1
|
França GDA, Nardinelli L, Giorgi RR, Pagliarini T, Guimarães Baiocchi OCC, Velloso EDRP, Jr WFDS, Magalhães Rego E, Bendit I. Assessing the genetic profile of cytochrome P450 and glutathione S-transferases of patients diagnosed with acute myeloid leukemia. Hematol Transfus Cell Ther 2025; 47:103759. [PMID: 40339533 DOI: 10.1016/j.htct.2025.103759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/27/2024] [Accepted: 12/13/2024] [Indexed: 05/10/2025] Open
Abstract
OBJECTIVE This study aimed to determine the frequency of genetic alterations as deletions and duplications in cytochrome P450 (CYP450) and glutathione S-transferases (GST) genes, as well as to investigate whether there is a relationship between these alterations and neutrophilic hematologic recovery in adult patients diagnosed with acute myeloid leukemia. METHOD DNA samples from 70 patients diagnosed with acute myeloid leukemia were evaluated using the Multiplex Ligation-dependent Probe Amplification technique. The presence or absence of polymorphisms was compared regarding the time to neutrophilic recovery (neutrophil count ≥1.0 × 109/L) using Kaplan-Meier curves, with the comparison between the curves being performed using the non-parametric log-rank test. RESULTS The median age of the participants was 57 years, with a higher proportion of females (57.2%) and white individuals (61.4%)'. A total of 76 polymorphisms (CYP450 + GST) were identified, comprising 38 deletions and 38 duplications. Kaplan-Meier curves revealed that the neutrophilic recovery time was longer for the group with polymorphisms (p-value = 0.0056). CONCLUSION The study demonstrated that CYP450 and GST genes are polymorphic, and these polymorphisms may lead to longer neutrophilic recovery after induction treatment of acute myeloid leukemia remission.
Collapse
Affiliation(s)
- Gilmar de Andrade França
- Department of Hematology, Transfusion and Cell Therapy, University of Sao Paulo Medical School (HCFMUSP), Sao Paulo, SP, Brazil; Laboratory of Medical Investigation in Pathogenesis and targeted therapy in Onco-Immuno-Hematology (LIM/31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Luciana Nardinelli
- Department of Hematology, Transfusion and Cell Therapy, University of Sao Paulo Medical School (HCFMUSP), Sao Paulo, SP, Brazil; Laboratory of Medical Investigation in Pathogenesis and targeted therapy in Onco-Immuno-Hematology (LIM/31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Ricardo Rodrigues Giorgi
- Laboratory of Medical Investigation in Pathogenesis and targeted therapy in Onco-Immuno-Hematology (LIM/31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Thiago Pagliarini
- Laboratory of Medical Investigation in Pathogenesis and targeted therapy in Onco-Immuno-Hematology (LIM/31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | | | - Elvira Deolinda Rodrigues Pereira Velloso
- Department of Hematology, Transfusion and Cell Therapy, University of Sao Paulo Medical School (HCFMUSP), Sao Paulo, SP, Brazil; Laboratory of Medical Investigation in Pathogenesis and targeted therapy in Onco-Immuno-Hematology (LIM/31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil; Department of Hematology, Cancer Institute of Sao Paulo, University of Sao Paulo Medical School (ICESP), Sao Paulo, SP, Brazil
| | - Wellington Fernandes da Silva Jr
- Department of Hematology, Transfusion and Cell Therapy, University of Sao Paulo Medical School (HCFMUSP), Sao Paulo, SP, Brazil; Laboratory of Medical Investigation in Pathogenesis and targeted therapy in Onco-Immuno-Hematology (LIM/31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil; Department of Hematology, Cancer Institute of Sao Paulo, University of Sao Paulo Medical School (ICESP), Sao Paulo, SP, Brazil
| | - Eduardo Magalhães Rego
- Department of Hematology, Transfusion and Cell Therapy, University of Sao Paulo Medical School (HCFMUSP), Sao Paulo, SP, Brazil; Laboratory of Medical Investigation in Pathogenesis and targeted therapy in Onco-Immuno-Hematology (LIM/31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil; Department of Hematology, Cancer Institute of Sao Paulo, University of Sao Paulo Medical School (ICESP), Sao Paulo, SP, Brazil
| | - Israel Bendit
- Department of Hematology, Transfusion and Cell Therapy, University of Sao Paulo Medical School (HCFMUSP), Sao Paulo, SP, Brazil; Laboratory of Medical Investigation in Pathogenesis and targeted therapy in Onco-Immuno-Hematology (LIM/31), Department of Hematology, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
2
|
Baranwal A, Hahn CN, Shah MV, Hiwase DK. Role of Germline Predisposition to Therapy-Related Myeloid Neoplasms. Curr Hematol Malig Rep 2022; 17:254-265. [PMID: 35986863 DOI: 10.1007/s11899-022-00676-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2022] [Indexed: 01/27/2023]
Abstract
PURPOSE OF REVIEW Therapy-related myeloid neoplasms (t-MNs) are aggressive leukemias that develop following exposure to DNA-damaging agents. A subset of patients developing t-MN may have an inherited susceptibility to develop myeloid neoplasia. Herein, we review studies reporting t-MN and their association with a germline or inherited predisposition. RECENT FINDINGS Emerging evidence suggests that development of t-MN is the result of complex interactions including generation of somatic variants in hematopoietic stem cells and/or clonal selection pressure exerted by the DNA-damaging agents, and immune evasion on top of any inherited genetic susceptibility. Conventionally, alkylating agents, topoisomerase inhibitors, and radiation have been associated with t-MN. Recently, newer modalities including poly (ADP-ribose) polymerase inhibitors (PARPi) and peptide receptor radionucleotide therapy (PRRT) are associated with t-MN. At the same time, the role of pathogenic germline variants (PGVs) in genes such as BRCA1/2, BARD1, or TP53 on the risk of t-MN is being explored. Moreover, studies have shown that while cytotoxic therapy increases the risk of developing myeloid neoplasia, it may be exposing the vulnerability of an underlying germline predisposition. t-MN remains a disease with poor prognosis. Studies are needed to better define an individual's inherited neoplastic susceptibility which will help predict the risk of myeloid neoplasia in the future. Understanding the genes driving the inherited neoplastic susceptibility will lead to better patient- and cancer-specific management including choice of therapeutic regimen to prevent, or at least delay, development of myeloid neoplasia after treatment of a prior malignancy.
Collapse
Affiliation(s)
- Anmol Baranwal
- Division of Hematology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55906, USA
| | - Christopher N Hahn
- Department of Genetics and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Mithun Vinod Shah
- Division of Hematology, Mayo Clinic, 200 1st Street SW, Rochester, MN, 55906, USA.
| | - Devendra K Hiwase
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia.
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia.
| |
Collapse
|
3
|
Keren-Froim N, Heering G, Sharvit G, Zlotnik M, Nagler A, Shimoni A, Avigdor A, Canaani J. ELN 2017 classification significantly impacts the risk of early death in acute myeloid leukemia patients receiving intensive induction chemotherapy. Ann Hematol 2022; 101:309-316. [DOI: 10.1007/s00277-021-04716-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 10/31/2021] [Indexed: 11/29/2022]
|
4
|
Megías-Vericat JE, Martínez-Cuadrón D, Herrero MJ, Rodríguez-Veiga R, Solana-Altabella A, Boluda B, Ballesta-López O, Cano I, Acuña-Cruz E, Cervera J, Poveda JL, Sanz MÁ, Aliño SF, Montesinos P. Influence of polymorphisms in anthracyclines metabolism genes in the standard induction chemotherapy of acute myeloid leukemia. Pharmacogenet Genomics 2021; 31:133-139. [PMID: 33675324 DOI: 10.1097/fpc.0000000000000431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Genetic variability in anthracycline metabolism could modify the response and safety of acute myeloid leukemia (AML) induction. METHODS Polymorphisms in genes that encodes enzymes of anthracyclines metabolic pathway (CBR3: rs1056892, rs8133052, NQO1: rs1800566, NQO2: rs1143684, NOS3: rs1799983, rs2070744) were evaluated in 225 adult de novo AML patients. RESULTS The variant CBR3 rs8133052 was associated with lower hepatotoxicity (P = 0.028). Wild-type genotype of NQO2 rs1143684 was related to higher complete remission (P = 0.014), and the variant allele with greater gastrointestinal toxicity (P = 0.024). However, the variant genotype of NQO1 rs1800566 was associated with mucositis (P = 0.018), but heterozygous genotype showed less gastrointestinal toxicity (P = 0.028) and thrombocytopenia (P = 0.009). Protective effects against nephrotoxicity and thrombocytopenia were reported with variant NOS3 rs1799983 (P = 0.006, P = 0.014), whereas carriers of NOS3 rs2070744 showed higher hepatotoxicity and thrombocytopenia (P = 0.017, P = 0.013). CONCLUSIONS This study supports the influence of genetic variability of idarubicin metabolizing could be critical in predicting anthracycline-induced toxicities.
