Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-x4r87 Total loading time: 0 Render date: 2024-04-25T10:33:23.984Z Has data issue: false hasContentIssue false

17 - Application of TK/PD modeling in predicting dose-limiting toxicity

from II - INTEGRATED APPROACHES OF PREDICTIVE TOXICOLOGY

Published online by Cambridge University Press:  06 December 2010

Jinghai J. Xu
Affiliation:
Merck Research Laboratory, New Jersey
Laszlo Urban
Affiliation:
Novartis Institutes for Biomedical Research, Massachusetts
Get access

Summary

INTRODUCTION

For the development of cytotoxic anticancer pharmaceuticals, the initial phase 1 clinical trials in patients with advanced cancer disease usually include determining a maximum tolerated dose (MTD) and identifying dose-limiting toxicity (DLT). The MTD is usually defined as the dose tolerated by five out of six patients with acceptable levels of DLT and dose levels above which result in unacceptable DLT in two or more patients. The dose–response curve for many pharmaceuticals in this class is steep. Traditionally, the dose recommended for phase 2 trials of cytotoxic agents is based on the MTD determined in phase 1. The inherent assumption in establishing and using an MTD is that the therapeutic effect and the associated toxicities are correlated and that the mechanism of action of both the toxic and therapeutic effects is the same – higher doses result in greater efficacy. Therefore, it is not uncommon for the efficacious dose of anticancer pharmaceuticals to be at or near the MTD. Predicting the dose and exposure at which DLT will likely occur in humans provides value in designing and conducting the first clinical trial for a cytotoxic anticancer pharmaceutical.

Before testing in patients, the toxicities of these pharmaceutical candidates are studied in nonclinical species in toxicology studies to determine the DLT. Toxicokinetics (TK) represent the assessment of systemic exposure in toxicity studies, in which pharmacokinetic data are generated, either from the same animals used in the main toxicology study (usually for larger species such as dogs or monkeys) or from a satellite group (usually for smaller species such as rats or mice) at each dose level used in the main toxicology study.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2010

