Pharmacogenetics - PowerPoint PPT Presentation

1 / 40
About This Presentation
Title:

Pharmacogenetics

Description:

Pharmacogenetics Plays a Role ... 5-Fluorouracil Catabolism ... DPD is responsible for catabolism of 5-fluorouracil. This accounts for 85% of drug metabolism ... – PowerPoint PPT presentation

Number of Views:673
Avg rating:3.0/5.0
Slides: 41
Provided by: RJ309
Category:

less

Transcript and Presenter's Notes

Title: Pharmacogenetics


1
Pharmacogenetics Chemotherapy
  • Jane S. Chawla, M.D.
  • January 30, 2009

2
Over the next 30 mintues
  • Intro to Pharmacogenetics
  • Some common polymorphisms
  • DPD
  • UGT1A1
  • TPMT
  • CYP2D6
  • Conclusions

3
Pharmacogenetics Plays a Role in Drug Toxicity
Efficacy
Many sources of interindividual variation in drug
toxicity and efficacy, both pharmacokinetic and
pharmacodynamic.
Deeken, J et al. Anti-cancer drugs, 2007.
4
Genetic Polymorphisms
  • Every gene contains some degree of polymorphism
  • Single nucleotide polymorphism (SNPs) occur every
    1000-3000 base pairs throughout the genome
  • Sometimes difficult to determine which are
    relevant to treatment of disease
  • Polymorphisms can occur in
  • Drug targets (including cell surface receptors
    and target proteins)
  • Hormonal-regulated enzymes
  • Genes involved in drug pharmacokinetics that
    impact drug absorption, distribution, metabolism,
    and excretion
  • Genetic polymorphisms can lead to
  • Severe toxicity when drugs not metabolized
    normally
  • Diminished or increased therapeutic affect

5
5-Fluorouracil DPD Deficiency
6
5-Fluorouracil Catabolism
  • DPD (Dihydropyrimidine dehydrogenase) is the 1st
    and rate-limiting enzyme in the pathway involved
    in the degradation of pyrimidine bases uracil and
    thymine
  • DPD is responsible for catabolism of
    5-fluorouracil
  • This accounts for gt85 of drug metabolism

7
5-FU-induced Toxicity
  • Inability to inactivate 5-FU can lead to
    neurological, hematopoietic, and GI toxicities
    that can be fatal
  • It is reported that about 31 of pts with
    advanced colorectal cancer who receive bolus 5-FU
    regimens have grades 3-4 hematologic toxicity
  • It is estimated that 40-60 of cancer pts with
    severe 5-FU toxicity have DPD-deficiency

8
DPD Deficiency
  • Variance in DPD is a common genetic polymorphism
  • 39 different mutations / polymorphisms identified
  • 14 of these in pts with excessive 5-FU toxicity
  • 3-5 of Caucasians heterozygous for DPD mutations
  • 0.1 of Caucasians homozygous for DPD mutations
  • DPD activity varies 8-21-fold among individuals

9
IVS14 1GgtA is the Most Common DPD Polymorphism
  • G?A transition at the 5 splice consensus
    sequence of exon 14
  • Exon 14 is deleted ? translation of
    non-functional enzyme
  • This polymorphism accounts for 50 of known
    nonfunctional DPD alleles

Deeken, J et al. Anit-Cancer Drugs, 2007.
Nature Reviews Cancer, 2001
10
Ethnic Frequency of DPD Deficiency IVS141GgtA
Variant
  • DPD deficiency seen in
  • Chinese, Indian, Malay, Japanese, Korean pts
  • DPD enzyme activity is lower in healthy African
    Americans compared to Caucasians
  • 8 versus 2.8

Ethnic frequency of IVS141GgtA variant
11
Is ? DPD Activity Predictive of Severe
5-FU-related Toxicity?
  • 131 Caucasian pts with severe 5-FU toxicity
  • DPD activity lower in pts with 5-FU toxicity
    compared to general cancer population (13 vs
    2.7)
  • The lower the DPD activity, the greater the
    grades of toxicity
  • DPD deficiency only partially explains
    5-FU-related toxicity
  • Of the 9 with lethal toxicity, 2 pts had normal
    DPD activity
  • Weak negative correlation b/w global toxicity
    score DPD activity
  • IVS141GgtA variant found in only 2 pts (2.2),
    both with ?? toxicity

