10.06.2013 Views

Issue 4 - August 2010 - Pacini Editore

Issue 4 - August 2010 - Pacini Editore

Issue 4 - August 2010 - Pacini Editore

SHOW MORE
SHOW LESS

You also want an ePaper? Increase the reach of your titles

YUMPU automatically turns print PDFs into web optimized ePapers that Google loves.

162<br />

15 Vlassenbroeck I, Califice S, Diserens AC, et al. Validation of realtime<br />

methylation-specific PCR to determine O6-methylguanine-DNA<br />

methyltransferase gene promoter methylation in glioma. J Mol Diagn<br />

2008;10(4):332-7.<br />

16 Roversi G, Pfundt R, Moroni RF, et al. Identification of novel genomic<br />

markers related to progression to glioblastoma through genomic profiling<br />

of 25 primary glioma cell lines. Oncogene 2006;25(10):1571-<br />

83.<br />

17 Yin D, Ogawa S, Kawamata N, et al. High-resolution genomic copy<br />

number profiling of glioblastoma multiforme by single nucleotide<br />

polymorphism DNA microarray. Mol Cancer Res 2009;7(5):665-77.<br />

18 von Deimling A, Louis DN, von Ammon K, et al. Association of epidermal<br />

growth factor receptor gene amplification with loss of chromosome<br />

10 in human glioblastoma multiforme. J Neurosurg 1992;77:295-<br />

301.<br />

19 Freire P, Vilela M, Deus H, et al. Exploratory analysis of the copy<br />

number alterations in glioblastoma multiforme. PLoS One 2008;3(12):<br />

e4076.<br />

20 Lo KC, Bailey D, Burkhardt T, et al. Comprehensive analysis of loss<br />

of heterozygosity events in glioblastoma using the 100K SNP mapping<br />

arrays and comparison with copy number abnormalities defined by<br />

BAC array comparative genomic hybridization. Genes Chromosomes<br />

Cancer 2008;47(3):221-37.<br />

21 Vlassenbroeck I, Califice S, Diserens AC, et al. Validation of realtime<br />

methylation-specific PCR to determine O6-methylguanine-DNA<br />

methyltransferase gene promoter methylation in glioma. J Mol Diagn<br />

2008;10(4):332-7.<br />

22 Brandes AA, Tosoni A, Franceschi E, et al. Recurrence pattern after<br />

temozolomide concomitant with and adjuvant to radiotherapy in newly<br />

diagnosed patients with glioblastoma: correlation With MGMT promoter<br />

methylation status. J Clin Oncol 2009;27(8):1275-9.<br />

23 Morandi L, Franceschi E, De Biase D, et al. Promoter methylation<br />

analysis of O6-methylguanine-DNA methyltransferase in glioblastoma:<br />

detection by locked nucleic acid based quantitative PCR<br />

using an imprinted gene (SNURF) as a reference. BMC Cancer<br />

<strong>2010</strong>;10:48<br />

24 Brandes AA, Franceschi E, Tosoni A, et al. O(6)-methylguanine DNAmethyltransferase<br />

methylation status can change between first surgery<br />

for newly diagnosed glioblastoma and second surgery for recurrence:<br />

clinical implications. Neuro Oncol <strong>2010</strong>;12(3):283-8.<br />

Pharmacogenetics in hematologic neoplasia<br />

S. Rasi, A. Bruscaggin, S. Franceschetti, R. Bruna, D. Rossi,<br />

G. Gaidano<br />

Division of Hematology, Department of Clinical and Experimental<br />

Medicine, Amedeo avogadro University of Eastern Piedmont, 28100<br />

Novara<br />

The field of pharmacogenetics investigates how genetic inheritance<br />

might influence the patients’ individual response to<br />

drugs. In fact, the majority of drugs exhibit large interindividual<br />

variability in their efficacy and toxicity, and this individual<br />

host response is influenced by genetic polymorphisms,<br />

in particular single nucleotide polymorphisms (SNPs).<br />

The main aim of pharmacogenetics is to optimize therapy<br />

based on the patient’s genotype, in order to maximize the<br />

therapeutic index of a given drug or regimen. Indeed, several<br />

pharmacogenetic studies have documented that host SNPs<br />

affecting genes involved in drug metabolism, detoxification,<br />

transport and targeting are in fact responsible, at least in part,<br />

for the interindividual variability in efficacy and toxicity of<br />

a given pharmacological treatment. Moreover, several drugs<br />

utilized in therapeutic treatment of hematologic neoplasms<br />

rely on DNA damage as part of their mechanisms of tumor<br />

cell killing. On these bases, treatment benefit and/or toxicities<br />

in patients may be modulated by the host DNA repair capacity.<br />

Also in this context, pharmacogenetic studies have shown<br />

that SNPs within genes of DNA repair pathways alter the host<br />

DNA repair capacity, thus affecting the individual response to<br />

drugs and the prognosis of the patients.<br />

5 th triennial congress of the italian society of anatomic Pathology and diagnostic cytoPathology<br />

