Skip to main content
Log in

Sulopenem: An Intravenous and Oral Penem for the Treatment of Urinary Tract Infections Due to Multidrug-Resistant Bacteria

  • Review Article
  • Published:
Drugs Aims and scope Submit manuscript

Abstract

Sulopenem (formerly known as CP-70,429, and CP-65,207 when a component of a racemic mixture with its R isomer) is an intravenous and oral penem that possesses in vitro activity against fluoroquinolone-resistant, extended spectrum β-lactamases (ESBL)-producing, multidrug-resistant (MDR) Enterobacterales. Sulopenem is being developed to treat patients with uncomplicated and complicated urinary tract infections (UTIs) as well as intra-abdominal infections. This review will focus mainly on its use in UTIs. The chemical structure of sulopenem shares properties of penicillins, cephalosporins, and carbapenems. Sulopenem is available as an oral prodrug formulation, sulopenem etzadroxil, which is hydrolyzed by intestinal esterases, resulting in active sulopenem. In early studies, the S isomer of CP-65,207, later developed as sulopenem, demonstrated greater absorption, higher drug concentrations in the urine, and increased stability against the renal enzyme dehydropeptidase-1 compared with the R isomer, which set the stage for its further development as a UTI antimicrobial. Sulopenem is active against both Gram-negative and Gram-positive microorganisms. Sulopenem’s β-lactam ring alkylates the serine residues of penicillin-binding protein (PBP), which inhibits peptidoglycan cross-linking. Due to its ionization and low molecular weight, sulopenem passes through outer membrane proteins to reach PBPs of Gram-negative bacteria. While sulopenem activity is unaffected by many β-lactamases, resistance arises from alterations in PBPs (e.g., methicillin-resistant Staphylococcus aureus [MRSA]), expression of carbapenemases (e.g., carbapenemase-producing Enterobacterales and in Stenotrophomonas maltophilia), reduction in the expression of outer membrane proteins (e.g., some Klebsiella spp.), and the presence of efflux pumps (e.g., MexAB-OprM in Pseudomonas aeruginosa), or a combination of these mechanisms. In vitro studies have reported that sulopenem demonstrates greater activity than meropenem and ertapenem against Enterococcus faecalis, Listeria monocytogenes, methicillin-susceptible S. aureus (MSSA), and Staphylococcus epidermidis, as well as similar activity to carbapenems against Streptococcus agalactiae, Streptococcus pneumoniae, and Streptococcus pyogenes. With some exceptions, sulopenem activity against Gram-negative aerobes was less than ertapenem and meropenem but greater than imipenem. Sulopenem activity against Escherichia coli carrying ESBL, CTX-M, or Amp-C enzymes, or demonstrating MDR phenotypes, as well as against ESBL-producing Klebsiella pneumoniae, was nearly identical to ertapenem and meropenem and greater than imipenem. Sulopenem exhibited identical or slightly greater activity than imipenem against many Gram-positive and Gram-negative anaerobes, including Bacteroides fragilis. The pharmacokinetics of intravenous sulopenem appear similar to carbapenems such as imipenem-cilastatin, meropenem, and doripenem. In healthy subjects, reported volumes of distribution (Vd) ranged from 15.8 to 27.6 L, total drug clearances (CLT) of 18.9–24.9 L/h, protein binding of approximately 10%, and elimination half-lives (t½) of 0.88–1.03 h. The estimated renal clearance (CLR) of sulopenem is 8.0–10.6 L/h, with 35.5% ± 6.7% of a 1000 mg dose recovered unchanged in the urine. An ester prodrug, sulopenem etzadroxil, has been developed for oral administration. Initial investigations reported a variable oral bioavailability of 20–34% under fasted conditions, however subsequent work showed that bioavailability is significantly improved by administering sulopenem with food to increase its oral absorption or with probenecid to reduce its renal tubular secretion. Food consumption increases the area under the curve (AUC) of oral sulopenem (500 mg twice daily) by 23.6% when administered alone and 62% when administered with 500 mg of probenecid. Like carbapenems, sulopenem demonstrates bactericidal activity that is associated with the percentage of time