Skip to main content

Glutathione S-transferase theta 1 (GSTT1) deletion polymorphism and susceptibility to head and neck carcinoma: a systematic review with five analyses

Abstract

Glutathione S-transferase theta 1 (GSTT1) enzyme plays a key role in the neutralization of electrophilic compounds such as carcinogens. Herein, we aimed to evaluate GSTT1 deletion polymorphism and susceptibility to head and neck carcinoma (HNC) according to 107 articles in a systematic review with five analyses. The databases of PubMed/Medline, Web of Science, Scopus, and Cochrane Library from the beginning of each database until June 21, 2023, with no restrictions to identify pertinent articles. The RevMan 5.3 software was used to calculate the effect sizes, which were displayed as the odds ratio (OR) along with a 95% confidence interval (CI). Both the publication bias and sensitivity analyses were performed using the CMA 3.0 software. A trial sequential analysis (TSA) was conducted. Of the 1966 records retrieved from four databases, 107 articles were included in the analysis. The combined analysis revealed that the pooled OR was 1.28 (95% CI: 1.14 to 1.44; p-value < 0.0001). The pooled OR was highest in mixed ethnicity. Nasopharyngeal cancer had the highest OR (1.84), followed by oral cancer (OR = 1.20), and laryngeal cancer (OR = 1.17). Studies with less than 200 samples had a higher OR compared to those with 200 or more samples. The studies with a quality score of 7 or more had a higher OR compared to those with a score of less than 7. When both age and sex are considered, while the OR of 1.42 is significant, the high heterogeneity suggests caution in interpreting these results. There is no evidence of publication bias. TSA reported that the study does not have sufficient statistical power. This comprehensive meta-analysis revealed a significant association between the GSTT1 null genotype and an increased risk of HNC, with variations based on factors such as ethnicity, cancer type, sample size, control source, and quality score.

Peer Review reports

Introduction

Head and neck cancer (HNC) is a group of cancers that starts within the nose, mouth, throat, sinuses, larynx, or salivary glands (1, 2). According to the most recent GLOBOCAN estimates from 2020, HNC ranks as the seventh most prevalent cancer worldwide. It is responsible for approximately 890,000 new cases annually, representing roughly 4.5% of all cancer diagnoses globally (3). Additionally, HNC leads to approximately 450,000 deaths each year, accounting for approximately 4.6% of total cancer-related deaths worldwide (3). Men are more prone to HNC than women, irrespective of their alcohol consumption or tobacco use habits. This gender disparity in the incidence of HNC becomes particularly noticeable in individuals in their 60s. The lower regions of the upper aerodigestive tract, such as the larynx and hypopharynx, are the most commonly affected areas (4). Although tobacco and alcohol are the main risk factors for the development of HNC, a significant correlation has been observed between a subset of HNC and the human papillomavirus in epidemiological studies (5). A number of single nucleotide polymorphisms can associate with the risk of HNC reported in recent meta-analyses (6–10).

Glutathione S-transferase theta 1 (GSTT1) gene produces an enzyme that plays a key role in the neutralization of electrophilic compounds. These compounds include carcinogens, therapeutic drugs, environmental toxins, and by-products of oxidative stress (11). GSTT1 is located at 22q11.23 with a 480 bp fragment using specific primers (12, 13). and the GSTT1 null genotype, which results from a homozygous deletion of the GSTT1 gene, leads to a lack of enzyme activity (14).

However, the association between GSTT1 deletion polymorphism and HNC remains unclear due to inconsistent findings among studies. Some studies have reported a significant association (15–17), while others have found no such link (18–21). These discrepancies could be due to differences in study design, sample size, population characteristics, or methods of genotyping.

Several meta-analyses (22–36) reported the association of GSTT1 deletion polymorphism and HNC susceptibility. Three meta-analyses (24, 28, 32) were published after 2015 and all three meta-analyses just reported GSTT1 deletion polymorphism in oral cancer. The last meta-analysis (32) included 36 studies.

This systematic review aimed to provide a more definitive answer to this question by combining the results of 107 articles (50 studies reported oral cancer).

Materials and methods

Study design and registration

The meta-analysis was conducted by the protocols of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) (37). The question posed in terms of PICO (population, intervention, comparison, and outcome) was (38): Does the deletion polymorphism of GSTT1 associate with the susceptibility to HNC in case-control studies? (Population (P): Patients with HNC. Intervention (I): GSTT1 deletion polymorphism. Comparison (C): Control subjects (non-HNC individuals). Outcome (O): Correlation with susceptibility to HNC in case-control studies). The study has not registered in any database.

Identification of articles

An exhaustive search was carried out by one author (M.S.) in the databases of PubMed/Medline, Web of Science, Scopus, and Cochrane Library from the beginning of each database until June 21, 2023, with no restrictions to identify pertinent articles. The titles/abstracts of the articles were evaluated by the same author (M.S), who also downloaded the full texts of the articles that satisfied the eligibility criteria. The search strategy encompassed: (“Glutathione S-transferase” OR “GSTT1” OR “GST”) AND (“head and neck” OR “oral” OR “OSCC” OR “tongue” OR “mouth” OR “HNSCC“ OR “nasopharyngeal” OR " oropharyngeal " OR “nasopharynx” OR “salivary gland” OR “hypopharyngeal” OR “pharyngeal” OR “pharynx” OR “oral cavity” OR “hypopharynx” OR “laryngeal” OR “larynx”) AND (“carcinoma*” OR “tumor*” OR “cancer*” OR “neoplasm*”) AND (“allele*” OR “variant*” OR “genotype*” OR “gene*” OR “polymorphism*”). To ensure no relevant study was overlooked, the reference lists of the articles were also scrutinized. The search and selection process was re-verified by another author (M.M.I.). In case of any disagreement between the two authors, a third author (M.S.) resolved it.

Inclusion and exclusion criteria

The inclusion criteria were as follows: (1) Studies of the case-control type that examined GSTT1 deletion polymorphism in patients with HNC and control subjects. (2) HNC was diagnosed clinically and pathologically. (3) Patients with HNC did not have any other systemic diseases and controls were either healthy or free from cancer. Conversely, the exclusion criteria included: review articles, meta-analyses, systematic reviews, articles that had incomplete data or lacked a control group, studies conducted on animals, conference papers, comment papers, duplicate studies, book chapters, studies that included controls with the disease, and studies that included cases under treatment.

Data summary

The information from the studies incorporated into the meta-analysis was independently gathered by two authors (M.S. and S.S.). Any disagreements were settled through discussion.

Quality evaluation

One author (M.S) performed the quality scoring using the Newcastle-Ottawa Scale (NOS) tool (39). This tool evaluates a study based on three broad perspectives: the selection of the study groups (4 scores), the comparability of the groups (2 scores), and the ascertainment of either the exposure or outcome of interest (3 scores) for case-control studies. The maximum possible score is nine, and a score of ≥ 7 is considered to be of high quality. Another author (N.K.) re-checked the scores. Disagreement between the authors was resolved by a short discussion.

Statistical analyses

The Review Manager 5.3 (RevMan 5.3) software was used to calculate the effect sizes, which were displayed as the odds ratio (OR) along with a 95% confidence interval (CI) for the prevalence of the null genotype of GSTT1 polymorphism in HNC patients and controls. The significance of the pooled OR was determined using the Z-test, with a two-sided p-value less than 0.05 deemed significant. A random-effects model (40) was employed if Pheterogeneity was < 0.10 (I2 > 50%), indicating significant heterogeneity. If the heterogeneity was not significant, a fixed-effect model (41) was applied.

A subgroup analysis was conducted to ascertain whether the combined effect sizes in these subgroups differed significantly from one another. Furthermore, a meta-regression analysis using a random-effects model was carried out to illustrate a linear correlation between auxiliary variables in the study and the effect size.

The extent of publication bias was assessed using the funnel plot and Egger’s regression test. The possibility of publication bias was evaluated using Begg’s funnel plot and Begg’s test, and the level of asymmetry was tested with Egger’s test. The p-values from both Egger’s and Begg’s tests were obtained, and a 2-sided p-value less than 0.10 indicated the existence of publication bias. In terms of sensitivity analysis, both “one-study-removed” (This is done to determine if any single study has a disproportionate impact on the overall estimate.) and “cumulative” (This is done to assess the impact of each additional study on the overall estimate.) analyses were employed to assess the stability and consistency of the pooled SMDs. Both the publication bias and sensitivity analyses were performed using the Comprehensive Meta-Analysis version 3.0 (CMA 3.0) software.

