57
Thiopurines 6TG 6MP Thiopurines are purine analogs WHO List of Essential Medicine, among the most important medications needed in a basic health system Indications: anti-cancer agents: acute lymphoblastic leukemia Immunosuppresive agents: inflammatory bowel diseases (Crohn’s disease and ulcerative colitis) AZA purine

Thiopurines - UMD 2... · 2020. 10. 23. · Thiopurines. 6TG. 6MP • Thiopurines are purine analogs • WHO List of Essential Medicine, among the most important medications needed

  • Upload
    others

  • View
    4

  • Download
    0

Embed Size (px)

Citation preview

  • Thiopurines

    6TG 6MP

    • Thiopurines are purine analogs• WHO List of Essential Medicine, among the most important

    medications needed in a basic health system• Indications:

    – anti-cancer agents: acute lymphoblastic leukemia– Immunosuppresive agents: inflammatory bowel diseases (Crohn’s disease

    and ulcerative colitis)

    AZApurine

  • Thiopurines are the Mainstay of ALL Therapy

    6TG

    6MP

    • 6MP/6TG is the most importantcomponents of curative ALL therapyin children and adults

    • Myelosuppression is the main sideeffect

    • Dose titration is done based onWBC/ANC but challenging– Too much dose reduction:less intense therapy, higher risk of relapse– Too little dose reduction:excessive toxicities, treatment disruption,ALL relapse

  • DNA incorporation (DNA-TG)

    DNA damage

    Cytotoxicity

    Thiopurine Drug Metabolism

    Modified from Stocc et al Clin PharmacolTher 2009

    TGTP

    PresenterPresentation NotesSo NUDT15 is another inactivation enzyme for 6MP, converting triphosphate metabolite to monophosphate metabolite and compromise the cytotoxic effects of this drug.

  • Weinshilboum Am J Hum Gen 1980Lennard Lancet 1990

    Anti-leukemic effect

    Myelosuppression(neutropenia)

    PresenterPresentation NotesThis is just a simplified scheme that shows TPMT is the sole source of MP inactivation in the hematopoietic system.

  • Human TPMT Gene and Mutant Alleles

    45 112 184 93 133 53 75 86 45 2075

    ATG

    TPMT*1(wild type)

    ATG

    TPMT*2( activity)

    G238C(AlaPro)

    ATG

    TPMT*3C( activity)

    A719G(TyrCys)

    ATG

    TPMT*3A( activity)

    G460A(AlaThr)

    A719G(TyrCys)

    P

    A

    C A

    Proc Natl Acad Sci USA 1995, DNA Cell Bio 1996, Am J Hum Genet 1996, Clin Pharmacol Ther 1997 and 1998

  • Hematological Toxicity Determined by TPMT genotype

    Relling et al, JNCI, 1999

    GG

    GC

    CC

    6MP TGN

    MeMPTPMT

    Relling et al, JNCI, 1999Relling et al., J Natl Cancer Inst 1999

    PresenterPresentation NotesFurther, we analyzed the MP-related toxicity in ALL patients and found that the incidence of 6-MP intolerance was significantly higher in patients with the CC genotype of TPMT. And there is a trend of decreasing toxicity from CC to GG genotype, with the heterozygotes exhibiting intermediate level of toxicity.

  • 0

    10

    20

    30

    40

    50

    60

    70

    80

    0

    1

    2

    3

    4

    5

    6

    7

    1 11 21 31 41 51 61 71

    Belinda, 5 year old, female Ethnic Chinese, Sarawak, Malaysia B-ALL, standard risk

    NUDT15 R139C

    Weeks in Maintenance Therapy

    Num

    ber

    of D

    ays

    (per

    Wee

    k)

    Norm

    alized 6MP D

    osage (mg/m

    2/day)

    50 mg/m2/day

    6MP dosageDays missed 6MPDays taken 6MP

    Meet Belinda, an Unexpected Case of 6MP toxicity

  • Two Loci Associated with 6MP Tolerance at Genome-wide Sig Level

    Discovery GWAS in COG AALL03N1 cohort (N=657), with independent validation (N=371) Illumina Exome array of 250K variants

    Each dot is a SNP and color indicates chromosome Inverse log-transformed P value on theY axis,The taller the peak, the smaller the P value

    Yang et al., J Clin Oncol 2015

    PresenterPresentation NotesSo there are two hits from the GWAS that reached genome-wide significance threshold, and the first one Is TPMT on chromosome 6.

  • NUDT15 C416T Variant is Strongly Associated with MP Intolerance

    T a

    llele

    % a

    t rs

    1168

    5523

    2

    Discovery GWAS (AALL03N1)

    Replication Cohort (St. Jude Total XV)

    NUDT15

    Arg→CysC415T

    PresenterPresentation NotesPatients homozygous for the risk allele at NUDT15 SNP were exquisitely sensitive to 6MP and tolerated only 8% of standard dose of 6MP, compared to pts with het or wt genotype. This is also true in the validation cohort at St. Jude. Luckily, this was also a missense variation changing Arg residue to cysteine.

