Glutathione S-Transferase pi (GSTP1)

 

Isabelle Meiers M.D.

Maidstone and Tunbridge Wells NHS Trust, Preston Hall Hospital,
Royal British Legion Village, Aylesford, Kent, ME20 7NJ, UK

 

February 2010

 

 

Running title: GSTP1 review

Key words: Glutathione S-transferase pi (GSTP1), cancer, methylation analysis, antioxidants.

Pi-class glutathione-S-transferase (GSTP1) located on chromosome 11q13 encodes a phase II metabolic enzyme that detoxifies reactive electrophilic intermediates. GSTP1 plays an important role in protecting cells from cytotoxic and carcinogenic agents and is expressed in normal tissues at variable levels in different cell types. Altered GSTP1 activity and expression have been reported in many tumors and this is largely due to GSTP1 DNA hypermethylation at the CpG island in the promoter-5'.

We review the potential novel role of glutathione S-transferase pi (GSTP1) and its related expression in miscellaneous cancers. We focus on the rationale for use of molecular assays for the detection of cancer, emphasizing the role of the identification of epigenetic alterations. Finally, we focus on the potential role of GSTP1 in the pathway of prostate cancer, the most GSTP1 DNA hypermethylation-related neoplasm studied to date.

Advances in the epigenetic characterization of cancers enabled the development of DNA methylation assays that may soon be used in diagnostic testing of serum and tissue for cancers. Inhibition of aberrant promoter methylation could theoretically prevent carcinogenesis.
Reactive oxygen species that are generated by physiologic processes such as cellular respiration, exposure to chemical agents, or exposure to ionizing radiation may overcome cellular antioxidant defense and cause DNA damage (Bostwick et al., 2000). Such damage may result in mutations and alteration of oncogenes or tumor suppressor genes. The cytosolic isoenzyme glutathione S-transferase pi (GSTP1) is an important multifunctional detoxifying enzyme within the glutathione S-transferase family enzymes that inactivates electrophilic carcinogens by conjugation with glutathione (Toffoli et al., 1992; Jerónimo et al., 2001). The regulatory sequence near the GST gene is commonly affected by hypermethylation during the early stages of carcinogenesis (Lee et al., 1994; Brooks et al., 1998; Cairns et al., 2001; Jerónimo et al., 2002; Henrique and Jerónimo, 2004).
Several classes of GST, including alpha, mu, pi, and theta, were previously found in human tissue. For example, compared with benign tissue, there is increased expression of GST pi in cancers of the breast, colon, stomach, pancreas, bladder, lung, head and neck, ovary, and cervix, as well as soft tissue sarcoma, testicular embryonal carcinoma, meningioma, and glioma (Niitsu et al., 1989; Randall et al., 1990; Kantor et al., 1991; Satta et al., 1992; Toffoli et al., 1992; Green et al., 1993; Inoue et al., 1995; Bentz et al., 2000; Tratche et al., 2002; Simic et al., 2005; Arai et al., 2006).
However, hypermethylation of the GSTP1 promoter has been associated with gene silencing in prostate cancer and kidney cancer (Lee et al., 1994; Brooks et al., 1998; Cairns et al., 2001; Jerónimo et al., 2002; Dulaimi et al., 2004). Similarly, expression of GSTP1 is lower in invasive pituitary tumors than in noninvasive pituitary tumors and methylation status correlates with significant downregulation of GSTP1 expression; the frequency of GSTP1 methylation being higher in invasive pituitary tumors with reduced-GSTP1 expression than in pituitary adenomas with normal or high GSTP1 expression. These data indicate that GSTP1 inactivation through CpG hypermethylation is common in pituitary adenomas and may contribute to aggressive pituitary tumor behavior (Yuan et al., 2008). More recently, a study showed a trend of increasing GSTP1 methylation frequency with increasing grade of mammary phyllodes tumors. The authors reported that GSTP1 promoter hypermethylation was associated with loss of GSTP1 expression. These results suggest that phyllodes tumors segregate into only two groups on the basis of their methylation profiles: the benign group and the combined borderline/malignant group (Kim et al., 2009). Other investigators studied the role of hypermethylation of the GSTP1 gene promoter region in endometrial carcinoma and found that reduced GSTP1 expression was associated with myometrial invasion potential (Chan et al., 2005).


