drug design lecture 1

Upload: mudit-misra

Post on 07-Apr-2018

216 views

Category:

Documents


0 download

TRANSCRIPT

  • 8/6/2019 Drug Design Lecture 1

    1/35

    Modern Methods of Drug Discovery

    Lecture 1: An Introduction

  • 8/6/2019 Drug Design Lecture 1

    2/35

    The Top 10 Pharmaceutical Companies

    19.4Bristol-Myers Squibb

    19.7Abbott Labs

    20.4Aventis-Sanofi

    21.4AstraZeneca

    22.9Merck

    24.5Hoffman LaRoche

    28.2Novartis

    39.0GlaxoSmithKline

    45.2Pfizer

    47.4Johnson & Johnson

    2004 sales figures(billions)

    15.3Bristol-Myers Squibb

    16.6Hoffman LaRoche

    20.3Novartis

    24.0AstraZeneca

    34.0Aventis-Sanofi

    22.3Johnson & Johnson

    19.7Abbott Labs

    21.9Merck

    34.0GlaxoSmithKline

    44.2Pfizer

    2005 sales figures(billions)

    14.8Eli Lilly

    26.6Hoffman LaRoche

    28.9Novartis

    25.7AstraZeneca

    35.6Aventis-Sanofi

    23.3Johnson & Johnson

    15.7Wyeth

    22.6Merck

    37.0GlaxoSmithKline

    45.1Pfizer

    2006 sales figures(billions)

  • 8/6/2019 Drug Design Lecture 1

    3/35

    Research based pharmaceutical companies, on average, spend

    about 20% of their sales on research and development (R&D).

    This percentage is significantly higher than in most other

    industries, including electronics, aerospace, automobiles, and

    computers.

    Since 1980 US pharmaceutical companies have practically doubled

    spending on R&D every 5 yrs.

  • 8/6/2019 Drug Design Lecture 1

    4/35

    Despite these enormous expenditures, there has been a steady

    decline in the number of drugs introduced each year into human

    therapy.

    70-100 in the 60s

    60-70 in the 70s~50 in the 80s

    ~40 in the 90s

    Innovation Deficit - coined in 1996 by Jurgen Drews, president

    of research at Hoffmann-LaRoche.

    - the gap between the number of new chemicalentities (NCEs) required to be launched in

    order to accomplish an annual 10% revenue

    increase and the actual number of NCEsintroduced in the market by the top 10

    pharmaceutical companies.

  • 8/6/2019 Drug Design Lecture 1

    5/35

    Reasons for the innovation deficit:

    a) increased demand on safety for drugs.

    - the average number of clinical trials per new drug application

    (NDA) increased from 30 in the 70s to 40 in the 80s, to 70 in the

    90s.- the increased demand on safety is also reflected in a prolonged

    duration of the drug development process.

    In the 60s, total development time was 8.1 yrs

    In the 70s, total development time was 11.8 yrs

    In the 80s, total development time was 14.2 yrs

    In the 90s, total development time was 14.9 yrsCurrently, total development time is ~16 yrs

    b) low hanging fruit have been picked.

  • 8/6/2019 Drug Design Lecture 1

    6/35

    A new drug today costs ~$880 million and takes ~15-16 yrs to

    develop.

    Allocation of R&D funds

    Allocation of R&D time

  • 8/6/2019 Drug Design Lecture 1

    7/35

    About 75% of this cost ($660 million) is attributable to failure

    during development.

    90% of all drug development candidates fail to make it to market.

    Methods that enhance the drug discovery process and reduce

    failure rates are highly desirable!

  • 8/6/2019 Drug Design Lecture 1

    8/35

    This course (Modern Methods of Drug Discovery) will focus on

    some of the key methods used in the first 4 steps.

    The Drug Discovery Process

    Target

    Validation

    Drug

    Target

    Identification

    Lead

    Compound

    Identification

    Lead

    Optimization

    Preclinical and

    Clinical

    Development

  • 8/6/2019 Drug Design Lecture 1

    9/35

    1) Drug Target Identification

    The identification of new, clinically relevant, molecular targets

    is of utmost importance to the discovery of innovative drugs.

    It has been estimated that up to 10 genes contribute to

    multifactoral diseases.

    Science 287:1960-1964 (2000)

    Typically these disease genes are linked to another 5 to 10

    gene products in physiological circuits which are also suitable

    for pharmaceutical intervention.

