1 / 21

Phase I studies in non-oncology: Dosing & Designs

Phase I studies in non-oncology: Dosing & Designs. April 18 th 2012 Sharon O ’ Byrne Senior Medical Director Genentech Research & Early Development. Overview of drug development process. Pharmacokinetics (PK) & Pharmacodynamics (PD). PK: What the body does to the drug (exposure-time).

elmer
Download Presentation

Phase I studies in non-oncology: Dosing & Designs

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. Phase I studies in non-oncology:Dosing & Designs April 18th 2012 Sharon O’Byrne Senior Medical Director Genentech Research & Early Development

  2. Overview of drug development process

  3. Pharmacokinetics (PK) & Pharmacodynamics (PD) • PK: What the body does to the drug (exposure-time). • PD: What the drug does to the body (response-time) • PKPD: PD response as a function of drug concentration and time (E-R) Site of action PK Clinical efficacy PD Blood MTC PKPD-Anti-CD11a vs CD11a expression on T-cells TW AUC Tmax

  4. Absorption Distribution Metabolism Excretion (ADME) Differences between monoclonal antibodies & small molecules • IV, SC, or IM only, no oral • Distribution limited to blood and interstitial spaces • Large CL capacity • Non-target mediated clearance • Tissue uptake endocytosis/pinocytosis • Lysosomal degradation if not recycled by FcRn (all cells) • Target mediated clearance • Endocytosis of receptor/antibody complex • Clearance of soluble ligand/antibody complex • Slow CL and long half life (days to weeks) • Low dose nonlinear, high dose linear • No excretion of intact mAb • Oral administration preferred • Typically all tissues accessible • Smaller CL capacity • Metabolism (liver; kidney) • Oxidation, conjugation, others • Active parent drug  active metabolite • Parent drug may be safe, metabolite toxic • Fast CL and short half life (hours) • Low dose linear and high dose nonlinear • Biliary and renal excretion (secretion) • Active or passive transporter

  5. Objectives of a First-in-Human study • To determine the acute safety profile of a study drug in man • Opportunity to detect a large safety signal • To characterize the pharmacokinetic profile of the study drug • Half-life helps to determine future dosing interval • To determine any pharmacodynamiceffects • Is the study drug acting as expected in humans ? • Helps to determine dose levels • Is there an opportunity to quantify clinical activity ? • Disease Activity Score in Rheumatoid Arthritis • Low Density Lipoprotein in Cardiovascular Disease • Glycosylated Hemoglobin levels in Diabetes Mellitus • Mayo Clinic Score in Ulcerative Colitis

  6. First-in-Human studiesGeneral considerations • Assessment of Risk • Related to mechanism of action • Totally novel • Target unstable (TGN1412 – anti-CD28)1 • Concern regarding the class of agent under study • May or may not apply to all therapeutics in the class • Related to target patient population • Acute coronary syndrome • Allergy • Pregnancy • Related to how well pre-clinical toxicology studies (in vitro & in vivo) predict toxicity in humans • Healthy Volunteer population vs Patient population • Focus on safety • (small n) • Conduct some early signal seeking for proof of concept • (expanded n in some cohorts) 1Stebbings et al Curr Opin Biotech 2009, 20:673-677

  7. First-in-HumanCalculation of maximum recommended starting dose (MRSD) • Starting dose based on • Pre-clinical safety studies • Large molecules – commonly Cynomologous monkeys • Small molecules – commonly Rodent, Dog • In vitro safety testing in a human cell-based system • Calculation of dose based on • No observed adverse effect level (NOAEL) • Highest dose in pre-clinical studies at which no biologically relevant safety events have occurred • Minimum anticipated biological effect level (MABEL) • Dose at the lowest end of the dose-response curve for biological effect • Estimated from all the in vivo and in vitro data, or what is available • EMEA now recommend use of MABEL for calculation of starting dose for all biologics* Use most appropriate species *http://www.dh.gov.uk/en/Publicationsandstatistics/Publications/PublicationsPolicyAndGuidance/DH_063117 *http://www.emea.europa.eu/pdfs/human/swp/2836707enfin.pdf