Epigenetic alterations: emerging molecular markers for cancer detection

Cancer is a process fuelled both by genetic alterations and epigenetic mechanisms. Epigenetics refer to changes in gene expression that can be mitotically inherited, but are not associated with the changes in the coding sequence of the affected genes. In other words, epigenetics refer to the inheritance of information based on gene expression levels, in contrast to genetics that refer to transmission of information based on gene sequence (Esteller et al., 2000). DNA methylation, the best understood mechanism in epigenetics, is an enzyme-mediated chemical modification that adds methyl (-CH3) groups at selected sites on DNA. In humans and most mammals, DNA methylation only affects the cytosine base (C), when it is followed by a guanosine (G). Methylation of the cytosine nucleotide residue located within the dinucleotide 5'-CpG-3' is the most frequent epigenetic alteration in humans. These CpG dinucleotides are not randomly distributed in the genome. Indeed, there are CpG-rich regions called "CpG islands" frequently associated with the 5' regulatory regions of genes, including the promoter. DNA methylation in the promoter regions is a powerful mechanism for the suppression of gene activity.


DNA methylation analysis: currently available methods

Methylation of CpG islands is of interest for diagnostic and prognostic reasons. Methylation of one or both alleles of a region can serve as a biomarker of cancer or silence gene expression when they are in a promoter region (Verma and Srivastava, 2002). Assays for methylation are appealing for translational research since they can utilize amplification techniques, such as methylation-specific polymerase chain reaction (PCR), and thereby utilize small amounts of samples.
Due to its relative simplicity, safety, and sensitivity, methylation-specific PCR is the most commonly employed method for methylation analysis (Herman et al., 1996).
The conventional methylation-specific PCR (CMSP) assay uses two sets of primers specifically designed to amplify the methylated or unmethylated sequence, and the PCR products are run in a gel (Herman et al., 1996). The results of CMSP at a particular DNA region are simply reported as methylated or unmethylated, not allowing quantitation or identification of partial methylation. The CMSP assay is mainly used for GSTP1 methylation detection in fluids. For instance, GSTP1 methylation in serum of men with localized prostate cancer prior to treatment carries a 4.4 fold increased risk of biochemical recurrence following surgery (Bastian et al., 2005).
The use of fluorescence-based real-time quantitative methylation-specific PCR (QMSP) assay improved the sensitivity of tumor detection. Continuous monitoring of fluorescent signals during the PCR process enabled quantification of methylated alleles of a single region amongst unmethylated DNA because the fluorescence emission of the reporter represents the number of generated DNA fragments (Heid et al., 1996).


