Testing paradigm for prediction of development-limiting barriers and human drug toxicity.

The financial investment grows exponentially as a new chemical entity advances through each stage of discovery and development. The opportunity exists for the modern toxicologist to significantly impact expenditures by the early prediction of potential toxicity/side effect barriers to development by aggressive evaluation of development-limiting liabilities early in drug discovery. Improved efficiency in pharmaceutical research and development lies both in leveraging "best in class" technology and integration with pharmacologic activities during hit-to-lead and early lead optimization stages. To meet this challenge, a discovery assay by stage (DABS) paradigm should be adopted. The DABS clearly delineates to discovery project teams the timing and type of assay required for advancement of compounds to each subsequent level of discovery and development. An integrative core pathology function unifying Drug Safety Evaluation, Molecular Technologies and Clinical Research groups that effectively spans all phases of drug discovery and development is encouraged to drive the DABS. The ultimate goal of such improved efficiency being the accurate prediction of toxicity and side effects that would occur in development before commitment of the large prerequisite resource. Good justification of this approach is that every reduction of development attrition by 10% results in an estimated increase in net present value by $100 million.

[1]  T. Sofuni,et al.  Validation study of the in vitro micronucleus test in a Chinese hamster lung cell line (CHL/IU). , 1999, Mutagenesis.

[2]  A M Richard,et al.  Structure-based methods for predicting mutagenicity and carcinogenicity: are we there yet? , 1998, Mutation research.

[3]  M. Fenech The in vitro micronucleus technique. , 2000, Mutation research.

[4]  E. Zeiger,et al.  Comparison of responses of base-specific Salmonella tester strains with the traditional strains for identifying mutagens: the results of a validation study. , 1998, Mutation research.

[5]  Peter C Jurs,et al.  Predicting the genotoxicity of polycyclic aromatic compounds from molecular structure with different classifiers. , 2003, Chemical research in toxicology.

[6]  G. Gibson Co-induction of cytochrome P4504A1 and peroxisome proliferation: a causal or casual relationship? , 1992, Xenobiotica; the fate of foreign compounds in biological systems.

[7]  C. Alden,et al.  The Tg rasH2 Mouse in Cancer Hazard Identification , 2002, Toxicologic pathology.

[8]  R G Ulrich,et al.  Clustering of hepatotoxins based on mechanism of toxicity using gene expression profiles. , 2001, Toxicology and applied pharmacology.

[9]  A. Sands,et al.  Predicting drug efficacy: knockouts model pipeline drugs of the pharmaceutical industry. , 2003, Current opinion in pharmacology.

[10]  R G Ulrich,et al.  Microarray analysis of hepatotoxins in vitro reveals a correlation between gene expression profiles and mechanisms of toxicity. , 2001, Toxicology letters.

[11]  D. Morris,et al.  Interspecies Differences in Renal Localization of Cyclooxygenase Isoforms: Implications in Nonsteroidal Antiinflammatory Drug-Related Nephrotoxicity , 1998, Toxicologic pathology.

[12]  Hannu Norppa,et al.  What do human micronuclei contain? , 2003, Mutagenesis.

[13]  J. Féher,et al.  Role of free-radical reactions in liver diseases. , 1992, Acta physiologica Hungarica.

[14]  J. Peters,et al.  Role of PPAR alpha in the mechanism of action of the nongenotoxic carcinogen and peroxisome proliferator Wy-14,643. , 1997, Carcinogenesis.

[15]  S. Irwin,et al.  Comprehensive observational assessment: Ia. A systematic, quantitative procedure for assessing the behavioral and physiologic state of the mouse , 1968, Psychopharmacologia.

[16]  J. Haseman,et al.  The role of transgenic mouse models in carcinogen identification. , 2002, Environmental health perspectives.

[17]  J. Sagartz,et al.  The Pathologist and Toxicologist in Pharmaceutical Product Discovery , 1999, Toxicologic pathology.

[18]  W. Haschek,et al.  Handbook of toxicologic pathology , 1991 .

[19]  R. W. Hansen,et al.  The price of innovation: new estimates of drug development costs. , 2003, Journal of health economics.

[20]  M. Rao,et al.  Peroxisome proliferation and hepatocarcinogenesis. , 1987, IARC scientific publications.

[21]  Stephen K. Durham,et al.  Predicting the Genotoxicity of Secondary and Aromatic Amines Using Data Subsetting To Generate a Model Ensemble , 2003, J. Chem. Inf. Comput. Sci..

[22]  V. Gervais,et al.  Assessment of the performance of the Ames II assay: a collaborative study with 19 coded compounds. , 2004, Mutation research.

[23]  S. Bursian,et al.  Underlying endotoxemia augments toxic responses to chlorpromazine: is there a relationship to drug idiosyncrasy? , 2002, The Journal of pharmacology and experimental therapeutics.

[24]  R. Macsween Pathology of the Liver , 1981 .

[25]  M. Hashimoto,et al.  [Draft ICH guideline S7B: guideline on safety pharmacology studies for assessing the potential for delayed ventricular repolarization (QT interval prolongation) by human pharmaceuticals]. , 2003, Nihon yakurigaku zasshi. Folia pharmacologica Japonica.

[26]  R. Snyder,et al.  Assessment of the sensitivity of the computational programs DEREK, TOPKAT, and MCASE in the prediction of the genotoxicity of pharmaceutical molecules , 2004, Environmental and molecular mutagenesis.

[27]  A. Sands,et al.  Knockouts model the 100 best-selling drugs—will they model the next 100? , 2003, Nature Reviews Drug Discovery.

[28]  Thierry Arnould,et al.  Use of a low-density microarray for studying gene expression patterns induced by hepatotoxicants on primary cultures of rat hepatocytes. , 2003, Toxicological sciences : an official journal of the Society of Toxicology.

[29]  D. Baltimore,et al.  Failure of lymphopoiesis after adoptive transfer of NF-kappaB-deficient fetal liver cells. , 1997, Immunity.

[30]  A. Camm,et al.  Relationships between preclinical cardiac electrophysiology, clinical QT interval prolongation and torsade de pointes for a broad range of drugs: evidence for a provisional safety margin in drug development. , 2003, Cardiovascular research.

[31]  G M Pearl,et al.  Integration of computational analysis as a sentinel tool in toxicological assessments. , 2001, Current topics in medicinal chemistry.

[32]  M. Meadows Why drugs get pulled off the market. , 2002, FDA consumer.

[33]  S. Wolfe,et al.  Timing of new black box warnings and withdrawals for prescription medications. , 2002, JAMA.

[34]  H. Stopper,et al.  Evaluation of the in vitro micronucleus test as an alternative to the in vitro chromosomal aberration assay: position of the GUM Working Group on the in vitro micronucleus test. Gesellschaft für Umwelt-Mutations-forschung. , 1998, Mutation research.

[35]  D. Waxman,et al.  P450 gene induction by structurally diverse xenochemicals: central role of nuclear receptors CAR, PXR, and PPAR. , 1999, Archives of biochemistry and biophysics.

[36]  I. Mcfarlane Pathogenesis of autoimmune hepatitis. , 1999, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie.

[37]  M. Sanguinetti,et al.  Molecular and Cellular Mechanisms of Cardiac Arrhythmias , 2001, Cell.

[38]  C. Alden,et al.  A Critical Appraisal of the Value of the Mouse Cancer Bioassay in Safety Assessment , 1996, Toxicologic pathology.

[39]  K. Petersen From Toxic Precursors to Safe Drugs , 2002, Arzneimittelforschung.