  8. First-in-HumanCalculation of maximum recommended starting dose (MRSD) using NOAEL • Calculate human equivalent dose (HED) of NOAEL from most appropriate species • Human equivalent dose (HED) is calculated using appropriate scaling factors • Based on body surface area (division factor of 3 for cynomolgus monkeys) • Dose of 1.0 mg/kg in monkeys ≅ 0.3 mg/kg in humans • Safety factors should be applied to the HED to ensure a safe starting dose • Safety factor applied depending on risk associated with target • In general safety factor of • At least 10 x should be applied for large molecules • At least 50 x should be applied for small molecules • Recommended starting dose • HED ÷ safety factor = maximum recommended starting dose (MRSD) • Safety factors may also be expressed in terms of • Exposure (AUC) or Concentration (Cmax) • The AUC or Cmax at the NOAEL • Using model simulation to determine dose that will provide human equivalent AUC or Cmax

  9. First-in-Human studiesEscalation of dose (amount of increase) • Require sufficient dose separation between doses to determine difference, however, not too large an increase • 2 x increase may be too little • 10 x increase may be too much • Initial dose escalation should be conservative based on knowledge of target • Molecule specific • Commonly used rule is 3 x increase to ensure safe separation of dose levels • Semilog increment • Depending on safety profile the degree of increment can be adjusted • < semilog increment

  10. Decision making in clinical trialsTerminology used • Adverse Event • An AE is any unfavorable and unintended sign, symptom, or disease temporally associated with the use of an investigational medicinal product (IMP; study drug) or other protocol‑imposed intervention, regardless of attribution • Serious Adverse Event – any AE that is any of the following: • Fatal (i.e., the AE actually causes or leads to death) • Life threatening (i.e., the AE, in the view of the investigator, places the subject at immediate risk of death) • Requires or prolongs inpatient hospitalization • Results in persistent or significant disability/incapacity (i.e., the AE results in substantial disruption of the subject’s ability to conduct normal life functions) • A congenital anomaly/birth defect in a neonate/infant born to a mother exposed to the investigational product(s) • Considered a significant medical event by the investigator (e.g., may jeopardize the subject or may require medical/surgical intervention to prevent one of the outcomes listed above) • Serious vs Severe • Terms not synonymous • Severity refers to intensity of AE – maybe relatively minor but severe e.g. backache • Seriousness is a Regulatory Authority definition • Refers to seriousness of threat to subject’s life or vital functions

  11. Decision making in clinical trialsClassification of severity of adverse event Adverse Event grade of severity • National Cancer Institute Common Terminology Criteria for Adverse Events (NCI CTCAE v.4)1 • Used in non-oncology for patients with moderate to severe disease • Mild, moderate to severe classification used (see examples in appendix) • In patients with less severe diseases • Healthy volunteers 1http://ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.htm#ctc_40

  12. First-in-Human studiesDecision making • Dose escalation rules • Define dose limiting toxicities (DLTs) • Adverse events that are pre-determined and defined according to grade of severity • Any study drug related Grade ≥ 3 adverse events occurring within 14 days of study drug administration • Any study drug related Grade ≥ 2 adverse events occurring within 14 days of study drug administration that are considered clinically significant as judged by the Investigator or Internal Safety Monitoring Committee • Specific related to target under study • Risk of neutropenia • Risk of infection • Define the number of DLTs at which dose escalation will stop • Study will cease completely • There will be further evaluation of safety and study will continue • Examples – 1-2 DLTs per cohort • Example of dose escalation rules • If 1 patient experiences a DLT, 5 additional patients will be randomized to the dose level at which the DLT was experienced • Of these 5 additional patients, 4 will receive active drug and 1 will receive placebo in a double‑blinded fashion. If the incidence of DLTs in the entire expanded cohort remains at 1 patient (1 of 8 patients treated with study drug), escalation will proceed to the next dose level • If 2 or more patients experience a DLT, enrollment will be stopped for evaluation of all available safety information

  13. Dose Limiting ToxicitiesProbability of detection • The sample size is based on the dose escalation rules • The number of subjects in each cohort should be sufficient to characterize the single‑dose safety and tolerability