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

LVaS, Rubinstein, Richard, M. Phase I Clinical Trial Design. In: Budman, DR, Calvert, AH, Rowinsky, EK, Hill, BT, eds. Handbook of Anticancer Drug Development. Baltimore: Lippincott Williams & Wilkins; 2003:297.Google Scholar
Kummar, S, Gutierrez, M, Doroshow, JH, et al. Drug development in oncology: classical cytotoxics and molecularly targeted agents. Br J Clin Pharmacol. 2006;62(1): 15–26.CrossRefGoogle ScholarPubMed
JMaEMJ, Collins. Measurements of Endpoints in Phase I Drug Design, Toxicity Versus Alternatives. In: Budman, DR, Calvert, AH, Rowinsky, EK, Hill, BT, ed. Handbook of Anticancer Drug Development. Baltimore: Lippincott Williams & Wilkins; 2003:319.Google Scholar
Haines, IE. Dose selection in phase I studies: Why we should always go for the most effective. J Clin Oncol. 2008;26(21):3650–3652; author reply 3652–3653.CrossRefGoogle ScholarPubMed
Bross, PF, Kane, R, Farrell, AT, et al. Approval summary for bortezomib for injection in the treatment of multiple myeloma. Clin Cancer Res. 2004;10(12 Pt 1):3954–3964.CrossRefGoogle ScholarPubMed
Goodman, VL, Rock, EP, Dagher, R, et al. Approval summary: Sunitinib for the treatment of imatinib refractory or intolerant gastrointestinal stromal tumors and advanced renal cell carcinoma. Clin Cancer Res. 2007;13(5):1367–1373.CrossRefGoogle ScholarPubMed
,FDA. Guideline for Industry – Toxicokinetics: The assessment of Systemic Exposure in Toxicity Studies. March 1995.
Bruinink, A. In Vitro Toxicokinetics and Dynamics: Modeling and Interpretation of Toxicity Data. In: Gad, SC, ed. Preclinical Development Handbook Toxicology: Wiley-Interscience; 2008:509–550.Google Scholar
Timm, J. Mathematical Models for Risk Extrapolation. In: Greim, HS, ed. Toxicology and Risk Assessment A Comprehensive introduction. Wiley; Chichester, 2008:479–494.Google Scholar
Krishnan, KA, Melvin, E. Physiologically-based pharmacokinetic modeling in toxicology. In: Hayes, AW, ed. Principles and Methods of Toxicology. 4th ed. Philadelphia: Taylor & Francis; 2001:193–241.Google Scholar
Diack, C, Bois, FY. Pharmacokinetic-pharmacodynamic models for categorical toxicity data. Regul Toxicol Pharmacol. 2005;41(1):55–65.CrossRefGoogle ScholarPubMed
Bongard, HJ, Sparidans, RW, Critchley, DJ, et al. Pharmacokinetic drug-drug interaction of the novel anticancer agent E7070 and acenocoumarol. Invest New Drugs. 2004;22(2):151–158.CrossRefGoogle ScholarPubMed
Wong, CM, Ko, Y, Chan, A. Clinically significant drug-drug interactions between oral anticancer agents and nonanticancer agents: Profiling and comparison of two drug compendia. Ann Pharmacother. 2008;42(12):1737–1748.CrossRefGoogle ScholarPubMed
Chen, N, Lau, H, Kong, L, et al. Pharmacokinetics of lenalidomide in subjects with various degrees of renal impairment and in subjects on hemodialysis. J Clin Pharmacol. 2007;47(12):1466–1475.CrossRefGoogle ScholarPubMed
Verbeeck, RK. Pharmacokinetics and dosage adjustment in patients with hepatic dysfunction. Eur J Clin Pharmacol. 2008;64(12):1147–1161.CrossRefGoogle ScholarPubMed
Kesteren, C, Mathot, RA, Beijnen, JH, et al. Pharmacokinetic-pharmacodynamic guided trial design in oncology. Invest New Drugs. 2003;21(2):225–241.CrossRefGoogle Scholar
Mandema, JW, Sansom, LN, Dios-Vieitez, MC, et al. Pharmacokinetic-pharmacodynamic modeling of the electroencephalographic effects of benzodiazepines. Correlation with receptor binding and anticonvulsant activity. J Pharmacol Exp Ther. 1991;257(1):472–478.Google ScholarPubMed
Webster, R, Allan, G, Anto-Awuakye, K, et al. Pharmacokinetic/pharmacodynamic assessment of the effects of E4031, cisapride, terfenadine and terodiline on monophasic action potential duration in dog. Xenobiotica. 2001;31(8–9):633–650.CrossRefGoogle Scholar