  • Cumulative Effects of NUDT15 and TPMT on 6MP Tolerance

    Yang et al., J Clin Oncol 2015 (ALL)

    Of patients requiring >80% dose reduction, 80% had risk variants at these two genes

    Yang et al., Nat Genet 2014 (IBD)

    PresenterPresentation NotesNUDT15 and TPMT independently influence 6MP toxicity, with the degree of toxicity correlated with the number of copies of the risk allele in these two genes. Patients heterozygous in either one of the two genes can tolerate abt 60% of the standard dose, those het for both gene further went down to 40% standard dose, and then those homozygous for either one of the to can tolerate only 6% of normal dose. All together, 80% of the most severe myelosuppression can be predicted by just 4 variants in these two genes, which is quire remarkable in my view.

    NUDT15-related thiopurine toxicity was also described in patients with inflammatory bowel diseases through an independent GWAS study.

  • NUDT15 DNA damage

    Cytotoxicity

    NUDT15 Inactivates TGTP and Reduces Cytotoxicity

    DNA incorporation (DNA-TG)

    TGTP

    Moriyama et al., Nat Genet 2016

    0 20 40 600

    500

    1000

    1500

    2000WTR139C

    TdGTP (uM)

    Velo

    city

    (pm

    ol P

    Pi/m

    in)

    PresenterPresentation NotesSo NUDT15 is another inactivation enzyme for 6MP, converting triphosphate metabolite to monophosphate metabolite and compromise the cytotoxic effects of this drug.

  • DN

    A-TG

    /MP

    dosa

    ge(fm

    ol/µ

    g D

    NA/

    mg

    MP)

    NUDT15diplotypes *1/*1

    *1/*3*1/*5

    *3/*5*3/*3

    N= 23 7 2

    NUDT15diplotypes *1/*1

    *1/*2*1/*3 *2/*3

    N= 22 9 1

    P=7.7 x 10-5D

    NA-

    TG/M

    P do

    sage

    (fmol

    /µg

    DN

    A/m

    g M

    P)P=2.9 x 10-4

    P=0.14

    P=0.039

    NUDT15 Diplotype and 6MP Metabolite (DNA-TG)

    6MP TGTP DNA-TGTGMP ApoptosisNUDT15

    Singapore Japan

  • 0.0 0.5 1.0 1.50

    2000

    4000

    6000

    8000

    10000WildtypeNUDT15 Deficient

    MP (µM)

    DN

    A-TG

    (fm

    ol/ µ

    g DN

    A)

    0.0 0.5 1.0 1.50

    2000

    4000

    6000

    8000

    10000WildtypeNUDT15 Deficient

    MP (µM)D

    NA-

    TG (f

    mol

    / µg

    DNA)

    NUDT15-guided Thiopurine Dose Adjustments?

  • Nudt15-/- Mice Experienced Bone Marrow Suppression with DNATG Accumulation

    Nishii et al., Blood 2018

    We subsequently developed a syngeneic mouse leukemia with Nudt15 deficiency Thiopurine dose reduction in Nudt15-/- mice mitigated toxicity without

    compromising anti-leukemia efficacy

    WT KONudt15 WT KO

    120MP Dosage(mg/kg)

    Neu

    trop

    hils

    (×1

    ,000

    cel

    ls/µ

    l)

    0

    0.25

    0.50

    0.75

    1.00

    1.25

    1.50

    WT KONudt15 WT KO

    120MP Dosage(mg/kg)

    Log1

    0 D

    NA

    -TG

    (fmol

    /µgD

    NA

    )

    2.0

    2.5

    3.0

    1.5

    PresenterPresentation NotesNow we can predict 6MP toxicity based on NUDT15 genotype, but the bigger question is whether we can use NUDT15 genotype to individualize 6MP therapy.

    To address this, we recently developed a Nudt15 knock out mouse model using CRISPR cas9 editing. Across 6MP doses, Nudt15 deficiency led to more severe neutropenia in vivo, and this is in line with increased DNA-TG metabolite accumulation.

    Interestingly, when we dropped the 6MP dose from 20mg to 1mg in the KO mice, their level of toxicity was essentially non-distinguishable from WT mice on normal dose. This 95% dose reduction also effectively normalized exposure to DNA-TG in vivo, shown on the right.

    Subsequently we developed a syngeneic mouse leukemia with Nudt15 deficiency and we showed that 6MP dose reduction can safely reduce host toxicity without compromising anti-leukemic efficacy of this important drug.

