 34:1–14. 10.1086/668770.
    1. Boral B, Unaldi Ö, Ergin A, Durmaz R, Eser ÖK, Acinetobacter Study Group. 2019. A prospective multicenter study on the evaluation of antimicrobial resistance and molecular epidemiology of multidrug-resistant Acinetobacter baumannii infections in intensive care units with clinical and environmental features. Ann Clin Microbiol Antimicrob 18:19. 10.1186/s12941-019-0319-8.
    1. Bulens SN, Yi SH, Walters MS, Jacob JT, Bower C, Reno J, Wilson L, Vaeth E, Bamberg W, Janelle SJ, Lynfield R, Vagnone PS, Shaw K, Kainer M, Muleta D, Mounsey J, Dumyati G, Concannon C, Beldavs Z, Cassidy PM, Phipps EC, Kenslow N, Hancock EB, Kallen AJ. 2018. Carbapenem-nonsusceptible Acinetobacter baumannii, 8 US Metropolitan areas, 2012–2015. Emerg Infect Dis 24:727–734. 10.3201/eid2404.171461.
    1. Gales AC, Seifert H, Gur D, Castanheira M, Jones RN, Sader HS. 2019. Antimicrobial susceptibility of Acinetobacter calcoaceticus-Acinetobacter baumannii complex and Stenotrophomonas maltophilia clinical isolates: results from the SENTRY Antimicrobial Surveillance Program (1997–2016). Open Forum Infect Dis 6:S34–S46. 10.1093/ofid/ofy293.
    1. LynchJP, III, Zhanel GG, Clark NM. 2017. Infections due to Acinetobacter baumannii in the ICU: treatment options. Semin Respir Crit Care Med 38:311–325. 10.1055/s-0037-1599225.
    1. Cheng A, Chuang Y-C, Sun H-Y, Sheng W-H, Yang C-J, Liao C-H, Hsueh P-R, Yang J-L, Shen N-J, Wang J-T, Hung C-C, Chen Y-C, Chang S-C. 2015. Excess mortality associated with colistin-tigecycline compared with colistin-carbapenem combination therapy for extensively drug-resistant Acinetobacter baumannii bacteremia: a multicenter prospective observational study. Crit Care Med 43:1194–1204. 10.1097/CCM.0000000000000933.
    1. Clark NM, Zhanel GG, Lynch JP. 2016. Emergence of antimicrobial resistance among Acinetobacter species: a global threat. Curr Opin Crit Care 22:491–499. 10.1097/MCC.0000000000000337.
    1. Lemos EV, de la Hoz FP, Einarson TR, McGhan WF, Quevedo E, Castañeda C, Kawai K. 2014. Carbapenem resistance and mortality in patients with Acinetobacter baumannii infection: systematic review and meta-analysis. Clin Microbiol Infect 20:416–423. 10.1111/1469-0691.12363.
    1. Spellberg B, Bonomo RA. 2015. Combination therapy for extreme drug-resistant (XDR) Acinetobacter baumannii: ready for prime-time? Crit Care Med 43:1332–1334. 10.1097/CCM.0000000000001029.
    1. Wong D, Nielsen TB, Bonomo RA, Pantapalangkoor P, Luna B, Spellberg B. 2017. Clinical and pathophysiological overview of Acinetobacter infections: a century of challenges. Clin Microbiol Rev 30:409–447. 10.1128/CMR.00058-16.
    1. Zilberberg MD, Kollef MH, Shorr AF. 2016. Secular trends in Acinetobacter baumannii resistance in respiratory and blood stream specimens in the United States, 2003 to 2012: a survey study. J Hosp Med 11:21–26. 10.1002/jhm.2477.
    1. Zilberberg MD, Nathanson BH, Sulham K, Fan W, Shorr AF. 2016. Multidrug resistance, inappropriate empiric therapy, and hospital mortality in Acinetobacter baumannii pneumonia and sepsis. Crit Care 20:221. 10.1186/s13054-016-1392-4.