GSTP1 hypermethylation: significance and incidence related to prostate cancer

Epigenetic silencing of gluthathione-S-transferase pi (GSTP1) is recognized as being a molecular hallmark of human prostate cancer. Methylation of CpG islands in the promoter of the pi class of glutathione S-transferase occurs in prostatic intraepithelial neoplasia (PIN) and cancer (Gonzalgo et al., 2004). Other hypermethylated regions relevant to prostate cancer include the retinoic acid receptor beta 2 (Bastian et al., 2007). These findings in prostate cancer suggest that DNA methylation is among the early events in tumorigenesis, but it remains to be seen whether DNA methylation is a necessary or permissive event in tumorigenesis.
The extensive methylation of deoxycytidine nucleotides distributed throughout the 5' "CG island" region of GSTP1 is not detected in benign prostatic epithelium, but has been detected in intraepithelial neoplasia, prostatic adenocarcinoma, and fluids (plasma, serum, ejaculate, and urine) of patients with prostate cancer by methylation-specific polymerase chain reaction assay, and may be useful as a cancer-specific molecular biomarker (Lee et al., 1994; Cairns et al., 2001; Henrique and Jerónimo, 2004; Crocitto et al., 2004; Perry et al., 2006; Hopkins et al., 2007; Cao and Yao, 2010).
Quantitative methylation-specific PCR (QMSP) reveals that the epigenetic silencing (loss of expression) of the GSTP1 gene is in fact the most common genetic alteration in prostate cancer (>90%) and high-grade prostatic intraepithelial neoplasia (PIN) (70%) (Lee et al., 1994; Brooks et al., 1998; Cairns et al., 2001; Harden et al., 2003; Henrique and Jerónimo, 2004) and this somatic inactivation ("silencing") of GSTP1 is directly associated with promoter methylation (Cairns et al., 2001; Jerónimo et al., 2002; Henrique and Jerónimo, 2004). Higher levels of GSTP1 promoter methylation is associated with the transition from prostatic intraepithelial neoplasia (PIN) to carcinoma (Henrique et al., 2006).
During cancer development, GSTP1 does not appear to function either as an oncogene or as a tumor suppressor gene, since induced GSTP1 expression in prostate cancer cell lines failed to suppress cell growth. Instead, GSTP1 was proposed to act as a "caretaker" gene. When GSTP1 is inactivated, prostate cells appear to become more vulnerable to somatic alterations upon chronic exposure to genome-damaging stresses as oxidants and electrophiles, that are contributed by environment and lifestyle (Kinzler and Vogelstein, 1997; Cairns et al., 2001).
The significance of absent GSTP1 (GSTP1 silencing) in high grade PIN and carcinoma is unclear. It may be an epiphenomenon, simply reflecting disruption of the basal cell layer with neoplastic progression. However, Lee et al. considered the likelihood of a more fundamental role (Lee et al., 1994). Two studies found that a small proportion (3.5-5%) of cases retained modest GSTP1 expression in carcinoma (Cookson et al., 1997; Moskaluk et al., 1997). Cookson et al. also recorded positivity in 1 of 17 cases of high grade PIN (Cookson et al., 1997) . Unlike genetic alterations that permanently and definitively change DNA sequence, promoter methylation is a potentially reversible modification. Hence, promoter methylation may be amenable to therapeutic intervention aimed at reactivating silenced cancer genes. This has important implications for chemoprevention because, as mentioned above, up to 70% of cases of high-grade PIN display GSTP1 promoter methylation (Brooks et al., 1998; Cairns et al., 2001; Jerónimo et al., 2002). Indeed, recently some authors have investigated the effects of green tea polyphenols (GTPs) on GSTP1 re-expression. They demonstrated that promoter demethylation by green tea polyphenols leads to re-expression of GSTP1 in human prostate cancer cells, therefore making green tea polyphenols excellent candidates for the chemoprevention of prostate cancer (Pandey et al., 2009).
Some investigators evaluated the impact of androgen deprivation therapy on the detection of GSTP1 hypermethylation in prostate cancer (Kollermann et al., 2006). In 87% (13/15) of the patients, there was no alteration in GSTP1 hypermethylation detection (Kollermann et al., 2006) and the authors suggested that the change from positive to negative GSTP1 hypermethylation status in two patients may point to partial androgen dependency (Kollermann et al., 2006). In addition to the supposed hormonal interaction, other possible explanations may be speculated to explain why prostate cancer loses GSTP1 hypermethylation after prolonged neoadjuvant hormonal therapy. First, the lack of GSTP1 hypermethylation may be attributable to technical problems (false negative results). Furthermore, the possibility that both tumors primarily lacked GSTP1 hypermethylation might be raised. However, further studies are necessary to assess the frequency and extent of hormonal interaction with GSTP1 hypermethylation.