  • 25 guidelines; 20 genes and > 60 drugs

    • TPMT, NUDT15– MP, TG, azathioprine

    • CYP2D6– Codeine, tramadol, hydrocodone,

    oxycodone, tricyclic antidepressant, tamoxifen, selective serotonin reuptake inhibitors, ondansetron, tropisetron, atomoxetine

    • CYP2C19– tricyclic antidepressant, clopidogrel,

    voriconazole, selective serotonin reuptake inhibitors, proton pump inhibitors

    • VKORC1– Warfarin

    • CYP2C9– Warfarin, phenytoin, NSAIDs

    • CYP4F2– Warfarin

    • HLA-B--Allopurinol, carbamazepine, Oxcarbazepine, abacavir, phenytoin

    • HLA-A– carbamazepine

    • CFTR-- Ivacaftor

    • DPYD– 5FU, capecitabine, tegafur

    • G6PD– Rasburicase

    • UGT1A1– Atazanavir

    • SLCO1B1– Simvastatin

    • IFNL3 (IL28B)– Interferon

    • CYP3A5– Tacrolimus

    • CYP2B6– Efavirenz

    • RYR1, CACNA1S– Inhaled anesthetics

    • mtRNR1 (in progress)– aminoglycosides

    Dose Adjustment Based on Pharmacogenetics13 Drugs with CPIC Recommendations

  • Pharmacogenomics seek to understand the genetic basis of inter-patient variability in drug response.

    Pharmacogenetic variants can directly alter drug metabolism,efficacy, and toxicity in patients, with TPMT and NUDT15 asexample in the context of thiopurine

    Thiopurine dosing can be individualized based on TPMT andNUDT15 genetics, highlighting the importance ofpharmacogenetics in drug dosing

    Currently, there are 13 drugs for which dose adjustment isrecommended based on pharmacogenetics, according to CPIC

    Summary

  • Acknowledgements

    Takaya MoriyamaChase Suiter Rina NishiiTing-Nien LinWentao YangTomoya Isobe

    Wenjian Yang

    Mary RellingWilliam Evans

    BiostatisticsCheng ChengXueyuan Cao

    Oncology/PathologyChing-Hon Pui Hiroto Inaba

    NUSAllen Yeoh

    JPLSGAtsushi Manabe, Hiroki Hori, Motohiro Kato

    UNOPFederico Antillon

    U CopenhagenKjeld Schmiegelow

    U TuebingenMatthias Schwab

  • Developing a reasonable approach for pediatric dose selection:

    Current and Future Approaches

  • Developing a reasonable approach for pediatric dose selection: Current and Future Approaches

    Jeffrey S. Barrett, PhD, FCPSenior Advisor, Quantitative Medicine

    Gauthier et. al, J. Pers. Med. 2011, 1(1), 5-16; https://doi.org/10.3390/jpm1010005

  • Outline

    Dose Selection Basics:• Data to make decisions

    - PK vs PK/PD• ExtrapolationCurrent Approaches:• Top-down and bottom-up approaches • Extrapolation• Combination ApproachesFuture Approaches:• Use of RWD• Synthetic data approaches• How good can we be• Necessity of data integration and planning

    3

  • Dose Selection Basics: Dose Finding or Equivalence?

    4

    • Dose Finding – PK/PD• A target / endpoint is known or theorized based on adult or other

    data; dose-exposure to be defined• Studies designed to explore dose-response relative to endpoint target

    • Equivalence Approach – PK only (typically)• Exposure requirements based on adult experience – “equivalent”

    safety and efficacy assumed by matching exposure• Studies designed to match exposure target

  • Dose Selection Basics: PK/PD Approach

    • Typically essential when adult experience and dose-exposure relationship is unlikely to be relevant for pediatric patients and/or adult endpoints are not relevant in children.

    • The use of a PD endpoint that has been validated for use in children should be a prerequisite (often use an unvalidated marker with the adult endpoint as a comparator)

    De Cock RF, Piana C, Krekels EH, Danhof M, Allegaert K, Knibbe CA. The role of population PK-PD modelling in paediatric clinical research. Eur J Clin Pharmacol. 2011 May;67 Suppl 1(Suppl 1):5-16. doi: 10.1007/s00228-009-0782-9. Epub 2010 Mar 26. PMID: 20340012; PMCID: PMC3082690.

  • Dose Selection Basics: PK/PD Approach

    • Requires prospective, dose-finding trials in the intended patient population (typically 3 or more doses – often just 2)

    • Sensitive analysis techniques requiring only small blood samples should be used even with optimal sampling schemes

    • All intended age strata of intended clinical use should be evaluated.

    • Staggered dosing (older children first) often recommended.

    Willmann, S., Thelen, K., Kubitza, D. et al. Pharmacokinetics of rivaroxaban in children using physiologically based and population pharmacokinetic modelling: an EINSTEIN-Jr phase I study. Thrombosis J 16, 32 (2018). https://doi.org/10.1186/s12959-018-0185-1

  • Dose Selection Basics: Equivalence Approach

    • Emphasis is based on the assumptions that therapeutic exposures attained in adult patients are relevant (appropriate) for the intended pediatric patient population(s)

    • Assumes that the underlying disease progressions are similar

    • Sensitive analysis techniques requiring only small blood samples should be used even with optimal sampling schemes

    • Designs are PK-centric without necessity of sampling for PD endpoints

    • All intended age strata of intended clinical use should be evaluated.

