Pharmacokinetics and Pharmacodynamics of Clofazimine for Treatment of Cryptosporidiosis

Cindy X Zhang, Melissa S Love, Case W McNamara, Victor Chi, Ashley K Woods, Sean Joseph, Deborah A Schaefer, Dana P Betzer, Michael W Riggs, Pui-Ying Iroh Tam, Wesley C Van Voorhis, Samuel L M Arnold, Cindy X Zhang, Melissa S Love, Case W McNamara, Victor Chi, Ashley K Woods, Sean Joseph, Deborah A Schaefer, Dana P Betzer, Michael W Riggs, Pui-Ying Iroh Tam, Wesley C Van Voorhis, Samuel L M Arnold

Abstract

Infection with Cryptosporidium spp. can cause severe diarrhea, leading to long-term adverse impacts and even death in malnourished children and immunocompromised patients. The only FDA-approved drug for treating cryptosporidiosis, nitazoxanide, has limited efficacy in the populations impacted the most by the diarrheal disease, and safe, effective treatment options are urgently needed. Initially identified by a large-scale phenotypic screening campaign, the antimycobacterial therapeutic clofazimine demonstrated great promise in both in vitro and in vivo preclinical models of Cryptosporidium infection. Unfortunately, a phase 2a clinical trial in HIV-infected adults with cryptosporidiosis did not identify any clofazimine treatment effect on Cryptosporidium infection burden or clinical outcomes. To explore whether clofazimine's lack of efficacy in the phase 2a trial may have been due to subtherapeutic clofazimine concentrations, a pharmacokinetic/pharmacodynamic modeling approach was undertaken to determine the relationship between clofazimine in vivo concentrations and treatment effects in multiple preclinical infection models. Exposure-response relationships were characterized using Emax and logistic models, which allowed predictions of efficacious clofazimine concentrations for the control and reduction of disease burden. After establishing exposure-response relationships for clofazimine treatment of Cryptosporidium infection in our preclinical model studies, it was unmistakable that the clofazimine levels observed in the phase 2a study participants were well below concentrations associated with anti-Cryptosporidium efficacy. Thus, despite a dosing regimen above the highest doses recommended for mycobacterial therapy, it is very likely the lack of treatment effect in the phase 2a trial was at least partially due to clofazimine concentrations below those required for efficacy against cryptosporidiosis. It is unlikely that clofazimine will provide a remedy for the large number of cryptosporidiosis patients currently without a viable treatment option unless alternative, safe clofazimine formulations with improved oral absorption are developed. (This study has been registered in ClinicalTrials.gov under identifier NCT03341767.).

Keywords: Cryptosporidium; PK/PD; cryptosporidiosis; gastrointestinal; gastrointestinal infection; infectious diseases; pharmacodynamics; pharmacokinetics.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