DPD Activity and Toxicity in 131 Patients Treated
with 5-FU
Magné et al. Br J of Clin Pharm, 2007.
12
Should We Test?
  • Screening for genetic mutations alone is not
    likely to effectively identify pts susceptible to
    lethal 5-FU toxicity
  • Genotype alone does not predict which pts will
    develop toxicity
  • Of Caucasians with DPD deficiency, only about 50
    have the most common IVS141GgtA variant
  • Peripheral blood testing for DPD activity in
    mononuclear cells is difficult and not conducive
    to wide use
  • Evidence of a circadian rhythm affecting DPD
    levels
  • About 50 of patients with severe 5-FU-related
    toxicity are NOT DPD deficient!
  • DPD deficiency only partially explains
    5-FU-related toxicity
  • Hypermethylation of DPYD promoter is associated
    with down-regulation of DPD activity in patients
  • Work being done on a rapid noninvasive 2-¹³C
    uracil breath test to determine DPD activity

13
FDA Label Recommendations
  • FDA mentions DPD deficiency as a cause of severe
    toxicity
  • No specific recommendations about dose-reductions
    or discontinuing the drug
  • No recommendations about genetic testing

14
Irinotecan UGT1A1
15
Irinotecan Metabolism
Competition for substrate b/w Activation
inactivation pathways
SN-38 activation Required for anti- Tumor effect
? To bile
1000x more Active than Irinotecan
UGT1A1 inactivates SN-38 by glucuronidation via
UDP glucuronosyltransferases
16
Polymorphisms in the UGT1A1 gene/promoter
  • 60 different genetic variations in UGT1A1 gene
    and promoter
  • Familial forms of hyperbilirubinemia due to
    UGT1A1 polymorphism
  • Crigler-Najjar type 1
  • Gilberts syndrome (3-10 of population)
  • Some genotypes associated with irinotecan
    toxicity
  • Promoter region of UGT1A128
  • Coding regions of UGT1A16, UGT1A127, UGT1A129,
    UGT1A17
  • Ethnic variation
  • Caucasians African Americans ? variation in the
    promoter region
  • Asians ? missense mutations in the coding region

17
UGT1A1
UGT1A1 promoter Contains TA repeats
Wild type has 6 TA repeats
7 TA repeats ? UGT1A1 enzyme expression
Glucuronidation makes SN-38 more soluble
allows it to be eliminated in bile
? inactivation of SN-38
Nature Reviews Cancer, 2001
18
Is the Presence of a UGT1A1 Polymorphism
Predictive of Toxicity?
  • Case-control, retrospective study by Ando et al
  • Presence of UGT1A1 polymorphisms evaluated in
    Japanese pts who previously received irinotecan
  • Severe toxicity grade 4 leukopenia and/or grade
    3 or 4 diarrhea
  • Of the 26 with severe toxicity
  • 15 homozygous for UGT1A128
  • 31 heterozygous for UGT1A128
  • Of the 92 without severe toxicity
  • 3 homozygous for UGT1A128
  • 11 heterozygous for UGT1A128
  • Those with UGT1A128 were 7x more likely to
    encounter toxicity
  • The frequency of the UGT1A128 allele was
    3.5-fold higher in those pts with grade 4
    leukopenia and/or grade 3 or 4 diarrhea

19
UGT1A128 Polymorphism Associated with Decreased
Glucuronidation of SN-38
  • Prospective study of 20 Caucasian pts treated
    with irinotecan for solid tumor malignancies
  • UGT1A128 polymorphism
  • 45 ? WT (TA6/TA6)
  • 35 ? Heterozygous (TA6/TA7)
  • 20 ? Homozygous (TA7/TA7)
  • Hetero / homozygous pts had ? glucuronidation of
    SN-38 ? SN-38 levels (3.9-fold lower)
  • Hetero / homozygous pts had ? grades of diarrhea
    neutro-penia (2.5-fold lower ANC post-tx)

Iyer, L et al. The Pharmacogenetics Journal, 2002.
20
Does UGT1A1 Genotyping Improve Outcomes?
  • Evidence-based review of outcomes in CRC pts
    treated with irinotecan found
  • Genetic testing has high sensitivity /specificity
    for the common genotypes
  • No prospective studies examining rate of adverse
    events in pts given dose reduced irinotecan
  • No direct evidence to show that dose
    modifications based on UGT1A1 genotype benefits
    or harms CRC pts
  • Homozygous UGT1A128 pts had improved survival
    statistically significant higher tumor response
    rates

21
FDA Label Recommendations
  • Individuals who are homozygous for the UGT1A128
    allele are at increased risk for neutropenia
  • FDA recommends a reduction in the starting dose
    in pts known to be homozygous for UGT1A128
  • Precise dose reduction is not known and should
    be considered based on individual patient
    tolerance to treatment
  • No recommendations for dose-reductions for
    hetero-zygous patients

22
Mercaptopurine TPMT
23
Thiopurines Thiopurine Methyltransferase (TPMT)
  • Thiopurines are purine analogs
  • Includes mercaptopurine (6-MP), thioguanine,
    and azathioprine
  • 6-MP has been used in the treatment of childhood
    ALL
  • This class of drugs is now used more for
    autoimmune disorders to prevent organ rejection
  • TPMT inactivates thiopurines