  • Dose Selection Basics: Equivalence Approach

  • Picking Starting Doses for Pediatric Trials

    Does a simple rule exist? NO

    Under-predicts across age strata

    Under-predicts infants and neonates

    Over-predicts age < 1y

    “Scaling using body weight alone may be safer in the neonatal and infant age range in terms of avoiding toxicity: the possibility exists that an under-dose will be administered, but this dose can then be titrated up according to clinical response. Scaling using the BSA or BW0.75 method would seem reasonable in children above 2 years of age, but even so should still be used with caution.”

    Conventional wisdom . . .?

    Johnson, T.N., The problems in scaling adult drug doses to children. Arch Dis Child, 2008. 93(3): p. 207-11

  • Current ApproachesTop Down and Bottom-up Approaches

    • The choice to use either method is typically based on the stage of development and the availability of adult data.

    • The two approaches are complimentary and generally support each other• Quite often, both are conducted

    10

    Top-Down Approach:(Adult-informed PPK Model)• Adult PPK model scaled to predict pediatric

    populations (size, maturation and ontogeny considerations)

    • Simulations conducted to assess dose requirements across age / weight strata relative to adult experience (PK and/or PK/PD accommodated as needed)

    Bottom-Up Approach:(PBPK Model)• Physiochemical and ADME data inputs to backbone of

    physiologic-represented model that can accommodate mechanistic relationship

    • Not reliant on adult data but can use adult data to refine /qualify model.

    • Can accommodate PK and/or PK/PD as well.• Simulations conducted to assess dose requirements

    across age / weight strata relative to adult experience

  • Current Approaches: Top-Down ApproachPicking Starting Doses – the usual procedure

    1. Evaluate doses or exposure profiles thought to yield equivalent exposures to adults

    2. Estimate sample size to detect a difference in key parameters between treatment groups, adults or historical controls

    3. Select sampling scheme that will yield “meaningful” parameter estimates in children

    • Generate PK distribution for intended age-weight subpopulations at all doses considered.

    • Compare overlap in ranges across populations and age groups• Compare with adult profiles

    • Same as above.• Derive metrics for individual simulated subjects (e.g., AUC,

    Cmax, CL)• Compare groups via ANOVA• Delta and variance to estimate sample size

    • Define critical time points for PK parameters based on adult model – D-optimal design.

    • Simulate pediatric profiles with various combinations of sampling time using pediatric-scaled model

    • Re-fit simulated data; compare precision and bias

  • Current Approaches: Top-Down ApproachPicking Starting Doses – the usual procedure

    CLGRP = CLSTD * * MF * OF

    CLGRP is the group clearance

    Wi is the is the individual total body weight

    CLstd is the clearance in a standardized individual of weight Wstd

    MF = Maturation Function

    OF = Organ Function

    Wstd

    Patient-specific factors related to their care or treatment

    * ???

    Scaling across age ranges . . .Customizing Expressions to Fit the Population

  • Current Approaches: Bottom-Up ApproachPicking Starting Doses – the usual procedure

    Blood

    Lung

    Rapidly perfused organs

    Slowly perfused organs

    Kidney

    Liver

    IntestinesBlood

    EliminationDosing

    ADME, PK, PD and MOA

    MetabolismActive transport

    Passive diffusionProtein binding

    Drug-Drug interactionsReceptor binding

    System component (drug-independent)

    PBPK MODEL

    A. Intrinsic/Extrinsic Factors B. PBPK Model Components

    Huang and Temple, 2008Individual or combined effects on human physiology

    EXTRINSIC

    INTRINSIC

    DDI

    Environment

    Medical Practice

    Regulatory Alcohol

    Smoking

    Diet

    Age

    Race

    Disease

    Gender

    Genetics

    Pregnancy

    Lactation

    Organ Dysfunction

    Drug-dependent component

    INTRINSIC

    Age

    Race

    Disease

    Gender

    Genetics

    Pregnancy

    Lactation

    Organ Dysfunction

    Zhao P. et al., Clin Pharmacol Ther, 2011

  • Approach:• Incorporate physiochemical and ADME data

    into PBPK model framework; refine with in vivo data (adult and /or pediatric data if available).

    • Devise dosing and sampling scenarios consistent with planned study constructs.

    • Recommend scenarios with highest PTOS.• Evaluate /refine against real-time, actual

    study data if possible.

    14Current Approaches: Bottom-Up ApproachPicking Starting Doses – the usual procedure

    Willmann S, Thelen K, Kubitza D, Lensing AWA, Frede M, Coboeken K, Stampfuss J, Burghaus R, Mück W, Lippert J. Pharmacokinetics of rivaroxaban in children using physiologically based and population pharmacokinetic modelling: an EINSTEIN-Jr phase I study. Thromb J. 2018 Dec 4;16:32. doi: 10.1186/s12959-018-0185-1. PMID: 30534008; PMCID: PMC6278136.