Chen, X, Cass, JD, Bradley, JA, et al. QT prolongation and proarrhythmia by moxifloxacin: Concordance of preclinical models in relation to clinical outcome. Br J Pharmacol. 2005;146(6):792–799.CrossRefGoogle ScholarPubMed
Hanada, K, Asano, K, Nishimura, T, et al. Use of a toxicity factor to explain differences in nephrotoxicity and myelosuppression among the platinum antitumour derivatives cisplatin, carboplatin and nedaplatin in rats. J Pharm Pharmacol. 2008;60(3):317–322.CrossRefGoogle ScholarPubMed
Grochow, LB, Rowinsky, EK, Johnson, R, et al. Pharmacokinetics and pharmacodynamics of topotecan in patients with advanced cancer. Drug Metab Dispos. 1992;20(5):706–713.Google ScholarPubMed
Boxenbaum, H. Interspecies scaling, allometry, physiological time, and the ground plan of pharmacokinetics. J Pharmacokinet Biopharm. 1982;10(2):201–227.CrossRefGoogle ScholarPubMed
Mahmood, I. Prediction of clearance, volume of distribution and half-life by allometric scaling and by use of plasma concentrations predicted from pharmacokinetic constants: A comparative study. J Pharm Pharmacol. 1999;51(8):905–910.CrossRefGoogle ScholarPubMed
Dedrick, R, Bischoff, KB, Zaharko, DS. Interspecies correlation of plasma concentration history of methotrexate (NSC-740). Cancer Chemother Rep. 1970;54(2):95–101.Google Scholar
Wajima, T, Yano, Y, Fukumura, K, et al. Prediction of human pharmacokinetic profile in animal scale up based on normalizing time course profiles. J Pharm Sci. 2004;93(7):1890–1900.CrossRefGoogle ScholarPubMed
Holford, NH, Guentert, TW, Dingemanse, J, et al. Pharmacodynamics of lazabemide, a reversible and selective inhibitor of monoamine oxidase B. Br J Clin Pharmacol. 1994;37(6):553–557.CrossRefGoogle ScholarPubMed
Jann, MW, Shirley, KL, Small, GW. Clinical pharmacokinetics and pharmacodynamics of cholinesterase inhibitors. Clin Pharmacokinet. 2002;41(10):719–739.CrossRefGoogle ScholarPubMed
Hosmer, DWL, Stanley, . Applied Logistic Regression. 2nd ed. New York: Wiley; 2000.CrossRefGoogle Scholar
Agresti, A. Categorical Data Analysis. New York: Wiley-Interscience; 2002.CrossRefGoogle Scholar
Nick, TG, Campbell, KM. Logistic regression. Methods Mol Biol. 2007;404:273–301.CrossRefGoogle ScholarPubMed
Ratain, MJ. Therapeutic relevance of pharmacokinetics and pharmacodynamics. Semin Oncol. 1992;19(Suppl 11):8–13.Google ScholarPubMed
Aarons, L, Graham, G. Methodological approaches to the population analysis of toxicity data. Toxicol Lett. 2001;120(1–3):405–410.CrossRefGoogle ScholarPubMed
Bonate, PL. Nonlinear Mixed Effects Models. In Bonate, PL, Pharmacokinetic-Pharmacodynamic Modeling and Simulation: Springer; New York, 2006.Google Scholar
Ette, EI, Williams, PJ. Population pharmacokinetics: II. Estimation methods. Ann Pharmacother. 2004;38(11):1907–1915.CrossRefGoogle ScholarPubMed
Ette, EI, Williams, PJ. Population pharmacokinetics: I, Background, concepts, and models. Ann Pharmacother. Oct 2004;38(10):1702–1706.CrossRefGoogle ScholarPubMed
Ette, EI, Williams, PJ, Lane, JR. Population pharmacokinetics: III. Design, analysis, and application of population pharmacokinetic Studies. Ann Pharmacother. 2004;38(12):2136–2144.CrossRefGoogle ScholarPubMed
Roy, A, Ette, EI. A pragmatic approach to the design of population pharmacokinetic studies. Aaps J. 2005;7(2):E408–420.CrossRefGoogle ScholarPubMed
Spiegelhalter, D. J., Best, N. G., Gilks, W. R., Inskip, H. (1995 c). Hepatitis: a case study in MCMC methods. In Markov chain Monte Carlo Methods in practice. (ed.\ W. R. Gilks, S. Richardson, and D. J. Spiegelhalter), pp. 21–43. Chapman and Hall, New York.Google Scholar