Collapse
Affiliation(s)
- Juan Eduardo Megías-Vericat
- Grupo de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento
- Servicio de Farmacia, Área del Medicamento
| | - David Martínez-Cuadrón
- Servicio de Hematología y Hemoterapia, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia
- CIBERONC, Instituto Carlos III, Madrid
| | - María José Herrero
- Grupo de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento
- Departamento Farmacología, Facultad de Medicina, Universidad de Valencia, Av. Blasco Ibáñez 15
| | - Rebeca Rodríguez-Veiga
- Servicio de Hematología y Hemoterapia, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia
- CIBERONC, Instituto Carlos III, Madrid
| | | | - Blanca Boluda
- Servicio de Hematología y Hemoterapia, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia
- CIBERONC, Instituto Carlos III, Madrid
| | | | - Isabel Cano
- Servicio de Hematología y Hemoterapia, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia
- CIBERONC, Instituto Carlos III, Madrid
| | - Evelyn Acuña-Cruz
- Servicio de Hematología y Hemoterapia, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia
- CIBERONC, Instituto Carlos III, Madrid
| | - José Cervera
- Servicio de Hematología y Hemoterapia, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia
- CIBERONC, Instituto Carlos III, Madrid
| | | | - Miguel Ángel Sanz
- Servicio de Hematología y Hemoterapia, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia
- CIBERONC, Instituto Carlos III, Madrid
| | - Salvador F Aliño
- Grupo de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento
- Departamento Farmacología, Facultad de Medicina, Universidad de Valencia, Av. Blasco Ibáñez 15
- Unidad de Farmacología Clínica, Área del Medicamento, Hospital Universitari I Politècnic La Fe, Av. Fernando Abril Martorell, Valencia, Spain
| | - Pau Montesinos
- Servicio de Hematología y Hemoterapia, Hospital Universitari i Politècnic La Fe. Av. Fernando Abril Martorell, Valencia
- CIBERONC, Instituto Carlos III, Madrid
| |
Collapse
|
5
|
Sophonnithiprasert T, Saelee P, Pongtheerat T. GSTM1 and GSTT1 copy number variants and the risk to Thai females of hepatocellular carcinoma. J Gastrointest Oncol 2019; 10:324-329. [PMID: 31032101 DOI: 10.21037/jgo.2018.09.14] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a common malignancy found throughout the world that most often occurs in males. The cancer is associated with many risk factors such as viral infection, cirrhosis, alcohol, smoking, and fungal toxins. GSTM1 and GSTT1 are detoxification enzymes activated by the cleansing of carcinogenic compounds. Low DNA copy numbers of Glutathione S-transferases M1 and T1 result in a loss of enzyme activity, which causes carcinogenesis factors. DNA copy number variants (CNVs) were determined to compare the differences between the frequencies of GSTM1 and GSTT1 in a control group and patients. Then, the association of these genes with the pathological/survival status of HCC patients was investigated. Methods Forty-nine Thai HCC patients' DNA and the genomic DNA of 66 healthy controls were investigated for GSTM1 and GSTT1 CNVs by real-time polymerase chain reaction (PCR). Then, the correlations between GSTM1 and GSTT1 patients' CNVs, the control group, and clinico-pathological parameters were determined. Results The results show that were no differences between the CNVs of GSTM1 and GSTT1 in the controls and patients (P≥0.05). Only GSTT1 genotypes 0/0 correlated to an increase in the risk of hepatocellular carcinogenesis (OR value was 1.88). GSTM1 CNVs were associated with the gender of patients (P=0.002). However, no correlations were found between GSTT1 CNVs and any of the clinico-pathological parameters. Conclusions The results suggest that only GSTT1 CNVs are associated with increased risk factors of HCC in Thais. GSTM1 copy numbers had a dominant correlation with female HCC patients.
Collapse
Affiliation(s)
- Thanet Sophonnithiprasert
- Unit of Biochemistry, Department of Medical Sciences, Faculty of Science, Rangsit University, Patumthani, Thailand
| | - Pensri Saelee
- Research Division, National Cancer Institute, Bangkok, Thailand
| | - Tanett Pongtheerat
- Unit of Biochemistry, Department of Medical Sciences, Faculty of Science, Rangsit University, Patumthani, Thailand
| |
Collapse
|
6
|
Megías-Vericat JE, Montesinos P, Herrero MJ, Bosó V, Martínez-Cuadrón D, Poveda JL, Sanz MÁ, Aliño SF. Pharmacogenomics and the treatment of acute myeloid leukemia. Pharmacogenomics 2016; 17:1245-1272. [DOI: 10.2217/pgs-2016-0055] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Acute myeloid leukemia (AML) is a clinically and biologically heterogeneous malignancy that is primarily treated with combinations of cytarabine and anthracyclines. Although this scheme remains effective in most of the patients, variability of outcomes in patients has been partly related with their genetic variability. Several pharmacogenetic studies have analyzed the impact of polymorphisms in genes encoding transporters, metabolizers or molecular targets of chemotherapy agents. A systematic review on all eligible studies was carried out in order to estimate the effect of polymorphisms of anthracyclines and cytarabine pathways on efficacy and toxicity of AML treatment. Other emerging genes recently studied in AML, such as DNA repair genes, genes potentially related to chemotherapy response or AML prognosis, have also been included.
Collapse
Affiliation(s)
- Juan Eduardo Megías-Vericat
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
- Servicio de Farmacia, Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - Pau Montesinos
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - María José Herrero
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
- Departamento Farmacología, Facultad de Medicina, Universidad de Valencia, Avda, Blasco Ibáñez 15, 46010 – Valencia, Spain
| | - Virginia Bosó
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
- Servicio de Farmacia, Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - David Martínez-Cuadrón
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - José Luis Poveda
- Servicio de Farmacia, Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - Miguel Ángel Sanz
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
| | - Salvador F Aliño
- Unidad de Farmacogenética, Instituto Investigación Sanitaria La Fe and Área del Medicamento, Hospital Universitario y Politécnico La Fe Avda, Fernando Abril Martorell 106, 46026 – Valencia, Spain
- Departamento Farmacología, Facultad de Medicina, Universidad de Valencia, Avda, Blasco Ibáñez 15, 46010 – Valencia, Spain
- Unidad de Farmacología Clínica, Área del Medicamento, Hospital Universitario y Politécnico La Fe. Avda. Fernando Abril Martorell 106, 46026 – Valencia, Spain
| |
Collapse
|
7
|
GSTT1 and GSTM1 polymorphisms predict treatment outcome for breast cancer: a systematic review and meta-analysis. Tumour Biol 2015; 37:151-62. [PMID: 26577857 DOI: 10.1007/s13277-015-4401-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/05/2015] [Indexed: 02/07/2023] Open
Abstract
Observational studies have reported controversial results on the association between GSTT1 and GSTM1 genotypes and treatment outcome of breast cancer. The purpose of this study is to evaluate the association between GSTT1 and GSTM1 and treatment outcome in breast cancer patients. Eligible studies were searched in PubMed, EMBASE, Cochrane Library, and China National Knowledge Infrastructure databases. A random-effect model or fixed-effect model was used to calculate the overall combined risk estimates. Twenty-one studies with a total of 4990 patients were included in this meta-analysis. The GSTM1 null genotype (odds ratio (OR) = 1.33, 95 % confidence interval (CI) 1.01-1.75, P = 0.046) and GSTT1/GSTM1 double null genotype (OR = 2.22, 95 % CI 1.02-4.84, P = 0.045) were significantly associated with an increased tumor response. A reduced overall survival (hazard ratio (HR) = 0.84, 95 % CI 0.72-0.98, P = 0.024) was observed in GSTM1 null genotype, especially in mixed descent (HR = 0.77, 95 % CI 0.61-0.96, P = 0.018) and large sample size (HR = 0.85, 95 % CI 0.72-0.99, P = 0.033). Evidence of publication bias was observed in GSTM1 genotype rather than in GSTT1 genotype. This meta-analysis suggests that GSTM1 null and GSTT1/GSTM1 double null polymorphisms might be significantly associated with an increased tumor response. However, the GSTM1 null genotype might be significantly associated with a reduced overall survival. Future studies are warranted to confirm these findings.