The Radial plot, also known as the Galbraith plot, was designed using the NCSS 2021 version 21.0.2 software. This plot displays the z-statistic (obtained by dividing by the standard error) on the vertical axis and the weight measurement on the horizontal axis (42). A p-value less than 0.05, indicates statistically significant heterogeneity.

To mitigate the risk of false-positive or negative conclusions from meta-analyses (43), a TSA was conducted using TSA software (version 0.9.5.10 beta) from the Copenhagen Trial Unit, Centre for Clinical Intervention Research, Rigshospitalet, Copenhagen, Denmark (44). TSA allows for the testing of a futility threshold to establish a result of no effect before reaching the necessary information size. The required information size (RIS) was computed with an alpha risk of 5%, a beta risk of 20%, and a two-sided boundary type. Heterogeneity (D2) was evaluated for the prevalence of the null genotype of GSTT1 polymorphism in HNC patients and controls. If the Z-curve reached the RIS line or traced the boundary line or futility area, it suggested that the studies included a sufficient number of cases and that the conclusions were reliable. If not, it indicated that the information available was insufficient and additional data was required.

Results

Study selection

A total of 1966 records were initially retrieved from four databases, along with 8 records from other electronic sources (Fig. 1). After the removal of duplicates, 1052 records remained and were screened. Of these, 887 records were deemed irrelevant and subsequently removed. This left 165 full-text articles that met the eligibility criteria. However, 58 of these were excluded for various reasons. Ultimately, 107 full-text articles were included in the analysis.

Fig. 1
figure 1

Flowchart of study selection for systematic review and meta-analysis

Study’s characteristics

Table 1 presents a comprehensive list of 107 articles (15–21, 45–142) including 109 studies conducted on the null genotype of GSTT1 polymorphism in HNC patients and controls. The studies span multiple countries and ethnicities, with a variety of cancer types and control sources. Each study includes the number of cases and controls, with some studies matching controls based on age and sex. The quality score of each study is also provided, offering insight into the reliability of the data.

Table 1 Characteristics of the studies

Meta-analysis

A forest plot analysis using a random-effects model was conducted to examine the association between GSTT1 polymorphism and the risk of HNC, as depicted in Fig. 2. The combined analysis revealed that the pooled OR was 1.28, with a 95% CI ranging from 1.14 to 1.44. This result was statistically significant with a p-value less than 0.0001. However, there was substantial heterogeneity among the studies, as indicated by an I2 value of 82%. The result suggests that there is a significant association between GSTT1 polymorphism and the risk of HNC, with the null genotype of GSTT1 associated with a 28% increased risk of HNC. However, due to the high heterogeneity (I2 = 83%), the results should be interpreted with caution as the studies included in the analysis may have varied in aspects such as study and population characteristics.

Fig. 2
figure 2

Forest plot analysis of the association between GSTT1 polymorphism and the risk of head and neck cancer

Subgroup analysis

Table 2 presents a subgroup analysis of the association between GSTT1 polymorphism and the risk of HNC. The subgroups are divided based on ethnicity, cancer type, sample size, control source, and quality score. In terms of ethnicity, the pooled OR was highest in Asian ethnicities (OR = 1.31), followed by Mixed (OR = 1.28), and Caucasians (OR = 1.17). For cancer type, nasopharyngeal cancer had the highest OR (1.84), followed by oral cancer with an OR of 1.20, and laryngeal cancer with an OR of 1.17. When considering sample size, studies with less than 200 samples had a higher OR (1.59) compared to those with 200 or more samples (OR = 1.23). The control source did not significantly affect the OR, with both population-based and hospital-based controls showing similar ORs of 1.29 and 1.24 respectively. The studies with a quality score of 7 or more had a higher OR (1.37) compared to those with a score of less than 7 (OR = 1.05). When considering age, the OR of 1.41 suggests a higher risk, but the high heterogeneity and p-value of 0.31 indicate that this result is not statistically significant. The analysis based on sex shows a significant association, with an OR of 1.36 and a p-value of 0.006. However, when both age and sex are considered, while the OR of 1.42 is significant, the high heterogeneity suggests caution in interpreting these results. Finally, in the group where neither age nor sex was considered, no significant association was found. These findings highlight the complexity of the relationship between the GSTT1 null genotype and HNC risk, and how it can be influenced by factors such as age and sex. It’s important to note that all these results should be interpreted with caution due to the high heterogeneity observed in most subgroups (I2 > 50%).

Table 2 Subgroup analysis

Meta-regression

Table 3 presents a meta-regression analysis of the variables: publication year, sample size, and quality score. For the publication year, the coefficient is ˗ 0.0003 with a p-value of 0.1213. For the sample size, the coefficient is -0.0002 with a p-value of 0.1965. For the quality score, the coefficient is 0.1283 with a p-value of 0.0147. In this case, only the quality score shows statistical significance as its p-value is less than 0.05. The results indicate that quality score increased, the effect size significantly increased.

Table 3 Meta-regression analysis

Sensitivity analysis

The sensitivity analysis, which included both one-study-removed and cumulative analyses, showed that the results were robust and reliable. In this case, the fact that the results did not change significantly in either analysis indicates that no single study unduly influenced the results and that the results were consistent across all studies. This adds to the validity and reliability of your findings.

Publication bias.

Figure 3 shows the funnel plot of the association between GSTT1 polymorphism and the risk of HNC. The p-values for both Egger’s test (0.895) and Begg’s test (0.108) are greater than 0.10. This suggests that there is no evidence of publication bias in the meta-analysis. Therefore, it can be interpreted that your results are likely not influenced by publication bias.

Fig. 3
figure 3

Funnel plot of the association between GSTT1 polymorphism and the risk of head and neck cancer

Heterogeneity analysis

Figure 4 identifies the radial plot of the association between GSTT1 polymorphism and the risk of HNC. The p-value of less than 0.0001 suggests that there is significant heterogeneity among the studies included in the meta-analysis. This means that there are substantial differences in the results of these studies that cannot be attributed to chance alone. The presence of outlier data in some studies could be contributing to this heterogeneity. It’s important to investigate these outliers further to understand their source and consider their impact on the overall results of the meta-analysis. Therefore, while your analysis shows a significant association between GSTT1 polymorphism and the risk of HNC, the high heterogeneity suggests that caution should be taken when interpreting these results. Further research may be needed to explore the sources of this heterogeneity.

Fig. 4
figure 4

Radial plot of the association between GSTT1 polymorphism and the risk of head and neck cancer

TSA

Figure 5 shows the TSA of the association between GSTT1 polymorphism and the risk of HNC (D2 = 85%, the incidence in the intervention arm (IIA) = 28.24%; the incidence in the control arm (ICA) = 25.76%). IIA is higher than ICA. This indicates that the occurrence of the GSTT1 null genotype under study is more frequent in the HNC group compared to the control group. The D2 value represents the diversity (or heterogeneity) of the study results. A high D2 value suggests a high degree of variability in the study results, which could be due to differences in study characteristics. The Z-curve crossing the boundary for harm suggests that the GSTT1 polymorphism being studied may have harmful effects. However, since the number of patients in the study (43,555) is less than the RIS (65,384), the study does not have sufficient statistical power. This means that the results should be interpreted with caution as they may be prone to random errors. In other words, while the current data suggests potential harm, it does not conclusively prove it due to insufficient information size. Therefore, more research or larger studies may be needed to conclusively determine whether the GSTT1 polymorphism is harmful.

Fig. 5
figure 5

A trial sequential analysis of the association between GSTT1 polymorphism and the risk of head and neck cancer

Discussion

A meta-analysis found that people with a certain gene variant (GSTT1 null) had a higher risk of HNC, especially nasopharyngeal cancer. However, the studies included in the meta-analysis were very different from each other and had some limitations. Subgroup analysis revealed differences in ORs based on factors such as ethnicity, cancer type, sample size, control source, and quality score. The quality score was found to significantly impact the effect size in the meta-regression analysis. Despite these findings, the high heterogeneity and the smaller sample size compared to the RIS suggest that these results should be interpreted cautiously.