FIG 1
FIG 1
Clofazimine in vivo efficacy in mouse model of Cryptosporidium infection. (A) Mean oocyst shedding profile for each clofazimine treatment group plotted over time. A single clofazimine oral dose of various amounts was administered 4 days after oral inoculation with C. parvum. Each dose group contained four mice, and the fecal oocyst concentration for each mouse was quantified by flow cytometry. (B) Rate of reduction in log-transformed fecal oocyst concentration from day 4 to day 6 postinfection, summarized by dose group. The tops and bottoms of the boxes are the 75th and 25th percentiles, respectively, and the center line is the median. The whiskers indicate the range of data distribution that are within 1.5-fold of the interquartile range (IQR). Points that lie out of the reach of the whiskers are outliers by the 1.5 IQR rule. (C) A simple Emax model (black solid line) with 90% confidence interval (gray shaded area) was fitted to describe the relationship between average clofazimine concentration over the 24 h postdosing (Cavg0–24) and the rate of oocyst reduction, with the model-predicted EC50 marked by the red dashed line. A dichotomous approach was also taken, with an event (“1”) being defined as having less than 10% of baseline oocyst load on day 6 postinfection. (D) Logistic regression model (blue solid line) with 90% confidence interval (gray shaded area), with the model predicted EC50 indicated by the red dashed line. (E and F) Last, a time-to-event outcome variable approach was employed. The number of days each mouse took to reach a 90% reduction in oocyst burden compared to the baseline oocyst concentration is summarized by dose group (E) or compared to Cavg0–24 (F). The box plots in panel E were generated in the same fashion as those in panel B. A simple Emax model (black solid line) with 90% confidence interval (gray shaded area) was built to describe the correlation between the number of days and clofazimine Cavg0–24 (F). The red dashed line represents EC50.
FIG 2
FIG 2
Clofazimine pharmacokinetics and pharmacodynamics in a calf model of cryptosporidiosis. Clofazimine was administered to calves with a dosing regimen of 30 mg/kg twice daily over 5 days for a total of 10 doses. (A) Up to 15 blood samples were collected from each calf (n = 6) over time, and mean and individual clofazimine concentrations were plotted. (B) Stool samples were collected every 24 h starting on day 3 postinfection for clofazimine- and vehicle control-treated calves, and fecal oocyst counts were quantified by real-time PCR. Mean and individual oocyst counts are plotted over time. (C to F) Other pharmacodynamic outcomes, including fecal volume (C), fecal consistency score (D), urine volume (E), and clinical evaluation score (F) were recorded daily and are shown as individual values and means (standard deviations are indicated by error bars). An asterisk indicates days on which the treatment group and the control group differed significantly (P ≤ 0.05) at the collection endpoint.
FIG 3
FIG 3
Relationship between clofazimine treatment and pharmacodynamic outcomes in calves. The oocyst count area under the curve (AUC) for the duration of the study was calculated for each calf. The association between oocyst count AUC24–192 and average clofazimine concentrations, Cavg0–24 (A) and Cavg96–108 (B), were investigated using a simple linear regression model, and the results are summarized in Table 2. Clinical outcome AUCs were also calculated for the duration of the study and compared between the treatment and control groups (C). The differences between the groups were not statistically significant after Bonferroni correction adjusting for multiple comparison. The top and bottom bars of the boxes represent the 75th and 25th percentiles, respectively, and the center lines mark the medians. The whiskers extend to the most extreme data point that is within 1.5 times the IQR away from the box. Points that fall beyond the end of the whiskers are outliers as defined by the 1.5 IQR rule.
FIG 4
FIG 4
Clofazimine pharmacokinetics and pharmacodynamics in phase 2a clinical trial. HIV-infected adults with cryptosporidiosis were administered clofazimine with a dosing regimen of 100 mg 3 times daily for participants who weighed ≥50 kg (n = 2 participants) or 50 mg 3 times daily for participants who weighed <50 kg (n = 10 participants) for a duration of 5 days. (A) Mean and individual clofazimine concentrations plotted over time. (B) Stool samples were collected and assessed three times a day, and oocyst shedding was quantified using qPCR. Mean daily oocyst shedding in the first stool was compared between clofazimine treatment group and the control group. (C) For each participant, the rate of reduction in daily oocyst shedding was calculated for the first 3 days of clofazimine treatment, with the result plotted against observed clofazimine Cavg0–24 to reveal a potential correlation between oocyst excretion rates and clofazimine concentrations. The dashed line marks no change in oocyst shedding rate. No correlation was clearly seen comparing the rate of reduction in oocyst shedding rates to clofazimine Cavg0–24. (D) A time-to-event approach was attempted, where the time to reach 90% oocyst reduction was compared to clofazimine Cavg0–24 and a significant correlation was not identified. (E) A Kaplan-Meier survival curve was used to compare the probability of achieving 90% reduction in oocyst count compared to the baseline count between clofazimine treatment group and the control group. A success was defined by having less than 10% of baseline oocyst count, and no significant difference was found, as shown by the log rank test (P = 0.47).