that free concentrations exceed the MIC (%f T > MIC). In animal models, bacteriostasis was associated with %f T > MICs ranging from 8.6 to 17%, whereas 2-log10 kill was seen at values ranging from 12 to 28%. No pharmacodynamic targets have been documented for suppression of resistance. Sulopenem concentrations in urine are variable, ranging from 21.8 to 420.0 mg/L (median 84.4 mg/L) in fasted subjects and 28.8 to 609.0 mg/L (median 87.3 mg/L) in those who were fed. Sulopenem has been compared with carbapenems and cephalosporins in guinea pig and murine systemic and lung infection animal models. Studied pathogens included Acinetobacter calcoaceticus, B. fragilis, Citrobacter freundii, Enterobacter cloacae, E. coli, K. pneumoniae, Proteus vulgaris, and Serratia marcescens. These studies reported that overall, sulopenem was non-inferior to carbapenems but appeared to be superior to cephalosporins. A phase III clinical trial (SURE-1) reported that sulopenem was not non-inferior to ciprofloxacin in women infected with fluoroquinolone-susceptible pathogens, due to a higher rate of asymptomatic bacteriuria in sulopenem-treated patients at the test-of-cure visit. However, the researchers reported superiority of sulopenem etzadroxil/probenecid over ciprofloxacin for the treatment of uncomplicated UTIs in women infected with fluoroquinolone/non-susceptible pathogens, and non-inferiority in all patients with a positive urine culture. A phase III clinical trial (SURE-2) compared intravenous sulopenem followed by oral sulopenem etzadroxil/probenecid with ertapenem in the treatment of complicated UTIs. No difference in overall success was noted at the end of therapy. However, intravenous sulopenem followed by oral sulopenem etzadroxil was not non-inferior to ertapenem followed by oral stepdown therapy in overall success at test-of-cure due to a higher rate of asymptomatic bacteriuria in the sulopenem arm. After a meeting with the US FDA, Iterum stated that they are currently evaluating the optimal design for an additional phase III uncomplicated UTI study to be conducted prior to the potential resubmission of the New Drug Application (NDA). It is unclear at this time whether Iterum intends to apply for EMA or Japanese regulatory approval. The safety and tolerability of sulopenem has been reported in various phase I pharmacokinetic studies and phase III clinical trials. Sulopenem (intravenous and oral) appears to be well tolerated in healthy subjects, with and without the coadministration of probenecid, with few serious drug-related treatment-emergent adverse events (TEAEs) reported to date. Reported TEAEs affecting ≥1% of patients were (from most to least common) diarrhea, nausea, headache, vomiting and dizziness. Discontinuation rates were low and were not different than comparator agents. Sulopenem administered orally and/or intravenously represents a potentially well tolerated and effective option for treating uncomplicated and complicated UTIs, especially in patients with documented or highly suspected antimicrobial pathogens to commonly used agents (e.g. fluoroquinolone-resistant E. coli), and in patients with documented microbiological or clinical failure or patients who demonstrate intolerance/adverse effects to first-line agents. This agent will likely be used orally in the outpatient setting, and intravenously followed by oral stepdown in the hospital setting. Sulopenem also allows for oral stepdown therapy in the hospital setting from intravenous non-sulopenem therapy. More clinical data are required to fully assess the clinical efficacy and safety of sulopenem, especially in patients with complicated UTIs caused by resistant pathogens such as ESBL-producing, Amp-C, MDR E. coli. Antimicrobial stewardship programs will need to create guidelines for when this oral and intravenous penem should be used.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Puttagunta S, Aronin S, Gupta V, Murray J, Dunne M. Impact of initial inappropriate antibiotic therapy on outcome for uncomplicated urinary tract infection due to fluoroquinolone non-susceptible Enterobacteriaceae [abstract plus poster]. American Society for Microbiology (ASM) MICROBE 2018. Atlanta, GA; 2018.