  • Future ApproachesUse of RWD – Value for Prescribing and Design of Future Trials

    • Both from the standpoint of guiding dosing practice and designing or complementing clinical trials, RWD has become and valued asset.

    • As pediatric research and development often operates with a deficit of data and relies heavily on the adult experience, there is great perceived opportunity.

    15

  • Future ApproachesUse of RWD – Still a few caveats

    • For many drugs valid measures of drug effect are not consistently documented (e.g., therapeutic response to medications for attention deficit hyperactivity disorder or depression) or not available for the full spectrum of pediatric patients.

    • Across all drugs, reliable documentation of adverse drug events is incomplete and represents an area of need for complete PD analysis. Long-term funding for multi-site collaborative networks is required to address the challenges, pool data, and validate findings.

    • Generation of high-quality, generalizable, and validated data will facilitate subsequent clinical implementation.

    16

    Van Driest SL and Choi L. Real World Data for Pediatric Pharmacometrics: Can We Upcycle Clinical Data for Research Use? Clin Pharmacol Ther. 2019 July ; 106(1): 84–86. doi:10.1002/cpt.1416.

  • 17

    QSP Modeling Application: Used in ERT program for extrapolation to pediatrics: Olipudase Example

    Figure 1, CPT Pharmacometrics Syst. Pharmacol. (2018)

    • Quantitative systems pharmacology (QSP) is a mechanistic modelling tool that links molecular mechanisms of disease and drug to biomarkers and clinical endpoints used for assessment of disease and therapeutic effect

    • QSP is suited to understanding the system-level response to treatment across multiple PD markers and clinical endpoints, and to assessing patient variability on a mechanistic basis

    • The QSP model for olipudase alfa links a reduced-order physiologically based pharmacokinetic (PBPK) model with molecular-, cellular-, and organ-level sub-models to understand treatment response patterns

    Kaddi CD, Niesner B, Baek R, Jasper P, Pappas J, Tolsma J, Li J, van Rijn Z, Tao M, Ortemann-Renon C, Easton R, Tan S, Puga AC, Schuchman EH, Barrett JS, and Azer K. Quantitative Systems Pharmacology Modeling of Acid Sphingomyelinase Deficiency and the Enzyme Replacement Therapy Olipudase Alfa Is an Innovative Tool for Linking Pathophysiology and Pharmacology. CPT Pharmacometrics Syst Pharmacol. 2018 Jul; 7(7): 442–452. PMCID: PMC6063739, PMID: 29920993

  • Pediatric extrapolation: Leveraging PBPK-QSP for Disease and Response Similarity Assessment – Olipudase Example (ASMD)

    Hypothetical pediatric scenarios

    Critical elements for pediatric extrapolation:

    • Selection of clinical endpoints or biomarkers for tracking disease: organ and sub-organ levels (multi-scale)

    • Genotype-phenotype mapping to create virtual populations commensurate with disease severity levels, substantiated by registry data

    • Virtual population can be genotype-prevalence weighted, based on registry prevalence distribution

    • Multi-organ/biomarker integration through model is critical to bridge together a multi-parameter distribution relationship

    HypotheticalExample

  • Future ApproachesSynthetic data approaches

    19

    Approach and Rationale:• Privacy restrictions limit access to

    protected patient-derived health information for research purposes.

    • Data anonymization is required to allow researchers data access for initial analysis before granting institutional review board approval.

    • Synthetic data generation seeks to mimic data from real electronic medical records, providing a synthetic patient dataset to analyze.

  • Future ApproachesNecessity of data integration and planning

    Planning• Starts at the Early Development teams and the TPP• Empower the pediatric section with key deliverables around dose projection• Obligate the PSP and PIP to include these deliverables• Delegate to the right skillsets.

    Data Integration• Landscape analysis on key data elements

    - Include RWD (prevalence, SOC, competitive intelligence data- Purchased data sources, literature data assembly

    • Generation of analysis datasets in advance of use• Scripting and coding templates where possible

    20

  • Future ApproachesHow good can we be?

    • Safety should still guide us – give a safe dose first

    • Still a disconnect between the prescription and the dosing

    • Plan for RWD evolution to influence precision dosing approaches in the future

    • Still need flexibility in the pediatric formulations themselves

    • The tools we leave behind from the effort to get the dose right in children become the starting point for model-informed precision dosing (MIPD) algorithms – hopefully, our future.

    We just need to be better!21

  • References:• Germovsek E, Barker CIS, Sharland M, Standing JF. Pharmacokinetic-Pharmacodynamic Modeling in Pediatric Drug Development, and the

    Importance of Standardized Scaling of Clearance. Clin Pharmacokinet. 2019 Jan;58(1):39-52. doi: 10.1007/s40262-018-0659-0. Erratum in: Clin Pharmacokinet. 2018 Dec 17;: PMID: 29675639; PMCID: PMC6325987.