24
6-MP Metabolism
MP is converted to (TGNs) thioguanine
nucleotides then incorporated into DNA
TPMT methylates MP prevents TGN formation
excessive drug toxicity
Antileukemic effects myelosupression
Nature Reviews Cancer, 2001
25
TPMT Polymorphisms Lead to Reduced Enzyme Activity
  • Variability between individuals in erythrocyte
    TPMT activity is a heritable autosomal
    co-dominate trait
  • SNPs ? amino acid substitutions ? TPMT enzyme is
    ubiquitylated destroyed
  • 8 TPMT alleles have been identified
  • TPMT2
  • TPMT3A
  • TPMT3C
  • As the of mutant alleles ?, TPMT activity in
    erythrocytes and leukemic blasts ? TGN
    concentration ?
  • Homozygous --gt ?? TPMT activity ?? TGN
  • Heterozygous ? ? TPMT activity ? TGN
  • Wild-type ? Normal TPMT activity TGN

These 3 account for gt95 of the
clinically- significant mutations resulting in
low or intermediate enzyme activity
26
Influence of Ethnicity on TPMT Mutation Frequency
Deeken, J et al. Anti-cancer drugs, 2007.
  • Frequency of reduced TPMT activity in
    Caucasians
  • 86.6 have high TPMT activity
  • 11.1 have intermediate activity
  • 0.3 have deficient TPMT activity

27
TPMT Polymorphisms Affect 6-MP Therapeutic
Efficacy Toxicity
  • TPMT Polymorphism therapeutic efficacy
  • Children with median TGN concentration less than
    the population median had worse RFS compared to
    those with concentration greater than the median
  • Patients with at least 1 mutant TPMT allele tend
    to have improved response to 6-MP compared to pts
    with both WT alleles
  • TPMT Polymorphism risk of toxicity
  • Reports of toxicity when full-dose 6-MP given to
    homozygous pts
  • As TGN ?, risk of leukopenia ? risk of
    relapse ?
  • Dose reductions in homozygous ALL pts ? ?TGN
    ? risk of relapse
  • TPMT genotype may influence risk of secondary
    malignancies (brain tumors AML)

28
TPMT Genotype Correlates Well With Phenotype
(enzyme activity)
TPMT phenotypes in thiopurine-intolerant
patients compared with the general population
  • Study of 23 children with ALL intolerant to 6-MP
  • 100 concordance b/w genotype phenotype in the
    homozygous deficient WT patients
  • Genotyping for common TPMT alleles can identify
    pts at risk for severe 6-MP toxicity
  • Dose reductions enabled re-initiation of therapy
  • Median dose? homozygous ? 90.8 heterozygous ?
    67

6-fold overrepresentation of TPMT- Deficient
patients heterozygotes in the
thiopurine-intolerant Group ( Plt0.001)
Evans, WE. J Clin Oncol, 2001.
29
How Do We Best Test for Variability in TPMT
Activity?
  • Measurement of TPMT activity RBC TGN levels is
    difficult
  • Labor-intensive assays
  • Laboratory-related variability
  • Multiple studies have shown correlation between
    TPMT genotype phenotype
  • Type of polymorphism number of affected alleles
    could help to guide dose reductions

30
FDA Label Recommendations
  • TPMT genotyping or phenotyping suggested for
    patients with severe toxicity
  • Dose reduction recommendations
  • Substantial dose reductions may be needed for
    homozygous patients to avoid life threatening
    bone marrow suppression
  • Specific dose adjustment not recommended
  • No dose adjustments recommended for heterozygous
    patients

31
Tamoxifen CYP2D6
32
CYP2D6 Polymorphisms and Tamoxifen
  • Tamoxifen is a prodrug that is converted to its
    active metabolites endoxifen and 4-OH-TAM
  • 4-OH-TAM is 30x more active
  • Endoxifen is 100x more active
  • CYP2D6 metabolizes Tamoxifen to endoxifen
  • Over 80 genetic variants of CYP2D6 have been
    described
  • Polymorphisms of CYP2D6 may produce a less active
    enzyme
  • CYP2D6 polymorphisms may affect response to
    tamoxifen

Dezentjé, V. Clinical Cancer Research, 2009.
33
Does CYP2D6 Polymorphism Affect Tamoxifen
Metabolism?
  • CYP2D6 polymorphisms affect drug metabolism
  • Based on mutation, enzyme activity can be normal
    to absent
  • Labeled as poor, intermediate, extensive, or
    ultrarapid metabolizer
  • Number of inactive alleles affects metobolism
  • Homozygous carriers (Ex 4/4) are poor
    metabolizers
  • Heterozygous carriers (Ex 1/4) are either
    intermediate or extensive metabolizers
  • There is concern that decreased CYP2D6 activity
    will lead to undertreatment with Tamoxifen