Anti-cancer effect of GSTP1 and future prospects

Increased levels of GST pi may protect human cancer cells against cytotoxic drugs. Several antineoplastic drugs, particularly reactive electrophilic alkylating agents, form conjugates with glutathione spontaneously and in GST-catalyzed reactions (Awasthi et al., 1996). The expression of particular subclasses of GST protects cells from the cytotoxicities of these cancer drugs, and overexpression of GST has been implicated in antineoplastic drug resistance (Morrow et al., 1998). Induction of the enzymes is thought to represent an adaptive response to stress, and may be triggered by exogenous chemical agents and probably also by reactive oxygen metabolites (Hayes and Pulford, 1995). GST enzymes have a broad substrate specificity that includes substances with known mutagenic properties. Elevated serum GST pi has been exploited as a serum tumor marker for gastrointestinal cancer (Niitsu et al., 1989) and non-Hodgkin's lymphoma (Katahira et al., 2004) as a method of predicting sensitivity to chemotherapy.
Inactivation of GSTP1 in prostate cancer occurs early during carcinogenesis, leaving prostate cells with inadequate defenses against oxidant and electrophile carcinogens. Epigenetic mechanisms (see above) are strongly implicated in progression (Rennie and Nelson, 1998). Unlike genetic alterations, changes in DNA methylation are potentially reversible. Thus, therapeutic interventions involving reversal of the methylation process of several key genes in prostate carcinogenesis might improve current therapeutic options, thereby enhancing the anti-cancer effect of GSTP1 gene in patients "at risk" with high-grade PIN or in men with established prostate cancer. Nucleoside-analogue inhibitors of DNA methyltransferases, such as 5-aza-2'-deoxycytidine, are able to demethylate DNA and restore silenced gene expression. Unfortunately, the clinical utility of these compounds has not yet been fully realized, mainly because of their side effects. The anti-arrhythmia drug procainamide, a nonnucleoside inhibitor of DNA methyltransferases (category of enzymes that catalyse DNA methylation during cell replication), reversed GSTP1 DNA hypermethylation and restorted GSTP1 expression in LNCaP human prostate cancer cells propagated in vitro or in vivo as xenograft tumors in athymic nude mice (Cairns et al., 2001). Some investigators tested the potential use of procaine, an anesthetic drug related to procainamide. Using the MCF-7 breast cancer cell line, they have found that procaine produced a 40% reduction in 5-methylcytosine DNA content as determined by high-performance capillary electrophoresis and total DNA enzyme digestion. Procaine can also demethylate densely hypermethylated CpG islands such as those located in the promoter region of the RAR beta 2 gene, restoring gene expression of epigenetically silenced genes. This property may be explained by binding of procaine to CpG-enriched DNA. Finally, procaine also has growth-inhibitory effects in these cancer cells, causing mitotic arrest (Villar-Garea et al., 2003). Thus, procaine and procainamide are promising candidate agents for future cancer therapies based on epigenetics. Li et al. reported that GSTP1 was upregulated in the stromal compartment of hormone-independent prostate cancer, which may contribute to chemoresistance of advanced prostate cancer (Morrow et al., 1998).
Epidemiologic evidence has shown a reduced risk of prostate cancer in men consuming selenium, suggesting a role for antioxidants in protection against prostate carcinogenesis. A systematic review and meta-analysis of the literature confirm that selenium intake may reduce the risk of prostate cancer (Etminan et al., 2005). Vitamin E intake also may decrease DNA damage and inhibit transformation through its antioxidant function. Long-term supplementation with alpha-tocopherol substantially reduced prostate cancer incidence and mortality in male smokers (Heinonen et al., 1998). Therapy directed at the induction or preservation of GSTP1 activity in benign prostatic epithelium may prevent or delay progression of prostatic cancer.
GSTP1 has a protective role as an antioxidant agent in transformation and progression of prostate cancer. The interplay between altered or impaired expression of GST appears to play a significant role in carcinogenesis in the prostate. Inhibition of aberrant promoter methylation could be an effective method of chemoprevention.