The relationship between GSTT1 and HNC can be influenced by several factors. A study presented the results of the analysis of the joint effects or interaction between tobacco use and GSTT1 null genotype (111). Another study (89) discovered that the GSTT1 null genotype was linked to a higher risk among individuals who had a longer history of smoking. In addition, interaction between GSTT1 polymorphism with other genes such as GSTs and CYPs can affect the risk of HNC (63, 129, 143, 144). The studies recommended also that occupational hazards can affect the association between GST polymorphisms and HNC risk (145–147). In this meta-analysis, we couldn’t check these factors due to a lack of sufficient data that future studies can check them in HNC.

GST is a family of enzymes that play an important role in detoxification by catalyzing the conjugation of many hydrophobic and electrophilic compounds with reduced glutathione (148, 149). They have been linked to the development of various cancers (150–153), but the specific role they play in HNC may require further research. The protection provided by the GSTT1 enzyme is viewed as more comprehensive, given that the gene is not only expressed in the liver but also erythrocytes. This results in a systemic action of the enzyme (154).

Research on the GSTT1 null genotype indicates that in the United States, the absence of GSTT1 is less prevalent compared to the GSTM1 deletion genotype. Among individuals of European descent, it’s found that 15–31% lack a functional GSTT1 enzyme. For African Americans, the frequencies range from 22 to 29%. Meanwhile, individuals of Hispanic origin exhibit GSTT1 deletions at a rate of 10–12% (155–158). In terms of ethnicity, Asians are more susceptible to HNC associated with GSTT1 null genotype, compared to their European and American counterparts (107). The present meta-analysis reported that GSTT1 polymorphism has an association with the risk of HNC in Asians and mixed ethnicities, not Caucasians. Therefore, the prevalence of GSTT1 null genotype may differ by geographic region (30).

In diabetic patients, the GSTM1 null genotype was found to be significantly more prevalent in the 24–36 year age group compared to other age groups (159). The present meta-analysis reported the relationship between the GSTT1 null genotype and HNC risk, and how it can be influenced by factors such as age and sex. Therefore, outliers based on radial plot, lack of sufficient cases based on TSA, variation in age range and sex percentage can be main factors for a high heterogeneity in this meta-analysis.

The present systematic review and meta-analysis included four limitations: (1) there was substantial heterogeneity among the studies included in the meta-analysis. (2) Due to the high heterogeneity, the results should be interpreted with caution. (3) the study may not have sufficient statistical power to detect small effect sizes or rare events. (4) the study was based on published data rather than individual patient data, which may limit the ability to control for potential confounding factors. The present systematic review and meta-analysis included four strengths: (1) the study included a comprehensive analysis of 107 full-text articles, providing a broad overview of the existing literature on the association between GSTT1 polymorphism and the risk of HNC. (2) Subgroup analysis allowed for a more nuanced understanding of how the factors might influence the association between GSTT1 polymorphism and HNC risk. (3) Meta-regression analysis provided insights into how these variables might impact the effect size. (4) the study found no evidence of publication bias, suggesting that the results are not skewed by the selective publication of studies with positive results.

Conclusions

This comprehensive meta-analysis revealed a significant association between the GSTT1 null genotype and an increased risk of HNC, with variations based on factors such as ethnicity, cancer type, sample size, control source, and quality score. Despite the robustness of the results, there was high heterogeneity among studies and limited statistical power due to a smaller number of cases. From a clinical perspective, these findings underscore the potential of the GSTT1 null genotype as a genetic marker for HNC susceptibility, which could have significant implications for early detection and prevention strategies. However, further research is needed to confirm these findings and elucidate the underlying mechanisms. This study sets the stage for future research in this area, highlighting the importance of considering factors such as ethnicity, cancer type, sample size, control source, and quality score in understanding the complex relationship between GSTT1 null genotype and HNC risk.

Data availability

All data generated or analyzed during this study are included in this published article and its Supplementary information files.

References

  • 1. Mutlu M, Korkmaz MH, Simsek E, Terzi E, Oz Bedir BE, Uysal TK, et al. Do CO2 and oxidative stress induce cancer?: a brief study about the evaluation of PON 1, CAT, CA and XO enzyme levels on head and neck cancer patients. Journal of Enzyme Inhibition and Medicinal Chemistry. 2019;34(1):459 − 64.

  • 2. Bose P, Brockton NT, Dort JC. Head and neck cancer: from anatomy to biology. International journal of cancer. 2013;133(9):2013-23.

  • 3. Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA: a cancer journal for clinicians. 2021;71(3):209 − 49.

  • 4. Park J-O, Nam I-C, Kim C-S, Park S-J, Lee D-H, Kim H-B, et al. Sex differences in the prevalence of head and neck cancers: a 10-year follow-up study of 10 million healthy people. Cancers. 2022;14(10):2521.

  • 5. Ragin C, Modugno F, Gollin S. The epidemiology and risk factors of head and neck cancer: a focus on human papillomavirus. Journal of dental research. 2007;86(2):104 − 14.

  • 6. Imani MM, Basamtabar M, Akbari S, Sadeghi E, Sadeghi M. Relationship between XPA, XPB/ERCC3, XPF/ERCC4, and XPG/ERCC5 Polymorphisms and the Susceptibility to Head and Neck Carcinoma: A Systematic Review, Meta-Analysis, and Trial Sequential Analysis. Medicina (Kaunas). 2024;60(3).

  • 7. Imani MM, Akbari S, Shalchi M, Sadeghi E, Sadeghi M. Relationship between ERCC1 and XPC polymorphisms and the susceptibility to head and neck carcinoma: A systematic review, meta-analysis, and trial sequential analysis. Arch Oral Biol. 2024;162:105955.

  • 8. Imani MM, Moradi MM, Rezaei F, Mozaffari HR, Sharifi R, Safaei M, et al. Association between alcohol dehydrogenase polymorphisms (rs1229984, rs1573496, rs1154460, and rs284787) and susceptibility to head and neck cancers: A systematic review and meta-analysis. Arch Oral Biol. 2024;160:105898.

  • 9. Mohammadi H, Momeni Roochi M, Rezaei F, Garajei A, Heidar H, Ghaderi B, et al. Association between the CYP1A1 MspI polymorphism and risk of head and neck cancer: a meta-analysis. Sci Rep. 2022;12(1):1527.

  • 10. Mohammadi H, Roochi MM, Sadeghi M, Garajei A, Heidar H, Ghaderi B, et al. Association of N-acetyltransferases 1 and 2 Polymorphisms with Susceptibility to Head and Neck Cancers-A Meta-Analysis, Meta-Regression, and Trial Sequential Analysis. Medicina (Kaunas). 2021;57(10).

  • 11. Carlsten C, Sagoo G, Frodsham A, Burke W, Higgins J. Glutathione S-transferase M1 (GSTM1) polymorphisms and lung cancer: a literature-based systematic HuGE review and meta-analysis. American journal of epidemiology. 2008;167(7):759 − 74.

  • 12. Parl FF. Glutathione S-transferase genotypes and cancer risk. Cancer letters. 2005;221(2):123-9.

  • 13. Ali NT, Ahmed OYM, Mohammed FO, Haroun HM. Prevalence of Glutathione S Transferase (GSTM1, GSTP1 and GSTT1) Genes Polymorphisms among Pediatric Sudanese Patients with Sickle Cell Anemia. Asian Hematology Research Journal. 2021;4(1):1–11.

  • 14. Sprenger R, Schlagenhaufer R, Kerb R, Bruhn C, Brockmöller J, Roots I, et al. Characterization of the glutathione S-transferase GSTT1 deletion: discrimination of all genotypes by polymerase chain reaction indicates a trimodular genotype–phenotype correlation. Pharmacogenetics and Genomics. 2000;10(6):557 − 65.

  • 15. Alamgir MM, Jamal Q, Mirza T. Genetic profiles of different ethnicities living in Karachi as regards to tobacco-metabolising enzyme systems and the risk of oral cancer. J Pak Med Assoc. 2022;72(6):1092-6.

  • 16. D’ Mello S, Bavle RM, Paremala K, Makarla S, Sudhakara M, Bhatt M. The synergy of tobacco and alcohol and glutathione S-transferase θ 1 gene deletion and oral squamous cell carcinoma. J Oral Maxillofac Pathol. 2016;20(3):348 − 53.

  • 17. Drummond SN, Gomez RS, Motta Noronha JC, Pordeus IA, Barbosa AA, De Marco L. Association between GSTT-1 gene deletion and the susceptibility to oral squamous cell carcinoma in cigarette-smoking subjects. Oral Oncol. 2005;41(5):515-9.