Collapse
|
8
|
Meyer SC, Levine RL. Translational implications of somatic genomics in acute myeloid leukaemia. Lancet Oncol 2014; 15:e382-94. [DOI: 10.1016/s1470-2045(14)70008-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
9
|
Xiao Q, Deng D, Li H, Ye F, Huang L, Zhang B, Ye B, Mo Z, Yang X, Liu Z. GSTT1 and GSTM1 polymorphisms predict treatment outcome for acute myeloid leukemia: a systematic review and meta-analysis. Ann Hematol 2014; 93:1381-90. [PMID: 24994605 DOI: 10.1007/s00277-014-2050-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2014] [Accepted: 03/03/2014] [Indexed: 01/01/2023]
Abstract
Glutathione S-transferases (GSTs) contribute to the metabolism of different xenobiotics and anticancer drugs and confer protection against oxidative stress thus may influence the treatment outcome of acute myeloid leukemia (AML). Studies regarding the association between GSTT1 and GSTM1 polymorphisms and treatment outcome in AML patients showed an inconsistent result. A systematic review and meta-analysis were performed to further explore this association. PubMed, Hartford User Group Exchange (HUGE), and China National Knowledge Infrastructure (CNKI) databases were searched for all related publications. Statistical analyses were analyzed by using RevMan 5.0 and Stata 9.0 softwares. A total of 1,837 patients in 11 studies were included. GSTT1 null genotype was found to be significantly associated with a reduced response after first course of induction chemotherapy (odds ratio (OR) = 0.894, 95 % confidence interval (CI) = 0.818-0.977, P = 0.013), progression-free survival (PFS; hazard ratio (HR) = 0.698, 95 % CI = 0.520-0.937, P = 0.017), and overall survival (OS; HR = 0.756, 95 % CI = 0.618-0.925, P = 0.007) in Asian population. GSTM1/GSTT1 double-null genotype was also identified to be significantly associated with response after the first course of induction chemotherapy (OR = 0.40, 95 % CI = 0.24-0.67, P = 0.0003). Our study suggested that GSTT1 null genotype and GSTT1/GSTM1 double-null genotype were associated with a worse treatment outcome for AML patients, especially in Asian population.
Collapse
Affiliation(s)
- Qiang Xiao
- Department of Hematology, The First Affiliated Hospital of Guangxi Medical University, Shuang Yong Rd. 6, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
|
11
|
Two minor NQO1 and NQO2 alleles predict poor response of breast cancer patients to adjuvant doxorubicin and cyclophosphamide therapy. Pharmacogenet Genomics 2012; 21:808-19. [PMID: 21946896 DOI: 10.1097/fpc.0b013e32834b6918] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE A SNP in the NQO1 gene has been implicated in the response of patients with breast cancer to anthracycline containing regimens. NQO1, and its homologue NQO2, share many substrates yet retain distinct functional differences, with NQO2 being a more permissive molecule for electron accepting substrates. We aimed to determine whether functional NQO2 variants are associated with altered response to adjuvant doxorubicin and cyclophosphamide therapy, with or without tamoxifen, in the treatment of breast cancer. METHODS Genomic DNA samples from 227 women with early breast cancer were genotyped for NQO1 and NQO2 polymorphisms. All participants were treated with an AC adjuvant therapy regimen. The functional implications of NQO2 polymorphisms were validated in in-vitro ectopic expression models. RESULTS The NQO1 SNP (rs1800566) was associated with a poorer outcome and a lower likelihood of having a treatment delay. Patients who had ER and PR negative disease and were wild type for both the NQO1 and an NQO2 SNP (rs1143684) had 100% 5-year overall survival compared with 88% for carriers of one minor allele and 70% for carriers of two or more minor alleles (P=0.018, log rank). Carriers of minor alleles of a triallelic NQO2 promoter polymorphism were more likely to be withdrawn from tamoxifen therapy prematurely due to intolerance (P=0.009, log rank). MCF-7 cells were sensitized to growth inhibition by doxorubicin and 4OH tamoxifen, but not cyclophosphamide, by ectopic expression of NQO2. CONCLUSION This study suggests that both NQO1 and NQO2 modulate the efficacy of AC therapy and that NQO2 is associated with tamoxifen toxicity.
Collapse
|
12
|
Maranville JC, Luca F, Richards AL, Wen X, Witonsky DB, Baxter S, Stephens M, Di Rienzo A. Interactions between glucocorticoid treatment and cis-regulatory polymorphisms contribute to cellular response phenotypes. PLoS Genet 2011; 7:e1002162. [PMID: 21750684 PMCID: PMC3131293 DOI: 10.1371/journal.pgen.1002162] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Accepted: 05/15/2011] [Indexed: 01/14/2023] Open
Abstract
Glucocorticoids (GCs) mediate physiological responses to environmental stress and are commonly used as pharmaceuticals. GCs act primarily through the GC receptor (GR, a transcription factor). Despite their clear biomedical importance, little is known about the genetic architecture of variation in GC response. Here we provide an initial assessment of variability in the cellular response to GC treatment by profiling gene expression and protein secretion in 114 EBV-transformed B lymphocytes of African and European ancestry. We found that genetic variation affects the response of nearby genes and exhibits distinctive patterns of genotype-treatment interactions, with genotypic effects evident in either only GC-treated or only control-treated conditions. Using a novel statistical framework, we identified interactions that influence the expression of 26 genes known to play central roles in GC-related pathways (e.g. NQO1, AIRE, and SGK1) and that influence the secretion of IL6.
Collapse
Affiliation(s)
- Joseph C. Maranville
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Francesca Luca
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Allison L. Richards
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Xiaoquan Wen
- Department of Statistics, The University of Chicago, Chicago, Illinois, United States of America
| | - David B. Witonsky
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Shaneen Baxter
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Matthew Stephens
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
- Department of Statistics, The University of Chicago, Chicago, Illinois, United States of America
| | - Anna Di Rienzo
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
13
|
Godley LA, Cunningham J, Dolan ME, Huang RS, Gurbuxani S, McNerney ME, Larson RA, Leong H, Lussier Y, Onel K, Odenike O, Stock W, White KP, Le Beau MM. An integrated genomic approach to the assessment and treatment of acute myeloid leukemia. Semin Oncol 2011; 38:215-24. [PMID: 21421111 DOI: 10.1053/j.seminoncol.2011.01.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Traditionally, new scientific advances have been applied quickly to the leukemias based on the ease with which relatively pure samples of malignant cells can be obtained. Currently, our arsenal of approaches used to characterize an individual's acute myeloid leukemia (AML) combines hematopathologic evaluation, flow cytometry, cytogenetic analysis, and molecular studies focused on a few key genes. The advent of high-throughput methods capable of full-genome evaluation presents new options for a revolutionary change in the way we diagnose, characterize, and treat AML. Next-generation DNA sequencing techniques allow full sequencing of a cancer genome or transcriptome, with the hope that this will be affordable for routine clinical care within the decade. Microarray-based testing will define gene and miRNA expression, DNA methylation patterns, chromosomal imbalances, and predisposition to disease and chemosensitivity. The vision for the future entails an integrated and automated approach to these analyses, bringing the possibility of formulating an individualized treatment plan within days of a patient's initial presentation. With these expectations comes the hope that such an approach will lead to decreased toxicities and prolonged survival for patients.