  2. Karlowsky J, Lagacé-Wiens P, Simner P, DeCorby M, Adam H, Walkty A, et al. Antimicrobial resistance in urinary tract pathogens in Canada from 2007 to 2009: CANWARD surveillance study. Antimicrob Agents Chemother. 2011;55:3169–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Dunne MW, Snow K, Mehta R. Failure of empiric treatment of uncomplicated urinary tract infection (UTI) associated with resistant pathogens [abstract no. 4561 plus poster]. American Society for Microbiology (ASM) MICROBE 2019. San Francisco, CA; 2019.

  4. Bader MS, Loeb M, Leto D, Brooks AA. Treatment of urinary tract infections in the era of antimicrobial resistance and new antimicrobial agents. Postgrad Med. 2020;132:234–50.

    Article  PubMed  Google Scholar 

  5. Puttagunta S, Aronin S, Gupta V, Murray J, Dunne M. Impact of initial inappropriate antibiotic therapy on outcome for uncomplicated urinary tract infection due to antibiotic non-susceptible Enterobacteriaceae [abstract plus poster]. American Society for Microbiology (ASM) MICROBE 2018. Atlanta, GA; 2018.

  6. Dunne M, Gupta V, Aronin S, Murray J, Puttagunta S. The prevalence of Enterobacteriaceae resistant to all major classes of oral antibiotics from outpatient urine cultures in the United States and effect on clinical outcomes [abstract plus poster]. IDWeek 2018. San Francisco, CA; 2018.

  7. Lagacé-Wiens P, Simner P, Forward K, Tailor F, Adam. HJ, Decorby M, et al. Analysis of 3789 in- and outpatient Escherichia coli isolates from across Canada—results of the CANWARD 2007-2009 study. Diagn Microbiol Infect Dis. 2011;69:314–9.

  8. Karlowsky JA, Adam HJ, Baxter MR, Denisuik AJ, Lagacé-Wiens PRS, Walkty AJ, et al. In vitro activity of sulopenem, an oral penem, against urinary isolates of Escherichia coli. Antimicrob Agents Chemother. 2019;63:e01832-e1918.

    Article  CAS  PubMed  Google Scholar 

  9. Dunne M, Aronin S, Gupta V, Murray J, Puttagunta S. Impact of ESBL-positivity and quinolone non-susceptibility on outcome for inpatients with complicated urinary tract infection: A multicenter evaluation in the U.S. [abstract plus poster]. In: 28th European Congress of Clinical Microbiology and Infectious Diseases. Madrid; 2018.

  10. Puttagunta S, Gupta V, Murray J, Dunne M. Prevalence of extended spectrum beta-lactamase producing and quinolone non-susceptible Enterobacteriaceae in inpatient and outpatient settings in the USA from 2011-2017 [abstract no. 400 plus poster]. IDWeek 2017. San Diego, CA; 2017.

  11. Denisuik AJ, Karlowsky JA, Adam HJ, Baxter MR, Lagacé-Wiens PRS, Mulvey MR, et al. Dramatic rise in the proportion of ESBL-producing Escherichia coli and Klebsiella pneumoniae among clinical isolates identified in Canadian hospital laboratories from 2007 to 2016. J Antimicrob Chemother. 2019;74:vi64-71.

    Article  Google Scholar 

  12. Centers for Disease Control and Prevention. Antibiotic Resistance. A.R. & Patient Safety Portal. Antibiotic Resistance and Patient Safety Portal. 2019. https://arpsp.cdc.gov/profile/antibiotic-resistance?tab=antibiotic-resistance. Accessed 18 Aug 2021.

  13. Dunne MW, Das A, Akinapelli K, Zelasky MT, Boucher HW, Aronin SI. Efficacy and safety of oral sulopenem etzadroxil/probenecid versus oral ciprofloxacin in the treatment of uncomplicated urinary uract infections (uUTI) in adult women: Results from the SURE-1 trial [abstract plus poster]. IDWeek 2020. Virtual Conference, Online; 2020.

  14. Dunne M, Aronin S. Efficacy and safety of intravenous sulopenem followed by oral sulopenem etzadroxil/probenecid versus intravenous ertapenem followed by oral ciprofloxacin or amoxicillin-clavulanate in the treatment of complicated urinary tract infections (cUTI): Results from the SURE-2 trial [abstract plus poster]. IDWeek 2020. Virtual Conference, Online; 2020.