  • 18. Cheng YJ, Chien YC, Hildesheim A, Hsu MM, Chen IH, Chuang J, et al. No association between genetic polymorphisms of CYP1A1, GSTM1, GSTT1, GSTP1, NAT2, and nasopharyngeal carcinoma in Taiwan. Cancer Epidemiol Biomarkers Prev. 2003;12(2):179 − 80.

  • 19. Ko Y, Abel J, Harth V, Bröde P, Antony C, Donat S, et al. Association of CYP1B1 codon 432 mutant allele in head and neck squamous cell cancer is reflected by somatic mutations of p53 in tumor tissue. Cancer Res. 2001;61(11):4398 − 404.

  • 20. Xie H, Hou L, Shields PG, Winn DM, Gridley G, Bravo-Otero E, et al. Metabolic polymorphisms, smoking, and oral cancer in Puerto Rico. Oncol Res. 2004;14(6):315 − 20.

  • 21. Yadav DS, Devi TR, Ihsan R, Mishra AK, Kaushal M, Chauhan PS, et al. Polymorphisms of glutathione-S-transferase genes and the risk of aerodigestive tract cancers in the Northeast Indian population. Genetic Testing and Molecular Biomarkers. 2010;14(5):715 − 23.

  • 22. Ye Z, Song H, Guo Y. Glutathione S-transferase M1, T1 status and the risk of head and neck cancer: a meta-analysis. Journal of medical genetics. 2004;41(5):360-5.

  • 23. Liu R-R, Chen J-C, Li M-D, Li T, Tan Y, Zhang M. A meta-analysis of glutathione S-transferase M1 and T1 genetic polymorphism in relation to susceptibility to nasopharyngeal carcinoma. International Journal of Clinical and Experimental Medicine. 2015;8(7):10626.

  • 24. Lopes A, Tacca A, Nogueira N, Sodario J, Vilanova-Costa C, Saddi V, et al. Evidence of association of glutathione-S transferase GSTT1 and GSTM1 null genotypes with susceptibility to oral cancer based on meta-analysis. Genet Mol Res. 2020;19.

  • 25. Zhang Z-J, Hao K, Shi R, Zhao G, Jiang G-X, Song Y, et al. Glutathione S-transferase M1 (GSTM1) and glutathione S-transferase T1 (GSTT1) null polymorphisms, smoking, and their interaction in oral cancer: a HuGE review and meta-analysis. American journal of epidemiology. 2011;173(8):847 − 57.

  • 26. Kumar V, Murthy AK, Suresh K. Glutathione S-transferase M1 and T1 status and the risk of laryngeal cancer: a meta-analysis. Asian Pac J Cancer Prev. 2011;12(9):2221-6.

  • 27. Li Q, Liu M. Glutathione S-transferase T1 null genotype and laryngeal cancer risk: a meta-analysis. Tumor Biology. 2014;35:8781-5.

  • 28. Li J-Y, Huang L-N, Xue H-L, Zhu Q-Q, Li C-H. Glutathione S-transferase mu-1, glutathione S-transferase theta-1 null genotypes, and oral cancer risk: A meta-analysis in the Chinese population. Journal of Cancer Research and Therapeutics. 2018;14(Suppl 5):S1052-S6.

  • 29. Zhuo X, Cai L, Xiang Z, Li Q, Zhang X. GSTMI and GSTTI polymorphisms and nasopharyngeal cancer risk: an evidence-based meta-analysis. Journal of Experimental & Clinical Cancer Research. 2009;28(1):1–8.

  • 30. Hashibe M, Brennan P, Strange RC, Bhisey R, Cascorbi I, Lazarus P, et al. Meta-and pooled analyses of GSTM1, GSTT1, GSTP1, and CYP1A1 genotypes and risk of head and neck cancer. Cancer Epidemiology Biomarkers & Prevention. 2003;12(12):1509-17.

  • 31. Murthy AK, Kumar V, Suresh K. Meta-analysis of GSTM1 and GSTT1 polymorphisms and risk of nasopharyngeal cancer. Asian Pacific Journal of Cancer Prevention. 2013;14(3):1697 − 701.

  • 32. Kumar KV, Goturi A, Nagaraj M, Goud ESS. Null genotypes of Glutathione S-transferase M1 and T1 and risk of oral cancer: A meta-analysis. Journal of Oral and Maxillofacial Pathology: JOMFP. 2022;26(4):592.

  • 33. Peng J, Liu H-Z, Zhu Y-J. Null Glutathione S-transferase T1 and M1 genotypes and oral cancer susceptibility in China and India-a meta-analysis. Asian Pacific Journal of Cancer Prevention. 2014;15(1):287 − 90.

  • 34. Zhang Y, Ni Y, Zhang H, Pan Y, Ma J, Wang L. Association between GSTM1 and GSTT1 allelic variants and head and neck squamous cell cancinoma. 2012.

  • 35. Dong G, Tian Y, Chen S, Xu X, Zheng J, Li T. Glutathione S-transferase T1 null genotype is associated with oral cancer susceptibility in Asian populations. Tumor Biology. 2013;34:1753-7.

  • 36. Wei Y, Zhou T, Lin H, Sun M, Wang D, Li H, et al. Significant associations between GSTM1/GSTT1 polymorphisms and nasopharyngeal cancer risk. Tumor Biology. 2013;34:887 − 94.

  • 37. Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. International journal of surgery. 2021;88:105906.

  • 38. Cumpston MS, McKenzie JE, Thomas J, Brennan SE. Current practice in systematic reviews including the ‘PICO for each synthesis’ and methods other than meta-analysis: protocol for a cross-sectional study [version 1; peer review: awaiting peer review]. F1000Research. 2020;9.

  • 39. Wells GA, Shea B, O’Connell D, Peterson J, Welch V, Losos M, et al. Newcastle-Ottawa Scale (NOS) for assessing the quality of nonrandomised studies in meta-analyses. Available online: http://wwwohrica/programs/clinical_epidemiology/oxfordasp (accessed on 12 January 2016). 2000.

  • 40. DerSimonian R, Laird N. Meta-analysis in clinical trials revisited. Contemporary clinical trials. 2015;45:139 − 45.

  • 41. Mantel N, Haenszel W. Statistical aspects of the analysis of data from retrospective studies of disease. Journal of the national cancer institute. 1959;22(4):719 − 48.

  • 42. Galbraith R. Graphical display of estimates having differing standard errors. Technometrics. 1988;30(3):271 − 81.

  • 43. Imberger G, Thorlund K, Gluud C, Wetterslev J. False-positive findings in Cochrane meta-analyses with and without application of trial sequential analysis: an empirical review. BMJ open. 2016;6(8):e011890.

  • 44. Wetterslev J, Jakobsen JC, Gluud C. Trial sequential analysis in systematic reviews with meta-analysis. BMC medical research methodology. 2017;17(1):1–18.

  • 45. Acar H, Ozturk K, Muslumanoglu MH, Yildirim MS, Cora T, Cilingir O, et al. Relation of glutathione S-transferase genotypes (GSTM1 and GSTT1) to laryngeal squamous cell carcinoma risk. Cancer Genet Cytogenet. 2006;169(2):89–93.

  • 46. Amtha R, Ching CS, Zain R, Razak IA, Basuki B, Roeslan BO, et al. GSTM1, GSTT1 and CYP1A1 polymorphisms and risk of oral cancer: a case-control study in Jakarta, Indonesia. Asian Pac J Cancer Prev. 2009;10(1):21 − 6.

  • 47. Anantharaman D, Chaubal PM, Kannan S, Bhisey RA, Mahimkar MB. Susceptibility to oral cancer by genetic polymorphisms at CYP1A1, GSTM1 and GSTT1 loci among Indians: tobacco exposure as a risk modulator. Carcinogenesis. 2007;28(7):1455-62.

  • 48. Anantharaman D, Samant TA, Sen S, Mahimkar MB. Polymorphisms in tobacco metabolism and DNA repair genes modulate oral precancer and cancer risk. Oral Oncol. 2011;47(9):866 − 72.

  • 49. Bathi RJ, Rao R, Mutalik S. GST null genotype and antioxidants: risk indicators for oral pre-cancer and cancer. Indian J Dent Res. 2009;20(3):298–303.

  • 50. Ben Chaaben A, Mariaselvam C, Salah S, Busson M, Dulphy N, Douik H, et al. Polymorphisms in oxidative stress-related genes are associated with nasopharyngeal carcinoma susceptibility. Immunobiology. 2015;220(1):20 − 5.

