Generic placeholder image

Recent Patents on Anti-Cancer Drug Discovery

Editor-in-Chief

ISSN (Print): 1574-8928
ISSN (Online): 2212-3970

Research Article

Enzyme and Transporter Kinetics for CPT-11 (Irinotecan) and SN-38: An Insight on Tumor Tissue Compartment Pharmacokinetics Using PBPK

Author(s): Yingfang Fan, Najia Mansoor*, Tasneem Ahmad, Zhuo X. Wu, Rafeeq A. Khan, Martin Czejka, Syed Sharib, Mansoor Ahmed, Zhe S. Chen and Dong H. Yang*

Volume 14, Issue 2, 2019

Page: [177 - 186] Pages: 10

DOI: 10.2174/1574892814666190212164356

Price: $65

Abstract

Background: Computational tools are becoming more and more powerful and comprehensive as compared to past decades in facilitating pharmaceutical, pharmacological and clinical practice. Anticancer agents are used either as monotherapy or in combination therapy to treat malignant conditions of the body. A single antineoplastic agent may be used in different types of malignancies at different doses according to the stage of the disease.

Objective: To study the behavior of CPT-11 (Irinotecan) and its metabolite SN-38 in tumor tissue compartment through the Whole Body-Physiologically Pharmacokinetics (WB-PBPK) and to determine the activity of metabolic enzymes and transporters participating in the disposition of CPT-11 and SN-38 working in their physiological environment inside the human body.

Methods: Whole body PBPK approach is used to determine the activity of different metabolic enzymes and transporters involved in the disposition of CPT-11 and its active metabolite, SN-38. The concentrations and pharmacokinetic parameters of the parent compound and its metabolite administered at clinically applicable dose via the intravenous route in the tumor tissue are predicted using this approach.

Results: The activity rate constants of metabolic enzymes and transporters of CPT-11 are derived at their natural anatomic locations. Concentration-time curves of CPT-11 and SN-38 with their 5th to 95th percentage range are achieved at the tumor tissue level. Mean tumor tissue pharmacokinetics of both compounds are determined in a population of 100 individuals.

Conclusion: Tumor tissue concentration-time curves of CPT-11 and SN-38 can be determined via PBPK modeling. Rate constants of enzymes and transporters can be shown for healthy and tumor bearing individuals. The results will throw light on the effective concentration of active compound at its target tissue at the clinically applied IV dose.

Keywords: ADME, CPT-11, neoplasia, pharmacokinetics, SN-38, solid tumors, therapeutic index, whole body-physiology based pharmacokinetics (WB-PBPK).