    • De Cock RF, Piana C, Krekels EH, Danhof M, Allegaert K, Knibbe CA. The role of population PK-PD modelling in paediatric clinical research. Eur J Clin Pharmacol. 2011 May;67 Suppl 1(Suppl 1):5-16. doi: 10.1007/s00228-009-0782-9. Epub 2010 Mar 26. PMID: 20340012; PMCID: PMC3082690.

    • Willmann, S., Thelen, K., Kubitza, D. et al. Pharmacokinetics of rivaroxaban in children using physiologically based and population pharmacokinetic modelling: an EINSTEIN-Jr phase I study. Thrombosis J 16, 32 (2018). https://doi.org/10.1186/s12959-018-0185-1

    • Reiner Benaim A, Almog R, Gorelik Y, Hochberg I, Nassar L, Mashiach T, Khamaisi M, Lurie Y, Azzam ZS, Khoury J, Kurnik D, Beyar R. Analyzing Medical Research Results Based on Synthetic Data and Their Relation to Real Data Results: Systematic Comparison From Five Observational Studies. JMIR Med Inform 2020;8(2):e16492, DOI: 10.2196/16492, PMID: 32130148, PMCID: 7059086

    • Van Driest SL and Choi L. Real World Data for Pediatric Pharmacometrics: Can We Upcycle Clinical Data for Research Use? Clin PharmacolTher. 2019 July ; 106(1): 84–86. doi:10.1002/cpt.1416.

    • Mulugeta LY, Yao L, Mould D, Jacobs B, Florian J, Smith B, Sinha V, Barrett JS. Leveraging Big Data in Pediatric Development Programs:Proceedings From the 2016 American College of Clinical Pharmacology Annual Meeting Symposium. Clin Pharmacol Ther. 2018 Jul;104(1):81-87. doi: 10.1002/cpt.975. Epub 2018 Jan 10. PMID: 29319159.

    • Kaddi CD, Niesner B, Baek R, Jasper P, Pappas J, Tolsma J, Li J, van Rijn Z, Tao M, Ortemann-Renon C, Easton R, Tan S, Puga AC, SchuchmanEH, Barrett JS, and Azer K. Quantitative Systems Pharmacology Modeling of Acid Sphingomyelinase Deficiency and the Enzyme Replacement Therapy Olipudase Alfa Is an Innovative Tool for Linking Pathophysiology and Pharmacology. CPT Pharmacometrics Syst Pharmacol. 2018 Jul; 7(7): 442–452. PMCID: PMC6063739, PMID: 29920993

    • Barbour AM, Fossler MJ, Barrett J. Practical considerations for dose selection in pediatric patients to ensure target exposure requirements. AAPS J. 2014 Jul;16(4):749-55. doi: 10.1208/s12248-014-9603-x. Epub 2014 May 20. PMID: 24841797; PMCID: PMC4070253.

    22

    https://doi.org/10.1186/s12959-018-0185-1

  • Questions:

    [email protected]

    23

  • Current and Future Pediatric Dosing Considerations from a Regulatory Viewpoint

    Lynne P. Yao, M.D.Director, Division of Pediatric and Maternal Health

    Office of Rare Diseases, Pediatrics, Urologic and Reproductive Medicine (ORPURM)

    Office of New DrugsCenter for Drug Evaluation and Research

    U.S. FDA

  • 2

    Disclosure Statement

    • I have no financial relationships to disclose relating to this presentation

    • The views expressed in this talk represent my opinions and do not necessarily represent the views of FDA

  • 3

    U.S. Evidentiary Standard for Approval• For approval, pediatric product development is held to same evidentiary

    standard as adult product development:• A product approved for children must:

    – Demonstrate substantial evidence of effectiveness/clinical benefit (21CFR 314.50)– Clinical benefit:

    • The impact of treatment on how patient feels, functions or survives• Improvement or delay in progression of clinically meaningful aspects of the disease

    • Evidence of effectiveness [FD&C 505(d) (21 U.S.C. § 355(d)].– Evidence consisting of adequate and well –controlled investigations on the basis of

    which it could fairly and responsibly be concluded that the drug will have the effect it purports to have under the conditions of use prescribed, recommended, or suggested in the labeling

    • Adequate safety information must be included in the application to allow for appropriate risk benefit analysis [FD&C 505(d)(1)]

    PresenterPresentation NotesHao already discussed

  • 4

    Special Considerations for Pediatric Product Development

    • Ethical considerations– Children should only be enrolled in a clinical trial if the scientific and/or public health

    objectives cannot be met through enrolling subjects who can provide informed consent personally (i.e., adults)

    – Absent a prospect of direct therapeutic benefit, the risks to which a child would be exposed in a clinical trial must be “low”

    – Children should not be placed at a disadvantage after being enrolled in a clinical trial, either through exposure to excessive risks or by failing to get necessary health care

    – Ethical considerations do play a role in the need to correctly apply pediatric extrapolation• Feasibility considerations

    – The prevalence and/or incidence of a condition is generally much lower compared to adult populations