  • 51. Bendjemana K, Abdennebi M, Gara S, Jmal A, Ghanem A, Touati S, et al. [Genetic polymorphism of gluthation-S transferases and N-acetyl transferases 2 and nasopharyngeal carcinoma: the Tunisia experience]. Bull Cancer. 2006;93(3):297–302.

  • 52. Bendjemana K, Douik H, Hamada Y, Fercha A, Bouakkaz A, Habibatni S, et al. GSTM1 and GSTT1 polymorphisms, tobacco use as risk factors for nasopharyngeal carcinoma in maghreb population—A case-control study. J Afr Cancer. 2014;6(11–16).

  • 53. Biselli JM, de Angelo Calsaverini Leal RC, Ruiz MT, Goloni-Bertollo EM, Maníglia JV, Rossit AR, et al. GSTT1 and GSTM1 polymorphism in cigarette smokers with head and neck squamous cell carcinoma. Braz J Otorhinolaryngol. 2006;72(5):654-8.

  • 54. Boccia S, Cadoni G, Sayed-Tabatabaei FA, Volante M, Arzani D, De Lauretis A, et al. CYP1A1, CYP2E1, GSTM1, GSTT1, EPHX1 exons 3 and 4, and NAT2 polymorphisms, smoking, consumption of alcohol and fruit and vegetables and risk of head and neck cancer. J Cancer Res Clin Oncol. 2008;134(1):93–100.

  • 55. Buch SC, Notani PN, Bhisey RA. Polymorphism at GSTM1, GSTM3 and GSTT1 gene loci and susceptibility to oral cancer in an Indian population. Carcinogenesis. 2002;23(5):803-7.

  • 56. Buch SC, Nazar-Stewart V, Weissfeld JL, Romkes M. Case–control study of oral and oropharyngeal cancer in whites and genetic variation in eight metabolic enzymes. Head & Neck: Journal for the Sciences and Specialties of the Head and Neck. 2008;30(9):1139-47.

  • 57. Cabelguenne A, Loriot MA, Stucker I, Blons H, Koum-Besson E, Brasnu D, et al. Glutathione-associated enzymes in head and neck squamous cell carcinoma and response to cisplatin-based neoadjuvant chemotherapy. Int J Cancer. 2001;93(5):725 − 30.

  • 58. Capoluongo E, Almadori G, Concolino P, Bussu F, Santonocito C, Vendittelli F, et al. GSTT1 and GSTM1 allelic polymorphisms in head and neck cancer patients from Italian Lazio Region. Clin Chim Acta. 2007;376(1–2):174-8.

  • 59. Chatterjee S, Dhar S, Sengupta B, Ghosh A, De M, Roy S, et al. Polymorphisms of CYP1A1, GSTM1 and GSTT1 Loci as the Genetic Predispositions of Oral Cancers and Other Oral Pathologies: Tobacco and Alcohol as Risk Modifiers. Indian J Clin Biochem. 2010;25(3):260 − 72.

  • 60. Chatzimichalis M, Xenellis J, Tzagaroulakis A, Sarof P, Banis K, Gazouli M, et al. GSTT1, GSTM1, GSTM3 and NAT2 polymorphisms in laryngeal squamous cell carcinoma in a Greek population. J Laryngol Otol. 2010;124(3):318 − 23.

  • 61. Chen MK, Tsai HT, Chung TT, Su SC, Kao TY, Tseng HC, et al. Glutathione S-transferase P1 and alpha gene variants; role in susceptibility and tumor size development of oral cancer. Head Neck. 2010;32(8):1079-87.

  • 62. Cheng L, Sturgis EM, Eicher SA, Char D, Spitz MR, Wei Q. Glutathione-S-transferase polymorphisms and risk of squamous-cell carcinoma of the head and neck. Int J Cancer. 1999;84(3):220-4.

  • 63. Choudhury JH, Singh SA, Kundu S, Choudhury B, Talukdar FR, Srivasta S, et al. Tobacco carcinogen-metabolizing genes CYP1A1, GSTM1, and GSTT1 polymorphisms and their interaction with tobacco exposure influence the risk of head and neck cancer in Northeast Indian population. Tumor Biology. 2015;36:5773-83.

  • 64. Cornean CI, Catana A, Maniu AA. Do Polymorphisms of the TERT, GSTM1, and GSTT1 Genes Increase Laryngeal Cancer Susceptibility in Smokers of Romanian Descent? Medicina (Kaunas). 2022;58(8).

  • 65. Deakin M, Elder J, Hendrickse C, Peckham D, Baldwin D, Pantin C, et al. Glutathione S-transferase GSTT1 genotypes and susceptibility to cancer: studies of interactions with GSTM1 in lung, oral, gastric and colorectal cancers. Carcinogenesis. 1996;17(4):881-4.

  • 66. Deng ZL, Wei YP, Ma Y. [Frequent genetic deletion of detoxifying enzyme GSTM1 and GSTT1 genes in nasopharyngeal carcinoma patients in Guangxi Province, China]. Zhonghua Zhong Liu Za Zhi. 2004;26(10):598–600.

  • 67. Deng Z, Wei Y, Luo W, Liao Z, Ma Y. Glutathione S-transferase M1 and T1 gene deletion associated with increased susceptibility to nasopharyngeal carcinoma. The Chinese-German Journal of Clinical Oncology. 2005;4:276-8.

  • 68. Dong TT, Wang LJ, Liu LZ, Ma SN. Susceptibility to oral squamous cell carcinoma: correlation with variants of CYP1A1-MspI, GSTT1, GSTM1, ALDH2, EC-SOD and Lifestyle factors. Balkan J Med Genet. 2016;19(2):61–70.

  • 69. Evans AJ, Henner WD, Eilers KM, Montalto MA, Wersinger EM, Andersen PE, et al. Polymorphisms of GSTT1 and related genes in head and neck cancer risk. Head Neck. 2004;26(1):63–70.

  • 70. Firigato I, de Toledo Gonçalves F, De Antonio J, Alberto Curioni O, Rocha Silva M, Jorge Fígaro Gattás G. How many copies of GSTM1 and GSTT1 are associated with head and neck cancer risk? Biomarkers. 2019;24(3):262-7.

  • 71. Firigato I, López RVM, Curioni OA, De Antonio J, Gattás GF, de Toledo Gonçalves F. Many hands make light work: CNV of GSTM1 effect on the oral carcinoma risk. Cancer Epidemiol. 2022;78:102150.

  • 72. Gajecka M, Rydzanicz M, Jaskula-Sztul R, Kujawski M, Szyfter W, Szyfter K. CYP1A1, CYP2D6, CYP2E1, NAT2, GSTM1 and GSTT1 polymorphisms or their combinations are associated with the increased risk of the laryngeal squamous cell carcinoma. Mutat Res. 2005;574(1–2):112 − 23.

  • 73. Gattás GJF, de Carvalho MB, Siraque MS, Curioni OA, Kohler P, Eluf-Neto J, et al. Genetic polymorphisms of CYP1A1, CYP2E1, GSTM1, and GSTT1 associated with head and neck cancer. Head & Neck: Journal for the Sciences and Specialties of the Head and Neck. 2006;28(9):819 − 26.

  • 74. Gaudet MM, Olshan AF, Poole C, Weissler MC, Watson M, Bell DA. Diet, GSTM1 and GSTT1 and head and neck cancer. Carcinogenesis. 2004;25(5):735 − 40.

  • 75. Goloni-Bertollo EM, Biselli JM, Corrêa LC, Maníglia JV, Rossit AR, Ruiz MT, et al. [Evaluation of the influence of GSTT1 and GSTM1 null genotypes in head and neck carcinogenesis]. Rev Assoc Med Bras (1992). 2006;52(5):365-8.

  • 76. Gronau S, Koenig-Greger D, Jerg M, Riechelmann H. Gene polymorphisms in detoxification enzymes as susceptibility factor for head and neck cancer? Otolaryngol Head Neck Surg. 2003;128(5):674 − 80.

  • 77. Gronau S, Koenig-Greger D, Jerg M, Riechelmann H. GSTM1 enzyme concentration and enzyme activity in correlation to the genotype of detoxification enzymes in squamous cell carcinoma of the oral cavity. Oral Dis. 2003;9(2):62 − 7.

