Skip to main content

Impact of the deletion glutathione S-transferase (class Mu) on lung cancer risk among smokers

Abstract

Background

The results of several studies assessing the effect of the glutathione S-transferase class Mu 1 (GSTM1) null variant on the genetic susceptibility of tobacco-related cancers have been conflicting. In this work, we aim to identify the impact of the deletion of GSTM1 on lung cancer risk among smokers.

Methods

This study was conducted on 20 patients diagnosed with primary lung cancer and 20 healthy individuals as a control group. They were subject to full medical history taking, complete clinical examination, and GSTM1 genotyping by PCR.

Results

Both studied groups were matched for age, sex, and smoking status. No statistically significant difference was exhibited between the frequency of GSTM1 positive and GSTM1 null in the studied population. No risk of lung cancer associated with GSTM1 null genotype was demonstrated between the patients and control group (n = 14/20 cases) (p = 0.110, OR = 2.852, 95% CI 0.777–10.467). Additionally, there was no association between the risk of lung cancer and the presence of the gene either in smokers (p = 1, OR = 1.8 and 95% CI 0.124–26.196) or non-smokers; (p = 0.063, OR = 4.4 and 95% CI 0.889–21.78). No statistically significant risk was found between the frequencies of GSTM1 null and the various histopathological types of lung malignancy.

Conclusion

The results of this work demonstrated no association between the occurrence of the GSTM1 null variant, even when stratified for smoking status, and the risk of lung cancer.

Background

Epidemiological studies have demonstrated that average tobacco smokers face approximately a 14-fold increased risk for the occurrence of lung malignancy. Additionally, individuals with a previous family history are present with about a 2.5-fold increase in risk of lung cancer development, even after accounting for tobacco smoke exposure [1, 2]. Noteworthy, tobacco consumption plays a negative role by causing the accumulation of genetic alterations eventually leading to lung cancer. Furthermore, lung malignancy follows a multistage process, occurring against a backdrop of increasing genomic instability [3, 4].

The role of GST (glutathione S-transferase) activity in metabolizing carcinogens has prompted numerous studies on the impact of GST polymorphisms on cancer susceptibility, particularly in tobacco-related cancers [5]. The GST gene family consists of four main classes: alpha (α), mu (μ), pi (π), and theta (θ) [6]. Most human tumor cell lines express significant levels of class pi GST, specifically, GSTP exon 5 polymorphisms have been linked to the development of lung malignancy [7], especially in adenocarcinoma, which was not significantly associated with smoking [8].

The GSTM1 gene, found on chromosome 1p13.3, is not manifested in approximately 50% of individuals due to a homozygous gene deletion, known as the GSTM1 null allele [6]. However, the results from several studies attempting to establish a relationship between the GSTM1 null genotype and the risk of incidence of lung cancer in various ethnic populations have been conflicting [9] which could suggest that the GSTM1 null variant has only a marginal impact on the genetic susceptibility to tobacco-related cancers [10].

This work aims to evaluate the role of the isoenzyme glutathione transferase gene deletion as a risk factor for the development of lung cancer among smokers.

Subjects and methods

This is a prospective case/control study that was performed on patients of the outpatient clinic of the chest department of Kasr El Ainy Hospital between September 2016 and March 2017.

Inclusion criteria

Patients consecutively included in the study, after their participation consent, were.

  1. 1

    - Those with confirmed histopathological diagnosis, obtained by tissue biopsy, of primary lung cancer

  2. 2

    - Not previously treated by radiotherapy and/or chemotherapy and categorized as the patient’s group

  3. 3

    - Not participating in another clinical trial.

  4. 4

    - The control group consisted of unrelated healthy subjects taken as control with no previous history of malignancy.

Exclusion criteria

  1. 1

    -  Patients with prior malignancy or present with metastatic malignant manifestations were not included in the present study.

  2. 2

    - Patients receiving radiotherapy and/ or chemotherapy

  3. 3

    - Patients with severe morbidity or unfit general condition

All patients were subject to complete medical history, full clinical examination, and GSTM1 genotyping by PCR.

GSTM1 genotyping

Peripheral blood sampling was done, and Genomic DNA was extracted from peripheral blood mononuclear cells using a QIAamp DNA extraction DNA mini kit (Catalog No. 51104) (QIAGEN, Germany) according to the manufacturer’s protocol. DNA was isolated from peripheral leukocytes using standard procedures with proteinase K digestion and extraction. GSTM1 gene deletion was determined by procedures described by [11] (Fig. 1).