  15. Chandra R, Hazra A, Skogerboe T, Labadie R, Kirby D, Soma K, et al. Pharmacokinetics (PK), safety and tolerability of single oral doses of PF-03709270, with and without co-administration of probenecid (abstract plus poster F1-353). In: 48th Annual ICAAC/IDSA 46th Annual Meeting. Washington, DC; 2008.

  16. Ednie LM, Appelbaum PC. Antianaerobic activity of sulopenem compared to six other agents. Antimicrob Agents Chemother. 2009;53:2163–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Dunne M, Huband M, Flamm R, Aronin S, Puttagunta S. Prediction of sulopenem activity against Enterobacteriaceae using ertapenem as a surrogate [abstract plus poster]. American Society for Microbiology (ASM) MICROBE 2018. Atlanta, GA; 2018.

  18. Zhanel GG, Adam H, Baxter M, Denisuik A, Lagace-Wiens P, Walkty A, et al. In vitro activity of sulopenem, an oral penem, against Escherichia coli isolated from urine specimens of patients across Canada in 2014-2016 [abstract plus poster SUNDAY-568]. American Society for Microbiology (ASM) MICROBE 2018. Atlanta, GA; 2018.

  19. Dunne M, Puttagunta S, Fisher J, Koeth L. In vitro activity of sulopenem against resistant Neisseria gonorrhoeae [abstract plus poster P1705]. In: 29th European Congress of Clinical Microbiology & Infectious Diseases. Amsterdam; 2019.

  20. Puttagunta S, Dunne M. Antimicrobial activity of sulopenem tested against Gram-positive and Gram-negative organisms from the United States and Europe (2013-2015) [abstract plus poster]. American Society for Microbiology (ASM) MICROBE 2017. New Orleans, LA; 2017.

  21. Aronin SI, Huband M, Flamm R, Puttagunta S, Dunne M. Sulopenem activity against Enterobacteriaceae isolates from patients with urinary tract infection [abstract plus poster]. American Society for Microbiology (ASM) MICROBE 2018. Atlanta, GA; 2018.

  22. Watanabe K, Kato N, Tanaka K, Tanaka Y, Kato H, Ueno K. In vitro activities of sulopenem, a new parenteral penem, against anaerobes. Jpn J Antibiot. 1996;49:367–76.

    CAS  PubMed  Google Scholar 

  23. Komoto A, Otsuki M, Nishino T. In vitro and in vivo antibacterial activities of sulopenem, a new penem antibiotic. Jpn J Antibiot. 1996;49:352–66.

    CAS  PubMed  Google Scholar 

  24. Inoue E, Komoto E, Taniyama Y, Mitsuhashi S. Antibacterial activity of sulopenem, a new parenteral penem antibiotic. Jpn J Antibiot. 1996;49:338–51.

    CAS  PubMed  Google Scholar 

  25. Yoshida T, Tateda E, Hiramatsu K, Yokota T. In vitro antibacterial activity of a new parenteral penem, sulopenem. Jpn J Antibiot. 1996;49:324–37.

    CAS  PubMed  Google Scholar 

  26. Nagashima M, Goto S, Yoshida T, Matsunaga T, Shimohira H, Ogawa M. In vitro and in vivo activities of sulopenem compared with those of imipenem and cephalosporins. Jpn J Antibiot. 1996;49:303–23.

    CAS  PubMed  Google Scholar 

  27. Brenek SJ, Caron S, Chisowa E, Delude MP, Drexler MT, Ewing MD, et al. Development of a practical and convergent process for the preparation of sulopenem. Org Process Res Dev Am Chem Soc. 2012;16:1348–59.

    Article  CAS  Google Scholar 

  28. Brenek SJ, Caron S, Chisowa E, Colon-Cruz R, Delude MP, Drexler MT, et al. Development of a second-generation process to antibacterial candidate sulopenem. Org Process Res Dev Am Chem Soc. 2012;16:1338–47.