[1]
Shou M, Hayashi M, Pan Y, et al. Modeling, prediction, and in vitro in vivo correlation of CYP3A4 induction. Drug Metab Dispos 2008; 36: 2355-70.
[2]
Kato M, Chiba K, Horikawa M, Sugiyama Y. The quantitative prediction of in vivo enzyme-induction caused by drug exposure from in vitro information on human hepatocytes. Drug Metab Pharmacokinet 2005; 20: 236-43.
[3]
Yamazaki S, Skaptason J, Romero D, Vekich S, Jones HM, Tan W, et al. Prediction of oral pharmacokinetics of cMet kinase inhibitors in humans: Physiologically based pharmacokinetic model versus traditional one-compartment model. Drug Metab Dispos 2011; 39: 383-93.
[4]
Zhu LQ, Yang JW, Zhang Y, Wang YM, Zhang JL, Zhao YY, et al. Prediction of pharmacokinetics and penetration of moxifloxacin in human with intra-abdominal infection based on extrapolated PBPK model. Korean J Physiol Pharmacol 2015; 19(2): 99-104.
[5]
Kuepfer L, Niederalt C, Wendl T, Schlender J, Willmann S, Lippert J, et al. Applied concepts in PBPK modeling: How to build a PBPK/PD model. CPT Pharmacometrics Syst Pharmacol 2016; 5: 516-31.
[6]
Jones H, Rowland-Yeo K. Basic concepts in physiologically based pharmacokinetic modeling in drug discovery and development. CPT Pharmacometrics Syst Pharmacol 2013; 2e63
[7]
Peters SA. Physiologically-Based Pharmacokinetic (PBPK) Modeling and simulations: Principles, Methods, and Applications in the Pharmaceutical Industry. John Wiley & Sons: Hoboken, NJ 2012.
[8]
Rostami-Hodjegan A, Tucker GT. Simulation and prediction of in vivo drug metabolism in human populations from in vitro data. Nat Rev Drug Discov 2007; 6: 140-8.
[9]
Vinks AA. The future of physiologically based pharmacokinetic modeling to predict drug exposure in pregnant women. CPT Pharmacometrics Syst Pharmacol 2013; 2e33
[10]
Tsamandouras N, Rostami-Hodjegan A, Aarons L. Combining the ‘bottomup’ and ‘topdown’ approaches in pharmacokinetic modelling: fitting pb pk models to observed clinical data. Br J Clin Pharmacol 2015; 79: 48-55.
[11]
Luzon E, Blake K, Cole S, Nordmark A, Versantvoort C, Berglund EG, et al. Physiologically based pharmacokinetic modeling in regulatory decision-making at the European Medicines Agency. Clin Pharmacol Ther 2016; 102(1): 98-105.
[12]
Wagner C, Zhao P, Pan Y, Hsu V, Grillo J, Huang SM, et al. Application of physiologically based pharmacokinetic (PBPK) modeling to support dose selection: Report of an FDA public workshop on PBPK. CPT Pharmacometrics Syst Pharmacol 2015; 4: 226-30.
[13]
Peters SA. Identification of intestinal loss of a drug through physiologically based pharmacokinetic simulation of plasma concentration-time profiles. Clin Pharm 2008; 47(4): 245-59.
[14]
Zhuang X, Lu C. PBPK modeling and simulation in drug research and development. Acta Pharm Sin B 2016; 6(5): 430-40.
[15]
Huang SM, Abernethy DR, Wang Y, Zhao P, Zineh I. The utility of modeling and simulation in drug development and regulatory review. J Pharm Sci 2013; 102(9): 2912-23.
[16]
Shugarts S, Benet LZ. The role of transporters in the pharmacokinetics of orally administered drugs. Pharm Res 2009; 26(9): 2039-54.
[17]
Mitch A, Phelps A. Sparreboom CPT-11 Pharmacogenetics: A finished puzzle? J Clin Oncol 2014; 32(22): 2287-9.
[18]
Ayrton A, Morgan P. Role of transport proteins in drug absorption, distribution and excretion. Xenobiotica 2001; 31(8-9): 469-97.
[19]
Dean M, Hamon Y, Chimini G. The human ATP-binding cassette (ABC) transporter superfamily. J Lipid Res 2001; 42(7): 1007-17.
[20]
Kim RB. Organic anion-transporting polypeptide (OATP) transporter family and drug disposition. Eur J Clin Invest 2003; 33(Suppl. 2): 1-5.
[21]
Pang KS. Enzyme and transporter-based drug-drug interactions. AAPS 2010. C