Fig. 1
figure 1

PCR analysis: First Lane: 100 bp ladder, Lanes 1–2 GSTM1positive genotype, lanes 3–4 GSTM1null genotype. Housekeeping gene (albumin 350 bp) is the internal control and is present in all samples

Ethical approval

This work was done according to the guidelines outlined in the Declaration of Helsinki. The Ethical Committee of the National Research Centre, Cairo, Egypt, reviewed and approved the study (number: 10088). each participant provided a written informed consent before their inclusion in the study.

Statistical analysis

Data were coded and entered using the statistical package SPSS version 23. Data was summarized using mean and standard deviation, median, minimum, and maximum for quantitative variables and frequencies (number of cases) and relative frequencies (percentages) for categorical variables. Comparisons between quantitative variables were done using the non-parametric Mann–Whitney test. For comparing categorical data, a chi-square (χ2) test was performed. The exact test was used instead when the expected frequency was less than 5. Genotype frequency was compared between the disease and the control groups using chi-square tests. An odds ratio (OR) with 95% confidence intervals was calculated. P values less than 0.05 were considered statistically significant.

Results

Twenty patients (10 males and 10 females with a mean age of 57.3 years) were included in this study of whom 8 of them were smokers and 12 were non-smokers with histologically confirmed primary lung cancer diagnosis. The control group consisted of 20 healthy individuals unrelated genetically to the patient group and consisted of 12 males and 8 females, 4 smokers and 16 non-smokers, with a mean age of 52.05 years.

The control and lung cancer groups were matched as regards the mean values of age, sex, and smoking status; p = 0.066, 0.525, and 0.168 respectively. The mean values of the smoking index (pack-years) were significantly higher in the patients (53.75 ± 15.98) compared to the control (16.50 ± 8.10); p = 0.006. Patients with SCLC were in the extensive stage of the disease. As their number was relatively small, they were added to stage IV NSCLC to be studied statistically. Stage II lung cancer represented 45% of cases (9/20 patients), stage III represented 5% of cases (1/20 patients) and 10/20 patients (50%) were at stage IV (Table 1).

Table 1 Clinical and demographic of the included subjects

All studied subjects (n = 40) showed no statistically significant difference between the GSTM1 positive and GSTM1 null as regards the frequency (17 and 23 respectively), the mean values of age, sex, smoking status, and smoking index; p = 0.110, 0.298, 0.131, 0.443, and 0.341 respectively (Table 2, Fig. 1).

Table 2 GSTM1 frequency in patients with lung carcinoma and healthy controls according to their age (years), sex, and smoking

There was no risk of lung cancer demonstrated between the patients and control group though the frequency of GSTM1 null was lower in patients; 45% (n = 9/20 cases) than in the control group 70% (n = 14/20 cases) (p = 0.110, OR = 2.852, 95% CI 0.777–10.467). The risk of lung cancer associated with the null genotype was examined by stratification for smoking status; although the frequency of deletion was higher in smokers patients (62.5%) than that of the positive genotype (37.5%); no risk of cancer was found to be associated with the gene among smokers (p = 1, OR = 1.8 and 95% CI 0.124–26.196) or non-smokers; (p = 0.063, OR = 4.4 and 95% CI 0.889–21.78) (Table 2).

The frequency of GSTM1 null was studied in the histopathological types of lung cancer; it was 33.3% of SCLC (n = 1/3 cases), and 47.1% of NSCLC cases 8/17) compared to the control group 70% null genotype (14/20), but no risk of SCLC was found to be associated with the gene deletion (p = 0.523, OR = 4.4 and 95% CI 0.319–60.614), or NSCLC (p = 0.208, OR = 2.475 and 95% CI 0.597–10.269) or histological subtypes of NSCLC (Table 3). There was no statistically significant difference found between the frequency of GSTM1 null in stage IV of the disease 40% (4/10) compared to 55.6% (n = 5/9) in stage II (Figs. 1 and 2).

Table 3 Frequencies of GSTM1 in different histopathological types of lung cancer
Fig. 2
figure 2

Frequencies of GSTM1 according to the stage of lung cancer

Discussion

In the present work, we observed that the frequency of the GSTM1 null genotype in the studied population was 57.5%. Interestingly, this frequency was lower in lung cancer patients (45%) compared to the control group (70%). These findings align with a previous study conducted in Egypt by Ramzy et al. [12]. In their study, which included 48 patients diagnosed with lung cancer and 42 healthy individuals, they reported nearly similar GSTM1 genotype distribution between the control group and lung cancer cases. Notably, the GSTM1 null allele was absent in 33.3% of controls and 31.25% of patients.