    Article  CAS  Google Scholar 

  29. Hujer K, Hamza N, Hujer A, Perez F, Helfand M, Bethel C, et al. Identification of a new allelic variant of the Acinetobacter baumannii cephalosporinase, ADC-7 beta-lactamase: defining a unique family of class C enzymes. Antimicrob Agents Chemother. 2005;49:2941–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Zhanel GG, Adam HJ, Baxter MR, Golden A, Lagacé-Wiens P, Walkty A, et al. In vitro activity of sulopenem and comparative agents against bacterial pathogens isolated from Canadian patients with urinary tract infections: CANWARD surveillance study 2014-2020 [abstract no. 4674 plus poster]. World Microbe Forum. Online Worldwide; 2021.

  31. Dunne M, Aronin S, Dunzo E, Puttagunta S. A phase 1, randomized, open-label, crossover study in healthy subjects under fasting conditions of orally administered sulopenem etzadroxil alone or with probenecid to determine the pharmacokinetics of sulopenem [abstract plus poster]. IDWeek 2018. San Francisco, CA; 2018.

  32. Dunne M, Dunzo E, Puttagunta S. A phase 1 study to assess the pharmacokinetics of sulopenem etzadroxil (PF-03709270) [abstract plus poster]. IDWeek 2017. San Diego, CA; 2017.

  33. Okamoto K, Gotoh N, Nishino T. Pseudomonas aeruginosa reveals high intrinsic resistance to penem antibiotics: Penem resistance mechanisms and their interplay. Antimicrob Agents Chemother. 2001;45:1964–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Dunne M, Puttagunta S, Aronin S, Huband M, Flamm R. Post-antibiotic and sub-inhibitory minimum inhibitory concentration effects of sulopenem [abstract plus poster P1992]. In: 29th European Congress of Clinical Microbiology & Infectious Diseases. Amsterdam; 2019.

  35. Dunne M, Morrissey I, de Piano C, Magnet S, Hawser S, Puttagunta S. Antimicrobial activity of sulopenem in the urine of healthy volunteers [abstract plus poster]. In: 28th European Congress of Clinical Microbiology and Infectious Diseases. Madrid; 2018.

  36. Kosowska-Shick K, Ednie LM, McGhee P, Appelbaum PC. Comparative antipneumococcal activities of sulopenem and other drugs. Antimicrob Agents Chemother. 2009;53:2239–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Puttagunta S, Aronin S, Huband M, Flamm R, Dunne M. Sulopenem activity against Enterobacteriaceae isolates from patients with urinary tract infection or intra-abdominal infection [abstract plus poster]. IDWeek 2018. San Francisco, CA; 2018.

  38. Minamimura M, Taniyama Y, Inoue E, Mitsuhashi S. In vitro antibacterial activity and β-lactamase stability of CP-70,429, a new penem antibiotic. Antimicrob Agents Chemother. 1993;37:1547–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Gootz T, Girard D, Schelkley W, Tensfeldt T, Foulds G, Kellogg M, et al. Pharmacokinetic studies in animals of a new parenteral penem CP-65,207 and its oral prodrug ester. J Antibiot. 1990;43:422–32.

    Article  CAS  Google Scholar 

  40. Hamilton-Miller JMT. Chemical and microbiologic aspects of penems, a distinct class of beta-lactams: focus on faropenem. Pharmacotherapy. 2003;23:1497–507.

    Article  CAS  PubMed  Google Scholar 

  41. Chatwin CL, Hamrick JC, Trout REL, Myers CL, Cusick SM, Weiss WJ, et al. Microbiological characterization of VNRX-5236, a broad-spectrum β-lactamase inhibitor for rescue of the orally bioavailable cephalosporin ceftibuten as a carbapenem-sparing agent against strains of Enterobacterales expressing extended-spectrum β-lactamases and serine carbapenemases. Antimicrob Agents Chemother. 2021;65:e0055221.