References

    1. Amadi B, Kelly P, Mwiya M, Mulwazi E, Sianongo S, Changwe F, Thomson M, Hachungula J, Watuka A, Walker-Smith J, Chintu C. 2001. Intestinal and systemic infection, HIV, and mortality in Zambian children with persistent diarrhea and malnutrition. J Pediatr Gastroenterol Nutr 32:550–554. 10.1097/00005176-200105000-00011.
    1. Mor SM, Tzipori S. 2008. Cryptosporidiosis in children in Sub-Saharan Africa: a lingering challenge. Clin Infect Dis 47:915–921. 10.1086/591539.
    1. Khalil IA, Troeger C, Rao PC, Blacker BF, Brown A, Brewer TG, Colombara DV, De Hostos EL, Engmann C, Guerrant RL, Haque R, Houpt ER, Kang G, Korpe PS, Kotloff KL, Lima AAM, Petri WA, Platts-Mills JA, Shoultz DA, Forouzanfar MH, Hay SI, Reiner RC, Mokdad AH. 2018. Morbidity, mortality, and long-term consequences associated with diarrhoea from Cryptosporidium infection in children younger than 5 years: a meta-analyses study. Lancet Glob Health 6:e758–e768. 10.1016/S2214-109X(18)30283-3.
    1. Guerrant DI, Moore SR, Lima AA, Patrick PD, Schorling JB, Guerrant RL. 1999. Association of early childhood diarrhea and cryptosporidiosis with impaired physical fitness and cognitive function four-seven years later in a poor urban community in northeast Brazil. Am J Trop Med Hyg 61:707–713. 10.4269/ajtmh.1999.61.707.
    1. Molbak K, Andersen M, Aaby P, Hojlyng N, Jakobsen M, Sodemann M, da Silva AP. 1997. Cryptosporidium infection in infancy as a cause of malnutrition: a community study from Guinea-Bissau, west Africa. Am J Clin Nutr 65:149–152. 10.1093/ajcn/65.1.149.
    1. Checkley W, Epstein LD, Gilman RH, Black RE, Cabrera L, Sterling CR. 1998. Effects of Cryptosporidium parvum infection in Peruvian children: growth faltering and subsequent catch-up growth. Am J Epidemiol 148:497–506. 10.1093/oxfordjournals.aje.a009675.
    1. Checkley W, Gilman RH, Epstein LD, Suarez M, Diaz JF, Cabrera L, Black RE, Sterling CR. 1997. Asymptomatic and symptomatic cryptosporidiosis: their acute effect on weight gain in Peruvian children. Am J Epidemiol 145:156–163. 10.1093/oxfordjournals.aje.a009086.
    1. Liu J, Platts-Mills JA, Juma J, Kabir F, Nkeze J, Okoi C, Operario DJ, Uddin J, Ahmed S, Alonso PL, Antonio M, Becker SM, Blackwelder WC, Breiman RF, Faruque AS, Fields B, Gratz J, Haque R, Hossain A, Hossain MJ, Jarju S, Qamar F, Iqbal NT, Kwambana B, Mandomando I, McMurry TL, Ochieng C, Ochieng JB, Ochieng M, Onyango C, Panchalingam S, Kalam A, Aziz F, Qureshi S, Ramamurthy T, Roberts JH, Saha D, Sow SO, Stroup SE, Sur D, Tamboura B, Taniuchi M, Tennant SM, Toema D, Wu Y, Zaidi A, Nataro JP, Kotloff KL, Levine MM, Houpt ER. 2016. Use of quantitative molecular diagnostic methods to identify causes of diarrhoea in children: a reanalysis of the GEMS case-control study. Lancet 388:1291–1301. 10.1016/S0140-6736(16)31529-X.
    1. Khalil IA, Troeger C, Blacker BF, Rao PC, Brown A, Atherly DE, Brewer TG, Engmann CM, Houpt ER, Kang G, Kotloff KL, Levine MM, Luby SP, MacLennan CA, Pan WK, Pavlinac PB, Platts-Mills JA, Qadri F, Riddle MS, Ryan ET, Shoultz DA, Steele AD, Walson JL, Sanders JW, Mokdad AH, Murray CJL, Hay SI, Reiner RC, Jr.. 2018. Morbidity and mortality due to Shigella and enterotoxigenic Escherichia coli diarrhoea: the Global Burden of Disease Study 1990–2016. Lancet Infect Dis 18:1229–1240. 10.1016/S1473-3099(18)30475-4.
    1. Adeyemo FE, Singh G, Reddy P, Bux F, Stenstrom TA. 2019. Efficiency of chlorine and UV in the inactivation of Cryptosporidium and Giardia in wastewater. PLoS One 14:e0216040. 10.1371/journal.pone.0216040.