Bibliography

Serum glutathione-S-transferase-pi as a tumor marker for gastrointestinal malignancies.
Niitsu Y, Takahashi Y, Saito T, Hirata Y, Arisato N, Maruyama H, Kohgo Y, Listowsky I.
Cancer. 1989 Jan 15;63(2):317-23.
PMID 2910437
 
Glutathione S-transferase (placental) as a marker of transformation in the human cervix uteri: an immunohistochemical study.
Randall BJ, Angus B, Akiba R, Hall A, Cattan AR, Proctor SJ, Jones RA, Horne CH.
Br J Cancer. 1990 Oct;62(4):614-8.
PMID 2223578
 
Monoclonal antibodies to glutathione S-transferase pi-immunohistochemical analysis of human tissues and cancers.
Kantor RR, Giardina SL, Bartolazzi A, Townsend AJ, Myers CE, Cowan KH, Longo DL, Natali PG.
Int J Cancer. 1991 Jan 21;47(2):193-201.
PMID 1703126
 
Expression of MDR1 and glutathione S transferase-pi genes and chemosensitivities in human gastrointestinal cancer.
Satta T, Isobe K, Yamauchi M, Nakashima I, Takagi H.
Cancer. 1992 Feb 15;69(4):941-6.
PMID 1735085
 
Expression of MDR1 and GST-pi in human soft tissue sarcomas: relation to drug resistance and biological aggressiveness.
Toffoli G, Frustaci S, Tumiotto L, Talamini R, Gherlinzoni F, Picci P, Boiocchi M.
Ann Oncol. 1992 Jan;3(1):63-9.
PMID 1606072
 
Glutathione S-transferase expression in benign and malignant ovarian tumours.
Green JA, Robertson LJ, Clark AH.
Br J Cancer. 1993 Aug;68(2):235-9.
PMID 8347477
 
Cytidine methylation of regulatory sequences near the pi-class glutathione S-transferase gene accompanies human prostatic carcinogenesis.
Lee WH, Morton RA, Epstein JI, Brooks JD, Campbell PA, Bova GS, Hsieh WS, Isaacs WB, Nelson WG.
Proc Natl Acad Sci U S A. 1994 Nov 22;91(24):11733-7.
PMID 7972132
 
The glutathione S-transferase supergene family: regulation of GST and the contribution of the isoenzymes to cancer chemoprotection and drug resistance.
Hayes JD, Pulford DJ.
Crit Rev Biochem Mol Biol. 1995;30(6):445-600. (REVIEW)
PMID 8770536
 
Glutathione S transferase Pi is a powerful indicator in chemotherapy of human lung squamous-cell carcinoma.
Inoue T, Ishida T, Sugio K, Maehara Y, Sugimachi K.
Respiration. 1995;62(4):223-7.
PMID 8578019
 
Interactions of melphalan with glutathione and the role of glutathione S-transferase.
Awasthi S, Bajpai KK, Piper JT, Singhal SS, Ballatore A, Seifert WE Jr, Awasthi YC, Ansari GA.
Drug Metab Dispos. 1996 Mar;24(3):371-4.
PMID 8820431
 
Real time quantitative PCR.
Heid CA, Stevens J, Livak KJ, Williams PM.
Genome Res. 1996 Oct;6(10):986-94.
PMID 8908518
 
Methylation-specific PCR: a novel PCR assay for methylation status of CpG islands.
Herman JG, Graff JR, Myohanen S, Nelkin BD, Baylin SB.
Proc Natl Acad Sci U S A. 1996 Sep 3;93(18):9821-6.
PMID 8790415
 
Glutathione S-transferase PI (GST-pi) class expression by immunohistochemistry in benign and malignant prostate tissue.
Cookson MS, Reuter VE, Linkov I, Fair WR.
J Urol. 1997 Feb;157(2):673-6.
PMID 8996396
 