    Article  PubMed  Google Scholar 

  42. Foulds G, Knirsch AK, Lazar JD, Tensfeldt TG, Gerber N. Pharmacokinetics of the penem CP-65,207 and its separate stereoisomers in humans. Antimicrob Agents Chemother. 1991;35:665–71.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Gootz T, Retsema J, Girard A, Hamanaka E, Anderson M, Sokolowski S. In vitro activity of CP-65,207, a new penem antimicrobial agent, in comparison with those of other agents. Antimicrob Agents Chemother. 1989;33:1160.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Kaczmarek FM, Dib-Hajj F, Shang W, Gootz TD. High-level carbapenem resistance in a Klebsiella pneumoniae clinical isolate is due to the combination of blaACT-1 β-lactamase production, porin OmpK35/36 insertional inactivation, and down-regulation of the phosphate transport porin PhoE. Antimicrob Agents Chemother. 2006;50:3396–406.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Iterum Therapeutics. Iterum Therapeutics announces topline results from phase 3 clinical trial of oral and IV sulopenem for the treatment of complicated intra-abdominal infections. 2019. https://www.iterumtx.com/news/press-releases/detail/29/iterum-therapeutics-announces-topline-results-from-phase-3. Accessed 11 Jul 2021.

  46. Theuretzbacher U, Bush K, Harbarth S, Paul M, Rex JH, Tacconelli E, et al. Critical analysis of antibacterial agents in clinical development. Nat Rev Microbiol. 2020;18:286–98.

    Article  CAS  PubMed  Google Scholar 

  47. Bush K, Macielag M. New β-lactam antibiotics and β-lactamase inhibitors. Expert Opin Ther Pat. 2010;20:1277–93.

    Article  CAS  PubMed  Google Scholar 

  48. Huband MD, Gootz TD, Mullins LM, McCurdy SP, Brennan LA, Penzien JB, et al. In vitro antibacterial activity of sulopenem: a new oral penem antimicrobial versus recent bacterial clinical isolates [abstract plus poster F1-344]. In: 48th Annual ICAAC/IDSA 46th Annual Meeting. Washington, DC; 2008.

  49. Girard D, Finegan S, O’Donnell JP. Pharmacokinetics and pharmacodynamics of sulopenem in preclinical species [abstract plus poster A-054]. In: 48th Annual ICAAC/IDSA 46th Annual Meeting. Washington, DC; 2008.

  50. Forrest A, Hazra A, Girard D, O’Donnell J, Hanna D, Ogden A, et al. PK/PD analysis to select IV sulopenem and oral PF-03709270 doses for phase 2 trials (abstract plus poster A-034). In: 48th Annual ICAAC/IDSA 46th Annual Meeting. Washington, DC; 2008.

  51. Schurek KN, Wiebe R, Karlowsky JA, Rubinstein E, Hoban DJ, Zhanel GG. Faropenem: review of a new oral penem. Expert Rev Anti Infect Ther. 2007;5:185–98.

    Article  CAS  PubMed  Google Scholar 

  52. Dalhoff A, Janjic N, Echols R. Redefining penems. Biochem Pharmacol. 2006;71:1085–95.

    Article  CAS  PubMed  Google Scholar 

  53. Zhanel GG, Wiebe R, Dilay L, Thomson K, Rubinstein E, Hoban DJ, et al. Comparative review of the carbapenems. Drugs. 2007;67:1027–52.

    Article  CAS  PubMed  Google Scholar 

  54. Zhanel GG, Johanson C, Embil JM, Noreddin A, Gin A, Vercaigne L, et al. Ertapenem: review of a new carbapenem. Expert Rev Anti Infect Ther. 2005;3:23–39.

    Article  CAS  PubMed  Google Scholar 

  55. Zhanel G, Lawrence C, Adam H, Schweizer F, Zelenitsky S, Zhanel M, et al. Imipenem–relebactam and meropenem–vaborbactam: two novel carbapenem-β-lactamase inhibitor combinations. Drugs. 2018;78:65–98.

    Article  CAS  PubMed  Google Scholar 

  56. Zhanel GG, Adam HJ, Baxter MR, Fuller J, Nichol KA, Denisuik AJ, et al. 42936 pathogens from Canadian hospitals: 10 years of results (2007–16) from the CANWARD surveillance study. J Antimicrob Chemother. 2019;74(Suppl 4):vi5-21.

    Article  Google Scholar 

  57. Zhanel GG, Simor AE, Vercaigne L, Mandell L. Imipenem and meropenem: comparison of in vitro activity, pharmacokinetics, clinical trials and adverse effects. Can J Infect Dis. 1998;9:215–28.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Hazra-Raybon A, O’Gorman M, Puttagunta S, Soma K, Skogerboe T, Fang J, et al. Effect of various degrees of renal impairment (RI) on the pharmacokinetics (PK) of sulopenem after IV and oral administration [abstract]. Clin Pharmacol Ther. 2011;89:S26.