  • 78. Guo X, O’Brien SJ, Zeng Y, Nelson GW, Winkler CA. GSTM1 and GSTT1 gene deletions and the risk for nasopharyngeal carcinoma in Han Chinese. Cancer Epidemiol Biomarkers Prev. 2008;17(7):1760-3.

  • 79. Hanna E, MacLeod S, Vural E, Lang N. Genetic deletions of glutathione-S-transferase as a risk factor in squamous cell carcinoma of the larynx: a preliminary report. Am J Otolaryngol. 2001;22(2):121-3.

  • 80. Harth V, Schafer M, Abel J, Maintz L, Neuhaus T, Besuden M, et al. Head and neck squamous-cell cancer and its association with polymorphic enzymes of xenobiotic metabolism and repair. J Toxicol Environ Health A. 2008;71(13–14):887 − 97.

  • 81. Hatagima A, Costa EC, Marques CF, Koifman RJ, Boffetta P, Koifman S. Glutathione S-transferase polymorphisms and oral cancer: a case-control study in Rio de Janeiro, Brazil. Oral Oncol. 2008;44(2):200-7.

  • 82. Hong YJ, Lee JK, Lee GH, Hong SI. Influence of glutathione S-transferase M1 and T1 genotypes on larynx cancer risk among Korean smokers. Clin Chem Lab Med. 2000;38(9):917-9.

  • 83. Huang P, An Y, Wu C, Meng X. GSTT1, GSTM1 polymorphisms, and oral and maxillofacial cancer. J Pract Oncol (Chin). 2006;21(1):39–42.

  • 84. Hung HC, Chuang J, Chien YC, Chern HD, Chiang CP, Kuo YS, et al. Genetic polymorphisms of CYP2E1, GSTM1, and GSTT1; environmental factors and risk of oral cancer. Cancer Epidemiol Biomarkers Prev. 1997;6(11):901-5.

  • 85. Jahnke V, Strange R, Matthias C, Fryer AA. [Initial results of glutathione-S-transferase GSTM1 and GSTT1 genotypes and genetic predisposition for laryngeal carcinoma]. Laryngorhinootologie. 1995;74(11):691-4.

  • 86. Jahnke V, Matthias C, Fryer A, Strange R. Glutathione S-transferase and cytochrome-P-450 polymorphism as risk factors for squamous cell carcinoma of the larynx. Am J Surg. 1996;172(6):671-3.

  • 87. Jaskula-Sztul R, Rienikainen M, Husgafvel-Pursiainen K, Szmeja Z, Szyfter W, Syfter K, et al. Glutathione S transferase M1 andT1 genotypes and susceptibility to smoking related larynx cancer. Biomarkers. 1998;3(2):149 − 55.

  • 88. Jiang Y, Li N, Dong P, Zhang N, Sun Y, Han M, et al. Polymorphisms in GSTM1, GSTTI and GSTP1 and nasopharyngeal cancer in the east of China: a case-control study. Asian Pac J Cancer Prev. 2011;12(11):3097 − 100.

  • 89. Jourenkova N, Reinikainen M, Bouchardy C, Dayer P, Benhamou S, Hirvonen A. Larynx cancer risk in relation to glutathione S-transferase M1 and T1 genotypes and tobacco smoking. Cancer Epidemiol Biomarkers Prev. 1998;7(1):19–23.

  • 90. Jourenkova-Mironova N, Voho A, Bouchardy C, Wikman H, Dayer P, Benhamou S, et al. Glutathione S-transferase GSTM1, GSTM3, GSTP1 and GSTT1 genotypes and the risk of smoking-related oral and pharyngeal cancers. Int J Cancer. 1999;81(1):44 − 8.

  • 91. Karen-Ng LP, Marhazlinda J, Rahman Z, Yang Y-H, Jalil N, Cheong SC, et al. Combined effects of isothiocyanate intake, glutathione S-transferase polymorphisms and risk habits for age of oral squamous cell carcinoma development. Asian Pac J Cancer Prev. 2011;12(5):1161-6.

  • 92. Katiyar T, Yadav V, Maurya SS, Ruwali M, Singh M, Hasan F, et al. Interaction of glutathione-s-transferase genotypes with environmental risk factors in determining susceptibility to head and neck cancer and treatment response and survival outcome. Environ Mol Mutagen. 2020;61(5):574 − 84.

  • 93. Katoh T, Kaneko S, Kohshi K, Munaka M, Kitagawa K, Kunugita N, et al. Genetic polymorphisms of tobacco- and alcohol-related metabolizing enzymes and oral cavity cancer. Int J Cancer. 1999;83(5):606-9.

  • 94. Kietthubthew S, Sriplung H, Au WW. Genetic and environmental interactions on oral cancer in Southern Thailand. Environ Mol Mutagen. 2001;37(2):111-6.

  • 95. Kondo S, Sturgis EM, Li F, Wei Q, Li G. GSTM1 and GSTT1 null polymorphisms and risk of salivary gland carcinoma. Int J Clin Exp Med. 2009;2(1):68–75.

  • 96. Krüger M, Pabst AM, Mahmoodi B, Becker B, Kämmerer PW, Koch FP. The impact of GSTM1/GSTT1 polymorphism for the risk of oral cancer. Clin Oral Investig. 2015;19(8):1791-7.

  • 97. Leme CV, Raposo LS, Ruiz MT, Biselli JM, Galbiatti AL, Maniglia JV, et al. GSTM1 and GSTT1 genes analysis in head and neck cancer patients. Rev Assoc Med Bras (1992). 2010;56(3):299–303.

  • 98. Li Q, Wang L, Chen Y, Du Y, Kong P, Li Y, et al. [Polymorphisms of GSTM1, GSTT1 and susceptibility of laryngeal and hypopharyngeal carcinomas]. Lin Chuang Er Bi Yan Hou Tou Jing Wai Ke Za Zhi. 2009;23(24):1105-7, 11.

  • 99. Liao ZL, Deng ZL, Wei YP, Xie KS, Zhang B, Dai XM, et al. Relationship of GSTT1 and GSTM1 gene polymorphisms with the development of nasopharyngeal carcinoma. J Guangxi Med Univ. 2005;22:372-4.

  • 100. Liu CJ, Chang CS, Lui MT, Dang CW, Shih YH, Chang KW. Association of GST genotypes with age of onset and lymph node metastasis in oral squamous cell carcinoma. J Oral Pathol Med. 2005;34(8):473-7.

  • 101. Losi-Guembarovski R, Cólus IM, De Menezes RP, Poliseli F, Chaves VN, Kuasne H, et al. Lack of association among polymorphic xenobiotic-metabolizing enzyme genotypes and the occurrence and progression of oral carcinoma in a Brazilian population. Anticancer Res. 2008;28(2a):1023-8.

  • 102. Lourenço GJ, Silva EF, Rinck-Junior JA, Chone CT, Lima CS. CYP1A1, GSTM1 and GSTT1 polymorphisms, tobacco and alcohol status and risk of head and neck squamous cell carcinoma. Tumour Biol. 2011;32(6):1209-15.

  • 103. Madhulatha G, Das S, Venkateswarlu N, Pujar A, Jyothy A, Munshi A. GSTM1 and GSTT1 null polymorphism and antioxidant levels in oral submucous fibrosis, leukoplakia and oral cancer patients among a South Indian Population. Journal of Oral and Maxillofacial Surgery, Medicine, and Pathology. 2018;30(2):169 − 74.

  • 104. Maniglia MP, Russo A, Biselli-Chicote PM, Oliveira-Cucolo JG, Rodrigues-Fleming GH, Maniglia JV, et al. Glutathione S-transferase Polymorphisms in Head and Neck Squamous Cell Carcinoma Treated with Chemotherapy and/or Radiotherapy. Asian Pac J Cancer Prev. 2020;21(6):1637-44.

  • 105. Marchioni DM, Gattás GJ, Curioni OA, Carvalho MB. [Interaction between dietary intake and GSTM1 and GSTT1 polymorphisms in head and neck cancer risk: a case-control study in São Paulo, Brazil]. Cad Saude Publica. 2011;27(2):379 − 87.

  • 106. Masood N, Kayani MA, Malik FA, Mahjabeen I, Baig RM, Faryal R. Genetic variation in carcinogen metabolizing genes associated with oral cancer in pakistani population. Asian Pac J Cancer Prev. 2011;12(2):491-5.