34
CYP2D6 Genetic Variants
Dezentjé, V. Clinical Cancer Research, 2009.
35
CYP2D6 Genotype Risk of Breast Cancer Recurrence
Dezentjé, V. Clinical Cancer Research, 2009.
36
How Do We Interpret These Conflicting Data?
  • 6 studies were evaluated in mainly Caucasian
    breast cancer population
  • 3 studies ? link b/w CYP2D6 genotype ? risk of
    recurrence
  • 2 studies ? no link b/w CYP2D6 genotype
    tamoxifen efficacy
  • 1 study ? link b/w CYP2D6 genotype ? risk for
    recurrence
  • Confounders
  • Heterogeneous populations of pts
  • Tumor grade, Her-2 status, pre/postmenopausal,
    tamoxifen dose tx length
  • Where to analyze data from heterozygous pts (w/
    WT or homozygous pts)?
  • No control for CYP2D6 inhibitors (SSRIs used in
    up to 30 of pts)
  • ? compliance in extensive metabolizers from side
    effects

37
Conclusions
  • Screening for these polymorphisms prior to
    initiating treatment is not recommended
  • Screening for DPD, UGT1A1, and TPMT polymorphisms
    can be done in the setting of severe toxicity
  • Most cancer pts are treated with a
    trial-and-error approach to adjustments in
    dosage
  • Most patients with severe toxicity will require
    dose adjust-ments regardless of presence /
    absence of polymorphism

38
Conclusions
  • Need more prospective studies examining affect of
    dose adjustments on patients with polymorphisms
    that alter drug metabolism
  • Does dose-reduction improve outcomes in patients
    with polymorphisms?
  • Delicate balance between decreasing toxicity with
    dose-reduction and increasing risk of relapse
  • As our understanding of the relationships between
    genotype phenotype increases, we may be able to
    treat our patients with a more tailored approach

39
Resources
  • Wei, X. et al. Molecular Basis of the Human
    Dihydropyrimidine Dehydrogenase Deficiency and
    5-Fluorouracil Toxicity. The Journal of Clinical
    Investigation 1996, 98 610-615.
  • Ando, Y et al. Polymorphisms of
    UDP-Glucuronosyltransferase Gene and Irinotecan
    Toxicity A Pharmacogenetic Analysis. Cancer
    Research 2000, 60 6621-6926.
  • Iyer, L. et al. UGT1A128 polymorphism as a
    determinant of irinotecan disposition and
    toxicity. The Pharmacogenomics Journal 2002, 2
    43-47.
  • Innocenti, F. et al. Genetic Variants in the
    UDP-glucuronosyltransferase 1A1 Gene Predict the
    Risk of Severe Neutropenia of Irinotecan.
    Journal of Clinical Oncology 2004, 22 1382-88.
  • Dezentjé, V. et al. Clinical Implications of
    CYP2D6 enotyping in Tamoxifen Treatment for
    Breast Cancer. Clinical Cancer Research 2009,
    15 15-21.
  • Magné, N. et al. Dihydropyrimidine dehydrogenase
    activity and the IVS141GgtA mutation in patients
    developing 5FU-related toxicity. British Journal
    of Clinical Pharmacology 2007, 64 237-240.
  • Mattison, L.K. et al. The uracil breath test in
    the assessment of dihydropyrimidine dehydrogenase
    activity pharmacokinetic relationship between
    expired 13CO2 and plasma 2-13Cdihydrouracil.
    Clinical Cancer Research 2006, 15 549-55.
  • Mattison, LK. Increased prevalence of
    dihydropyrimidine dehydrogenase deficiency in
    African-Americans compared with Caucasians.
    Clinical Cancer Research 2006, 12 5491-5.
  • Evans, W.E. Preponderance of Thiopurine
    S-Methyltransferase Deficiency and Heterozygosity
    Among Patients Intolerant to Mercaptopurine or
    Azathioprine. J of Clinical Oncology 2001, 19
    2293-2301.
  • Relling, MV et al. Pharmacogenetics and Cancer
    Therapy. Nature Reviews Cancer 2001, 1
    99-108.
  • Maitland, ML et al. TPMT, UGT1A1, and DPYD
    genotyping to ensure safer cancer therapy?
    Trends in Pharmacological Sciences 2006, 27
    432-437.
  • Deeken, JF et al. Toward Individualized
    Treatment Prediction of Anticancer Drug
    Disposition and Toxicity with Pharmacogenetics.
    Anti-Cancer Drugs 2007, 18 111-126.

40
(No Transcript)
Write a Comment
User Comments (0)
About PowerShow.com