Collapse
Affiliation(s)
- Lucy A Godley
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA. lgodley@medicine
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Abstract
BACKGROUND Osteosarcoma is the most common malignant bone tumor in children and adolescents. Multidrug resistance and poor clinical outcome are the problems that still affect osteosarcoma patients. The glutathione S-transferase supergene family includes several genes that encode enzymes involved in the detoxification of many xenobiotic agents, including carcinogens and anticancer drugs. The polymorphisms in these genes have already been associated both with cancer susceptibility and anticancer drugs resistance. OBJECTIVES This study aims to investigate the genotype frequencies of GSTM1, GSTT1 and GSTM3 genes in 80 osteosarcoma patients and 160 normal control participants, and also the influence of these polymorphisms in the clinical outcome of osteosarcoma patients. METHODS GSTM1 and GSTT1 deletion polymorphisms were examined through a multiplex-PCR and the GSTM3 polymorphism of three base pair-deletion at intron 6 using PCR-restriction fragments length polymorphism method. RESULTS We found that GSTM1 null genotype is correlated to poor clinical outcome characterized by the increased lung relapse occurrence [odds ratio (OR)=2.71, P=0.036], while the presence of at least one GSTM1 allele is associated with a good response to treatment and better survival (OR=4.28, P=0.020 and hazards ratio=4.09, P=0.0078, respectively). The GSTT1 null genotype was correlated with a better overall survival (hazards ratio=7.15, P=0.0247), whereas GSTM3*B allele was associated with metastasis at diagnosis (OR=2.83, P=0.028). CONCLUSION The findings of this study suggest that GST polymorphisms may have a role in treatment response and osteosarcoma progression.
Collapse
|
15
|
Yang JJ, Mehta PA, Relling MV, Davies SM. Pharmacogenetic and Pharmacogenomic Considerations in the Biology and Treatment of Childhood Leukemia. CHILDHOOD LEUKEMIA 2011. [DOI: 10.1007/978-3-642-13781-5_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
16
|
Oliveira AL, Rodrigues FFO, Santos RE, Aoki T, Rocha MN, Longui CA, Melo MB. GSTT1, GSTM1, and GSTP1 polymorphisms and chemotherapy response in locally advanced breast cancer. GENETICS AND MOLECULAR RESEARCH 2010; 9:1045-53. [PMID: 20568049 DOI: 10.4238/vol9-2gmr726] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The glutathione S-transferase (GST) family consists of phase II detoxification enzymes that catalyze the conjugation of toxic substances, such as chemotherapeutic agents, to glutathione. We examined whether GSTT1/GSTT1"null", GSTM1/GSTM1"null" and GSTP1Ile105Ile/GSTP1Ile105Val polymorphisms are associated with different response rates to neoadjuvant chemotherapy in the treatment of stage II and III breast cancer. Forty Brazilian women with invasive ductal adenocarcinoma of the breast submitted to neoadjuvant chemotherapy, using 5-fluorouracil, epirubicin and cyclophosphamide, were genotyped for the GSTT1, GSTM1 and GSTP1 genes. Clinical response was assessed by RECIST criteria. Comparisons were made for the three genes alone and in pairs, as polymorphic and as wild-type combinations and polymorphic/wild-type combinations. We analyzed all possible combinations and their response rate. Patients with the GSTT1/GSTP1105Ile combination were found to have a significantly better response than GSTT1"null"/GSTP1105Val (P = 0.0209) and GSTT1/GSTM1 (P = 0.0376) combinations. Analysis of all possible combinations showed the GSTM1"null" polymorphic genotype to be present in four, and the wild-type GSTP1105Ile in six of the combinations associated with the largest number of responding patients. We found that patients with the GSTT1/GSTP1105Ile wild-type combination had a significantly higher response rate to chemotherapy than patients with the respective polymorphic GSTT1"null"/GSTP1105Val combination or patients with the wild-type GSTT1/GSTM1. The six gene combinations associated with the largest number of responding patients were found to contain the wild-type GSTP1105Ile and the polymorphic-type GSTM1"null". These specific combinations were virtually absent in the combinations with few responding patients.
Collapse
Affiliation(s)
- A L Oliveira
- Departamento de Ginecologia e Obstetrícia, Irmandade da Santa Casa de Misericórdia de São Paulo, Faculdade de Ciências Médicas, São Paulo, SP, Brasil.
| | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of leukemias that result from clonal transformation of hematopoietic precursors through the acquisition of chromosomal rearrangements and multiple gene mutations. As a result of highly collaborative clinical research by pediatric cooperative cancer groups worldwide, disease-free survival has improved significantly during the past 3 decades. Further improvements in outcomes of children who have AML probably will reflect continued progress in understanding the biology of AML and the concomitant development of new molecularly targeted agents for use in combination with conventional chemotherapy drugs.
Collapse
|
18
|
Kilburn L, Okcu MF, Wang T, Cao Y, Renfro-Spelman A, Aldape KD, Gilbert MR, Bondy M. Glutathione S-transferase polymorphisms are associated with survival in anaplastic glioma patients. Cancer 2010; 116:2242-9. [PMID: 20187096 PMCID: PMC2861043 DOI: 10.1002/cncr.25006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Glutathione S-transferases (GSTs) are polymorphic enzymes that are responsible for glutathione conjugation of alkylators and scavenging of free radicals created by radiation. GST polymorphisms may result in altered or absent enzyme activity and have been associated with survival in patients with cancer. The authors of this report hypothesized that patients with anaplastic glioma (AG) who have GST genotypes that encode for lower activity enzymes will have longer survival than similar patients who have higher activity genotypes. The current study was performed to investigate the role of GST enzyme polymorphisms in predicting the survival of patients with AG. METHODS The medical records of 207 patients with AG from a single cancer center were reviewed retrospectively. Polymorphisms for the GST micro1 (GSTM1), GST theta1 (GSTT1), and GST pi1 (GSTP1) enzymes were identified. Overall survival was compared using the Kaplan-Meier method and Cox proportional hazards analyses adjusting for age, sex, histology, and therapy. RESULTS Among the patients with oligodendroglial tumors (n = 94), patients who had the GSTT1 null genotype had a 2.9 times increased risk of death (95% confidence interval [CI], 1.3-6.3) compared with patients who had the GSTT1 non-null genotype. Adjustment for 1p/19q status did not change the finding. In the patients who had anaplastic astrocytoma (n = 113), the patients with all GSTP1 genotypes except GSTP1 *B/*B had a 3.8 times increased risk of death (95% CI, 0.5-29.6) compared with patients who had the GSTP1 *B/*B genotype. CONCLUSIONS In patients with anaplastic oligodendroglial tumors, the GSTT1 null genotype may be associated with poor survival, possibly because of modifications in therapy secondary to increased toxicity. This hypothesis is under investigation. In patients with anaplastic astrocytoma, the GSTP1 *B/*B genotype may confer a survival advantage.