  • 107. Masood N, Kayani MA. Protection against laryngeal and pharyngeal carcinoma: Heterozygous vs. homozygous deletions of GSTM1 and GSTT1. Genet Mol Biol. 2013;36(1):1–6.

  • 108. Matthias C, Jahnke V, Hand P, Fryer AA, Strange RC. [Immunohistologic and molecular genetic studies of the effect of glutathione-S-transferases on the development of squamous epithelial carcinomas in the area of the head-neck]. Laryngorhinootologie. 1999;78(4):182-8.

  • 109. McWilliams JE, Evans AJ, Beer TM, Andersen PE, Cohen JI, Everts EC, et al. Genetic polymorphisms in head and neck cancer risk. Head Neck. 2000;22(6):609 − 17.

  • 110. Mondal R, Ghosh SK, Choudhury JH, Seram A, Sinha K, Hussain M, et al. Mitochondrial DNA copy number and risk of oral cancer: a report from Northeast India. PloS one. 2013;8(3):e57771.

  • 111. Olshan AF, Weissler MC, Watson MA, Bell DA. GSTM1, GSTT1, GSTP1, CYP1A1, and NAT1 polymorphisms, tobacco use, and the risk of head and neck cancer. Cancer Epidemiol Biomarkers Prev. 2000;9(2):185 − 91.

  • 112. Oude Ophuis MB, van Lieshout EM, Roelofs HM, Peters WH, Manni JJ. Glutathione S-transferase M1 and T1 and cytochrome P4501A1 polymorphisms in relation to the risk for benign and malignant head and neck lesions. Cancer. 1998;82(5):936 − 43.

  • 113. Patel SK, Sinha M, Mitra M. Glutathione-S-transferase M1 and T1 gene Polymorphism as Risk Factors of Oral Squamous Cell Carcinoma: A Preliminary Investigation. Research Journal of Pharmacy and Technology. 2012;5(7):918 − 20.

  • 114. Peters ES, McClean MD, Marsit CJ, Luckett B, Kelsey KT. Glutathione S-transferase polymorphisms and the synergy of alcohol and tobacco in oral, pharyngeal, and laryngeal carcinoma. Cancer Epidemiol Biomarkers Prev. 2006;15(11):2196 − 202.

  • 115. Rao AK, Parameswar P, Majumdar S, Uppala D, Kotina S, Vennamaneni NH. Evaluation of Genetic Polymorphisms in Glutathione S-Transferase Theta1, Glutathione S-Transferase Mu1, and Glutathione S-Transferase Mu3 in Oral Leukoplakia and Oral Squamous Cell Carcinoma with Deleterious Habits using Polymerase Chain Reaction. Int J Appl Basic Med Res. 2017;7(3):181-5.

  • 116. Reszka E, Czekaj P, Adamska J, Wasowicz W. Relevance of glutathione S-transferase M1 and cytochrome P450 1A1 genetic polymorphisms to the development of head and neck cancers. Clinical chemistry and laboratory medicine. 2008;46(8):1090-6.

  • 117. Risch A, Ramroth H, Raedts V, Rajaee-Behbahani N, Schmezer P, Bartsch H, et al. Laryngeal cancer risk in Caucasians is associated with alcohol and tobacco consumption but not modified by genetic polymorphisms in class I alcohol dehydrogenases ADH1B and ADH1C, and glutathione-S-transferases GSTM1 and GSTT1. Pharmacogenetics. 2003;13(4):225 − 30.

  • 118. Russo A, Francelin PR, Galbiatti AL, Raposo LS, Maníglia JV, Pavarino EC, et al. Association between GSTP1, GSTM1 and GSTT1 polymorphisms involved in xenobiotic metabolism and head and neck cancer development. Mol Biol Rep. 2013;40(7):4181-8.

  • 119. Ruwali M, Singh M, Pant MC, Parmar D. Polymorphism in glutathione S-transferases: susceptibility and treatment outcome for head and neck cancer. Xenobiotica. 2011;41(12):1122-30.

  • 120. Sam SS, Thomas V, Reddy KS, Surianarayanan G, Chandrasekaran A. Gene-environment interactions associated with CYP1A1 MspI and GST polymorphisms and the risk of upper aerodigestive tract cancers in an Indian population. J Cancer Res Clin Oncol. 2010;136(6):945 − 51.

  • 121. Sánchez-Siles M, Pelegrín-Hernández JP, Hellin-Meseguer D, Guerrero-Sánchez Y, Corno-Caparrós A, Cabezas-Herrera J, et al. Genotype of Null Polymorphisms in Genes GSTM1, GSTT1, CYP1A1, and CYP1A1*2A (rs4646903 T > C)/CYP1A1*2 C (rs1048943 A > G) in Patients with Larynx Cancer in Southeast Spain. Cancers (Basel). 2020;12(9).

  • 122. Saravani S, Miri-Moghaddam M, Bazi A, Miri-Moghaddam E. Association of glutathione-S-transferases M1 and T1 deletional variants with development of oral squamous cell carcinoma: A study in the South-East of Iran. Asian Pacific journal of cancer prevention: APJCP. 2019;20(6):1921.

  • 123. Senthilkumar KP, Thirumurugan R. Risk modulation of GSTM1–GSTT1 interactions to head and neck cancer in tobacco users. Mol Biol Rep. 2014;41(9):5635-44.

  • 124. Sharma A, Mishra A, Das BC, Sardana S, Sharma JK. Genetic polymorphism at GSTM1 and GSTT1 gene loci and susceptibility to oral cancer. Neoplasma. 2006;53(4):309 − 15.

  • 125. Sikdar N, Paul RR, Roy B. Glutathione S-transferase M3 (A/A) genotype as a risk factor for oral cancer and leukoplakia among Indian tobacco smokers. Int J Cancer. 2004;109(1):95–101.

  • 126. Silva T, Marques C, Marques Filho M, Marques A, Di Pietro G, Rios-Santos F. Association of the GSTT1 polymorphism in upper aerodigestive tract cancer with tobacco smoking. Genet Mol Res. 2014;13(1):528 − 37.

  • 127. Singh M, Shah PP, Singh AP, Ruwali M, Mathur N, Pant MC, et al. Association of genetic polymorphisms in glutathione S-transferases and susceptibility to head and neck cancer. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis. 2008;638(1–2):184 − 94.

  • 128. Singh RD, Haridas N, Shah FD, Patel JB, Shukla SN, Patel PS. Gene polymorphisms, tobacco exposure and oral cancer susceptibility: a study from Gujarat, West India. Oral Dis. 2014;20(1):84–93.

  • 129. Singh SA, Ghosh SK. Metabolic Phase I (CYPs) and Phase II (GSTs) Gene Polymorphisms and Their Interaction with Environmental Factors in Nasopharyngeal Cancer from the Ethnic Population of Northeast India. Pathol Oncol Res. 2019;25(1):33–44.

  • 130. Soucek P, Ústav SZ, Susova S, Mohelnikova-Duchonova B, Vodicka P. Polymorphisms in metabolizing enzymes and the risk of head and neck squamous cell carcinoma in the Slavic population of. Neoplasma. 2010;57:5.

  • 131. Soya S, Vinod T, Reddy K, Gopalakrishnan S, Adithan C. Genetic polymorphisms of glutathione-S-transferase genes (GSTM1, GSTT1 and GSTP1) and upper aerodigestive tract cancer risk among smokers, tobacco chewers and alcoholics in an Indian population. European Journal of Cancer. 2007;43(18):2698 − 706.

  • 132. Sreelekha TT, Ramadas K, Pandey M, Thomas G, Nalinakumari KR, Pillai MR. Genetic polymorphism of CYP1A1, GSTM1 and GSTT1 genes in Indian oral cancer. Oral Oncol. 2001;37(7):593-8.

  • 133. Sugimura T, Kumimoto H, Tohnai I, Fukui T, Matsuo K, Tsurusako S, et al. Gene–environment interaction involved in oral carcinogenesis: molecular epidemiological study for metabolic and DNA repair gene polymorphisms. Journal of oral pathology & medicine. 2006;35(1):11 − 8.

  • 134. Surit R, Kumar S, Sinha DK, Shekhar R. Genomic Pattern of GSTM1 and T1 Gene Null polymorphism of Head and Neck Cancer Patients in Eastern India. Asian Pac J Cancer Prev. 2022;23(8):2655-9.