Cancer-susceptibility genes. Gatekeepers and caretakers.
Kinzler KW, Vogelstein B.
Nature. 1997 Apr 24;386(6627):761, 763.
PMID 9126728
 
Immunohistochemical expression of pi-class glutathione S-transferase is down-regulated in adenocarcinoma of the prostate.
Moskaluk CA, Duray PH, Cowan KH, Linehan M, Merino MJ.
Cancer. 1997 Apr 15;79(8):1595-9.
PMID 9118044
 
CG island methylation changes near the GSTP1 gene in prostatic intraepithelial neoplasia.
Brooks JD, Weinstein M, Lin X, Sun Y, Pin SS, Bova GS, Epstein JI, Isaacs WB, Nelson WG.
Cancer Epidemiol Biomarkers Prev. 1998 Jun;7(6):531-6.
PMID 9641498
 
Coordinated action of glutathione S-transferases (GSTs) and multidrug resistance protein 1 (MRP1) in antineoplastic drug detoxification. Mechanism of GST A1-1- and MRP1-associated resistance to chlorambucil in MCF7 breast carcinoma cells.
Morrow CS, Smitherman PK, Diah SK, Schneider E, Townsend AJ.
J Biol Chem. 1998 Aug 7;273(32):20114-20.
PMID 9685354
 
Epigenetic mechanisms for progression of prostate cancer.
Rennie PS, Nelson CC.
Cancer Metastasis Rev. 1998-1999;17(4):401-9. (REVIEW)
PMID 10453284
 
Prostate cancer and supplementation with alpha-tocopherol and beta-carotene: incidence and mortality in a controlled trial.
Heinonen OP, Albanes D, Virtamo J, Taylor PR, Huttunen JK, Hartman AM, Haapakoski J, Malila N, Rautalahti M, Ripatti S, Maenpaa H, Teerenhovi L, Koss L, Virolainen M, Edwards BK.
J Natl Cancer Inst. 1998 Mar 18;90(6):440-6.
PMID 9521168
 
Glutathione S-transferase pi in squamous cell carcinoma of the head and neck.
Bentz BG, Haines GK 3rd, Radosevich JA.
Laryngoscope. 2000 Oct;110(10 Pt 1):1642-7.
PMID 11037818
 
Antioxidant enzyme expression and reactive oxygen species damage in prostatic intraepithelial neoplasia and cancer.
Bostwick DG, Alexander EE, Singh R, Shan A, Qian J, Santella RM, Oberley LW, Yan T, Zhong W, Jiang X, Oberley TD.
Cancer. 2000 Jul 1;89(1):123-34.
PMID 10897009
 
Epigenetic lesions causing genetic lesions in human cancer: promoter hypermethylation of DNA repair genes.
Esteller M.
Eur J Cancer. 2000 Dec;36(18):2294-300. (REVIEW)
PMID 11094302
 
Molecular detection of prostate cancer in urine by GSTP1 hypermethylation.
Cairns P, Esteller M, Herman JG, Schoenberg M, Jeronimo C, Sanchez-Cespedes M, Chow NH, Grasso M, Wu L, Westra WB, Sidransky D.
Clin Cancer Res. 2001 Sep;7(9):2727-30.
PMID 11555585
 
Quantitation of GSTP1 methylation in non-neoplastic prostatic tissue and organ-confined prostate adenocarcinoma.
Jeronimo C, Usadel H, Henrique R, Oliveira J, Lopes C, Nelson WG, Sidransky D.
J Natl Cancer Inst. 2001 Nov 21;93(22):1747-52.
PMID 11717336
 
Quantitative GSTP1 hypermethylation in bodily fluids of patients with prostate cancer.
Jeronimo C, Usadel H, Henrique R, Silva C, Oliveira J, Lopes C, Sidransky D.
Urology. 2002 Dec;60(6):1131-5.
PMID 12475696
 