[http://dx.doi.org/10.1007/978-1-4419-0840-7_2]
[22]
Mohelnikova-Duchonova B, Melichar B, Soucek P. FOLFOX/FOLFIRI pharmacogenetics: The call for a personalized approachin colorectal cancer therapy. World J Gastroenterol 2014; 20(30): 10316-30.
[23]
US Cancer Statistics Working Group United States Cancer Statistics: 1999-2013 Incidence and Mortality Web-based Report. Atlanta, GA: Department of Health and Human Services, Centers for Disease Control and Prevention, and National Cancer Institute 2016.
[24]
Center MM, Jemal A, Smith RA. Worldwide variations in colorectal cancer. CA Cancer J Clin 2009; 59: 366-78.
[25]
Ferlay J, Soerjomataram I, Ervik M. Globo Can 2012 v1.0, Cancer Incidence and Mortality Worldwide: IARC Cancer Base No.11. Lyon, France: International Agency for Research on Cancer, 2013; 64(3): 381-7.
[26]
Rothenberg ML. CPT-11(CPT-11): Recent developments and future directions-colorectal cancer and beyond. Oncologist 2001; 6: 66-80.
[27]
Kohne CH, Thuss-Patience P, Catane R. A phase II trial of CPT-11 in patients (pts) with advanced gastric carcinoma (AGC). Ann of Oncol 1998; 9: 46-6.
[28]
Sevinc A, Kalender ME, Altinbas M, Ozkan M, Dikilitas M, Camci C. Anatolian Society of Medical Oncology (ASMO) irinotecan as a second-line monotherapy for small cell lung cancer. Asian Pac J Cancer Prev 2011; 12(4): 1055-9.
[29]
Noble CO, Krauze MT, Drummond DC, Yamashita Y, Saito R, Berger MS, et al. Novel nanoliposomal CPT-11 infused by convection-enhanced delivery in intracranial tumors: Pharmacology and efficacy. Cancer Res 2006; 66: 2801-6.
[30]
Bayever E, Fitzgerald JB, Kim J, Klinz S. Treatment of breast cancer with liposomal CPT-11. WO2016094402 (2016).
[31]
Heinrich G, Kerb R. Use of irinotecan for improved treatment of cancer based on MDR1. WO2003013535 (2003).
[32]
Bayever E, Dhindsa N, Fitzgerald J B, Laivins P, Moyo V, Niyikiza C. Methods for treating pancreatic cancer using combination therapies comprising liposomal irinotecan. US9339497 (2016).
[33]
Bayever E, Dhindsa N, Fitzgerald JB, Laivins P, Moyo V, Niyikiza C. Treatment of pancreatic cancer with liposomal irinotecan. US20170202840 (2017).
[34]
Chen J, Higgins B, Kolinsky K. Combined treatment with irinotecan and an epidermal growth factor receptor kinase inhibitor. US20050272737 (2005).
[35]
Govindarajan R, Zeitlin A. Methods of using thalidomide in combination with irinotecan. US7479499 (2009).
[36]
Bissery MC, Chiron-Blondel M, Lejeune P, Vrignaud P. Antitumor combinations containing a VEGf inhibiting agent and irinotecan. EP2173349 (2011).
[37]
Emanuel D, Ramachandra S. Combination of irinotecan and revimid for the treating multiple myeloma. WO2004100953 (2004).
[38]
Marsh S, Hoskins JM. Irinotecan pharmacogenomics. APJCP 2010; 11(7): 1003-10.
[39]
Hatfield MJ. Organ-specific carboxylesterase profiling identifies the small intestine and kidney as major contributors of activation of the anticancer prodrug CPT-11. Biochem Pharmacol 2001; 81(1): 24-31.
[40]
Wallace BD. Alleviating cancer drug toxicity by inhibiting a bacterial enzyme. Science 2010; 330(6005): 831-5.
[41]
Imran A, Aquilur R, Zhang J A. SN-38 lipid complexes and their methods of use. US7390502 (2008).
[42]
Govindan SV, Gale JB, Holman NJ, Goldenberg DM. Antibody- SN-38 Immunoconjugates with a CL2A linker. US, US9629926 (2017).
[43]
Paulik A, Grim J, Filip S. Predictors of irinotecan toxicity and efficacy in treatment of metastatic colorectral cancer. Acta Med (Hradec Kralove) 2012; 55(4): 153-9.
[44]
Innocenti F, Kroetz DL, Schuetz E, Dolan ME, Ramírez J, Relling M, et al. Comprehensive pharmacogenetic analysis of irinotecan neutropenia and pharmacokinetics. J Clin Oncol 2009; 27(16): 2604-14.
[45]
