html5-img
1 / 81

Non-Clinical Drug Development

Non-Clinical Drug Development. Chris H. Takimoto, MD, PhD Institute for Drug Development San Antonio Cancer Institute University of Texas Health Science Center San Antonio, TX. Drug Development. Drug discovery & screening Non-clinical development Animal scale up Phase I studies

teal
Download Presentation

Non-Clinical Drug Development

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Non-Clinical Drug Development Chris H. Takimoto, MD, PhD Institute for Drug Development San Antonio Cancer Institute University of Texas Health Science Center San Antonio, TX

  2. Drug Development • Drug discovery & screening • Non-clinical development • Animal scale up • Phase I studies • Phase II studies • Phase III studies Specific examples from anticancer drug development

  3. Overview of Anticancer Drug Development NDA IND Chemical Synthesis and Formulation Development Animal Models for Efficacy Assay Development Large Efficacy Trials with PK Screen Dose Escalation and Initial PK Proof of Concept and Dose Finding Animal PK and PD PK/PD Studies in Special Populations PHASE I PHASE II PHASE III Pre-Clinical Development Clinical Development

  4. Goals of Non-Clinical Testing of Small Molecule Drugs and Biologicals • To characterize potential adverse drug effects • Define end organ toxicities • Define reversibility of toxicity • To characterize pharmacokinetic profile • To characterize beneficial pharmacodynamic effects • Proof of principle • To guide safe use in human clinical studies • To determine a safe & reasonable starting dose • Provide monitoring guidelines for the clinical study • Provide sufficient data to conclude that patients are not exposed to unreasonable risks • Potential for benefit must also exist

  5. Oncology drug development is changing in the new era of targeted cancer therapies

  6. Conventional Wisdom in the New Age of Modern Drug Development • Targeted therapies are unique and distinct from classic cytotoxic chemotherapies • Classical chemotherapy = poisons • All new agents entering the clinical have well defined molecular targets • Conventional clinical study designs are outdated, outmoded, and poorly-suited to develop targeted therapies • Biomarkers rule!

  7. What are Targeted Therapies?The NCI’s Definition • From the NCI’s internet fact sheet • Targeted cancer therapies use drugs that block the growth and spread of cancer. • They interfere with specific molecules involved in carcinogenesis … and tumor growth. • Because scientists call these molecules “molecular targets,” these therapies are sometimes called “molecularly-targeted therapies.” • By focusing on molecular and cellular changes that are specific to cancer, targeted cancer therapies may be more effective than current treatments and less harmful to normal cells. (http://www.cancer.gov/cancertopics/factsheet/Therapy/targeted)

  8. Imatinib (Gleevec™) Poster Child for Targeted Therapies Imatinib (Gleevec™) • Prototypical targeted agent is imatinib • A selective inhibitor of BCR-Abl tyrosine kinase in CML or c-Kit in GIST

  9. Chronic Myelogenous Leukemia and the Ph Chromosome • Abnormal Philadelphia (Ph) chromosome identified in most patients with chronic myelogenous leukemia (CML) • Identified in over 90% of CML, 20% of adult ALL and 5% of pediatric ALL patients • Piece of chromosome 9 is abnormally linked to chromosome 22 • 9:22 translocation • c-Abl, the cellular homologue of the transforming retrovirus oncogene (v-Abl), is located on chromosome 9 • Activation of c-Abl signals the cell to proliferate and grow

  10. Targeted Therapy with Imatinib • Imatinib is a potent inhibitor of the BCR-Abl and the c-Kit tyrosine kinases • Generates marked growth inhibition of CML cells and Gastrointestinal Stromal Cell Tumors (GIST) • Early Phase I data in CML • Hematological response in 98% and cytogenetic remissions in 13% of patients treated in Phase I • Substantial single agent activity in GIST tumors

  11. Targeted Therapies & Preclinical Development(adapted from Paoletti 2005)

  12. Targeted Therapies & Phase I Trials(adapted from Paoletti 2005)

  13. Components of Non-Clinical Drug Development • In vitro studies: Cell lines, cell-free systems (drug screening) • Drug formulation • Chemistry, Manufacturing, and Controls: Drug supply & quality • In vivo efficacy studies: Animal models and proof of principle 5. Non-clinical safety studies

  14. In Vitro Study Goals: Define the Drug’s Pharmacology • Molecular mechanism of action and specific drug targets • Molecular pharmacology • Determinants of response • Intracellular pharmacodynamics • Mechanisms of drug resistance

  15. In Vitro Study Systems • Cell-free assay for specific molecular effects • Enzyme inhibition, receptor blockade, etc. • Yeast-based screening in genetically defined target • Mammalian cell lines: (murine, human, etc.)