    Google Scholar 

  59. Iterum Therapeutics. Iterum Therapeutics announces topline results from its phase 3 clinical trial of oral sulopenem for the treatment of uncomplicated urinary tract infections. 2020. https://www.iterumtx.com/news/press-releases/detail/40/iterum-therapeutics-announces-topline-results-from-its. Accessed 5 Aug 2021.

  60. Robbins N, Koch SE, Tranter M, Rubinstein J. The history and future of probenecid. Cardiovasc Toxicol. 2011;12:1–9.

    Article  Google Scholar 

  61. Wu C-C, Pai T-Y, Hsiao F-Y, Shen L-J, Wu F-LL. The effect of different carbapenem antibiotics (ertapenem, imipenem/cilastatin, and meropenem) on serum valproic acid concentrations. Ther Drug Monit. 2016;38:587–92.

    Article  CAS  PubMed  Google Scholar 

  62. A study to investigate the pharmacokinetics, safety and tolerability of an intravenous and oral form of a compound in subjects with varying degrees of renal impairment and normal renal function—study results. ClinicalTrials.gov; 2016. https://clinicaltrials.gov/ct2/show/results/NCT00759564?cond=NCT00759564&draw=2&rank=1&view=results. Accessed 6 Sep 2021.

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to George G. Zhanel.

Ethics declarations

Conflicts of interest

George G. Zhanel has received research grants from Iterum Pharmaceuticals. Marianna Pozdirca, Alyssa Golden, Courtney K. Lawrence, Sheryl Zelenitsky, Liam Berry, Frank Schweizer, Denice Bay, Heather J. Adam, Michael A. Zhanel, Philippe Lagacé-Wiens, Andrew Walkty, Neal Irfan, Kurt Naber, Joseph P. Lynch III and James A. Karlowsky have declared no conflicts of interest.

Funding

The authors are grateful to Iterum Pharmaceuticals for their assistance with literature retrieval and an unrestricted research grant to aid in funding Marianna Pozdirca.

Author contributions

GGZ: design and concept of the entire manuscript and drafting all sections; senior and corresponding author. MP: design and concept of the entire manuscript and drafting all sections. AG: design and concept of the entire manuscript and drafting all sections. CKL: design and concept of the entire manuscript and drafting the Pharmacokinetic/Pharmacodynamic section. SZ: design and concept of the entire manuscript and drafting the Pharmacokinetic/Pharmacodynamic section. LB: design and concept of the entire manuscript and drafting the Chemistry and Microbiology sections. FS: design and concept of the entire manuscript and drafting the Chemistry and Microbiology sections. DB: design and concept of the entire manuscript and drafting the Introduction, Mechanisms of Action and Mechanisms of Resistance sections. HJA: design and concept of the entire manuscript and drafting the Microbiology section. MAZ: design and concept of the entire manuscript and drafting the Introduction, Mechanisms of Action, Mechanisms of Resistance, Chemistry and Microbiology sections. PL-W: design and concept of the entire manuscript and drafting the Animal Models section. AW: design and concept of the entire manuscript and drafting the Clinical Trials, Adverse Effects and Place in Therapy sections. NI: design and concept of the entire manuscript and drafting the Adverse Effects, Drug Interactions and Place in Therapy sections. KN: design and concept of the entire manuscript and drafting the Clinical Trials section. JPLIII: design and concept of the entire manuscript and drafting the Clinical Trials and Place in Therapy sections. JAK: design and concept of the entire manuscript and drafting all sections.

Ethical approval

Not applicable.

Availability of Data and Material

Not applicable.

Consent for publication

Not applicable.

Consent for participate

Not applicable.

Code availability

Not applicable.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhanel, G.G., Pozdirca, M., Golden, A.R. et al. Sulopenem: An Intravenous and Oral Penem for the Treatment of Urinary Tract Infections Due to Multidrug-Resistant Bacteria. Drugs 82, 533–557 (2022). https://doi.org/10.1007/s40265-022-01688-1

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40265-022-01688-1

Navigation