Increased expression of alpha-1-antitrypsin, glutathione S-transferase pi and vascular endothelial growth factor in human pancreatic adenocarcinoma.
Trachte AL, Suthers SE, Lerner MR, Hanas JS, Jupe ER, Sienko AE, Adesina AM, Lightfoot SA, Brackett DJ, Postier RG.
Am J Surg. 2002 Dec;184(6):642-7; discussion 647-8.
PMID 12488200
 
Epigenetics in cancer: implications for early detection and prevention.
Verma M, Srivastava S.
Lancet Oncol. 2002 Dec;3(12):755-63. (REVIEW)
PMID 12473517
 
Quantitative GSTP1 methylation clearly distinguishes benign prostatic tissue and limited prostate adenocarcinoma.
Harden SV, Guo Z, Epstein JI, Sidransky D.
J Urol. 2003 Mar;169(3):1138-42.
PMID 12576869
 
Procaine is a DNA-demethylating agent with growth-inhibitory effects in human cancer cells.
Villar-Garea A, Fraga MF, Espada J, Esteller M.
Cancer Res. 2003 Aug 15;63(16):4984-9.
PMID 12941824
 
Prostate cancer molecular markers GSTP1 and hTERT in expressed prostatic secretions as predictors of biopsy results.
Crocitto LE, Korns D, Kretzner L, Shevchuk T, Blair SL, Wilson TG, Ramin SA, Kawachi MH, Smith SS.
Urology. 2004 Oct;64(4):821-5.
PMID 15491741
 
Promoter hypermethylation profile of kidney cancer.
Dulaimi E, Ibanez de Caceres I, Uzzo RG, Al-Saleem T, Greenberg RE, Polascik TJ, Babb JS, Grizzle WE, Cairns P.
Clin Cancer Res. 2004 Jun 15;10(12 Pt 1):3972-9.
PMID 15217927
 
Detection of GSTP1 methylation in prostatic secretions using combinatorial MSP analysis.
Gonzalgo ML, Nakayama M, Lee SM, De Marzo AM, Nelson WG.
Urology. 2004 Feb;63(2):414-8.
PMID 14972513
 
Molecular detection of prostate cancer: a role for GSTP1 hypermethylation.
Henrique R, Jeronimo C.
Eur Urol. 2004 Nov;46(5):660-9; discussion 669. (REVIEW)
PMID 15474280
 
Plasma glutathione S-Transferase P1-1 as a prognostic factor in patients with advanced non-Hodgkin's lymphoma (stages III and IV).
Katahira T, Takayama T, Miyanishi K, Hayashi T, Ikeda T, Takahashi Y, Takimoto R, Matsunaga T, Kato J, Niitsu Y.
Clin Cancer Res. 2004 Dec 1;10(23):7934-40.
PMID 15585627
 
Preoperative serum DNA GSTP1 CpG island hypermethylation and the risk of early prostate-specific antigen recurrence following radical prostatectomy.
Bastian PJ, Palapattu GS, Lin X, Yegnasubramanian S, Mangold LA, Trock B, Eisenberger MA, Partin AW, Nelson WG.
Clin Cancer Res. 2005 Jun 1;11(11):4037-43.
PMID 15930338
 
Promoter methylation and differential expression of pi-class glutathione S-transferase in endometrial carcinoma.
Chan QK, Khoo US, Chan KY, Ngan HY, Li SS, Chiu PM, Man LS, Ip PP, Xue WC, Cheung AN.
J Mol Diagn. 2005 Feb;7(1):8-16.
PMID 15681469
 
Intake of selenium in the prevention of prostate cancer: a systematic review and meta-analysis.
Etminan M, FitzGerald JM, Gleave M, Chambers K.
Cancer Causes Control. 2005 Nov;16(9):1125-31.
PMID 16184479
 
