Quantitative systems toxicology (QST) reproduces species differences in PF-04895162 liver safety due to combined mitochondrial and bile acid toxicity

Grant Generaux, Vinal V Lakhani, Yuching Yang, Sashi Nadanaciva, Luping Qiu, Keith Riccardi, Li Di, Brett A Howell, Scott Q Siler, Paul B Watkins, Hugh A Barton, Michael D Aleo, Lisl K M Shoda, Grant Generaux, Vinal V Lakhani, Yuching Yang, Sashi Nadanaciva, Luping Qiu, Keith Riccardi, Li Di, Brett A Howell, Scott Q Siler, Paul B Watkins, Hugh A Barton, Michael D Aleo, Lisl K M Shoda

Abstract

Many compounds that appear promising in preclinical species, fail in human clinical trials due to safety concerns. The FDA has strongly encouraged the application of modeling in drug development to improve product safety. This study illustrates how DILIsym, a computational representation of liver injury, was able to reproduce species differences in liver toxicity due to PF-04895162 (ICA-105665). PF-04895162, a drug in development for the treatment of epilepsy, was terminated after transaminase elevations were observed in healthy volunteers (NCT01691274). Liver safety concerns had not been raised in preclinical safety studies. DILIsym, which integrates in vitro data on mechanisms of hepatotoxicity with predicted in vivo liver exposure, reproduced clinical hepatotoxicity and the absence of hepatotoxicity observed in the rat. Simulated differences were multifactorial. Simulated liver exposure was greater in humans than rats. The simulated human hepatotoxicity was demonstrated to be due to the interaction between mitochondrial toxicity and bile acid transporter inhibition; elimination of either mechanism from the simulations abrogated injury. The bile acid contribution occurred despite the fact that the IC50 for bile salt export pump (BSEP) inhibition by PF-04895162 was higher (311 µmol/L) than that has been generally thought to contribute to hepatotoxicity. Modeling even higher PF-04895162 liver exposures than were measured in the rat safety studies aggravated mitochondrial toxicity but did not result in rat hepatotoxicity due to insufficient accumulation of cytotoxic bile acid species. This investigative study highlights the potential for combined in vitro and computational screening methods to identify latent hepatotoxic risks and paves the way for similar and prospective studies.

Keywords: DILIsym; PBPK; PF‐04895162; QSP; QST; bile acid transporters; drug‐induced liver injury; mechanistic; mitochondria; species translation.

Conflict of interest statement

The authors report that this research did not receive external public or private foundation funding. The study was sponsored by Pfizer. Authors are former or current employees of Pfizer or DSS or an advisory board member of DSS (PBW) and may continue to hold stock or other equity positions with the companies.

© 2019 The Authors. Pharmacology Research & Perspectives published by John Wiley & Sons Ltd, British Pharmacological Society and American Society for Pharmacology and Experimental Therapeutics.