In various ethnic populations, the prevalence of polymorphic genes was extensively studied [12,13,14,15,16]. Notably, the GSTM1-null genotype has been a focus of investigation. In the Turkish population, the GSTM1-null variant was expressed in around half of the patients with lung cancer compared to a slightly higher prevalence of 52.6% in the control group [17]. Among the Brazilian population, the percentage was 45.5% for the GSTM1 deletion in patients with lung cancer and 48.1% in their control group [18], in Caucasians, GSTM1 frequencies vary between 42 and 60% [19]. GSTM1 null allele has been observed in approximately 31% to 66% of Asian and Indian populations [20,21,22]. Alternatively, for African Americans, the GSTM1 deletion polymorphism was found to be 23 to 35% [23] and Among Chileans, the GSTM1 deletion frequency was 21% [24].

In the current study, we observed that the expression of the GSTM1 null genotype was not correlated to the susceptibility of lung tumors. Even when stratified by tobacco consumption, no increased cancer risk was found among tobacco consumers with the GSTM1 deletion. Interestingly, Ramzy et al. reached a similar conclusion in Egyptian patients, indicating that the GSTM1 null genotype alone does not impact lung cancer risk, however, when combined with GSTP1 polymorphism, there appears to be an increased risk for lung cancer [12].

Likewise, in Bangladeshi individuals, the GSTM1 null genotype does not appear to significantly increase the risk of developing lung cancer. Moreover, the risk of lung malignancy was not observed among both tobacco smokers and nonsmokers who carried either the null or present genotypes of GSTM1 [25]. These results are consistent with the conclusion reached by Masood et al. which suggests that the deletion of GSTM1 is not linked with the incidence of lung malignancy [26].

Additionally, an earlier meta-analysis reported that even though the number of patients carrying GSTM1 null genotype was higher in those with lung cancer diagnosis, no causal relationship between carrying this genotype and an increased susceptibility to lung cancer could be demonstrated [27]. Kalikaki et al. also outlined the absence of a significant correlation between the presence of GSTM1 gene polymorphism and the overall survival of patients with advanced NSCLC [28].

On the other hand, contrasting findings related to the GSTM1 null polymorphism and its association with lung cancer risk were observed in the Turkish population. Pinarbasi and colleagues [29] reported a significant correlation between GSTM1 and lung cancer (p = 0.0001) while other trials showed the opposite (p > 0.05) [20]. GSTM1 has been implicated in cancer etiology in multiple studies with some even noting a significantly increased risk associated with GSTM1 deletion [30,31,32,33] consistent with large Asian studies that observed an obvious relationship of the GSTM1 null genotype with lung cancer [10, 34] in accordance with an earlier meta-analysis, revealing that the GSTM null allele was a risk factor for lung cancer development [32]. These diverse findings underscore the complexity of genetic factors in lung cancer susceptibility and emphasize the necessity for further research to unravel their underlying mechanisms.

Moreover, it was reported in both Caucasians and Indians, that a notable link exists between lung adenocarcinoma and the GSTM1 null genotype [33, 35, 36]. Additionally, Liu and colleagues showed that Asians carrying the GSTM1 null variant are considered more predisposed to lung cancer and when stratified by smoking status, this genotype increases the risk of both adenocarcinoma and squamous cell carcinoma in both smokers and non-smokers [31]. However, in addition to the previous findings of the current work, no association was exhibited between the GSTM1 gene deletion and the histological types of lung cancer.

Despite the results of the present work, the authors recognize that the study is limited by being single-centered and that further large more diverse multicentric studies are needed to extrapolate these data on the general population along with extensive research to fully elucidate the complex relationships between genetic interactions and disease susceptibility.

Conclusion

According to our findings, we conclude that GSTM1 null, even when stratified for smoking status, is not related to an increased risk of incidence of lung cancer.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

DNA:

Deoxyribonucleic acid

GST:

Glutathione S-transferase

GSTM1:

Glutathione S-transferase class Mu 1

GSTP1:

Glutathione S-transferase class Pi 1

GSTT1:

Glutathione S-transferase class Theta1

NSCLC:

Non-small cell lung cancer

PAH:

Polycyclic aromatic hydrocarbons

PCR:

Polymerase chain reaction

SCLC:

Small cell lung cancer

References

  1. Nitadori JI, Inoue M, Iwasaki M, Otani T, Sasazuki S, Nagai K et al (2006) Association between lung cancer incidence and family history of lung cancer: data from a large-scale population-based cohort study, the JPHC study. Chest 130(4):968–975