Glutathione S-transferase T1-1 activity upregulated in transitional cell carcinoma of urinary bladder.
Simic T, Mimic-Oka J, Savic-Radojevic A, Opacic M, Pljesa M, Dragicevic D, Djokic M, Radosavljevic R.
Urology. 2005 May;65(5):1035-40.
PMID 15882759
 
Association of GSTP1 CpG islands hypermethylation with poor prognosis in human breast cancers.
Arai T, Miyoshi Y, Kim SJ, Taguchi T, Tamaki Y, Noguchi S.
Breast Cancer Res Treat. 2006 Nov;100(2):169-76. Epub 2006 Jun 22.
PMID 16791478
 
Epigenetic heterogeneity of high-grade prostatic intraepithelial neoplasia: clues for clonal progression in prostate carcinogenesis.
Henrique R, Jeronimo C, Teixeira MR, Hoque MO, Carvalho AL, Pais I, Ribeiro FR, Oliveira J, Lopes C, Sidransky D.
Mol Cancer Res. 2006 Jan;4(1):1-8.
PMID 16446401
 
Impact of hormonal therapy on the detection of promoter hypermethylation of the detoxifying glutathione-S-transferase P1 gene (GSTP1) in prostate cancer.
Kollermann J, Kempkensteffen C, Helpap B, Schrader M, Krause H, Muller M, Miller K, Schostak M.
BMC Urol. 2006 Jun 27;6:15.
PMID 16803634
 
The emerging roles of DNA methylation in the clinical management of prostate cancer.
Perry AS, Foley R, Woodson K, Lawler M.
Endocr Relat Cancer. 2006 Jun;13(2):357-77. (REVIEW)
PMID 16728568
 
Prognostic value of CpG island hypermethylation at PTGS2, RAR-beta, EDNRB, and other gene loci in patients undergoing radical prostatectomy.
Bastian PJ, Ellinger J, Heukamp LC, Kahl P, Muller SC, von Rucker A.
Eur Urol. 2007 Mar;51(3):665-74; discussion 674. Epub 2006 Aug 23.
PMID 16956712
 
DNA methylation of GSTP1 as biomarker in diagnosis of prostate cancer.
Hopkins TG, Burns PA, Routledge MN.
Urology. 2007 Jan;69(1):11-6. (REVIEW)
PMID 17270599
 
Reduction of GSTP1 expression by DNA methylation correlates with clinicopathological features in pituitary adenomas.
Yuan Y, Qian ZR, Sano T, Asa SL, Yamada S, Kagawa N, Kudo E.
Mod Pathol. 2008 Jul;21(7):856-65. Epub 2008 Apr 18.
PMID 18425080
 
Borderline and malignant phyllodes tumors display similar promoter methylation profiles.
Kim JH, Choi YD, Lee JS, Lee JH, Nam JH, Choi C, Park MH, Yoon JH.
Virchows Arch. 2009 Dec;455(6):469-75. Epub 2009 Nov 19.
PMID 19924440
 
Advances in biomarkers for the early diagnosis of prostate cancer.
Cao DL, Yao XD.
Chin J Cancer. 2010 Feb;29(2):229-33.
PMID 20109357
 
Promoter demethylation and chromatin remodeling by green tea polyphenols leads to re-expression of GSTP1 in human prostate cancer cells.
Pandey M, Shukla S, Gupta S.
Int J Cancer. 2010 Jun 1;126(11):2520-33.
PMID 19856314
 
Written2010-02Isabelle Meiers
Tunbridge Wells NHS Trust, Preston Hall Hospital, Royal British Legion Village, Aylesford, Kent, ME20 7NJ, UK

Citation

This paper should be referenced as such :
Meiers, I
Glutathione S-Transferase pi (GSTP1)
Atlas Genet Cytogenet Oncol Haematol. 2010;14(12):1181-1185.
Free journal version : [ pdf ]   [ DOI ]
On line version : http://AtlasGeneticsOncology.org/Deep/GSTP1inCancerID20084.htm