Figures

Figure 1
Figure 1
Examples of in vitro data collected to quantitatively characterize the relationship between PF‐04895162 and mechanisms of toxicity. (A) PF‐04895162 interaction with human BSEP in vesicular transport assays (SB‐BSEP‐HEK293), using 0.2 µmol/L taurocholate as the probe substrate. PF‐04895162 inhibited BSEP‐mediated taurocholate transport by 38% at 250 µmol/L (maximum tested concentration). (B) PF‐04895162 interaction with rat Bsep in vesicular transport assays (SB‐ratBsep‐HEK293), using 2 µmol/L taurocholate as the probe substrate. The calculated IC50 value was 229.6 µmol/L. (C) Oxygen consumption rate (OCR) following 24‐hour incubation of primary human hepatocytes with PF‐04895162. Basal OCR was measured in a Seahorse XF Analyzer for 40 minutes, followed by injection of FCCP (3 µmol/L) and measurement of the increase in OCR as an indicator of spare respiratory capacity. Two independent studies were conducted, with similar results. (D) OCR following 1‐hour incubation of rat primary hepatocytes with PF‐04895162. Basal OCR and spare respiratory capacity were measured using a Seahorse XF Analyzer. Two independent studies were conducted, with similar results. Studies using 24‐hour incubation with PF‐04895162 were also conducted, with similar results
Figure 2
Figure 2
Comparisons of PBPK sub‐model simulations against measured data. The rat PBPK sub‐model was simultaneously optimized against multiple dosing protocols. (A) Simulated rat plasma PF‐04895162 (ng/mL) following single oral administration of 3, 30, or 100 mg/kg was compared with measured data for the same protocols. (B) Simulated rat plasma PF‐04895162 (ng/mL) for study day 28 following repeat daily dosing of 3, 30, or 100 mg/kg was compared with measured data for the same protocols. (C) Simulated human plasma PF‐04895162 (ng/mL) following optimization against data for a single 300‐mg po dose. (D) Simulated human plasma PF‐04895162 (ng/mL) from day 14, following 300‐mg BID dosing, as compared with data. The human repeat dosing simulation result did not require further changes to the human PBPK sub‐model optimized to a single 300‐mg dose. (E) Simulated rat liver concentrations on day 28 following administration of 100 mg/kg/day. (F) Simulated human liver concentrations on day 14 following administration of 300‐mg BID for 14 days
Figure 3
Figure 3
Simulation of PF‐04895162 (100 mg/kg/day for 28 days) in rat SimPops (n = 294) does not result in hepatotoxicity. Evaluation of drug‐induced serious hepatotoxicity (eDISH) plot for rat SimPops results, illustrating peak ALT (x‐axis) vs peak total bilirubin (y‐axis) for each individual. Each star represents peak ALT and total bilirubin for an individual rat. Vertical lines correspond to 3x ULN for ALT and 2x ULN for total bilirubin
Figure 4
Figure 4
Subclinical simulated indicators of mechanisms of toxicity in rat SimPops. (A) Liver CDCA‐amide across the 28‐day dosing period (100 mg/kg/day). Each line represents an individual rat. (B) Liver average ATP across the 28‐day dosing period (100 mg/kg/day)
Figure 5
Figure 5
Simulation of PF‐04895162 (300‐mg BID for 14 days) in human SimPops (n = 285) results in hepatotoxicity. Each star represents peak ALT and total bilirubin for an individual human subject in the eDISH plot
Figure 6
Figure 6
Simulated time courses for ALT in human SimPops treated with PF‐04895162 (300‐mg BID for 14 days and followed for an additional 14 days). Each line represents an individual subject
Figure 7
Figure 7
Comparison of time to peak ALT in human SimPops vs measured clinical data. Simulated individuals are represented in red. Clinical measurements for subjects in the Phase I study are represented in black. The median value is indicated by a line
Figure 8
Figure 8
Subclinical simulated indicators of mechanisms of toxicity in human SimPops. (A) Liver CDCA‐amide across the 14‐day dosing period (300‐mg BID), with 14‐day follow‐up. Each line represents an individual subject. (B) Liver average ATP across the 14‐day dosing period (300‐mg BID), with 14‐day follow‐up