  16. Preclinical PharmacologyIn Vitro Studies of Cancer Agents (1) • Define anticancer effects • Growth inhibition, differentiation, apoptosis, etc • Impact on defined biochemical and molecular pathways • RNA, DNA and protein biosynthesis, signaling kinases, etc • Spectrum of antitumor activity • Human tumor cell lines

  17. Preclinical PharmacologyIn Vitro Studies of Cancer Agents (2) • Cellular uptake and membrane transport • MDR, MRP, etc • Mechanisms of resistance • In vitro drug metabolism • P450 isoenzymes • Effects on hERG channels (prolonged QT interval risk) • Preliminary protein binding studies

  18. Components of Non-Clinical Drug Development • In vitro studies: Cell lines, cell-free systems (drug screening) • Drug formulation • Chemistry, Manufacturing, and Controls: Drug supply & quality • In vivo efficacy studies: Animal models and proof of principle 5. Non-clinical safety studies

  19. Drug Supply and Formulation • Drug supply: bulk chemical synthesis, natural product isolation, etc. • Good Manufacturing Practice (GMP) guidelines for pharmaceutical product manufacturing • Formulation for clinical delivery of drug: vehicles for intravenous or other routes of administration

  20. Drug Supply Issues • Paclitaxel source from the bark and wood of the Pacific Yew tree • Early drug supply limited the amount available for initial clinical trials • Newer semisynthetic production from the needles of the Yew tree (renewable)

  21. Drug Formulation Issues • Poor water solubility of natural products • Paclitaxel formulation in Cremophore EL™ (increased toxicity?) • Camptothecin derivatives formulated in a dimethylacetamide, polyethylene glycol and phosphoric acid vehicle • Later formulated as a lipid colloidal dispersion

  22. Components of Non-Clinical Drug Development • In vitro studies: Cell lines, cell-free systems (drug screening) • Drug formulation • Chemistry, Manufacturing, and Controls: Drug supply & quality • In vivo efficacy studies: Animal models and proof of principle 5. Non-clinical safety studies

  23. In Vivo Study Goals:Animal Models • Efficacy: Proof of therapeutic principle • Toxicology: Toxicity profile • Practical Issues: • Animal pharmacokinetics and pharmacodynamics • Starting dose and schedule for clinical trials

  24. Animal ModelsProof of Principle • Animal screening is too expensive for routine use • Efficacy in animal models of specific disease states occurs after in vitro studies • Evaluation of therapeutic index • Toxicity versus efficacy

  25. Ideal Animal Model • Validity • Selectivity • Predictability • Reproducibility “There is no perfect tumor model”

  26. Endostatin: An Endogenous Inhibitor of Angiogenesis and Tumor GrowthO'Reilly et al, Cell 88:277-285 (1997)

  27. Animal Models in Cancer • Spontaneous tumors • Idiopathic • Carcinogen-induced • Transgenic/gene knockout animals: p53, RB, etc • Transplanted tumors • Animal tumors: Lewis lung, S180 sarcoma, etc • Human tumor xenografts: human tumor lines implanted in immunodeficient mice (current NCI standard in vivo efficacy testing system) • Human tumors growing in vivo in implantable hollow fibers

  28. Human Tumor Xenografts • Athymic “nude”mice developed in 1960’s • Mutation in nu gene on chromosome 11 • Phenotype: retarded growth, low fertility, no fur, immunocompromised • Lack thymus gland, T-cell immunity • First human tumor xenograft of colon adenocarcinoma by Rygaard & Poulson, 1969

  29. Athymic Nude Mice

  30. Murine Xenograft Sites • Subcutaneous tumor (NCI method of choice) with IP drug administration • Intraperitoneal • Intracranial • Intrasplenic • Renal subcapsule • Site-specific (orthotopic) organ inoculation

  31. Xenograft Study Endpoints • Toxicity Endpoints: • Drug related death • Net animal weight loss • Efficacy Endpoints • Clonogenic assay • Tumor growth assay (corrected for tumor doubling time) • Treated/control survival ratio • Tumor weight change

  32. Xenograft Tumor Weight Change • Tumor weight change ratio (used by the NCI in xenograft evaluation) • Defined as: treated/control x 100% • Tumor weight in mg = (a x b2)/2 • a = tumor length • b = tumor width • T/C < 40-50% is considered significant

  33. Xenograft Advantages • Many different human tumor cell lines transplantable • Wide representation of most human solid tumors • Allows for evaluation of therapeutic index • Good correlation with drug regimens active in human lung, colon, breast, and melanoma cancers

  34. Xenograft Disadvantages • Brain tumors difficult to model • Different biological behavior, metastases rare • Survival not an ideal endpoint: death from bulk of tumor, not invasion • Shorter doubling times than original growth in human • Less necrosis, better blood supply • Difficult to maintain animals due to infection risks • Host directed therapies (angiogenesis, immune modulation) may not be applicable • Human vs. murine effects