One of the most consolidated pharmacogenetic model in<br />

hematological-oncology is represented by the case of acute<br />

lymphoblastic leukemia (ALL) of childhood. Among hematological<br />

neoplasms, ALL of childhood is the sole disease<br />

whose pharmacogenetics has been characterized in detail,<br />

and has entered clinical practice. The enzyme thiopurine Smethyltransferase<br />

(TPMT), classified as a phase II enzyme,<br />

metabolizes chemotherapeutic agents, and in particular is<br />

responsible for the metabolism of 6-mercaptopurine. The<br />

activity of the TPMT enzyme displays marked variability in<br />

the population, being influenced by SNPs of the TPMT gene<br />

that determine a reduction of the cellular content of TPMT.<br />

In particular, three allelic variants (TPMT*2, TPMT*3A,<br />

and TPMT*3C) are responsible for more than 90% of cases<br />

with an intermediate- or low-enzyme activity. ALL patients<br />

with a wild-type TPMT allele (TPMT*1 or TPMT*1S) and<br />

a non functional variant allele (TPMT*2, TPMT*3A, and<br />

TPMT*3C) have an intermediate activity of TPMT, while<br />

patients with two non functional variant alleles are TPMT<br />

deficient. In the context of ALL of childhood, the TPMT<br />

genotype identifies patients who are at risk of hematopoietic<br />

toxicity after thiopurine therapy. In fact, patients who are<br />

homozygous carriers of these SNPs and are therefore devoid<br />

of TPMT activity, are at higher risk of hematological toxicity<br />

when treated with 6-mercaptopurine. Instead, patients who<br />

are heterozygous carriers of these SNPs have an intermediate<br />

risk of dose-limiting toxicity. A 6-mercaptopurine dose reduction<br />

of 90% is generally required for homozygous-TPMT<br />

deficiency patients, whereas the TPMT heterozygous carriers<br />

require a mean dose reduction only of 35%.<br />

Instead, scant information is available about the impact of<br />

pharmacogenetics in predicting outcome and toxicity in the<br />

context of non-Hodgkin lymphoma (NHL) and available information<br />

is restricted to a limited number of studies. Studies<br />

in follicular lymphoma and diffuse large B cell lymphoma<br />

(DLBCL) aimed at assessing the association between antilymphoma<br />

efficacy of rituximab and SNPs of Fcγ receptors<br />

have reported conflicting results. Fcγ receptors are are involved<br />

in rituximab antibody-dependent cellular toxicity. A<br />

few studies have identified SNPs located in the Fcγ receptors<br />

(FcγRIIIa 158V/F and FcγRIIa 131H/R) as being associated<br />

with tumor response in patients treated with rituximab as<br />

first-line therapy. On the other hand, both in the context of<br />

follicular lymphoma and in DLBCL, other studies failed to<br />

identify an association between prognosis and SNPs of Fcγ<br />

receptors.<br />

In the context of DLBCL, the host pharmacogenetic background<br />

predicts efficacy of R-CHOP21 (R=rituximab,<br />

C=cyclophosphamide, H=doxorubicin, O=vincristine and<br />

P=prednisone) chemotherapy program, that is considered the<br />

standard treatment for this disease. In fact, SNPs modulating<br />

gene expression and/or function of enzymes involved<br />

in R-CHOP pharmacogenetics may contribute to prognostic<br />

stratification and prediction of toxicity in DLBCL patients<br />

treated with R-CHOP21. In particular, host SNPs affecting<br />

alkylating agent detoxification (GSTA1 rs3957357) and<br />

doxorubicin pharmacodynamics (CYBA rs4673) may predict<br />

survival in DLBCL treated with R-CHOP21. GSTA1 encodes<br />

a glutathione S-transferase that catalyses the conjugation of<br />

cyclophosphamide with glutathione to facilitate excretion.<br />

In particular, the GSTA1 rs3957357T minor allele associates<br />

with reduced levels of GSTA1 enzyme in healthy individuals,<br />

and predicts for reduced detoxification of alkylating agents,<br />

thus increasing tumor cell exposure to drug. In fact, DL-<br />

BCL patients carrying GSTA1 rs3957357 CT/TT genotypes

Hooray! Your file is uploaded and ready to be published.

Saved successfully!

Ooh no, something went wrong!