Human Hepatic Cell Cultures: In Vitro and In Vivo Drug Metabolism

Drug metabolism is the major determinant of drug clearance, and the factor most frequently responsible for inter-individual differences in drug pharmacokinetics. The expression of drug metabolising enzymes shows significant interspecies differences, and variability among human individuals (polymorphic or inducible enzymes) makes the accurate prediction of the metabolism of a new compound in humans difficult. Several key issues need to be addressed at the early stages of drug development to improve drug candidate selection: a) how fast the compound will be metabolised; b) what metabolites will be formed (metabolic profile); c) which enzymes are involved and to what extent; and d) whether drug metabolism will be affected directly (drug-drug interactions) or indirectly (enzyme induction) by the administered compound. Drug metabolism studies are routinely performed in laboratory animals, but they are not sufficiently accurate to predict the metabolic profiles of drugs in humans. Many of these issues can now be addressed by the use of relevant human in vitro models, which speed up the selection of new candidate drugs. Human hepatocytes are the closest in vitro model to the human liver, and they are the only model which can produce a metabolic profile of a drug which is very similar to that found in vivo. However, the use of human hepatocytes is restricted, because limited access to suitable tissue samples prevents their use in high throughput screening systems. The pharmaceutical industry has made great efforts to develop fast and reliable in vitro models to overcome these drawbacks. Comparative studies on liver microsomes and cells from animal species, including humans, are very useful for demonstrating species differences in the metabolic profile of given drug candidates, and are of great value in the judicious and justifiable selection of animal species for later pharmacokinetic and toxicological studies. Cytochrome P450 (CYP)-engineered cells (or microsomes from CYP-engineered cells, for example, Supersomes™) have made the identification of the CYPs involved in the metabolism of a drug candidate more straightforward and much easier. However, the screening of compounds acting as potential CYP inducers can only be conducted in cellular systems fully capable of transcribing and translating CYP genes.

[1]  X. Ponsoda,et al.  Comparative metabolism of the nonsteroidal antiinflammatory drug, aceclofenac, in the rat, monkey, and human. , 1996, Drug metabolism and disposition: the biological fate of chemicals.

[2]  C. Ioannides,et al.  Cytochromes P450 and species differences in xenobiotic metabolism and activation of carcinogen. , 1998, Environmental health perspectives.

[3]  X. Ponsoda,et al.  Metabolism of aceclofenac in humans. , 1996, Drug metabolism and disposition: the biological fate of chemicals.

[4]  C. Rodríguez-Antona,et al.  Quantitative RT-PCR measurement of human cytochrome P-450s: application to drug induction studies. , 2000, Archives of biochemistry and biophysics.

[5]  R. Ulrich,et al.  Cultured hepatocytes as investigational models for hepatic toxicity: practical applications in drug discovery and development. , 1995, Toxicology letters.

[6]  A. Seidel,et al.  Cytochrome P450 mediated reactions studied in genetically engineered V79 Chinese hamster cells. , 1995, Pharmacogenetics.

[7]  J. Castell,et al.  Cytochrome P450 regulation by hepatocyte nuclear factor 4 in human hepatocytes: A study using adenovirus‐mediated antisense targeting , 2001, Hepatology.

[8]  M. Gómez-Lechón,et al.  6 – Isolation, Culture and Use of Human Hepatocytes in Drug Research , 1997 .

[9]  C. Guguen-Guillouzo,et al.  Expression and induction of a large set of drug-metabolizing enzymes by the highly differentiated human hepatoma cell line BC2. , 2001, European journal of biochemistry.

[10]  U. Meyer,et al.  Molecular mechanisms of genetic polymorphisms of drug metabolism. , 1997, Annual review of pharmacology and toxicology.

[11]  M. T. Donato,et al.  Effect of xenobiotics on monooxygenase activities in cultured human hepatocytes. , 1990, Biochemical pharmacology.

[12]  A. Li,et al.  Preclinical evaluation of drug-drug interaction potential: present status of the application of primary human hepatocytes in the evaluation of cytochrome P450 induction. , 1997, Chemico-biological interactions.

[13]  M. T. Donato,et al.  Characterization of drug metabolizing activities in pig hepatocytes for use in bioartificial liver devices: comparison with other hepatic cellular models. , 1999, Journal of hepatology.

[14]  J. Castell,et al.  Re‐expression of C/EBPα induces CYP2B6, CYP2C9 and CYP2D6 genes in HepG2 cells , 1998 .

[15]  Rodrigues Ad,et al.  Integrated Cytochrome P450 Reaction Phenotyping: Attempting to Bridge the Gap Between cDNA-expressed Cytochromes P450 and Native Human Liver Microsomes , 1999 .

[16]  R. Edwards,et al.  Cytochrome P450 expression in human hepatocytes and hepatoma cell lines: molecular mechanisms that determine lower expression in cultured cells , 2002, Xenobiotica; the fate of foreign compounds in biological systems.

[17]  X. Ponsoda,et al.  Drug biotransformation by human hepatocytes. In vitro/in vivo metabolism by cells from the same donor. , 2001, Journal of hepatology.

[18]  A. Pfeifer,et al.  18 – Drug Metabolism and Carcinogen Activation Studies with Human Genetically Engineered Cells , 1996 .

[19]  M. Ranis,et al.  Induction of Human Drug-Metabolizing Enzymes: Mechanisms and Implications , 1999 .

[20]  I. Gut,et al.  P450 in the rat and man: methods of investigation, substrate specificities and relevance to cancer. , 1994, Xenobiotica; the fate of foreign compounds in biological systems.

[21]  M. T. Donato,et al.  Effect of model inducers on cytochrome P450 activities of human hepatocytes in primary culture. , 1995, Drug metabolism and disposition: the biological fate of chemicals.

[22]  F. Gonzalez,et al.  Molecular genetics of the P-450 superfamily. , 1990, Pharmacology & therapeutics.

[23]  R. Langenbach,et al.  Human cell lines, derived from AHH-1 TK+/- human lymphoblasts, genetically engineered for expression of cytochromes P450. , 1993, Toxicology.