    If these numbers are multiplied with the number of diseases

    that pose a major medical problem in the industrial world,

    then there are ~5000 to 10000 potential drug targets.

  • 8/6/2019 Drug Design Lecture 1

    10/35

    Current therapy is based upon less than 500 molecular targets

    45% of which are G-protein coupled receptors

    28% are enzymes11% are hormones and factors

    5% ion channels

    2% nuclear receptors

    Therefore, many more drug targets exist! How to identify them?

    Besides classical methods of cellular and molecular biology, newtechniques of target identification are becoming increasingly

    important. These include:

    a) genomics (Biotechniques 31: 626-630 2001)

    b) bioinformatics (Drug Discovery Today 7:315-323 2002)

    c) proteomics (J. Pharmacol. Toxicol. Methods 44:291-300

    2000; Biopolymers 60:206-211 2001)

  • 8/6/2019 Drug Design Lecture 1

    11/35

    a) genomics

    term was coined in the mid 80s.

    evolved from 2 independent advances:

    1) Automation resulting in a significant increase in the

    number of experiments that could be constructed

    in a given time. (eg. DNA sequencing)

    $22.5 MillionUS $300,000 each

  • 8/6/2019 Drug Design Lecture 1

    12/35

    2) Informatics- the ability to transform raw data into meaningful

    information by applying computerized techniques for managing,

    analyzing, and interpreting data.

    the identification of new biological targets has benefited from the

    genomics approach:

    eg. The sequencing of the human genome

    Nature 409:860-921 2001;

    Science 291:1304-1351 2001 blueprint of all proteins

    bioinformatics methods are used to transform the raw

    sequence into meaningful information (eg. genes and theirencoded proteins) and to compare whole genomes (disease vs.

    not).

    Drug Discovery Today 5:135-143 2000

  • 8/6/2019 Drug Design Lecture 1

    13/35

    Sequencing of microbial genomes will enable the identification of

    novel drug targets, especially when comparing to the human genome

    (eg MEP pathway).

    b) Bioinformatics the in silico identification of novel drug targets

    is now feasible by systematically searching for paralogs (related

    proteins within an organism) of known drug targets (eg. may be

    able to modify an existing drug to bind to the paralog).

    Can compare the entire genome of pathogenic and non-

    pathogenic strains of a microbe and identify genes/proteinsassociated with pathogenism.

    Current Opin. Microbiol 1:572-579 1998

  • 8/6/2019 Drug Design Lecture 1

    14/35

    Using gene expression microarrays and gene chip technologies,

    a single device can be used to evaluate and compare the expression

    of up to 20000 genes of healthy and diseased individuals at once.Trends Biotechnol 19:412-415 2001

    c) Proteomics concerning expression analysis, it has been shown

    that the correlation between RNA and protein expression is weak

    and ranges in yeast from 10-40% for lower abundance proteins to

    up to 94% for higher abundance proteins.

    Molecular Cell Biol 19:1720-1730 1999

    It is becoming increasingly evident that the complexity of biological

    systems lies at the level of the proteins, and that genomics alone will

    not suffice to understand these systems.

    It is also at the protein level that disease processes become

    manifest, and at which most (91%) drugs act.

  • 8/6/2019 Drug Design Lecture 1

    15/35

    Therefore, the analysis of proteins (including protein-protein,

    protein-nucleic acid, and protein-ligand interactions) will beutmost importance to target discovery.

    Proteomics is the systematic high-throughput separation and

    characterization of proteins within biological systems.

    Target identification with proteomics is performed by comparing

    the protein expression levels in normal and diseased tissues.

    2D PAGE is used to separate the proteins, which are subsequently

    identified and fully characterized with LC-MS/MS.

  • 8/6/2019 Drug Design Lecture 1

    16/35

    Target

    Validation

    Drug

    Target

    Identification

    Lead

    Compound

    Identification

    Lead

    Optimization

    Preclinical and

    Clinical

    Development

    The Drug Discovery Process

    2) Target Validation

    Involves demonstrating the relevance of the target protein in a

    disease process/pathogenicity and ideally requires both gain and

    loss of function studies.

    This is accomplished primarily with knock-out or knock-in animal

    models, small molecule inhibitors/agonists/antagonists, antisense

    nucleic acid constructs, ribozymes, and neutralizing antibodies.

  • 8/6/2019 Drug Design Lecture 1

    17/35

    Since strong interactions between a protein and its ligand are

    characterized by a high degree of complementarity, knowledge ofthe protein three dimensional structure will enable the prediction of

    druggability of the protein.