    1. Carpenter C, Fayer R, Trout J, Beach MJ. 1999. Chlorine disinfection of recreational water for Cryptosporidium parvum. Emerg Infect Dis 5:579–584. 10.3201/eid0504.990425.
    1. Peeters JE, Mazas EA, Masschelein WJ, Villacorta Martiez de Maturana I, Debacker E. 1989. Effect of disinfection of drinking water with ozone or chlorine dioxide on survival of Cryptosporidium parvum oocysts. Appl Environ Microbiol 55:1519–1522. 10.1128/aem.55.6.1519-1522.1989.
    1. Pumipuntu N, Piratae S. 2018. Cryptosporidiosis: a zoonotic disease concern. Vet World 11:681–686. 10.14202/vetworld.2018.681-686.
    1. Hlavsa MC, Cikesh BL, Roberts VA, Kahler AM, Vigar M, Hilborn ED, Wade TJ, Roellig DM, Murphy JL, Xiao L, Yates KM, Kunz JM, Arduino MJ, Reddy SC, Fullerton KE, Cooley LA, Beach MJ, Hill VR, Yoder JS. 2018. Outbreaks associated with treated recreational water—United States, 2000–2014. MMWR Morb Mortal Wkly Rep 67:547–551. 10.15585/mmwr.mm6719a3.
    1. Painter JE, Gargano JW, Yoder JS, Collier SA, Hlavsa MC. 2016. Evolving epidemiology of reported cryptosporidiosis cases in the United States, 1995–2012. Epidemiol Infect 144:1792–1802. 10.1017/S0950268815003131.
    1. Rossignol JF, Kabil SM, el-Gohary Y, Younis AM. 2006. Effect of nitazoxanide in diarrhea and enteritis caused by Cryptosporidium species. Clin Gastroenterol Hepatol 4:320–324. 10.1016/j.cgh.2005.12.020.
    1. Amadi B, Mwiya M, Musuku J, Watuka A, Sianongo S, Ayoub A, Kelly P. 2002. Effect of nitazoxanide on morbidity and mortality in Zambian children with cryptosporidiosis: a randomised controlled trial. Lancet 360:1375–1380. 10.1016/S0140-6736(02)11401-2.
    1. Amadi B, Mwiya M, Sianongo S, Payne L, Watuka A, Katubulushi M, Kelly P. 2009. High dose prolonged treatment with nitazoxanide is not effective for cryptosporidiosis in HIV positive Zambian children: a randomised controlled trial. BMC Infect Dis 9:195. 10.1186/1471-2334-9-195.
    1. Love MS, McNamara CW. 2021. Phenotypic screening techniques for Cryptosporidium drug discovery. Expert Opin Drug Discov 16:59–74. 10.1080/17460441.2020.1812577.
    1. Pushpakom S, Iorio F, Eyers PA, Escott KJ, Hopper S, Wells A, Doig A, Guilliams T, Latimer J, McNamee C, Norris A, Sanseau P, Cavalla D, Pirmohamed M. 2019. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov 18:41–58. 10.1038/nrd.2018.168.
    1. Love MS, Beasley FC, Jumani RS, Wright TM, Chatterjee AK, Huston CD, Schultz PG, McNamara CW. 2017. A high-throughput phenotypic screen identifies clofazimine as a potential treatment for cryptosporidiosis. PLoS Negl Trop Dis 11:e0005373. 10.1371/journal.pntd.0005373.
    1. Arbiser JL, Moschella SL. 1995. Clofazimine: a review of its medical uses and mechanisms of action. J Am Acad Dermatol 32:241–247. 10.1016/0190-9622(95)90134-5.
    1. Cholo MC, Steel HC, Fourie PB, Germishuizen WA, Anderson R. 2012. Clofazimine: current status and future prospects. J Antimicrob Chemother 67:290–298. 10.1093/jac/dkr444.
    1. Iroh Tam PY, Arnold SLM, Barrett LK, Chen CR, Conrad TM, Douglas E, Gordon MA, Hebert D, Henrion M, Hermann D, Hollingsworth B, Houpt E, Jere KC, Lindblad R, Love MS, Makhaza L, McNamara CW, Nedi W, Nyirenda J, Operario DJ, Phulusa J, Quinnan GV, Sawyer LA, Thole H, Toto N, Winter A, Van Voorhis WC. 2021. Clofazimine for treatment of cryptosporidiosis in human immunodeficiency virus-infected adults: an experimental medicine, randomized, double-blind, placebo-controlled phase 2a trial. Clin Infect Dis 73:183–191. 10.1093/cid/ciaa421.