Collapse
Affiliation(s)
- Lindsay Kilburn
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Childhood Cancer Prevention and Epidemiology Center, 6621 Fannin Street, CC1510.00, Houston, TX 77030
- Texas Children’s Cancer Center, Baylor College of Medicine, 6621 Fannin Street, CC1410.00, Houston, TX 77030
| | - M. Fatih Okcu
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Department of Epidemiology, Division of Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
- Childhood Cancer Prevention and Epidemiology Center, 6621 Fannin Street, CC1510.00, Houston, TX 77030
- Texas Children’s Cancer Center, Baylor College of Medicine, 6621 Fannin Street, CC1410.00, Houston, TX 77030
| | - Tao Wang
- Division of Biostatistics, Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, BCMM600, Houston, TX 77030
| | - Yumei Cao
- Department of Epidemiology, Division of Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
- Childhood Cancer Prevention and Epidemiology Center, 6621 Fannin Street, CC1510.00, Houston, TX 77030
| | - Amy Renfro-Spelman
- Department of Epidemiology, Division of Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
- Childhood Cancer Prevention and Epidemiology Center, 6621 Fannin Street, CC1510.00, Houston, TX 77030
| | - Kenneth D. Aldape
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd., Box 85, Houston, TX 77030
| | - Mark R. Gilbert
- Department of Neuro-oncology, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd., Box 431, Houston, TX 77030
| | - Melissa Bondy
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Department of Epidemiology, Division of Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
- Childhood Cancer Prevention and Epidemiology Center, 6621 Fannin Street, CC1510.00, Houston, TX 77030
- Texas Children’s Cancer Center, Baylor College of Medicine, 6621 Fannin Street, CC1410.00, Houston, TX 77030
| |
Collapse
|
19
|
Cho HJ, Eom HS, Kim HJ, Kim IS, Lee GW, Kong SY. Glutathione-S-transferase genotypes influence the risk of chemotherapy-related toxicities and prognosis in Korean patients with diffuse large B-cell lymphoma. ACTA ACUST UNITED AC 2010; 198:40-6. [DOI: 10.1016/j.cancergencyto.2009.12.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2009] [Revised: 11/25/2009] [Accepted: 12/07/2009] [Indexed: 11/25/2022]
|
20
|
Hahn T, Zhelnova E, Sucheston L, Demidova I, Savchenko V, Battiwalla M, Smiley SL, Ambrosone CB, McCarthy PL. A deletion polymorphism in glutathione-S-transferase mu (GSTM1) and/or theta (GSTT1) is associated with an increased risk of toxicity after autologous blood and marrow transplantation. Biol Blood Marrow Transplant 2010; 16:801-8. [PMID: 20074657 DOI: 10.1016/j.bbmt.2010.01.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Accepted: 01/05/2010] [Indexed: 10/20/2022]
Abstract
Toxicity after blood and marrow transplantation (BMT) has interindividual variability that may be explained by common genetic polymorphisms in critical pathways. The glutathione-S-transferase (GST) isoenzymes detoxify the reactive oxygen species generated by chemotherapy agents and radiation. We investigated whether deletion polymorphisms in 2 GST genes (GSTM1 and GSTT1) were associated with toxicity after autologous or allogeneic BMT. The study population was selected from 699 consecutive BMT patients from 2 centers in Buffalo, NY, and Moscow, Russia, of whom 321 (203 autologous, 118 allogeneic BMT) had available banked samples and amplifiable DNA. Fifty percent of patients were homozygous null for GSTM1, which did not vary by center; however, the GSTT1 homozygous null deletion polymorphism occurred more frequently in patients treated in Moscow (38% versus 18%, P < .001). Overall grade 2-4 regimen-related toxicity occurred in 56%, with nearly 1 in 5 patients having 2 or more organ systems affected. Among autologous BMT patients, a deletion polymorphism in 1 or both genes was significantly associated with increased occurrence of overall toxicity (71% versus 56%, P = .034) and mucositis (74% versus 55%, P = .006). GSTM1 and/or GSTT1 deletion polymorphisms were not associated with toxicity after allogeneic BMT. Future studies may allow for individualized genetic risk stratification.
Collapse
Affiliation(s)
- Theresa Hahn
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York 14263, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
Acute myeloid leukemia (AML) in adults is a heterogeneous malignant pathology with a globally unfavorable prognosis. The classification of AML allows identification of subgroups with favorable prognosis. However, besides these specific subgroups, most patients will have an intermediate or unfavorable prognosis often resulting in induction failure, probably due to drug resistance of the leukemic blasts, and more frequently resulting in early relapse after achieving complete remission. This unfavorable situation leads to a strong need to develop new diagnostic and therapeutic options. However, development of these therapies and their efficient use requires a better understanding of the biology and the molecular pathogenesis of AML. Pharmacogenomics focuses on the genetic variation of drug-metabolizing enzymes, targets and transporters, and how these genetic variations interact to produce specific drug-related phenotypes. Potential genetic markers may serve to functionally subclassify patients by their disease and therefore influence the nature and intensity of treatment. This review summarizes important aspects of and recent advances in the field of pharmacogenomics in AML.
Collapse
Affiliation(s)
| | - Meyling H Cheok
- Jean-Pierre Aubert Research Center, INSERM U837, Institute for Cancer Research, 1 Place de Verdun, F-59045 Lille Cedex, France
| |
Collapse
|
22
|
Das P, Shaik AP, Bammidi VK. Meta-analysis study of glutathione-S-transferases (GSTM1,GSTP1, andGSTT1) gene polymorphisms and risk of acute myeloid leukemia. Leuk Lymphoma 2009; 50:1345-51. [DOI: 10.1080/10428190903003236] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
23
|
Clinical significance of GSTM1 and GSTT1 polymorphisms in younger patients with acute myeloid leukemia of intermediate-risk cytogenetics. Leuk Res 2009; 33:426-33. [DOI: 10.1016/j.leukres.2008.07.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2008] [Revised: 07/19/2008] [Accepted: 07/21/2008] [Indexed: 11/21/2022]
|
24
|
Ekhart C, Rodenhuis S, Smits PH, Beijnen JH, Huitema AD. An overview of the relations between polymorphisms in drug metabolising enzymes and drug transporters and survival after cancer drug treatment. Cancer Treat Rev 2009; 35:18-31. [DOI: 10.1016/j.ctrv.2008.07.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Revised: 07/09/2008] [Accepted: 07/22/2008] [Indexed: 11/16/2022]
|
25
|
Barahmani N, Carpentieri S, Li XN, Wang T, Cao Y, Howe L, Kilburn L, Chintagumpala M, Lau C, Okcu MF. Glutathione S-transferase M1 and T1 polymorphisms may predict adverse effects after therapy in children with medulloblastoma. Neuro Oncol 2008; 11:292-300. [PMID: 18952980 DOI: 10.1215/15228517-2008-089] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Glutathione S-transferases (GSTs) are polymorphic enzymes that catalyze the glutathione conjugation of alkylating agents, platinum compounds, and free radicals formed by radiation used to treat medulloblastoma. We hypothesized that GST polymorphisms may be responsible, in part, for individual differences in toxicity and responses in pediatric medulloblastoma. We investigated the relationship between GSTM1 and GSTT1 polymorphisms and survival and toxicity in 42 children with medulloblastoma diagnosed and treated at the Texas Children's Cancer Center. We conducted Kaplan-Meier analyses to determine if the GST polymorphisms were related to progression-free survival (PFS) and performed logistic regression to explore associations between GST polymorphisms and occurrence of grade 3 or greater (> or =Gr 3) myelosuppression, ototoxicity, nephrotoxicity, neurotoxicity, and intellectual impairment. Patients with at least one null genotype had a 4.3 (95% confidence interval, 1.1-16.8), 3.7 (1-13.6), and 6.4 (1.2-34) times increased risk for any > or =Gr 3 toxicity, any > or =Gr 3 toxicity excluding peripheral neuropathy, and any > or =Gr 3 toxicity requiring omission or cessation of chemotherapy, respectively. Compared with all others, patients with at least one null genotype had, on average, 27.2 (p x= 0.0002), 29 (p = 0.0004), and 21.7 (p = 0.002) lower full-scale, performance, and verbal intelligence quotient (IQ) scores, respectively. GSTM1 and GSTT1 polymorphisms may predict adverse events, including cognitive impairment after therapy, in patients with medulloblastoma. A larger study to validate these findings is under way.
Collapse
Affiliation(s)
- Nadia Barahmani
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Xiao Z, Yang L, Xu Z, Zhang Y, Liu L, Nie L, Li L, Wang J, Hao Y. Glutathione S-transferases (GSTT1 and GSTM1) genes polymorphisms and the treatment response and prognosis in Chinese patients with de novo acute myeloid leukemia. Leuk Res 2008; 32:1288-91. [DOI: 10.1016/j.leukres.2007.10.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2007] [Revised: 10/15/2007] [Accepted: 10/15/2007] [Indexed: 10/22/2022]
|
27
|
Huang RS, Duan S, Kistner EO, Bleibel WK, Delaney SM, Fackenthal DL, Das S, Dolan ME. Genetic variants contributing to daunorubicin-induced cytotoxicity. Cancer Res 2008; 68:3161-8. [PMID: 18451141 PMCID: PMC2714371 DOI: 10.1158/0008-5472.can-07-6381] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Identifying heritable genetic variants responsible for chemotherapeutic toxicities has been challenging due in part to its multigenic nature. To date, there is a paucity of data on genetic variants associated with patients experiencing severe myelosuppression or cardiac toxicity following treatment with daunorubicin. We present a genome-wide model using International HapMap cell lines that integrate genotype and gene expression to identify genetic variants that contribute to daunorubicin-induced cytotoxicity. A cell growth inhibition assay was used to measure variations in the cytotoxicity of daunorubicin. Gene expression was determined using the Affymetrix GeneChip Human Exon 1.0ST Array. Using sequential analysis, we evaluated the associations between genotype and cytotoxicity, those significant genotypes with gene expression and correlated gene expression of the identified candidates with cytotoxicity. A total of 26, 9, and 18 genetic variants were identified to contribute to daunorubicin-induced cytotoxicity through their effect on 16, 9, and 36 gene expressions in the combined, Centre d' Etude du Polymorphisme Humain (CEPH), and Yoruban populations, respectively. Using 50 non-HapMap CEPH cell lines, single nucleotide polymorphisms generated through our model predicted 29% of the overall variation in daunorubicin sensitivity and the expression of CYP1B1 was significantly correlated with sensitivity to daunorubicin. In the CEPH validation set, rs120525235 and rs3750518 were significant predictors of transformed daunorubicin IC(50) (P = 0.005 and P = 0.0008, respectively), and rs1551315 trends toward significance (P = 0.089). This unbiased method can be used to elucidate genetic variants contributing to a wide range of cellular phenotypes.
Collapse
Affiliation(s)
| | - Shiwei Duan
- Department of Medicine, University of Chicago, Chicago, Illinois
| | - Emily O. Kistner
- Department of Health Studies, University of Chicago, Chicago, Illinois
| | - Wasim K. Bleibel
- Department of Medicine, University of Chicago, Chicago, Illinois
| | | | | | - Soma Das
- Department of Human Genetics, University of Chicago, Chicago, Illinois
| | - M. Eileen Dolan
- Department of Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
28
|
Abstract
Acute myeloid leukemia (AML) is a heterogeneous group of leukemias that result from clonal transformation of hematopoietic precursors through the acquisition of chromosomal rearrangements and multiple gene mutations. As a result of highly collaborative clinical research by pediatric cooperative cancer groups worldwide, disease-free survival has improved significantly during the past 3 decades. Further improvements in outcomes of children who have AML probably will reflect continued progress in understanding the biology of AML and the concomitant development of new molecularly targeted agents for use in combination with conventional chemotherapy drugs.
Collapse
Affiliation(s)
- Jeffrey E Rubnitz
- Department of Oncology, St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | | | | |
Collapse
|
29
|
Ashton LJ, Murray JE, Haber M, Marshall GM, Ashley DM, Norris MD. Polymorphisms in genes encoding drug metabolizing enzymes and their influence on the outcome of children with neuroblastoma. Pharmacogenet Genomics 2007; 17:709-17. [PMID: 17700360 DOI: 10.1097/fpc.0b013e3280e1cc92] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Although several studies have shown that drug metabolizing enzyme gene polymorphisms may influence the impact of therapy in childhood leukemia, no comprehensive investigations have been carried out in children with neuroblastoma. The aim of this study was to identify polymorphisms in the genes encoding phase I and II drug metabolizing enzymes associated with the risk of relapse or death in a cohort of 209 children with neuroblastoma. METHODS Real-time PCR allelic discrimination was used to characterize the presence of polymorphisms in DNA from children with neuroblastoma. Three broad gene categories were examined: cytochrome P450, glutathione-S-transferase and N-acetyltransferase. Cumulative event-free survival was computed by the Kaplan-Meier method. The influence of selected factors on event-free survival was tested using the Cox proportional hazards model. RESULTS As previously reported, amplification of MYCN (hazards ratio=4.25, 95% confidence interval=2.76-6.56, P<0.001), unfavorable stage (hazard ratio=4.14, 95% confidence interval=2.3-7.47, P<0.001) or age more than 1 year at diagnosis (hazard ratio=1.86, 95% confidence interval=1.19-2.92, P=0.007) were all associated with an increased risk of relapse or death. Carriers of a NAT1*11 allele variant were significantly less likely to relapse or die compared with those with NAT1*10 or other NAT1 allele variants (P<0.001). In multivariate analysis, children who were GSTM1 null were more likely to relapse or die during follow-up after adjusting for MYCN amplification, stage and age at diagnosis (hazard ratio=1.6, 95% confidence interval=1.02-2.9, P=0.04). CONCLUSIONS These observations suggest that the NAT1*11 variant and the GSTM1 wild-type genotype contribute to a more favorable outcome in patients treated for neuroblastoma and are the first to demonstrate a relationship between NAT1 and GSTM1 genotypes in childhood neuroblastoma.
Collapse
Affiliation(s)
- Lesley J Ashton
- Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia.
| | | | | | | | | | | |
Collapse
|
30
|
Board PG. The use of glutathione transferase-knockout mice as pharmacological and toxicological models. Expert Opin Drug Metab Toxicol 2007; 3:421-33. [PMID: 17539748 DOI: 10.1517/17425255.3.3.421] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
ADME/Tox studies are of increasing importance because of the necessity to eliminate poor drug candidates early in the development pipeline. The glutathione S-transferases (GSTs) are a family of phase II enzymes that have been shown to play a significant role in the disposition of a wide range of drugs and other xenobiotics. Several GST-knockout mice strains have been developed that can potentially be used in ADME/Tox studies. So far, mice have been generated with deficiencies of mGSTP1/2, mGSTA4-4, mGSTZ1-1, mGSTM1-1, mGSTO1-1 and mGSTS1-1, but studies of drug metabolism in these strains have been limited. As there are 21 recognised GST genes in mice there is potential for many more strains to be made. However, a review of the available data suggests that because of differences in the evolution of the GST gene family between rodents and humans, only some knockout strains can provide insights relevant to human drug metabolism. It is concluded that, of the strains generated so far, only those deficient in mGSTP1-1, mGSTA4-4, mGSTO1-1 and mGSTZ1-1 have direct human orthologues and can be considered as human models. In contrast, there may not be appropriate orthologues of some enzymes, such as hGSTM1-1, that are known to be of relevance in drug metabolism.
Collapse
Affiliation(s)
- Philip G Board
- Australian National University, Molecular Genetics Group, John Curtin School of Medical Research, PO Box 334, Canberra ACT 2601, Australia.
| |
Collapse
|
31
|
Abstract
Pharmacogenomics provides knowledge regarding how genetic polymorphisms affect treatment responses. Such an approach is particularly needed in cancer therapy, as most chemotherapeutics drugs affect both tumor and normal cells, are ineffective in many patients and exhibit serious side effects. Leukemia exists in two different forms, myeloid and lymphoid. Acute lymphoblastic leukemia more frequently occurs in children, whereas the risk of acute myeloid leukemia is more common in adults. Despite significant progress in the treatment of these diseases, therapy is still unsuccessful in many patients. Prognosis is particularly poor in adult acute myeloid leukemia. Treatment failure in childhood acute lymphoblastic leukemia due to drug resistance remains the leading cause of cancer-related death in children. Here, we provide an overview of pharmacogenetics studies carried out in children and adults with acute lymphoblastic leukemia and acute myeloid leukemia, attempting to find the associations between treatment responses and polymorphisms in the genes whose products are needed for metabolism, and effects of drugs used in the treatment of leukemia.