  • 8/6/2019 Drug Design Lecture 1

    18/35

    Target

    Validation

    Drug

    Target

    Identification

    Lead

    Compound

    Identification

    Lead

    Optimization

    Preclinical and

    Clinical

    Development

    The Drug Discovery Process

    3) Lead Compound Identification

    Compounds are identified which interact with the target protein

    and modulate its activity.

    Compounds are mainly identified using random (screening) or

    rational (design) approaches.

  • 8/6/2019 Drug Design Lecture 1

    19/35

    A) High-throughput Screening

    Used to test large numbers of compounds for their ability to affect

    the activity of target proteins.

    Natural product and synthetic compound libraries with millions

    of compounds are screened using a test assay.Curr Opin Chem Biol 4:445-451 2000

    There are concerns with the numbers approach to screening for

    a lead molecule. In theory generating the entire chemical space

    for drug molecules and testing them would be an elegant approach

    to drug discovery.

    In practice, this isnt feasible.

    Drug Discovery Today 5:2-4 2000

  • 8/6/2019 Drug Design Lecture 1

    20/35

    Therefore, concepts are needed to synthesize and select biologically

    relevant compounds.

    One solution may be to accumulate as much knowledge as possible

    on biological targets (eg. structure, function, interactions, ligands)

    and choose targeted approaches to chemical synthesis.

    Another crucial point for reliable high-throughput screening

    results is the robustness and quality of the biological test assays.

  • 8/6/2019 Drug Design Lecture 1

    21/35

    B) Structure Based Drug Design

    Three dimensional structures of compounds from virtual or

    physically existing libraries are docked into binding sites oftarget proteins with known or predicted structure.

    Scoring functions evaluate the steric and electrostatic

    complementarity between compounds and the target protein.

    The highest ranked compounds are then suggested for

    biological testing.

    Drug Discovery Today 7:64-70 2002

  • 8/6/2019 Drug Design Lecture 1

    22/35

    Once hits (compounds that elicit a positive response in an assay)

    have been identified via the screening approach, these are validated

    by re-testing them and checking the purity and structure of thecompounds.

    Only if the hits fulfill certain criteria are they regarded as leads.

    The criteria can originate from:

    1) Pharmacodynamic properties - efficacy, potency, selectivity

    2) Physiochemical properties - water solubility, chemicalstability, Lipinskis rule-of-five.

    3) Pharmacokinetic properties - metabolic stability and

    toxological aspects.4) Chemical optimization potential - ease of chemical synthesis

    and derivatization.

    5) Patentability

  • 8/6/2019 Drug Design Lecture 1

    23/35

    Target

    Validation

    Drug

    Target

    Identification

    Lead

    Compound

    Identification

    Lead

    Optimization

    Preclinical and

    Clinical

    Development

    The Drug Discovery Process

    4) Lead Optimization

    Molecules are chemically modified and subsequently characterized

    in order to obtain compounds with suitable properties to become a

    drug.

    Leads are characterized with respect to pharmacodynamic

    properties such as efficacy and potency in vitro and in vivo,

    physiochemical properties, pharmacokinetic properties, and

    toxicological aspects.

  • 8/6/2019 Drug Design Lecture 1

    24/35

    Efficacy vs PotencyPotency refers to the amount of drug required for its specific effect

    to occur; it is measured simply as the inverse of the EC50 for thatdrug.

    Efficacy measures the maximum strength of the effect itself, at

    saturating drug concentrations.

    Drug Red exceeds drug Black in potency, while the opposite istrue of the efficacy.

  • 8/6/2019 Drug Design Lecture 1

    25/35

    Pharmacokinetics - determining the fate of xenobiotics.- what the body does to the drug.

    Often divided into areas examining the extent and rate

    of adsorption, distribution, metabolism, and excretion(ADME).

    Pharmacodynamics - determining the biochemical and physiological

    effects of drugs, the mechanism of drug action,

    and the relationship between drug

    concentration and effect.

    - what the drug does to the body

  • 8/6/2019 Drug Design Lecture 1

    26/35

    This is often the tightest bottleneck in drug discovery.

    This process ideally requires the simultaneous optimization ofmultiple parameters and is thus a time consuming and costly step.

    Hints on how to modify a lead compound can originate from

    molecular modeling, quantitative structure-activity relationships,and from structural biology (structure-based drug design).