Ratain MJ. Methods and compositions for predicting irinotecan toxicity. WO2004108954 (2006).
[46]
Govindan SV, Goldenberg DM. Dosages of immunoconjugates of antibodies and SN-38 for improved efficacy and decreased toxicity. US9493574 (2016).
[47]
Iusuf D, Ludwig M, Elbatsh A, van Esch A, van de Steeg E, Wagenaar E, et al. OATP1A/1B transporters affect irinotecan and SN-38 pharmacokinetics and carboxylesterase expression in knockout and humanized transgenic mice. Mol Cancer Ther 2014; 13(2): 492-503.
[48]
Li M, Seiser EL, Baldwin RM. ABC transporter polymorphisms are associated with irinotecan pharmacokinetics and neutropenia. Pharmacog J 2016.
[http://dx.doi.org/10.1038/tpj.2016.75]
[49]
Rowland M, Balant L, Peck C. Physiologically based pharmacokinetics in drug development and regulatory science: A workshop report. AAPS J 2004; 6: 56-67.
[50]
Gerlowski LE, Jain RK. Physiologically based pharmacokinetic modeling: Principles and applications. J Pharm Sci 1983; 72(10): 1103-27.
[51]
Gospavic R, Knoll P, Mirzaei S, Popov V. Physiologically Based Pharmacokinetic (PBPK) model for biodistribution of radiolabeled peptides in patients with neuroendocrine tumours. Asia Ocean J Nucl Med Biol 2016; 4(2): 90-8.
[52]
Jadhav PR, Cook J, Sinha V, Zhao P, Rostami‐Hodjegan A, Sahasrabudhe V, et al. A proposal for scientific framework enabling specific population drug dosing recommendations. J Clin Pharmacol 2015; 55(10): 1073-8.
[53]
Jamei M, Dickinson GL, Rostami-Hodjegan A. A framework for assessing inter-individual variability in pharmacokinetics using virtual human populations and integrating general knowledge of physical chemistry, biology, anatomy, physiology and genetics: A tale of ‘bottom-up’ vs ‘top-down’ recognition of covariates. Drug Metab Pharmacokinet 2009; 24(1): 53-75.
[54]
Chandrani G. Drug metabolism and pharmacokinetics in drug discovery: A primer for bio-analytical chemists. Curr Sep 2001; 19: 3-11.
[55]
Zhao J, Cao Y, Jusko WJ. Across-species scaling of monoclonal antibody pharmacokinetics using a minimal PBPK model. Pharm Res 2015; 32(10): 3269-81.
[56]
Clewell HJ, Reddy MB, Lave T, Andersen ME. Physiologically Based Pharmacokinetic Modeling. In: Gad SC, Ed. Preclinical Drug Development Handbook. Hoboken: John Wiley & Sons 2008; pp. 1167-227.
[57]
Rivory LP, Haaz MC, Canal P, Lokiec F, Armand JP, Robert J. Pharmacokinetic interrelationships of irinotecan (CPT-11) and its three major plasma metabolites in patients enrolled in Phase I/II trials. Clin Cancer Res 1997; 3: 1261-6.
[58]
Kimie S, Naoko K. A new metabolite of CPT-11 in which formation is mediated by human hepatic cytochrome p-450 3a4. Drug Metab Dispos 2001; 29(11): 1505-13.
[59]
Dodds HM, Haaz MC, Riou JF, Robert J, Rivory LP. Identification of a new metabolite of CPT-11 (irinotecan): Pharmacological properties and activation to SN-38. J Pharmacol Exp Ther 1998; 286: 578-83.
[60]
Leslie E, Carlini N, Meropol J. UGT1A7 and UGT1A9 polymorphisms predict response and toxicity in colorectal cancer patients treated with capecitabine/irinotecan. Clin Cancer Res 2005; 11(3): 1226-36.
[61]
Goodman G. Pharmacokinetics and pharmacodynamics 11 Ed. Chapter 1. The McGraw-Hill Companies, Inc; 2005. The Pharmacological Basis of Therapeutics. ISBN:0-07-142280-3.
[62]
Fan Y, Mansoor N, Ahmad T. Physiologically based pharmacokinetic modeling for predicting Irinotecan exposure in human body. Oncotarget 2017; 8(29): 48178-85.
[63]
Takimoto CH. Pharmacokinetics and pharmacodynamic biomarkers in early oncology drug development. Eur J Cancer 2009; 45: 436-8.
[64]
Yap TA, Sandhu SK, Workman P, De Bono JS. Envisioning the future of early anticancer drug development. Nat Rev Cancer 2010; 10(7): 514-9.

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy