Therapeutic Bench to Beside: Art & Science Of Drug Discovery and Development (and everyone’s role in it) Theodore F. Reiss, M.D. 1 Nancy Whorf 2 Message/Objectives • Therapeutic innovation : impactful, practical, real world application – Integrated, complex system, non-linear thinking, iterative, learning, collaborative, team centered. • An overview approach – Model – Some specifics – Challenges and opportunities for the future 3 The 3T’s Dougherty, D. et al. JAMA 2008;299:2319-2321 The Innovation Interface • Basic/clinical science • Technology focus • Public health • Policy/ Culture Copyright restrictions may apply. “Language” of Biomedical Innovation •Certainty of Benefit/Risk --vs•Partially justified new knowledge “Bench To Bedside” : Interdependence • Centered on collaborative, synergistic scientific efforts • Resource intensive, necessitating efficiency Innovation or Stagnation: FDA March 2004 [----------------------”Bench to Bedside”------------------] 5 Traditional Concept: Novel Mechanism Discovery Pre-clinical Toxicology • In vitro • Understand pathobiology • Target ID/validation • In vivo • Molecule ID • Initial regulatory interactions • Start formulation development Phase II • POC • Dose selection • Safety in patients • Initial benefit/risk profile • Formulation completed • Finalize alignment with regulatory agencies Phase III Phase I • SD/MD safety • PK/PD characterization • Target engaged • Biologic effects Phase IV/V • Post marketing requirements • Efficacy confirmation • Post marketing safety • Outcomes surveillance • Safety confirmation • Detailed benefit/risk profile • New indications • Health economic data for reimbursement authorization 6 Strategic Development Metaphor: Financial Planning • Define the goal • Identify the component parts • Develop a plan working backwards from the goal • Plan depends on many factors: – Define level of benefit/risk – Determine interim steps that have to be achieved – Flexibility to adjust to environmental changes 7 “Bench to Bedside”: Principles • Neither simple nor linear • Begin with the goal: Unmet medical needs-public health value – Data: Clinical, regulatory, and health economic • Demonstrate clear, population specific, benefit / risk – Efficient and timely as possible • Dynamic, responding to new knowledge 8 Principles: Optimize Potential for Success • Disease area focus – Multiple targets and/or molecules within a target • Strategic scientific development plan – Begin with goal and design backwards • Failure the norm – Go/no go criteria to exit early if risk/benefit unacceptable – Kill early • Critical importance of predictive safety & efficacy biomarkers – Patient identification – Response prediction • Apply learning iteratively 9 Drug Development Paradigm (Better!) Discovery • Toxicology Phase I Phase II Phase III Goal “System” Approach: – – – – – Neither simple, nor linear Each component is part of a “whole” strategy “Goal” driving earlier development steps: Iterative • Address unmet medical need • Demonstrate clear, population specific benefit/risk • Efficient and timely as possible Dynamic, responding to new knowledge Collaborative : Integrated Project Team - Many Experts “Thinking as One” 10 C O M M E R C I A L I Z A T I O N “Begin With The End (Goal) In Mind” First Principle – Clinical Vision • • • • • • • Patient population (or sub population) Efficacy “threshold” Tolerability profile Method of delivery Potency Dosing interval Drug Interaction Create public health value with optimum benefit /risk in a defined patient population Could refocus goal further during Phase I-III For example: optimal responder population identified 11 Efficiency Gain From A Thoughtful Scientific/Regulatory Strategy Unoptimized Strategy Discovery Preclinical Preclinical Toxicology Toxicology Phase Phase I I Phase II Phase II Phase III Phase III Time (Years) Regulatory Filing Patent life 12 Efficiency Gain From A Thoughtful End-to-End Strategy: Kill Early Discovery Preclinical Toxicology Phase I Phase II Phase III Time (Years) If don’t meet POC or ePOC Kill Regulatory Filing Patent life 13 Less Optimal Approaches • Therapeutic goal not clear • No planning: Stumbling forward one experiment to the next • Single molecule focused only Why? • Time/resource limitations • Design to goal – Solving unmet need with acceptable benefit/risk – Molecules are means to ends 14 Advanced Concepts • Development strategy optimizes for: – Fewest resources – Least time – Most information (scientific, clinical, regulatory, health economic) Milestones • Go/no go – Scientific probability of achieving “goal” – Investment decision points • Key regulatory agency interactions – Strategy aligned with regulatory agency perspective 15 Conceptual Model Linking Drug Development with Value PUBLIC HEALTH Unmet Medical Needs Cost vs. Effectiveness (or Utilities) EXTERNAL BIOLOGY DISCOVERY PH I PRECLINICAL TOXICOLOGY PH III GOAL TPP VALUE PH II INTERNAL NO GO NO GO NPV 16 Advanced Concepts: Target Discovery • What makes development programs efficient? • Why are some disease areas easer than others for drug discovery and development? • What are optimal characteristics of a “molecular target” for a therapy? 17 Pathobiology Of Disease: Clear Understanding Critical To Optimize Development • Foundation for basic-preclinical-clinical consistency • Facilitate safety and efficacy prediction – Provides potential for clearly defining “subpopulations” • If clear: – Target rich (osteoporosis) • If speculative: – Targets are high risk – Predicting efficacy and safety and defining “subpopulations” much more challenging 18 Osteoporosis: Available Knowledge Allows for “Efficient” Development • • • • Pathobiology of disease reasonably well understood Animal models available Animal models predictive of human disease Natural history of disease known – Large cohort studies – Clinically relevant endpoint defined and outcomes known – Establish and improving clinical disease biomarkers • Investigational therapies affect biology of disease “Efficient” system to develop therapies, investigate new tools to predict response, and identify sub-populations 19 Respiratory Development Is Challenging • Pathobiology is less well understood (relative to osteoporosis) • Disease definition is imprecise • Animal models are imprecisely predictive • Natural history, outcomes less are defined • Criteria defining clinical response difficult to determine 20 Example: Failure of Prediction DP1 Antagonist in Asthma/Allergic Rhinitis • Receptor for PGD2 • PGD2 released from mast cells with histamine/CysLT’s – Pharmacologic activity in airways of asthmatics • DP1 knock-out – blocks response in mouse OVA model • DP1 blockade inhibits inflammation in guinea pigs • Polymorphism in DP1 receptor associated with asthma • However: – No effect of receptor antagonist in asthma/allergic rhinitis 21 Target Identification and Validation • Address unmet medical needs? • Understand pathobiology/pathway regulation – Pathology, genetic pathways linked – Pharmacologic studies in disease models • Knockouts, transgenic, pathway interruption (siRNA, antibodies, small molecules) – Predict benefit/risk • Link to man for prediction (altered expression of target or biomarker) • Probability of activity different from the target –Safety – Biomarker development • Determine molecular approach and delivery optimization – Clone target /tractable • Design molecules – Iterative process 22 Pre-Clinical Biology Target Pre-clinical Discovery Toxicology • Generate and select optimal compound – Potency – PK in animals – “Probe” safety study – Determine if formulation possible • Unique challenges by type and route • Difficulty under appreciated • Alternate strategy: unoptimized molecule to POC in man ASAP – Other strategies 23 Pre-clinical Toxicology General Concepts • In vitro and in vivo studies to predict tolerability in man • Examples: In vitro toxicology – Carcinogenic potential / metabolic profile/ P450 studies • Examples: In vivo (at least two species) – Single dose, multiple dose: 1 week – 1 year, carcinogenicity studies – Dose or time related toxicities • Component of strategic development plan – Facilitate strategy: Phase I-III trials (length, sequence) – Must provide adequate dose exposure margin 24 Phase I – IIa: Critical Bridge - Iterative Optimization • Demonstrate: – PK, target engagement, biological activity, initial clinical benefit /risk – Go/no go • “Learning phase” – Clinical experimental models to optimize target molecules – “Hypothesis” generating trial(s) to: • Optimize subsequent clinical experiments • Identify and validate predictive markers or sub-populations • Time intensive, speed not primary concern – Biomarker(s) to optimize dose selection/prediction of benefit or risk • Target engagement (example: receptor occupancy: NK1) • Target engagement and biologic effect (example: urinary LTE4: 5LO) • Target engagement and biologic effect and clinical surrogate endpoint (example: reticulocytes - EPO) 25 Imaging as a Biomarker Target Engagement and Dose NK1 Antagonist Binding of PET tracer to NK1 receptors Blockade of NK1 receptors after aprepitant dosing Brain NK1 Receptor Occupancy (%) Mean (± SE) Plasma Trough Concentrations 40/25 125/80 375/125 100 90 80 70 60 50 40 30 20 10 0 0 1 10 100 1000 10000 Aprepitant Plasma Trough Concentration (ng/mL) Tracer Binding Low Hargreaves J Clin Psych 63: (suppl 11): 18-24, 2003 High 26 LTE4 % Predose (Mean ± SEM) Example: Biomarker 5-LO FLAP Inhibition Urinary LTE4 140 120 Placebo 25 mg 50 mg 125 mg 250 mg 500 mg 100 80 60 40 20 0 0-3 3-6 6-9 9-12 24 Hours Post dose 36 48 72 27 Dose Selection: The Phase II Activity • Identify dose-response relationship • Benefit/risk: must be determined in a defined population, through an adequate dose range - Must demonstrate minimal or no effect • Goals: Identify minimal dose achieving maximal response without evidence of dose limiting toxicity Response 100 Efficacy Safety 50 0 Dose 28 Example: Biomarker Leading to Dose Ranging CysLT1 Antagonist Clinical Pharmacology & Therapeutics 1997; 61:(1) 83-92 29 Phase III: Characteristics • Program design considerations – Sufficient to address clinical questions in targeted population (use in clinical practice) • Multiple or few trials? If worldwide, special considerations • One dose optimal – All measurements must have been previously “validated” and “qualified” according to stringent standards – Placebo vs. comparative designs – Tolerability: Pooling data – continued “signal” detection – Endpoint: outcome or surrogate? (CASS Study example) • Health economic data: public health / reimbursement issues • Cost - Effectiveness/ Cost - Utility 30 Regulatory Considerations • Consultations – “Buy-in” to Phase III plans before starting • Endpoints/validation/statistical plan • Data submissions – Worldwide submissions to regulatory agencies • Content: scientific/clinical rationale, individual trial data, “integrated summaries” (safety and efficacy) – Biggest grant proposal you ever submitted! (5-15 trials [4,000-20,000 patients]) – Draft label included – Recommendation for patient information/post marketing surveillance • Labeling – Scientific and risk/benefit data summarized – Negotiated separately with agencies worldwide – Draft label early in development: target product profile/promotion 31 Post Marketing Safety Surveillance • Major emphasis for the future: – Example: “Sentinel” initiative • Spontaneous reports – Different rigor among countries – Claims databases – Data very difficult to interpret • Causal relationships difficult to determine • New methods of “signal detection” • Pooled clinical trial database (including Phase IV) 32 Efficient, Timely Execution Of Development Programs: Project Team • Forms early: target discovery – Describe goal, develop plan, iteratively manage plan – Efficient, fast, scientifically excellent – Effectiveness: integration of scientific/regulatory information in a hypothesis driven, sequenced plan (No go decision points) • High performance team (synergism) • Individually experts in separate disciplines – Broad scientific/regulatory knowledge • • • • Members know all roles and responsibilities Anticipate others’ needs/thoughts Always think two steps ahead for self and other team members Leadership 33 Research & Discovery Process It takes ~10-15 years and $802 million to develop one new medicine1 PostMarketing FDA Review 1 2 5 Compounds 6 250 Compounds 1.5 Phase III n=1000-5000 Clinical Trials Phase II n=100-500 Phase I n=20-100 Preclinical Drug Discovery 5,000 – 10,000 Compounds 5 Years 1DiMasi JA, Hansen RW, Grabowski HG Journal of Health Economics2003, 22, 151-185 34 19 8 19 4 8 19 5 8 19 6 8 19 7 8 19 8 8 19 9 9 19 0 9 19 1 9 19 2 9 19 3 9 19 4 9 19 5 9 19 6 9 19 7 9 19 8 9 20 9 0 20 0 0 20 1 0 20 2 0 20 3 0 20 4 0 20 5 0 20 6 0 20 7 20 0 09 8 (E ) 50 #'s NCE's Approved 40 40 30 30 20 20 10 10 0 Source: PhRMA, FDA No. NCE's approved 60 No. NCE s Approved ’ R & D Spend- PhRMA Members (in $B) R&D Expenditure PhRMA Members (in $B) Despite Substantial Investment, New Products Infrequent NCE's approved and R&D Costs since 1984 R&D Spend (in $B) 60 50 0 35 What are Today’s Challenges? • Greater, efficient output – Increasingly complex system/environment • No consensus on efficient process improvement • Poor predictive ability to identify targets with adequate benefit/risk • Development knowledge/world view is siloed • Art of development, regulatory, translational science not broadly known • Collaborative efforts stifled • Investment capital disappearing Some Causes For Fewer New Therapies • Simple, “low hanging ” targets have been developed – Biology is complex – Multiple, difficult to dissect pathways • Accurate prediction of safety and efficacy based on the molecular target has proven elusive • Better benefit/risk required, difficult to predict – Unmet medical needs more narrowly defined - phenotypes – To demonstrate efficacy, “Clinical Outcome” frequently required • Rather than surrogate endpoint • Greater need for validation – Less tolerance of side effects • Common and infrequent • Need to demonstrate value to multiple stakeholders (payers) 37 Challenges In Discovery & Development • Greater efficacy and better safety necessitate: – Pathobiology of disease must be accurately understood (pick the right targets) – New tools for better safety and efficacy prediction • Must increase probability of success at each step – Larger trials required • Costs increasing rapidly, resources limited • Demonstrate cost/effectiveness benefit – Specific “sub-populations” to enhance benefit risk • Population identification • Response prediction 38 How Should “Bench to Bedside” Evolve And Who Will Lead The Way? • Bayh-Dole Act – Allowed University to patent research discoveries – Potential to create “wealth” for the universities (institutes) – Have had a effect on the conceptual models of the future of translational science 39 Path Forward: Improving “Bench to Bedside” – Position Statements • National Institute of Medicine – Clinical Research Round Table • NIH – “Road map for medical research” • National Academy of Sciences – “Exploring strategies for future research” • FDA – Critical Path Initiative 40 Key Elements of Position Statements • Collaboration among Pharma, academic, government – Share knowledge (FDA, NIH & Pharma) to develop new science through consortiums – Extensive and complementary databases – Similar initiatives overseas • “Bench to Bedside” is complex, true progress will require integrated solutions • Need to teach team collaboration science 41 Conflict of Interest (COI) • COI issues are real – Focus on financial only – Tone: non – collaborative – Unintended consequences – effecting collaborative efforts • Debate needs broader view: goal - improve public health • Alternative position – – – – Include all types of conflicts Improve public health is a primary interest Checks and balances in place Trust can’t be developed unless balance achieved • Collaboration/ cooperation/ learning /teaching • Examples in the public good: Bell Labs / IMI 42 Where are We Going? • Vision? • Scientific “paradigm shift”? • Increasing realization that institutions can’t go it alone • Collaborative efforts in complex systems • Translational development science • Focus on unmet medical need/public health value • Sharing the rewards – Bayh/Dole • New commercial models Future Vision: Integration/Collaboration • Sci Transl Med 7 April 2010: Vol. 2, Issue 26, p. 26cm12 Models and Policy Choices • • • • • • • • Shared science Cross-institution development & business models Shared continuous improvement Broad as well as deep scientific knowledge More transparency More phenotype specific therapies Cost /effectiveness Broader, balanced ethics discussions Discovery & Development • Complex system, goal oriented, integrated – Optimally, create public health value & maximize benefit/ risk – Non-linear – Iterative/responsive to new information • Many moving parts: constant problem-solving/management • Optimally, development phases predicatively linked • Resource intensive: optimally efficient, disciplined; predictable biomarkers & surrogate outpatients to determine POS and Go/No Rules • Collaborative, experienced project team • Progress will require cross-institutional collaboration 46 Nancy Whorf 47 Concepts: Reviewed • Neither simple nor linear • Goal: Unmet medical needs – public health value – Data: Clinical, regulatory and health economic – Demonstrate clear, population specific benefit / risk – Efficient and timely as possible • Dynamic, responding to new knowledge • Disease area focus – Multiple targets and/or molecules within a target • Strategic scientific development plan – Begin with goal and design backwards • Failure the norm – Go/no go criteria to exit early if risk/benefit unacceptable – Kill early • Critical importance of predictive safety & efficacy biomarkers – Patient identification – Response prediction • Apply learning iteratively 48 Formulation Research and Development • Is it feasible to synthesize the compound and develop a formulation? • Difficulty under appreciated • Unique challenges: By type: “small molecule”, vaccine, protein antibody, siRNA, gene By route: Oral, inhaled, IV, topical, etc. 49 Phase I-II: General Concepts • Phase I: – Tolerability/pharmacokinetics/efficacy • Tolerability issues affect benefit /risk or limit dose determination - Possible No Go – Biomarker(s) to optimize dose selection/prediction of benefit or risk • Target engagement (example: receptor occupancy: NK1) • Target engagement and biologic effect (example: urinary LTE4: 5LO) • Target engagement and biologic effect and clinical surrogate endpoint (example: reticulocytes - EPO) • Phase II – Significant strategy choices: selection of surrogate biomarkers and endpoints – Help in bracketing doses for final clinical dose ranging study – Predictive of ultimate clinical endpoint (outcome) – Help to identify responder populations and biomarkers – Experimental models in humans 50 Project Teams: Additional Principles • High performance team – In order to facilitate collaboration/synergism • Members know all roles and responsibilities • Anticipate others’ needs/thoughts • Always think two steps ahead for self and other team members – Listen to others’ points of view – Debate and dialogue respected and valued 51 Future Approaches: Systems Biology Nature Genetics 2005: 37; 710-17 52 Dose Ranging: M3 Antagonist Example European Respiratory Journal 2006; 28: p772-780 53 Phase I – IIa: Critical Bridge Pharmacodynamics/Proof Of Concept • Demonstrate: – • Identify benefit/risk – • Go/no go Understanding safety profile important – • PK, target engagement, biological activity, initial clinical benefit Strategy about commitment illness, medicines Significant strategy choices: selection of surrogate biomarkers and endpoints – – – – Help in bracketing doses for final clinical dose ranging study Predictive of ultimate clinical endpoint (outcome) Help to identify responder populations and biomarkers Experimental models in humans 54 Major Goals: Phase III and Beyond • In population(s) of interest – Document tolerability in more patients for longer time – Confirm benefit/risk – Potentially outcome / health economic data • Plan for post approval safety monitoring • Plan for and execute new indication(s) 55 Dose Selection: Phase II Activity • Necessary inputs to formal dose ranging study(ies) – Tolerability response curve or known limitations (Phase I/IIa) – Pharmacokinetics, biomarker of target engagement or activity – Dose spacing • Strategic options to obtain data – Standard dose ranging study/dose adaptive design/modeling • Other strategic options – Combine dose ranging and POC – Dose ranging on surrogate endpoint or outcome 56 Other Phase III Considerations • High costs – Example: 15K patient outcome study: $400M over 4 years – Resources becoming limited • Recruitment of patients globally – Different approaches to medicine • What is the basis for a Phase III Program “go” – Strategic options • Importance of achieving profile • Partnering • Funding 57 Example: Serendipity Anti-Leukotriene (CysLT1) Program • History: Nobel prize: classical physiology, pharmacology • Many attempts before final candidate “evolved” – Receptor Antagonists • 100 Compounds • 10 Animal Toxicology • 5 Compounds in Man • 5-LO Program (Both Direct and Indirect Inhibitors) – 50 Compounds – 5 Animal Toxicology – 2 Compounds in Man • Took too long/too many iterations • Efficient development program 58 Example: Dose Ranging - CysLT1 Antagonist European Respiratory Journal 1998 11(6) 1232-1239 59 Phase III: Characteristics (con’t) • Trial design consideration – Usually single dose from Phase IIb – Statistically rigorous: clear hypothesis, pre-established data analysis plan, no post hoc data dredging – “Clinically important” treatment effect targeted – Safety monitoring boards becoming standard – Non-inferiority designs – Real world designs/health economic data – Inclusion criteria (generalizability) – Exposure to as many patients and for as long as possible 60