    1. Walters MS, Grass JE, Bulens SN, Hancock EB, Phipps EC, Muleta D, Mounsey J, Kainer MA, Concannon C, Dumyati G, Bower C, Jacob J, Cassidy PM, Beldavs Z, Culbreath K, PhillipsWE, Jr, Hardy DJ, Vargas RL, Oethinger M, Ansari U, Stanton R, Albrecht V, Halpin AL, Karlsson M, Rasheed JK, Kallen A. 2019. Carbapenem-resistant Pseudomonas aeruginosa at US emerging infections program sites, 2015. Emerg Infect Dis 25:1281–1288. 10.3201/eid2507.181200.
    1. Thaden JT, Park LP, Maskarinec SA, Ruffin F, FowlerVG, Jr, van Duin D. 2017. Results from a 13-year prospective cohort study show increased mortality associated with bloodstream infections caused by Pseudomonas aeruginosa compared to other bacteria. Antimicrob Agents Chemother 61:e02671-16. 10.1128/AAC.02671-16.
    1. Zhang Y, Chen XL, Huang AW, Liu SL, Liu WJ, Zhang N, Lu XZ. 2016. Mortality attributable to carbapenem-resistant Pseudomonas aeruginosa bacteremia: a meta-analysis of cohort studies. Emerg Microbes Infect 5:e27. 10.1038/emi.2016.22.
    1. Amaya-Villar R, Garnacho-Montero J. 2019. How should we treat Acinetobacter pneumonia? Curr Opin Crit Care 25:465–472. 10.1097/MCC.0000000000000649.
    1. Piperaki ET, Tzouvelekis LS, Miriagou V, Daikos GL. 2019. Carbapenem-resistant Acinetobacter baumannii: in pursuit of an effective treatment. Clin Microbiol Infect 25:951–957. 10.1016/j.cmi.2019.03.014.
    1. Sheu CC, Chang YT, Lin SY, Chen YH, Hsueh PR. 2019. Infections caused by carbapenem-resistant Enterobacteriaceae: an update on therapeutic options. Front Microbiol 10:80. 10.3389/fmicb.2019.00080.
    1. Abdul-Mutakabbir JC, Nguyen L, Stamper K, Maassen P, Lev K, Morrisette T, Kebriaei R, Rybak MJ. 2020. Activity of SPR206, a polymyxin derivative, compared to colistin alone and in combination against multidrug-resistant Pseudomonas aeruginosa strains. Open Forum Infect Dis 7:S662–S663. 10.1093/ofid/ofaa439.1478.
    1. Arends SJR, Rhomberg PR, Lister T, Cotroneo N, Rubio A, Flamm RK, Mendes RE. 2018. Activity of an investigational polymyxin-b-like compound (SPR206) against a set of Enterobacteriaceae organisms responsible for human infections, poster 81. Abstr ESCMID-ASM.
    1. Arends SJR, Rhomberg PR, Lister T, Cotroneo N, Rubio A, Flamm RK, Mendes RE. 2019. Activity of an investigational polymyxin-b-like compound (SPR206) against a set of gram-negative bacilli responsible for human infections, poster AAR-796. Abstr ASM Microbe.
    1. Arends SJR, Rhomberg PR, Lister T, Cotroneo N, Rubio A, Flamm RK, Mendes RE. 2018. In vitro activity evaluation of a next-generation polymyxin, SPR206, against non-fermentative gram-negative bacilli responsible for human infections, poster 80. Abstr ESCMID-ASM.
    1. Cotroneo N, Tomich A, Zou Y, Lister T, Brown P, Dawson M. 2019. In vitro bactericidal activity of next-generation polymyxin SPR206 against susceptible and multidrug-resistant (MDR) Acinetobacter baumannii, Pseudomonas aeruginosa, Escherichia coli, and Klebsiella pneumoniae as compared to levofloxacin and meropenem, poster AAR-797. Abstr ASM Microbe.
    1. Zhang Y, Zhao C, Wang Q, Wang X, Chen H, Li H, Zhang F, Wang H. 2020. Evaluation of the in vitro activity of new polymyxin B analogue SPR206 against clinical MDR, colistin-resistant and tigecycline-resistant Gram-negative bacilli. J Antimicrob Chemother 75:2609–2615. 10.1093/jac/dkaa217.