    1. Gomes M, Ribeiro I, Warsame M, Karunajeewa H, Petzold M. 2008. Rectal artemisinins for malaria: a review of efficacy and safety from individual patient data in clinical studies. BMC Infect Dis 8:39. 10.1186/1471-2334-8-39.
    1. Bendel D, Rulisa S, Ansah P, Sirima S. 2015. Efficacy of a novel sublingual spray formulation of artemether in African children with Plasmodium falciparum malaria. Antimicrob Agents Chemother 59:6930–6938. 10.1128/AAC.00243-15.
    1. Schaefer DA, Betzer DP, Smith KD, Millman ZG, Michalski HC, Menchaca SE, Zambriski JA, Ojo KK, Hulverson MA, Arnold SL, Rivas KL, Vidadala RS, Huang W, Barrett LK, Maly DJ, Fan E, Van Voorhis WC, Riggs MW. 2016. Novel bumped kinase Inhibitors are safe and effective therapeutics in the calf clinical model for cryptosporidiosis. J Infect Dis 214:1856–1864. 10.1093/infdis/jiw488.
    1. Irwin SM, Gruppo V, Brooks E, Gilliland J, Scherman M, Reichlen MJ, Leistikow R, Kramnik I, Nuermberger EL, Voskuil MI, Lenaerts AJ. 2014. Limited activity of clofazimine as a single drug in a mouse model of tuberculosis exhibiting caseous necrotic granulomas. Antimicrob Agents Chemother 58:4026–4034. 10.1128/AAC.02565-14.
    1. Everitt D. 2012. Clofazimine in clinical trials for tuberculosis: resurrecting clofazimine. TB alliance, Global Alliance for TB drug development.
    1. Arnold SLM, Choi R, Hulverson MA, Schaefer DA, Vinayak S, Vidadala RSR, McCloskey MC, Whitman GR, Huang W, Barrett LK, Ojo KK, Fan E, Maly DJ, Riggs MW, Striepen B, Van Voorhis WC. 2017. Necessity of bumped kinase inhibitor gastrointestinal exposure in treating Cryptosporidium infection. J Infect Dis 216:55–63. 10.1093/infdis/jix247.
    1. Tzipori S, Griffiths JK. 1998. Natural history and biology of Cryptosporidium parvum. Adv Parasitol 40:5–36. 10.1016/s0065-308x(08)60116-5.
    1. Huang BQ, Chen XM, LaRusso NF. 2004. Cryptosporidium parvum attachment to and internalization by human biliary epithelia in vitro: a morphologic study. J Parasitol 90:212–221. 10.1645/GE-3204.
    1. Arnold SLM, Choi R, Hulverson MA, Whitman GR, McCloskey MC, Dorr CS, Vidadala RSR, Khatod M, Morada M, Barrett LK, Maly DJ, Yarlett N, Van Voorhis WC. 2019. P-glycoprotein-mediated efflux reduces the in vivo efficacy of a therapeutic targeting the gastrointestinal parasite Cryptosporidium. J Infect Dis 220:1188–1198. 10.1093/infdis/jiz269.
    1. Yawalkar SJ, Vischer W. 1979. Lamprene (clofazimine) in leprosy. Basic information. Lepr Rev 50:135–144. 10.5935/0305-7518.19790020.
    1. Zhang Y, Feng J, McManus SA, Lu HD, Ristroph KD, Cho EJ, Dobrijevic EL, Chan HK, Prud'homme RK. 2017. Design and solidification of fast-releasing clofazimine nanoparticles for treatment of cryptosporidiosis. Mol Pharm 14:3480–3488. 10.1021/acs.molpharmaceut.7b00521.
    1. National Research Council. 2011. Guide for the care and use of laboratory animals, 8th ed. National Academies Press, Washington, DC.
    1. Federation of Animal Science Societies. 2020. Guide for the care and use of agricultural animals in research and teaching, 3rd ed. Federation of Animal Science Societies, Champaign, IL.
    1. Imboden M, Schaefer DA, Bremel RD, Homan EJ, Riggs MW. 2012. Antibody fusions reduce onset of experimental Cryptosporidium parvum infection in calves. Vet Parasitol 188:41–47. 10.1016/j.vetpar.2012.02.014.

Source: PubMed

3
購読する