  35. Other Animal Models • Orthotopic animal models: Tumor cell implantation in target organ • Metastatic disease models • Transgenic Animal Models • P53 or other tumor suppressor gene knockout animals • Endogenous tumor cell development • May be of high value for mAb therapies

  36. Non-Clinical Efficacy TestingThe FDA Perspective(J. Leighton, FDA ODAC Meeting, March 13, 2006) • Pharmacological activity assessed by models of disease are generally of low relevance to safety (IND) and efficacy (NDA) decisions • Efficacy in vivo and in vitro from non-clinical studies may not dependably predict clinical efficacy • Heterogeneity of disease • Interspecies differences in ADME • Role of immune system • Pharmacology studies are useful for: • Assessing an appropriate schedule (daily, weekly, q3wks) • Justification for a drug combination • Understanding effect at a molecular target • Examine receptor specificity • Identifying and evaluating biomarkers

  37. Components of Non-Clinical Drug Development • In vitro studies: Cell lines, cell-free systems (drug screening) • Drug formulation • Chemistry, Manufacturing, and Controls: Drug supply & quality • In vivo efficacy studies: Animal models and proof of principle 5. Non-clinical safety studies

  38. Non-Clinical Safety Studies • Safety pharmacology • Toxicokinetics & pharmacokinetic studies • Single dose toxicity studies • Repeated dose toxicity studies

  39. Safety Pharmacology • Assessment of drug on vital functions • Examples: • Cardiovascular: heart rate, BP, ECG, QT interval • Central nervous system: locomotor activity, coordination, proconvulsive effects, analgesic effects • Respiratory system: respiratory rate, tidal and minute volumes • Should complete prior to FIH studies • May be separate or a component of toxicity studies

  40. Pharmacokinetic & Toxicokinetic Studies • Analytic assay development and testing • Preclinical PK/PD efficacy and toxicity relationships • Initial drug formulation testing • Testing of different schedules and routes of administration • Animal ADME

  41. Non-Clinical Toxicology Studies • GLP Toxicology is expected • Use the clinical schedule, route, and formulation • Single dose acute toxicity studies required in 2 mammalian species prior to FIH studies • Classically rat and dog for small molecules • Non-human primates for biologicals • Repeat dose toxicology required for anticipated duration of clinical use for most non-oncology agents • 3 mo. toxicology for ≤ 3 mo. clinical study • Recommendations for agents used in the treatment of advanced cancer differs

  42. Expected Toxicology Testing for Phase I Oncology Drug Studies(J. Leighton, FDA ODAC Meeting, March 13, 2006) * Study schedule does not include a a recovery period -- 28 day toxicology is generally sufficient for DRUG trials extending beyond 28 days

  43. Non-Clinical Toxicology Studies For Oncology Drug Combinations • May not be necessary for testing in advanced cancer patients • May exclude if: • No PK, PD, or metabolic interactions anticipated • Drugs are not packaged as a combination • All components well studied individually

  44. Single Dose Toxicity Studies • Dose escalation study may be an alternative to a single dose design • Dose range should include maximally tolerated dose (MTD) and no adverse effect level (NOAEL) • Standard design • Early sacrifice at 24 to 48 hr and after 14 days

  45. Repeated Dose Toxicity Studies • Duration of repeated dose studies related to duration of anticipated clinical use • Use same schedule and duration • Typically 14-28 days • Should include recovery group • Use can support repeat dose clinical studies

  46. Non-Clinical Toxicology Ongoing Endpoints • Ongoing • Clinical signs, behavior • Body weights and food consumption • Clinical pathology (in larger species) • Hematology • Chemistry panels • Toxicokinetics • End of Study • Macroscopic changes at necropsy • Organ weights • Histopathology of all organs

  47. Other Toxicology Studies • Local tolerance studies • If warranted by route of administration • Genotoxicity studies • Reproductive Toxicity studies • Carcinogenicity studies

  48. Genotoxicity studies • General • Normally done prior to FIH studies, but not required prior to phase I studies in oncology patients • Standard battery of genotoxicity tests required prior to initiation of phase II • Specific genotoxicity studies • In vitro bacterial reverse mutation assays: Ames test, point mutation test • In vitro chromosome damage tests in mammalian cells: metaphase cell analysis, murine lymphoma gene mutation assays • In vivo chromosomal damage assays: rodent micronucleus tests

  49. Reproductive Toxicity Studies • Men • May include in Phase I/II after relevant repeated dose toxicity studies • Male fertility study should be completed prior to initiation of Phase III • Women not of childbearing potential • May include in clinical trials after relevant repeated dose toxicity studies • Women of childbearing potential • May include in carefully monitored early studies with precautions • Fertility and embryo-fetal toxicity studies should be completed prior to entry of women into phase III trials • Pregnant women • All reproductive toxicity and genotoxicity studies must be completed prior to entry of these women in trials

More Related