    1. Grosser L, Heang K, Teague J, Warn P, Corbett D, Dawson MJ, Rubio A. 2018. In vivo efficacy of SPR206 in murine lung and thigh infection models caused by multidrug resistant pathogens Pseudomonas aeruginosa and Acinetobacter baumannii, poster 139. Abstr ESCMID-ASM.
    1. Grosser L, Lister T, Heang K, Brown P, Dawson M, Rubio A. 2019. In vivo efficacy of next-generation polymyxin SPR206 in an immunocompetent murine ascending UTI infection model caused by Escherichia coli, poster AAR-798. Abstr ASM Microbe.
    1. Grosser L, Lister T, Heang K, Warn P, Corbett D, Dawson MJ, Rubio A. 2019. In vivo efficacy of SPR206 in murine lung and thigh infection models caused by multidrug resistant pathogens Pseudomonas aeruginosa and Acinetobacter baumannii, poster AAR-799. Abstr ASM Microbe.
    1. Brown P, Abbott E, Abdulle O, Boakes S, Coleman S, Divall N, Duperchy E, Moss S, Rivers D, Simonovic M, Singh J, Stanway S, Wilson A, Dawson MJ. 2019. Design of next generation polymyxins with lower toxicity: the discovery of SPR206. ACS Infect Dis 5:1645–1656. 10.1021/acsinfecdis.9b00217.
    1. Brown P, Boakes S, Duperchy E, Rivers D, Singh J, Dawson MJ. 2018. Understanding the SAR interplay for kidney exposure and cytotoxicity facilitates the design of improved polymyxin derivatives—identification of SPR206 as a development candidate, poster 145. Abstr ESCMID-ASM.
    1. Falagas ME, Kasiakou SK. 2006. Toxicity of polymyxins: a systematic review of the evidence from old and recent studies. Crit Care 10:R27. 10.1186/cc3995.
    1. Houghton JL, Green KD, Chen W, Garneau-Tsodikova S. 2010. The future of aminoglycosides: the end or renaissance? Chembiochem 11:880–902. 10.1002/cbic.200900779.
    1. Nation RL, Rigatto MHP, Falci DR, Zavascki AP. 2019. Polymyxin acute kidney injury: dosing and other strategies to reduce toxicity. Antibiotics (Basel) 8:E24. 10.3390/antibiotics8010024.
    1. Zavascki AP, Nation RL. 2017. Nephrotoxicity of polymyxins: is there any difference between colistimethate and polymyxin B? Antimicrob Agents Chemother 61:e02319-16. 10.1128/AAC.02319-16.
    1. Jenkins A, Thomson AH, Brown NM, Semple Y, Sluman C, MacGowan A, Lovering AM, Wiffen PJ, BSAC Working Party on Therapeutic Drug Monitoring. 2016. Amikacin use and therapeutic drug monitoring in adults: do dose regimens and drug exposures affect either outcome or adverse events? A systematic review. J Antimicrob Chemother 71:2754–2759. 10.1093/jac/dkw250.
    1. Tsuji BT, Pogue JM, Zavascki AP, Paul M, Daikos GL, Forrest A, Giacobbe DR, Viscoli C, Giamarellou H, Karaiskos I, Kaye D, Mouton JW, Tam VH, Thamlikitkul V, Wunderink RG, Li J, Nation RL, Kaye KS. 2019. International consensus guidelines for the optimal use of the polymyxins: endorsed by the American College of Clinical Pharmacy (ACCP), European Society of Clinical Microbiology and Infectious Diseases (ESCMID), Infectious Diseases Society of America (IDSA), International Society for Anti-infective Pharmacology (ISAP), Society of Critical Care Medicine (SCCM), and Society of Infectious Diseases Pharmacists (SIDP). Pharmacotherapy 39:10–39. 10.1002/phar.2209.

Source: PubMed

3
Se inscrever