Clinical Research Methods



Clinical Research Methods





PHASE I CLINICAL TRIALS

Connie Lee Batlevi

David R. Spriggs


Phase 0 Trials



  • Proof of concept trials designed to be first in human (Nat Rev Cancer 2007;7:131) administered while toxicology studies are being performed for IND submission


  • Single doses of new drug given at low concentrations not expected to cause tox: Correlated w/biomarker assay, imaging technique, of drug activity w/drug administration in vivo


Phase I Trials



  • One of the initial phases of clinical experimentation



    • First-in-human trials: Promising preclinical data, clinical efficacy unproven novel Rx combinations of existing and/or new agents (eg, carboplatin/paclitaxel + PI3K inhibitor)


  • Primary objectives: Identify drug tox, describe drug pharmacology (PK, PD), identify RP2D


  • Population: Heterogeneous (eg, nonhistology specific), newer trials of molecular agents may select pts based on genotype, often includes pts who have failed multiple lines of prior tx


  • Starting dose: Dose chosen to treat first cohort of pts based on animal studies, commonly 1/10-1/3 of mouse LD10 (dose resulting in death of 10% of mice), other animal species may be used










































Definition of Terms


Dose-limiting toxicity (DLT)


Unacceptable tox that limit further dose escalation, protocol-specific, defined based on CTCAE criteria (note: Not all grade 3 tox are dose-limiting), occur w/in a specific time-frame (eg, w/in first cycle)


Maximum tolerated dose (MTD)


Highest dose level at which a defined proportion of pts (eg, 33%) experience a DLT


Target tox level


Typically 20-33%, maximum probability of DLT considered acceptable


Recommended phase 2 dose (RP2D)


Dose recommended for subsequent phase II trials: Either the MTD or one dose level below MTD


Optimal biologic dose (OBD)


Dose a/w most desirable prespecified effect on a biomarker


Dose-efficacy curve


Reflects relationship between dose & probability of efficacy for a specific agent


Dose-tox curve


Reflects relationship between dose & probability of tox for a specific drug


PK


Drug effects on absorption, distribution, metabolism, & excretion, several PK levels taken along study course


PD


Drug effects on host processes (eg, molecular correlates or biomarkers, imaging endpoints, includes hematologic & nonhematologic tox)


Therapeutic index


Dose required to produce critical tox divided by dose yielding defined antitumor effect


(Adapted from J Natl Cancer Inst 2009;101:708)



Types of Phase I Designs



  • Rule-based designs



    • Egs: Standard 3+3, AT, PGDE


    • Traditional phase I designs w/no prior assumptions of dose-tox curve, allow dose escalation & reduction, easily implemented w/o need for special mathematical software → relatively inefficient at establishing MTD, RP2D based on current dose level & does not incorporate all tox data


  • Model-based designs



    • Egs: CRM, escalation w/O/D control, TITE-CRM, EffTox, TriCRM


    • Statistical model: Starts w/initial dose-tox curve → model modified by tox experienced by pts on trial; allows the incorporation of all available tox data, RP2D estimated w/confidence interval



Traditional 3+3



  • Classic phase I clinical trial design where pts are treated initially in cohorts of 3s at escalating dose levels, dose level escalation or de-escalation follows modified Fibonacci sequence: 100% → 67% → 50% → 40% → 33%, intra-pt dose escalation is typically not allowed


  • Relatively simple to implement, provides more info on PK inter-pt variability per dose level, defined sequence of escalation & de-escalation

























    3+3 Design Based on Number of Patients Experiencing DLT Per Cohort


    Cohort


    Dose Escalate (↑ to next higher dose level)


    Expand Cohort (↑ cohort by 3 pts)


    Dose De-escalate (↓ to next lower dose level)


    3-Pts


    0 DLTs


    1 DLT


    ≥2 DLTs


    6-Pts (expanded)


    1 DLT


    N/A


    ≥2/6 DLTs



  • MTD definition = Highest dose where 0-1/16 pts w/DLT (estimates MTD at 33%) or next higher dose level


  • Pitfalls: Grossly underestimate MTD, dose escalation is slow, many pts treated at subtherapeutic (below RP2D) dose levels


Accelerated Titration (J Natl Cancer Inst 1997;89:1138)



  • Affords rapid initial dose escalation, wide variety of designs, intra-pt dose escalation permitted in some designs


  • Design eg: Initial dose-escalation phase w/single-pt cohorts, dose is escalated rapidly at 96-100% of previous dose → defined stopping rule is hit (eg, 1 DLT or 2 mod. tox observed) → reverts to 3+3 design w/40% dose increments between cohorts


  • MTD is estimated using all tox data collected during trial, some designs may use 3+3

    rules to determine MTD


  • Advantages: Reduces number of pts treated at subtherapeutic doses, designs w/intra-pt dose escalation affords tx at higher & potentially more effective doses, disadvantages: Intra-pt dose escalation may mask cumulative/delayed tox, aggressive & may expose more pts to higher doses, requires acute tox (chronic/cumulative tox not captured)


Pharmacologically-Guided Dose Escalation (J Natl Cancer Inst 1990;82:1321)



  • Assumes DLT predictable by plasma drug concentrations: Plasma exposure defined by AUC extrapolated from preclinical data


  • Design: Single-pt dose escalation (based on PK data, typically 100%) → stopping rule hit (eg, target AUC reached or DLTs occur) → switch to 3+3


  • Provides rapid dose escalation but not widely used due to difficulty obtaining real-time PKs, dose escalation may be hampered by inter-pt variability


Continual Reassessment Method (Biometrics 1990;46:33)



  • Bayesian approach: Starts w/defined acceptable probability of DLT (20-33%), 1-3

    pts selected per cohort depending on accrual & desired DLT rate, starts at dose thought to be close to MTD


  • Mathematical model for dose-tox curve continuously modified as each enrolled pt experiences a DLT → dose is then escalated & de-escalated accordingly (next pt is treated at new estimated dose close to MTD) → trial stopped if prespecified condition met or precision in probability of DLT at estimated MTD level is achieved


  • Requires intensive statistical assistance, thought to provide more precise MTD estimate, may be safer w/fewer pts treated at toxic doses


Other Model-Based Designs



  • Modified CRM design: Escalation w/O/D control (Stat Med 1998;17:1103)


  • TITE-CRM (Biometrics 2000;56:1177): Incorporates time-to-event of tox for each pt, assumes hazard of tox remains constant


  • Efficacy & tox models: Eff Tox defines acceptable dose combination based on trade-offs between probabilities of efficacy & tox, TriCRM w/3 potential trial outcomes (no efficacy no tox, efficacy only, tox only)



PHASE II and III CLINICAL TRIALS

Connie Lee Batlevi

Robert J. Motzer































Definition of Terms


Historical controls


Pt prev treated (or not) used as standard for comparison to pt being treated in current experimental design


Type I error (α)


FP, ↓ type I error by increasing sample size


Type II error (β)


FN, ↓ type II error by increasing sample size


Effect size


Difference between null & alternative hypothesis values


Binomial proportion test


Assuming equal distribution, determines statistical significance of deviations from expected distribution


Hazard ratio


Ratio of standard/experimental tx failure rates


Logrank test


Nonparametric test (assumes data is skewed) establish efficacy of new tx vs. control when measurement is time-to-event

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Aug 17, 2016 | Posted by in ONCOLOGY | Comments Off on Clinical Research Methods

Full access? Get Clinical Tree

Get Clinical Tree app for offline access