  • 8/6/2019 Drug Design Lecture 1

    27/35

    In parallel to compound characterization with respect to potencyand selectivity, in vitro assays for the prediction of pharmacokinetic

    properties should be performed.

    Curr Drug Metab 2:299-314 2001

    Once compounds with desirable in vitro profiles have been

    identified, these are characterized using in vivo models.

    Compounds that do not fulfill the requirements for a successful

    drug development candidate have to be optimized through the

    synthesis of better suited derivatives.

  • 8/6/2019 Drug Design Lecture 1

    28/35

    It is vital to conceive lead optimization as a simultaneous

    multidimensional process rather than a sequential one. Optimizingleads first with respect to pharmacodynamic properties (potency,

    selectivity, etc) and looking at pharmacokinetic parameters of these

    optimized compounds later guarantees frustration in the lateoptimization phase.

  • 8/6/2019 Drug Design Lecture 1

    29/35

    Target

    Validation

    Drug

    Target

    Identification

    Lead

    Compound

    Identification

    Lead

    Optimization

    Preclinical and

    Clinical

    Development

    The Drug Discovery Process

    Preclinical studies involve in vitro studies and trials on animal

    populations.

    Wide ranging dosages of the compounds are introduced to the

    cell line or animal in order to obtain preliminary efficacy and

    pharmacokinetic information.

  • 8/6/2019 Drug Design Lecture 1

    30/35

    The NIH organizes clinical trials into 5 different types:

    1) Treatment trials: test experimental treatments or a new

    combination of drugs.

    2) Prevention trials: look for ways to prevent a disease or prevent

    it from returning.

    3) Diagnostic trials: find better tests or procedures for diagnosinga disease.

    4) Screening trials: test methods of detecting diseases.

    5) Quality of Life trials: explore ways to improve comfort and

    quality of life for individuals with a chronic illness.

    Cli i l T i l

  • 8/6/2019 Drug Design Lecture 1

    31/35

    Clinical Trials

    Phase 0 - a recent designation for exploratory, first-in-human trials.

    Designed to expedite the development of promising

    therapeutic agents by establishing early on whether the agent

    behaves in human subjects as was anticipated from

    preclinical studies.

    Pharmaceutical clinical trials are commonly classified into 4 phases:

    (as of 2006, there are now 5)

    Phase I - a small group of healthy volunteers (20-80) are selected

    to assess the safety, tolerability, pharmacokinetics, and

    pharmacodynamics of a therapy.

    - normally include dose ranging studies so that doses forclinical use can be set/adjusted.

    New Scientist, March 2006,

    Catastrophic immune response may have caused

    drug trial horror

  • 8/6/2019 Drug Design Lecture 1

    32/35

    Phase I - there are 3 common kinds of phase I trials:

    1) Single Ascending Dose (SAD) studies- a small group

    of patients are given a single dose of the drug and thenare monitored over a period of time. If they do not exhibit

    any adverse side effects, the dose is escalated and a new

    group of patients is given the higher dose.

    2) Multiple Ascending Dose (MAD) studies- a group of

    patients receives multiple low doses of the drug, while

    blood (and other fluids) are collected at various time

    points and analyzed to understand how the drug is

    processed within the body. The dose is subsequently

    escalated for further groups.

    3) Food effect- designed to investigate any differences in

    absorption caused by eating before the dose is given.

  • 8/6/2019 Drug Design Lecture 1

    33/35

    80% of drugs fail the Phase I clinical trial.

  • 8/6/2019 Drug Design Lecture 1

    34/35

    Phase II - performed on larger groups (20-300) and are designed

    to assess the activity of the therapy, and continue Phase I

    safety assessments.

    Phase III - randomized controlled trials on large patient groups

    (hundreds to thousands) aimed at being the definitiveassessment of the efficacy of the new therapy, in comparison

    with standard therapy. Side effects are also monitored.

    -it is typically expected that there be at least two successfulphase III clinical trials to obtain approval from the FDA.

    Once a drug has proven acceptable, the trial results are combined

    into a large document which includes a comprehensive description

    of manufacturing procedures, formulation details, shelf life, etc.

    This document is submitted to the FDA for review.

  • 8/6/2019 Drug Design Lecture 1

    35/35

    Phase IV - post-launch safety monitoring and ongoing technical

    support of a drug.

    - may be mandated or initiated by the pharmaceuticalcompany.

    - designed to detect rare or long term adverse effects over

    a large patient population and timescale than was possibleduring clinical trials.

    USA Today 10/12/2004 How did Vioxx debacle happen?