Goldsmith, MA, Slavik, M, Carter, SK. Quantitative prediction of drug toxicity in humans from toxicology in small and large animals. Cancer Res. 1975;35(5):1354–1364.Google ScholarPubMed
Friberg, , Freijs, A, Sandstrom, M, et al. Semiphysiological model for the time course of leukocytes after varying schedules of 5-fluorouracil in rats. J Pharmacol Exp Ther. 2000;295(2):734–740.Google ScholarPubMed
Gourdeau, H, Leblond, L, Hamelin, B, et al. Species differences in troxacitabine pharmacokinetics and pharmacodynamics: implications for clinical development. Clin Cancer Res. 2004;10(22):7692–7702.CrossRefGoogle ScholarPubMed
Hassan, SB, Haglund, C, Aleskog, A, et al. Primary lymphocytes as predictors for species differences in cytotoxic drug sensitivity. Toxicol In Vitro. 2007;21(6):1174–1181.CrossRefGoogle ScholarPubMed
Hochhaus, G, Barrett, JS, Derendorf, H. Evolution of pharmacokinetics and pharmacokinetic/dynamic correlations during the 20th century. J Clin Pharmacol. 2000;40(9):908–917.CrossRefGoogle ScholarPubMed
Cavero, I. Using pharmacokinetic/pharmacodynamic modelling in safety pharmacology to better define safety margins: a regional workshop of the Safety Pharmacology Society. Expert Opin Drug Saf. 2007;6(4):465–471.CrossRefGoogle ScholarPubMed
Machado, SG, Miller, R, Hu, C. A regulatory perspective on pharmacokinetic/pharmacodynamic modelling. Stat Methods Med Res. 1999;8(3):217–245.CrossRefGoogle ScholarPubMed
Rajman, I. PK/PD modelling and simulations: utility in drug development. Drug Discov Today. 2008;13(7–8):341–346.CrossRefGoogle ScholarPubMed
Mutlib, AE, Gerson, RJ, Meunier, PC, et al. The species-dependent metabolism of efavirenz produces a nephrotoxic glutathione conjugate in rats. Toxicol Appl Pharmacol. 2000;169(1):102–113.CrossRefGoogle ScholarPubMed
Yu, LJ, Bulychev, A, O'Brien, L, et al. Pharmacokinetics and Pharmacodynamics of a Selective Proteasome Inhibitor MLN9708 in Nonclinical Species Following either Intravenous or Oral Administration. Proc Am Assoc Cancer Res. 2009;50.Google Scholar
Aitken, MM. Species differences in pharmacodynamics: some examples. Vet Res Commun. Dec 1983;7(1–4):313–324.CrossRefGoogle ScholarPubMed
Greaves, P, Williams, A, Eve, M . First dose of potential new medicines to humans: How animals help. Nat Rev Drug Discov. 2004;3(3):226–236.CrossRefGoogle ScholarPubMed
Kupperman, E, Lee, EC, Cao, Y, et al. Evaluation of the proteasome inhibitor MLN9708 in preclinical models of human cancer. Cancer Res. 2010;70(5): 1970–1980.Google ScholarPubMed
Simonsen, , Wahlby, U, Sandstrom, M, et al. Haematological toxicity following different dosing schedules of 5-fluorouracil and epirubicin in rats. Anticancer Res. 2000;20(3A):1519–1525.Google ScholarPubMed
Schurig, JE, Florczyk, AP, Bradner, WT. The mouse as a model for predicting the myelosuppressive effects of anticancer drugs. Cancer Chemother Pharmacol. 1986;16(3):243–246.CrossRefGoogle ScholarPubMed
Karlsson, MO, Molnar, V, Bergh, J, et al. A general model for time-dissociated pharmacokinetic-pharmacodynamic relationship exemplified by paclitaxel myelosuppression. Clin Pharmacol Ther. 1998;63(1):11–25.CrossRefGoogle ScholarPubMed
Pessina, A, Albella, B, Bayo, M, et al. Application of the CFU-GM assay to predict acute drug-induced neutropenia: An international blind trial to validate a prediction model for the maximum tolerated dose (MTD) of myelosuppressive xenobiotics. Toxicol Sci. 2003;75(2):355–367.CrossRefGoogle ScholarPubMed
Gabrielsson, JL, Weiner, DL. Methodology for pharmacokinetic/pharmacodynamic data analysis. Pharm Sci Technol Today. 1999;2(6):244–252.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×