  • 135. Tata NH, Kshirsagar A, Nangare N. Characterization of genetic polymorphisms in oral cancer-related genes pertaining to oxidative stress, carcinogen detoxifying, and DNA repair: A case–control study. Journal of Cancer Research and Therapeutics. 2022;18(4):1023-9.

  • 136. To-Figueras J, Gené M, Gómez-Catalán J, Piqué E, Borrego N, Caballero M, et al. Microsomal epoxide hydrolase and glutathione S-transferase polymorphisms in relation to laryngeal carcinoma risk. Cancer Lett. 2002;187(1–2):95–101.

  • 137. Trizna Z, Clayman GL, Spitz MR, Briggs KL, Goepfert H. Glutathione s-transferase genotypes as risk factors for head and neck cancer. Am J Surg. 1995;170(5):499–501.

  • 138. Unal M, Tamer L, Ateş NA, Akbaş Y, Pata YS, Vayisoğlu Y, et al. Glutathione S-transferase M1, T1, and P1 gene polymorphism in laryngeal squamous cell carcinoma. Am J Otolaryngol. 2004;25(5):318 − 22.

  • 139. Wei Y, Long X, Liu Z, Ma Y, Deng Z. Genetic polymorphism of glutathione-S-transferase M1 and T1 in associated with carcinogenesis of hepatocellular carcinoma and nasopharyngeal carcinoma. The Chinese-German Journal of Clinical Oncology. 2012;11:138 − 41.

  • 140. Yaghmaei B, Yaghmaei K, Jafarian M, Golmohammadi S. Genetic polymorphisms of glutathione S-transferase Mu 1, glutathione S-transferase theta 1, and glutathione S-transferase P1 in oral squamous cell carcinoma: A case-control study in Iranian population. Journal of Orofacial Sciences. 2015;7(2):108 − 12.

  • 141. Zakiullah, Ahmadullah, Khisroon M, Saeed M, Khan A, Khuda F, et al. Genetic susceptibility to oral cancer due to combined effects of GSTT1, GSTM1 and CYP1A1 gene variants in tobacco addicted patients of Pashtun ethnicity of Khyber Pakhtunkhwa province of Pakistan. Asian Pac J Cancer Prev. 2015;16(3):1145-50.

  • 142. Zakiullah, Saeed M, Javed N, Khuda F, Ovais M. Association of nasopharyngeal cancer risk with genetic polymorphisms of drug-metabolizing enzyme genes GSTM1, GSTT1 and CYP1A1 (rs4646903 variant), in tobacco addicted patients of Pashtun ethnicity of Khyber Pakhtunkhwa Province of Pakistan. Pak J Pharm Sci. 2019;32(5):2107-16.

  • 143. Khlifi R, Messaoud O, Rebai A, Hamza-Chaffai A. Polymorphisms in the human cytochrome P450 and arylamine N-acetyltransferase: susceptibility to head and neck cancers. BioMed research international. 2013;2013.

  • 144. Cadoni G, Boccia S, Petrelli L, Di Giannantonio P, Arzani D, Giorgio A, et al. A review of genetic epidemiology of head and neck cancer related to polymorphisms in metabolic genes, cell cycle control and alcohol metabolism. ACTA otorhinolaryngologica italica. 2012;32(1):1.

  • 145. Minina VI, Soboleva OA, Glushkov AN, Voronina EN, Sokolova EA, Bakanova ML, et al. Polymorphisms of GSTM1, GSTT1, GSTP1 genes and chromosomal aberrations in lung cancer patients. Journal of Cancer Research and Clinical Oncology. 2017;143:2235-43.

  • 146. Eshkoor SA, Marashi S, Ismail P, Rahman S, Mirinargesi M, Adon M, et al. Association of GSTM1 and GSTT1 with ageing in auto repair shop workers. Genetics and molecular research [electronic resource]: GMR. 2012;11(2):1486-96.

  • 147. Shaikhova D, Amromina A, Sitnikov I, Sutunkova M, Gurvich V, Astakhova S. Features of GSTM1, GSTT1 and GSTP1 Genetic Polymorphism in Nizhny Tagil Metallurgical Plant Workers with Cardiovascular Diseases. Public Health and Life Environment–PH&LE. 2021;1(12):36–40.

  • 148. Tew KD. Glutathione-associated enzymes in anticancer drug resistance. Cancer research. 1994;54(16):4313-20.

  • 149. Pool-Zobel B, Veeriah S, Böhmer F-D. Modulation of xenobiotic metabolising enzymes by anticarcinogens—focus on glutathione S-transferases and their role as targets of dietary chemoprevention in colorectal carcinogenesis. Mutation Research/Fundamental and Molecular Mechanisms of Mutagenesis. 2005;591(1–2):74–92.

  • 150. Murata M, Watanabe M, Yamanaka M, Kubota Y, Ito H, Nagao M, et al. Genetic polymorphisms in cytochrome P450 (CYP) 1A1, CYP1A2, CYP2E1, glutathione S-transferase (GST) M1 and GSTT1 and susceptibility to prostate cancer in the Japanese population. Cancer letters. 2001;165(2):171-7.

  • 151. Hernandez A, Xamena N, Surralles J, Galofre P, Velazquez A, Creus A, et al. Role of GST and NAT2 polymorphisms in thyroid cancer. Journal of Endocrinological Investigation. 2008;31:1025-31.

  • 152. McCarty KM, Santella RM, Steck SE, Cleveland RJ, Ahn J, Ambrosone CB, et al. PAH–DNA adducts, cigarette smoking, GST polymorphisms, and breast cancer risk. National Institute of Environmental Health Sciences; 2009.

  • 153. Tsuchida S, Sekine Y, Shineha R, Nishihira T, Sato K. Elevation of the placental glutathione S-transferase form (GST-π) in tumor tissues and the levels in sera of patients with cancer. Cancer research. 1989;49(18):5225-9.

  • 154. Landi S. Mammalian class theta GST and differential susceptibility to carcinogens: a review. Mutation Research/Reviews in Mutation Research. 2000;463(3):247 − 83.

  • 155. Cotton S, Sharp L, Little J, Brockton N. Glutathione S-transferase polymorphisms and colorectal cancer: a HuGE review. American journal of epidemiology. 2000;151(1):7–32.

  • 156. Rebbeck TR, Walker AH, Jaffe JM, White DL, Wein AJ, Malkowicz SB. Glutathione S-transferase- (GSTM1) and-θ (GSTT1) genotypes in the etiology of prostate cancer. Cancer Epidemiology Biomarkers & Prevention. 1999;8(4):283-7.

  • 157. Gertig DM, Stampfer M, Haiman C, Hennekens CH, Kelsey K, Hunter DJ. Glutathione S-transferase GSTM1 and GSTT1 polymorphisms and colorectal cancer risk: a prospective study. Cancer epidemiology, biomarkers & prevention: a publication of the American Association for Cancer Research, cosponsored by the American Society of Preventive Oncology. 1998;7(11):1001-5.

  • 158. Crump C, Chen C, Appelbaum FR, Kopecky KJ, Schwartz SM, Willman CL, et al. Glutathione S-transferase theta 1 gene deletion and risk of acute myeloid leukemia. Cancer Epidemiology Biomarkers & prevention. 2000;9(5):457 − 60.

  • 159. Al-Badran AI, Al-Mayah MK. Association between GSTT1 and GSTM1 genes polymorphisms Type II diabetes miletus patients in Basra Iraq. Int J Curr Microbiol App Sci. 2014;3(11):288 − 99.

Download references

Funding

There is no funding.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, S.S. and M.S.; methodology, M.S.; software, M.S.; validation, M.M.I. and S.S.; formal analysis, M.S.; investigation, M.S. and S.S.; resources, M.M.I.; data curation, M.S.; writing—original draft preparation, M.M.I.; writing—review and editing, S.S., M.S., S.M.M.K. and P.C.; visualization, P.C. and S.M.M.K.; supervision, S.S.; project administration, M.M.I; funding acquisition, S.S. All authors have read and agreed to the published version of the manuscript.

Corresponding author

Correspondence to Mohammad Moslem Imani.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Institutional review board statement

Not applicable.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary Material 1

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sadafi, S., Choubsaz, P., Kazemeini, S.M.M. et al. Glutathione S-transferase theta 1 (GSTT1) deletion polymorphism and susceptibility to head and neck carcinoma: a systematic review with five analyses. BMC Cancer 24, 885 (2024). https://doi.org/10.1186/s12885-024-12618-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12885-024-12618-7

Keywords