  • 5

    Statutory Requirement

    • Pediatric Research Equity Act (PREA) requires that a pediatric assessment “shall contain data. . .to support dosing and administration for each pediatric subpopulation for which the drug or the biological product is safe and effective.” [505B(a)(2)(A)(ii)]

    • No clear requirement to establish an “optimal” dose• Unsuccessful dose selection methods lead to failed pediatric

    trials

  • 6

    Extrapolation of Efficacy:Disease/response “similarity” is a continuum

    Significant overlap; no known significant differences

    between adult and pediatric condition

    Large degree of overlap with some differences between

    adult and pediatric condition

    Some degree of overlap with significant differences

    between adult and pediatric condition

    No overlap between adult and pediatric condition

    Different Dissimilar Similar Same

    Increasing relevance of adult information to pediatric population with increasing confidence in similarity between adult and pediatric condition

    Exposure matchingPediatric RCT(s)

    Pharmacodynamic markers, Bayesian methodologies, etc.

  • 7

    Apparent Exposure Response in Pediatrics

    • Pediatric exposure response studies are difficult to design to truly evaluate exposure response– Ethically difficult to assign patients to a dose that would be considered

    “ineffective”– Assignment to one dose in the effective range (“flat part”) of the curve may

    lead to incorrect conclusions about differences in exposure response– Consider evaluation of dose-response or “dose-exposure-response”

    • Use of biomarkers to better understand response and guide dose selection

    Excerpted from Marc Gastonguay Presentation FDA/UMD CERSI pJIA Drug Development Workshop October 2nd, 2019

  • 8

    Pediatric Dose Selection Scenarios

    Assume similar dose-exposure-response (DER) compared to adults• Requires confidence in DER in

    adults• Methods used to match exposures

    will depend upon age groups to be studied

    • Confirmation of model-based predictions is needed

    Cannot assume similar dose-exposure-response (DER) in adults• Will need to conduct dose-ranging

    studies• May use PK/PD modeling but will

    need to have PD marker available• Generally will need evaluation of

    the PD marker in adult populations• Confirmation of dose regardless of

    method of selection will also be needed

    PresenterPresentation NotesPoint counter point Alice KePBPK and allometric scaling/ DDI and formulation and disease effect(can’t necessarily use POPPk)When is allometric scaling sufficient? Drugs with linear PKHow do deal with variability and small populations? Is variability really true? Joga talkFiguring out when to use the right toolNot necessarily PBPK is not needed but when to use it appropriately and when okConfusion: what is the modeling used for?Allometric Scaling/Maturational (age) Models with POPpk vs. PBPKNonlinear PK: When does that occur? Use PBPK?Are Pop PK and PBPK the same? Depends on the equations in the model? Do the PBPK models help to explain variability? HIDE variability??Should it be the opposite? USE PBPK when there are little data; Is this a problem?

  • 9

    Incorporation of Available Data

    • Real World Data

    • Nonclinical Data

    • Natural History Data

    • Clinical Trial Data (adult and pediatric data)

    Modeling and Simulation

    Quantitative Systems

    Pharmacology

    Clinical Trial Simulations

    Other Mathematical

    Models

  • 10

    A Regulator’s Perspective

    Mor

    e Da

    ta/K

    now

    ledg

    eLess Data/Know

    ledge

    • Takes more time• Requires early

    planning• Difficult to obtain in

    certain age groups• Ultimately could

    support more streamlined approaches

    • Takes less time• Often includes

    numerous assumptions• False assumptions will

    lead to incorrect conclusions

    • More difficult to obtain regulatory acceptance

    PresenterPresentation NotesHow to put it all togetherIrony—the need to use modeling generally means less dataThe more data the more comfortable (but takes years—decades)What happens when variability is high? Do we discount the model? No it seems like we try to make the data fit.How do we guard against too much confidence in a model with very little data to confirm

  • 11

    Summary• Our goal is to increase the availability of approved products for

    use in pediatric patients• Dose selection in pediatric development programs depends upon

    the available knowledge• Optimizing the use of available data

    – Appropriate use of modeling and simulation– Transparency in model development and confirmation– Innovative statistical approaches– Appropriate use of biomarkers

    • Improved communication of dosing information in labeling

    PresenterPresentation NotesBernd Meibohm: biologicsDosing: lack of biomarkers to assess disposition; high tolerability (can’t dose to MTD) and limited off target effectsHow to express dosing in labeling even if you have known difference in exposure/response (what is the bottom line for dosing in labeling)

  • Thank you

  • Pediatric Dose SelectionFDA/MCERSI Workshop – Wrap Up

    Gilbert J. Burckart, Pharm.D.Associate Director for Pediatrics Office of Clinical Pharmacology

    OTS, CDER, FDA

    Disclaimer: The comments and concepts presented are those of the speaker and should not necessarily be interpreted as the position of the US FDA

  • 2

    Thanks!