  14. First-in-HumanSafety monitoring • Recommend dosing of first subjects at least 24 hours apart • First dose cohort • First couple of subjects in each increasing dose cohort • Starting dose route of administration • Intravenous infusion over 1-2 hours for totally novel target, or where there are concerns for safety • Considered safer as infusion can be turned off if AE occurs during infusion • Subcutaneous route can be used if the target is not novel http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/ucm078932.pdf

  15. First-in-HumanSafety monitoring • Timing of dose escalation (SAD/MAD) • 14 days generally considered acceptable length of time to determine acute toxicity • Single Ascending Dose stage – 14 days after single dose • Multiple Ascending Dose stage – 14 days after second or third dose • Depending on likely toxicity of study drug, this time interval can be adjusted • Duration of follow-up of patients for adverse event monitoring • ~ 5 half-lives for clearance of therapeutic under study • Sponsor’s internal safety monitoring committee • Unblinded to treatment assignment to quickly review any SAEs etc • Treating physician and patient • Blinded to eliminate bias • Serious adverse events, adverse events Grade ≥ 2/3 and all events of special interest • Reported to the Sponsor within 24 hours

  16. First-in-Human Study Design Example 1 6:1 Cohort H: 300 mg FD SC (4.0 mg/kg) 6:1 Cohort E: 450 mg (6.0 mg/kg)SC Cohort C: 50 mg (0.67 mg/kg)SC Cohort G:100 mg FD SC (1.3 mg/kg) d14 d14 4:1 4:1 Active:Placebo 6:1 Cohort D: 150 mg (2.0 mg/kg)SC Cohort F: 30 mg FD SC (0.4 mg/kg) 4:1 d14 d14 Cohort B: 15 mg (0.2 mg/kg)SC 4:1 d14 d14 Cohort A: 5 mg (0.07 mg/kg) SC 2:1 Dosing Randomized, Double Blind within Cohort, Placebo-controlled, Dose Escalating Study Await full assessment of safety from SAD Phase Ia – SAD Phase Ib – MAD Main objective of Phase I is to seek an early safety signal Assuming body weight of 75 Kg

  17. First-in-Human Study DesignExample 2 Cohort E: 450 mg (6.0 mg/kg)SC Cohort C: 50 mg (0.67 mg/kg)SC 4:1 4:1 6:1 Cohort D: 150 mg (2.0 mg/kg)SC 4:1 d14 d14 Cohort B: 15 mg (0.2 mg/kg)SC 4:1 d14 d14 Cohort A: 5 mg (0.07 mg/kg) SC 2:1 Dosing Randomized, Double Blind within Cohort, Placebo-controlled, Dose Escalating Study Phase Ia – SAD Phase Ib – MAD Cohort H: 300 mg FD SC (4.0 mg/kg) - Real time safety reporting - Cohort E & F start at the same time 6:1 Active: Placebo Cohort G:100 mg FD SC (1.3 mg/kg) D14 from 2nd dose D14 from 2nd dose 6:1 Cohort F: 30 mg FD SC (0.4 mg/kg) Wk 0 Wk 2 Wk 4 • All patients dosed in Cohort A dosed on a separate day • First 2 patients in each subsequent cohort in SAD dosed on separate days Assuming average body weight of 75 Kg

  18. First-in-Human Study DesignExample 3 Cohort F: 3.0 SC 4:1 d14 d14 d14 = dosing; IV = intravenous; SC = subcutaneous Randomized, Double Blind within Cohort, Placebo-controlled, Dose Escalating Study Multiple dose stage (MD; Phase Ib) Dosing q4wks: Cohort J: 4.0 IV Single ascending dose stage (SAD; Phase Ia) 4:1 Dose mg/kg Cohort D: 10.0 IV 4:1 Cohort I: 3.0 SC 4:1 Cohort C: 3.0 IV Analysis of PK + clinical safety from SD DE Cohort H: 1.5 SC 4:1 Cohort B: 1.0 IV 4:1 4:1 Cohort G: 0.5 SC Cohort A: 0.3 IV 4:1 Active:Placebo 4:1 Time

  19. Appendix

  20. Examples of Grading for Adverse EventsSystem based

  21. Neonatal Fc receptor for IgG (FcRn) FcRn Recycles IgG Via pH Dependent Binding & Release pH 7.4 pH 6 V. Ghetie & E.S. Ward, Ann. Rev. Immunol. 2000

More Related