Collapse
Affiliation(s)
- Marc Ansari
- Research Center Charles Bruneau, CHU Sainte-Justine, Montréal, Quebec, H3T 1C5, Canada
| | - Maja Krajinovic
- Research Center Charles Bruneau, CHU Sainte-Justine, Montréal, Quebec, H3T 1C5, Canada
- University of Montreal, Department of Pediatrics, Montréal, Québec, H3T 1C5, Canada
- University of Montreal, Department of Pharmacology, Montréal, Québec, H3T 1J4, Canada
| |
Collapse
|
32
|
Kishi S, Cheng C, French D, Pei D, Das S, Cook EH, Hijiya N, Rizzari C, Rosner GL, Frudakis T, Pui CH, Evans WE, Relling MV. Ancestry and pharmacogenetics of antileukemic drug toxicity. Blood 2007; 109:4151-7. [PMID: 17264302 PMCID: PMC1885506 DOI: 10.1182/blood-2006-10-054528] [Citation(s) in RCA: 151] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Treatment-related toxicity in acute lymphoblastic leukemia (ALL) can not only be life threatening but may also affect relapse risk. In 240 patients, we determined whether toxicities were related to 16 polymorphisms in genes linked to the pharmacodynamics of ALL chemotherapy, adjusting for age, race (self-reported or via ancestry-informative markers), sex, and disease risk group (lower- vs higher-risk therapy). Toxicities (gastrointestinal, infectious, hepatic, and neurologic) were assessed in each treatment phase. During the induction phase, when drugs subject to the steroid/cytochrome P4503A pathway predominated, genotypes in that pathway were important: vitamin D receptor (odds ratio [OR], 6.85 [95% confidence interval [CI], 1.73-27.0]) and cytochrome P4503A5 (OR, 4.61 [95% CI, 1.11-19.2]) polymorphisms were related to gastrointestinal toxicity and infection, respectively. During the consolidation phase, when antifolates predominated, the reduced folate carrier polymorphism predicted gastrointestinal toxicity (OR, 10.4 [95% CI, 1.35-80.4]) as it also did during continuation (OR, 2.01 [95% CI, 1.06-4.11]). In all 3 treatment phases, a glucuronosyltransferase polymorphism predicted hyperbilirubinemia (P = .017, P < .001, and P < .001) and methotrexate clearance (P = .028), which was also independently associated with hyperbilirubinemia (P = .026). The genotype-phenotype associations were similar whether analyses were adjusted by self-reported race or ancestry-informative genetic markers. Germ-line polymorphisms are significant determinants of toxicity of antileukemic therapy.
Collapse
Affiliation(s)
- Shinji Kishi
- Department of Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, TN 38105-2794, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Barragan E, Collado M, Cervera J, Martin G, Bolufer P, Roman J, Sanz MA. The GST deletions and NQO1*2 polymorphism confers interindividual variability of response to treatment in patients with acute myeloid leukemia. Leuk Res 2006; 31:947-53. [PMID: 17118447 DOI: 10.1016/j.leukres.2006.10.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Revised: 09/29/2006] [Accepted: 10/03/2006] [Indexed: 11/23/2022]
Abstract
Functional polymorphisms in the genes encoding detoxification enzymes could modify the response to treatment in acute myeloid leukemia and therefore affect the final clinical outcome. In the present study, we genotyped 153 patients diagnosed with de novo acute myeloid leukemia (AML) to clarify the influence of the genetic polymorphisms CYP1A1*2A, CYP3A4*1B, CYP2E1*5B, del{GSTT1}, del{GSTM1}, and NQO1*2 on disease outcome. The del{GSTM1} showed a higher frequency in females (62%) than in males (41%) (P=0.01). The number of functional NQO1 alleles influenced the response to induction therapy; 81% (55/68) NQO1-negative patients, 69% (28/41) heterozygous patients, and 27% (2/7) homozygous patients achieved complete remission (CR) (P=0.04). The presence of GST deletions was associated with a lower probability of disease-free survival (DFS) and this effect was more relevant in male patients. Males with del{GSTM1} showed a 28% DFS versus 57% DFS for undeleted GSTM1 (P=0.04). Similarly, males with undeleted GSTM1 and GSTT1 showed a 64% DFS versus 34% DFS for males with at least one GST deletion (P=0.05). This study suggests that the NQO1*2 polymorphism is relevant to the patient's response to induction therapy and that GST deletions influence treatment outcome after chemotherapy, especially in male patients.
Collapse
Affiliation(s)
- Eva Barragan
- Molecular Biology Laboratory, (Dpt. Medical Pathology), Spain.
| | | | | | | | | | | | | |
Collapse
|
34
|
Weiss JR, Kopecky KJ, Godwin J, Anderson J, Willman CL, Moysich KB, Slovak ML, Hoque A, Ambrosone CB. Glutathione S-transferase (GSTM1, GSTT1 and GSTA1) polymorphisms and outcomes after treatment for acute myeloid leukemia: pharmacogenetics in Southwest Oncology Group (SWOG) clinical trials. Leukemia 2006; 20:2169-71. [PMID: 17008887 DOI: 10.1038/sj.leu.2404421] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
35
|
Abstract
Over the past four decades, treatment of acute leukemia in children has made remarkable progress, from this disease being lethal to now achieving cure rates of 80% for acute lymphoblastic leukemia and 45% for acute myeloid leukemia. This progress is largely owed to the optimization of existing treatment modalities rather than the discovery of new agents. However, the annual number of patients with leukemia who experience relapse after initial therapy remains greater than that of new cases of most childhood cancers. The aim of pharmacogenetics is to develop strategies to personalize medications and tailor treatment regimens to individual patients, with the goal of enhancing efficacy and safety through better understanding of the person's genetic makeup. In this review, we summarize recent pharmacogenomic studies related to the treatment of pediatric acute leukemia. These include work using candidate-gene approaches, as well as genome-wide studies using haplotype mapping and gene expression profiling. These strategies illustrate the promise of pharmacogenomics to further advance the treatment of human cancers, with childhood leukemia serving as a paradigm.
Collapse
Affiliation(s)
- Meyling H Cheok
- St. Jude Children's Research Hospital, Department of Pharmaceutical Sciences, Memphis, TN 38105, USA.
| | | | | |
Collapse
|
36
|
Gasparini G, Longo R, Torino F, Gattuso D, Morabito A, Toffoli G. Is tailored therapy feasible in oncology? Crit Rev Oncol Hematol 2006; 57:79-101. [PMID: 16337806 DOI: 10.1016/j.critrevonc.2005.07.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2004] [Revised: 07/01/2005] [Accepted: 07/27/2005] [Indexed: 11/18/2022] Open
Abstract
Tailored therapy aims to cure a patient who suffers from a specific disease with an effective and safe drug, based on the complex interactions among patient's characteristics, disease physiopathology and drug metabolism. Genomic and proteomic technologies represent promising new useful tools to understand cancer biology and molecular basis of interindividual differences of anticancer drugs efficacy. Genomic profiling seems to be able to re-classifying cancer into new molecular and prognostic homogeneous subgroups. By individual polymorphisms it is possible to identify the patients at higher risk for severe toxicity from those that may gain benefit from a particular treatment. The clinical use of targeted therapy is hampered by several questions, including: optimal biological dose, availability of surrogate biomarkers predictive of activity, schedule of administration, tumor histotype and stage to treat and modalities of combination with chemo/radiotherapy. In addition, further efforts are needed to improve the reliability of genomic and proteomic technologies. These unsolved issues presently make tailored therapy an open challenge.
Collapse
Affiliation(s)
- Giampietro Gasparini
- Division of Medical Oncology, Azienda Complesso Ospedaliero di Rilevanza Nazionale S. Filippo Neri Hospital, via G. Martinotti 20, 00135 Rome, Italy.
| | | | | | | | | | | |
Collapse
|
37
|
Abstract
The same doses of medication cause considerable heterogeneity in efficacy and toxicity across human populations. Genetic factors are thought to represent important determinants of drug efficacy and toxicity. Pharmacogenetics focuses on the prediction of the response of tumor and normal tissue to standard therapy by genetic profiling and, thereby, to select the most appropriate medication at optimal doses for each individual patient. In the present review, we discuss the relevance of single nucleotide polymorphisms (SNP) in genes, whose gene products act upstream of the actual drug target sites, that is, drug transporters and drug metabolizing phase I and II enzymes, or downstream of them, that is, apoptosis-regulating genes and chemokines. SNPs in relevant genes, which encode for proteins that interact with anticancer drugs, were also considered, that is, enzymes of DNA biosynthesis and metabolism, DNA repair enzymes, and proteins of the mitotic spindle. A significant body of evidence supports the concept of predicting drug efficacy and toxicity by SNP genotyping. As the efficacy of cancer chemotherapy, as well as the drug-related toxicity in normal tissues is multifactorial in nature, sophisticated approaches such as genome-wide linkage analyses and integrate drug pathway profiling may improve the predictive power compared with genotyping of single genes. The implementation of pharmacogenetics into clinical routine diagnostics including genotype-based recommendations for treatment decisions and risk assessment for practitioners represents a challenge for the future.
Collapse
Affiliation(s)
- Thomas Efferth
- German Cancer Research Center, M070, Im Neuenheimer Feld 282, 69120 Heidelberg, Germany.
| | | |
Collapse
|
38
|
Robert J, Morvan VL, Smith D, Pourquier P, Bonnet J. Predicting drug response and toxicity based on gene polymorphisms. Crit Rev Oncol Hematol 2005; 54:171-96. [PMID: 15890268 DOI: 10.1016/j.critrevonc.2005.01.005] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2004] [Revised: 01/01/2005] [Accepted: 01/28/2005] [Indexed: 12/16/2022] Open
Abstract
The sequencing of the human genome has allowed the identification of thousands of gene polymorphisms, most often single nucleotide polymorphims (SNP), which may play an important role in the expression level and activity of the corresponding proteins. When these polymorphisms occur at the level of drug metabolising enzymes or transporters, the disposition of the drug may be altered and, consequently, its efficacy may be compromised or its toxicity enhanced. Polymorphisms can also occur at the level of proteins directly involved in drug action, either when the protein is the target of the drug or when the protein is involved in the repair of drug-induced lesions. There again, these polymorphisms may lead to alterations in drug efficacy and/or toxicity. The identification of functional polymorphisms in patients undergoing chemotherapy may help the clinician prescribe the optimal drug combination or schedule and predict with more accuracy the response to these prescriptions. We have recorded in this review the polymorphisms that have been identified up till now in genes involved in anticancer drug activity. Some of them appear especially important in predicting drug toxicity and should be determined in routine before drug administration; this is the case of the most common variations of thiopurine methyltransferase for 6-mercaptopurine and of dihydropyrimidine dehydrogenase for fluorouracil. Other appear determinant for drug response, such as the common SNPs found in glutathione S-transferase P1 or xereoderma pigmentosum group D enzyme for the activity of oxaliplatin. However, confusion factors may exist between the role of gene polymorphisms in cancer risk or overall prognosis and their role in drug response.
Collapse
Affiliation(s)
- Jacques Robert
- Institut Bergonié and Université Victor Segalen Bordeaux 2, 229 cours de l'Argonne, 33076 Bordeaux-Cedex, France.
| | | | | | | | | |
Collapse
|
39
|
Vande Woude GF, Kelloff GJ, Ruddon RW, Koo HM, Sigman CC, Barrett JC, Day RW, Dicker AP, Kerbel RS, Parkinson DR, Slichenmyer WJ. Reanalysis of cancer drugs: old drugs, new tricks. Clin Cancer Res 2004; 10:3897-907. [PMID: 15173099 DOI: 10.1158/1078-0432.ccr-03-0786] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
40
|
Okcu MF, Selvan M, Wang LE, Stout L, Erana R, Airewele G, Adatto P, Hess K, Ali-Osman F, Groves M, Yung AWK, Levin VA, Wei Q, Bondy M. Glutathione S-transferase polymorphisms and survival in primary malignant glioma. Clin Cancer Res 2004; 10:2618-25. [PMID: 15102663 DOI: 10.1158/1078-0432.ccr-03-0053] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE The purpose of this research was to investigate the relationship between glutathione S-transferase (GST) polymorphisms and survival, and chemotherapy-related toxicity in 278 glioma patients. EXPERIMENTAL DESIGN We determined genetic variants for GSTM1, GSTT1, and GSTP1 enzymes by PCR and restriction fragment length polymorphisms. We conducted Kaplan-Meier and Cox-proportional hazard analyses to examine whether the GST polymorphisms are related to overall survival, and logistic regression analysis to explore whether the GST polymorphisms are associated with toxicity. RESULTS For patients with anaplastic astrocytoma, anaplastic oligodendroglioma, anaplastic oligoastrocytoma, and anaplastic ependymoma (n = 78), patients with GSTP1*A/*A-M1 null genotype survived longer than did the rest of the group (median survival "not achieved," and 41 months, respectively; P = 0.06). Among patients treated with nitrosoureas (n = 108), those with GSTP1*A/*A and GSTM1 null genotype were 5.7 times (95% confidence interval, 0.9-37.4) more likely to experience an adverse event secondary to chemotherapy, compared with the others. CONCLUSIONS In patients with anaplastic astrocytoma, anaplastic oligodendroglioma, and anaplastic oligoastrocytoma, combination of germ-line GSTP1*A/*A and GSTM1 null genotype confers a survival advantage. Patients with this genotype also have an increased risk of adverse events secondary to chemotherapy that primarily comprised nitrosourea alkylating agents.
Collapse
Affiliation(s)
- M Fatih Okcu
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Sugimura J, Foster RS, Cummings OW, Kort EJ, Takahashi M, Lavery TT, Furge KA, Einhorn LH, Teh BT. Gene Expression Profiling of Early- and Late-Relapse Nonseminomatous Germ Cell Tumor and Primitive Neuroectodermal Tumor of the Testis. Clin Cancer Res 2004; 10:2368-78. [PMID: 15073113 DOI: 10.1158/1078-0432.ccr-03-0341] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To better understand the molecular mechanisms that underlay the development and progression of nonseminomatous germ cell tumor of testis (NSGCTT) as well as malignant transformation of teratoma and primitive neuroectodermal tumor (PNET). EXPERIMENTAL DESIGN We studied the gene expression profiles of 17 retroperitoneal NSGCTTs (10 yolk sac tumors, 3 embryonal carcinomas, 4 teratomas) and 2 PNETs obtained from patients with two clinical outcomes. Tissue samples were obtained from the Indiana University. One group of NSGCTT and PNET patients developed metastases within 2 years (early-relapse) of initial successful treatment, and the other group developed metastases after 2 years (late-relapse). Gene expression in these groups of patients was quantified using cDNA microarrays and real-time relative quantitative PCR. RESULTS We demonstrate that the gene expression profiles of these tumors correlate with histological type. In addition, we identify type-specific genes that may serve as novel diagnostic markers. We also identify a gene set that can distinguish between early-relapse and late-relapse yolk sac tumors. The expression differences of these genes may underlie the differences in clinical outcome and drug response of these tumors. CONCLUSION This is the first study that used gene expression profiling to examine the molecular characteristics of the NSGCTTs and drug response in early- and late-relapse tumors. These results suggest that two molecularly distinct forms of NSGCTTs exist and that the integration of expression profile data with clinical parameters could enhance the diagnosis and prognosis of NSGCTTs. More importantly, the identified genes provide insight into the molecular mechanisms of aggressive NSGCTTs and suggest intervention strategies.
Collapse
Affiliation(s)
- Jun Sugimura
- Laboratory of Cancer Genetics, Van Andel Research Institute, Grand Rapids, Michigan 49503, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Ross D, Siegel D. NAD(P)H:quinone oxidoreductase 1 (NQO1, DT-diaphorase), functions and pharmacogenetics. Methods Enzymol 2004; 382:115-44. [PMID: 15047100 DOI: 10.1016/s0076-6879(04)82008-1] [Citation(s) in RCA: 194] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- David Ross
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Colorado, Denver 80262, USA
| | | |
Collapse
|
43
|
Pui CH, Relling MV, Evans WE. Role of pharmacogenomics and pharmacodynamics in the treatment of acute lymphoblastic leukaemia. Best Pract Res Clin Haematol 2002; 15:741-56. [PMID: 12617874 DOI: 10.1053/beha.2002.0225] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Pharmacodynamic studies have been used to establish the relationships between the administered dosage and the concentration of drugs and metabolites in the blood or tissues and that between these concentrations and pharmacological effects. Polymorphisms in the genes that encode drug-metabolizing enzymes, drug transporters and drug targets can affect a person's response to therapy and may affect the development of de novo or therapy-related leukaemias. The burgeoning field of pharmacogenomics elucidates inherited differences in drug metabolism and treatment response. Increasingly, pharmacodynamic and pharmacogenomic studies are being used to individualize therapy to enhance efficacy and reduce toxicity.
Collapse
Affiliation(s)
- Ching Hon Pui
- St Jude Children's Research Hospital, and the Colleges of Medicine and Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | | | | |
Collapse
|
44
|
Recent publications in hematology oncology. Hematol Oncol 2002; 20:147-54. [PMID: 12360948 DOI: 10.1002/hon.692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|