    • Many thanks again to:– All of the speakers who gave their time and effort to make

    the workshop a success!– The University of Maryland Center for Excellence in

    Regulatory Science and Innovation (MCERSI)– The US FDA Office of Clinical Pharmacology

    • What comes next?

    2

  • 3

    The future challenge is to create a structuredapproach to determining pediatric doses for newtherapeutic agents (CPT Commentary, 2010)

    - This workshop is a step in the direction of developinga structured approach.

  • 4

    Workshop Published Supplement – The Journal of Clinical Pharmacology, March 2021

    • Aaron Pawlyk - A Call for Objective Dose Selection to Increase Success in Pediatric Clinical Trials

    •• Gil Burckart/John van den Anker – A Decision Tree for Pediatric

    Dose Selection – Workshop Summary•• John van den Anker – Dose Selection for Premature Infants•• Gil Burckart - Methods Used for Pediatric Dose Selection in Drug

    Development Programs Submitted to the US FDA 2012-2019•• Efthymios Manolis - The EMA experience with pediatric dose

    selection•• Alice Ke - PBPK modeling and allometric scaling in pediatric drug

    development: where do we draw the line?•• Joga Gobburu – Pediatric Therapeutics Management with Clinical

    Decision Support Systems•• Hao Zhu - Confirming Extrapolation of Efficacy: Atypical

    Antipsychotic Dose-Selection in Adolescents with Schizophrenia and Bipolar I Disorder

    •• Youwei Bi - Use of MIDD in Dose Selection in Pediatric Clinical Trials•• Jian Wang – Evaluation of Exposure-Response Similarity Between

    Pediatrics and Adults in Drug Development•

    • Bernd Meibohm – Pediatric Dose Selection for Therapeutic Proteins•• Danny Gonzalez - Pediatric Drug-Drug Interaction Evaluation: Drug,

    Patient Population, and Methodological Considerations•• Mona Khurana – Renal Impairment Dosage Recommendations for

    Pediatric Patients•• Andre Dallmann – Pediatric Drug Absorption and Physiologically-

    Based Pharmacokinetic Modeling•• Andre Dallmann - Predictive performance of PBPK dose estimates

    for pediatric trials•• Jun Yang - The Role of Pharmacogenomics in Drug Dosing in

    Children•• Jeff Barrett – Precision Dosing in Children•• Sander Vinks – Model-informed Pediatric Dosing•• Karel Allegaert – Dose related adverse drug-related events in

    neonates: severity assessment and recognition•• Gil Burckart – The Role of Drug Safety in Pediatric Dose Section•

    Guest Editors: John van den Anker, Gilbert Burckart

  • ADP5B39.tmpSlide Number 1Slide Number 2OutlineDose Selection Basics: Dose Finding or Equivalence?Dose Selection Basics: PK/PD ApproachDose Selection Basics: PK/PD ApproachDose Selection Basics: Equivalence ApproachDose Selection Basics: Equivalence ApproachPicking Starting Doses for Pediatric TrialsCurrent Approaches�Top Down and Bottom-up ApproachesCurrent Approaches: Top-Down Approach�Picking Starting Doses – the usual procedureCurrent Approaches: Top-Down Approach�Picking Starting Doses – the usual procedureCurrent Approaches: Bottom-Up Approach�Picking Starting Doses – the usual procedureCurrent Approaches: Bottom-Up Approach�Picking Starting Doses – the usual procedureFuture Approaches�Use of RWD – Value for Prescribing and Design of Future TrialsFuture Approaches�Use of RWD – Still a few caveatsSlide Number 17Pediatric extrapolation: Leveraging PBPK-QSP for Disease and Response Similarity Assessment – Olipudase Example (ASMD)Future Approaches�Synthetic data approachesFuture Approaches�Necessity of data integration and planningFuture Approaches�How good can we be?References:Questions:

    ADPB2D1.tmpSlide Number 1Thanks!Slide Number 3Workshop Published Supplement – The Journal of Clinical Pharmacology, March 2021Slide Number 5

    Jun Yang (1).pdfSlide Number 4Slide Number 5Thiopurine Drug MetabolismSlide Number 7Slide Number 8Slide Number 9Slide Number 10Two Loci Associated with 6MP Tolerance at Genome-wide Sig LevelNUDT15 C416T Variant is Strongly Associated with MP IntoleranceCumulative Effects of NUDT15 and TPMT on 6MP ToleranceSlide Number 14Slide Number 15Slide Number 16Slide Number 1725 guidelines; 20 genes and > 60 drugsSlide Number 19Acknowledgements

    Lynne Yao.pdfCurrent and Future Pediatric Dosing Considerations from a Regulatory ViewpointDisclosure StatementU.S. Evidentiary Standard for ApprovalSpecial Considerations for Pediatric Product DevelopmentStatutory RequirementSlide Number 6Apparent Exposure Response in Pediatrics Pediatric Dose Selection ScenariosIncorporation of Available DataA Regulator’s PerspectiveSummaryThank you