References

    1. Battista C, Howell BA, Siler SQ, Watkins PB. An introduction to DILIsym® software, a mechanistic mathematical representation of drug‐induced liver injury In: Chen M, Will Y, eds. Drug‐induced liver toxicity. New York, NY: Springer; 2018:101–121.
    1. Longo DM, Generaux GT, Howell BA, et al. Refining liver safety risk assessment: application of mechanistic modeling and serum biomarkers to cimaglermin alfa (GGF2) clinical trials. Clin Pharmacol Ther. 2017;102:961‐969.
    1. Longo DM, Woodhead JL, Walker P, et al. Quantitative systems toxicology analysis of in vitro mechanistic assays reveals importance of bile acid accumulation and mitochondrial dysfunction in TAK‐875‐induced liver injury. Toxicol Sci Off J Soc Toxicol. 2019;167:458‐467.
    1. Woodhead JL, Yang K, Oldach D, et al. Analyzing the mechanisms behind macrolide antibiotic‐induced liver injury using quantitative systems toxicology modeling. Pharm Res. 2019;36:48.
    1. Battista C, Yang K, Stahl SH, et al. Using quantitative systems toxicology to investigate observed species differences in CKA-mediated hepatotoxicity. Toxicol Sci Off J Soc Toxicol. 2018;166:123–130.
    1. Howell BA, Yang Y, Kumar R, et al. In vitro to in vivo extrapolation and species response comparisons for drug‐induced liver injury (DILI) using DILIsymTM: a mechanistic, mathematical model of DILI. J Pharmacokinet Pharmacodyn. 2012;39:527‐541.
    1. Yang K, Woodhead JL, Watkins PB, Howell BA, Brouwer K. Systems pharmacology modeling predicts delayed presentation and species differences in bile acid‐mediated troglitazone hepatotoxicity. Clin Pharmacol Ther. 2014;96:589‐598.
    1. Kasteleijn‐Nolst Trenité D, Biton V, French JA, et al. Kv7 potassium channel activation with ICA‐105665 reduces photoparoxysmal EEG responses in patients with epilepsy. Epilepsia. 2013;54:1437‐1443.
    1. Roeloffs R, Wickenden AD, Crean C, et al. In vivo profile of ICA‐27243 [N‐(6‐chloro‐pyridin‐3‐yl)‐3,4‐difluoro‐benzamide], a potent and selective KCNQ2/Q3 (Kv7.2/Kv7.3) activator in rodent anticonvulsant models. J Pharmacol Exp Ther. 2008;326:818‐828.
    1. Aleo MD, Aubrecht J, Bonin D, et al. Phase I study of PF‐04895162, a Kv7 channel opener, reveals unexpected hepatotoxicity in healthy subjects, but not rats or monkeys: clinical evidence of disrupted bile acid homeostasis. Pharmacol Res Perspect. 2019;7:e00467.
    1. Bialer M, Johannessen SI, Levy RH, Perucca E, Tomson T, White HS. Progress report on new antiepileptic drugs: a summary of the Eleventh Eilat Conference (EILAT XI). Epilepsy Res. 2013;103:2‐30.
    1. Bhattacharya S, Shoda L, Zhang Q, et al. Modeling drug‐ and chemical‐induced hepatotoxicity with systems biology approaches. Front. Physiol. 2012;3:462.
    1. Shoda L, Woodhead JL, Siler SQ, Watkins PB, Howell BA. Linking physiology to toxicity using DILIsym®, a mechanistic mathematical model of drug‐induced liver injury. Biopharm Drug Dispos. 2014;35:33‐49.
    1. Watkins PB. The DILI‐sim initiative: insights into hepatotoxicity mechanisms and biomarker interpretation. Clin Transl Sci. 2019;12:122‐129.
    1. Woodhead JL, Howell BA, Yang Y, et al. An analysis of N‐acetylcysteine treatment for acetaminophen overdose using a systems model of drug‐induced liver injury. J Pharmacol Exp Ther. 2012;342:529‐540.
    1. Longo DM, Yang Y, Watkins PB, Howell BA, Siler SQ. Elucidating differences in the hepatotoxic potential of tolcapone and entacapone with DILIsym(®), a mechanistic model of drug‐induced liver injury. CPT Pharmacomet Syst Pharmacol. 2016;5:31‐39.
    1. Woodhead JL, Yang K, Siler SQ, et al. Exploring BSEP inhibition‐mediated toxicity with a mechanistic model of drug‐induced liver injury. Front Pharmacol. 2014;5:240.
    1. Woodhead JL, Brock WJ, Roth SE, et al. Application of a mechanistic model to evaluate putative mechanisms of tolvaptan drug‐induced liver injury and identify patient susceptibility factors. Toxicol Sci Off J Soc Toxicol. 2017;155:61‐74.
    1. Eakins J, Bauch C, Woodhouse H, et al. A combined in vitro approach to improve the prediction of mitochondrial toxicants. Toxicol Vitro Int J Publ Assoc BIBRA. 2016;34:161‐170.
    1. Nadanaciva S, Rana P, Beeson GC, et al. Assessment of drug‐induced mitochondrial dysfunction via altered cellular respiration and acidification measured in a 96‐well platform. J Bioenerg Biomembr. 2012;44:421‐437.
    1. Yang Y, Nadanaciva S, Will Y, et al. MITOsym®: a mechanistic, mathematical model of hepatocellular respiration and bioenergetics. Pharm Res. 2015;32:1975‐1992.
    1. Watkins PB, Desai M, Berkowitz SD, et al. Evaluation of drug‐induced serious hepatotoxicity (eDISH): application of this data organization approach to phase III clinical trials of rivaroxaban after total hip or knee replacement surgery. Drug Saf. 2011;34:243‐252.
    1. Schulz S, Schmitt S, Wimmer R, et al. Progressive stages of mitochondrial destruction caused by cell toxic bile salts. Biochim Biophys Acta. 2013;1828:2121‐2133.
    1. Kenna JG, Taskar KS, Battista C, et al. Can bile salt export pump inhibition testing in drug discovery and development reduce liver injury risk? An international transporter consortium perspective. Clin Pharmacol Ther. 2018;104:916‐932.
    1. Aleo MD, Luo Y, Swiss R, Bonin PD, Potter DM, Will Y. Human drug‐induced liver injury severity is highly associated with dual inhibition of liver mitochondrial function and bile salt export pump. Hepatology. 2014;60:1015‐1022.
    1. Kullak‐Ublick GA, Andrade RJ, Merz M, et al. Drug‐induced liver injury: recent advances in diagnosis and risk assessment. Gut. 2017;66:1154‐1164.
    1. Morgan RE, van Staden CJ, Chen Y, et al. A multifactorial approach to hepatobiliary transporter assessment enables improved therapeutic compound development. Toxicol Sci Off J Soc Toxicol. 2013;136:216‐241.
    1. Morgan RE, Trauner M, van Staden CJ, et al. Interference with bile salt export pump function is a susceptibility factor for human liver injury in drug development. Toxicol Sci Off J SocToxicol. 2010;118:485‐500.
    1. Michel MC, Radziszewski P, Falconer C, Marschall‐Kehrel D, Blot K. Unexpected frequent hepatotoxicity of a prescription drug, flupirtine, marketed for about 30 years. Br J Clin Pharmacol 2012;73:821‐825.
    1. Han D, Ybanez MD, Johnson HS, et al. Dynamic adaptation of liver mitochondria to chronic alcohol feeding in mice: biogenesis, remodeling, and functional alterations. J Biol Chem 2012;287:42165‐42179.

Source: PubMed

3
Abonnere