    Article  PubMed  Google Scholar 

  2. de Alencar VTL, Formiga MN, de Lima VCC (2020) Inherited lung cancer: a review. Ecancermedicalscience 14:1008

    PubMed  PubMed Central  Google Scholar 

  3. Wu N, Liu S, Li J, Hu Z, Yan S, Duan H et al (2021) Deep sequencing reveals the genomic characteristics of lung adenocarcinoma presenting as ground-glass nodules (GGNs). Transl Lung Cancer Res 10(3):1239–1255

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Li L, Zhang X, Tian T, Pang L (2019) Mathematical modelling the pathway of genomic instability in lung cancer. Sci Rep 9:14136

    Article  PubMed  PubMed Central  Google Scholar 

  5. Cui J, Li G, Yin J, Li L, Tan Y, Wei H et al (2020) GSTP1 and cancer: expression, methylation, polymorphisms and signaling (Review). Int J Oncol 56(4):867–878

    PubMed  CAS  Google Scholar 

  6. Grussy K, Łaska M, Moczurad W, Król-Kulikowska M, Ściskalska M (2023) The importance of polymorphisms in the genes encoding glutathione S-transferase isoenzymes in development of selected cancers and cardiovascular diseases. Mol Biol Rep 50(11):9649–9661

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Wang Y, Ren B, Zhang L, Guo Z (2015) Correlation between metabolic enzyme GSTP1 polymorphisms and susceptibility to lung cancer. Exp Ther Med 10(4):1521–1527

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Ezzeldin N, Shalaby A, Ashur W, El Lebedy D, Farouk H, Kandil DM (2011) Glutathione S-transferase P1 gene Ile105Val polymorphism and risk of lung cancer. Medi J Cairo Univ 79(1):245–251

    Google Scholar 

  9. Liu T, Liu WZ, Sun Y, Bi XH, Zhou HF (2020) An updated meta-analysis of the relationship between glutathione S-transferase T1 null/presence gene polymorphism and the risk of lung cancer. J Cancer Res Ther 16(4):718–725

    Article  PubMed  CAS  Google Scholar 

  10. Hu Q, Li C, Huang Y, Wei Z, Chen L, Luo Y et al (2024) Effects of Glutathione S-Transferases (GSTM1, GSTT1 and GSTP1) gene variants in combination with smoking or drinking on cancers: a meta-analysis. Medicine (Baltimore) 103(14):e37707

    Article  PubMed  CAS  Google Scholar 

  11. Bell DA, Taylor JA, Paulson DF, Robertson CN, Mohler JL, Lucier GW (1993) Genetic risk and carcinogen exposure: a common inherited defect of the carcinogen-metabolism gene glutathione S-transferase M1 (GSTM1) that increases susceptibility to bladder cancer. J Natl Cancer Inst 85(14):1159–1164

    Article  PubMed  CAS  Google Scholar 

  12. Ramzy M, Solliman MED, Abdel-Hafiz H, Salah R (2011) Genetic polymorphism of GSTM1 and GSTP1 in lung cancer in Egypt. Int J Collab Res Intern Med Public Health. 3:41

    Google Scholar 

  13. Gao Y, Gao F, Hu TT, Li G, Sui YX (2017) Combined effects of glutathione S-transferase M1 and T1 polymorphisms on risk of lung cancer: evidence from a meta-analysis. Oncotarget 8(17):28135–28143

    Article  PubMed  PubMed Central  Google Scholar 

  14. Li XM, Yu XW, Yuan Y, Pu MZ, Zhang HX, Wang KJ et al (2015) Glutathione S-transferase P1, gene-gene interaction, and lung cancer susceptibility in the Chinese population: An updated meta-analysis and review. J Cancer Res Ther 11(3):565–570

    Article  PubMed  CAS  Google Scholar 

  15. Kiliç M, Ada AO, Oğuztüzün S, Demirağ F, Çelik S, Biçakçioğlu P et al (2017) Polymorphisms and protein expressions of glutathione S-transferase M1 and T1 in non-small cell lung cancer. Turk J Pharm Sci 14(3):237–242

    Article  PubMed  PubMed Central  Google Scholar 

  16. Adibhesami G, Shahsavari GR, Amiri A, Emami Razavi AN, Shamaei M, Birjandi M (2018) Glutathione S-transferase M1 (GSTM1) and T1 (GSTT1) Polymorphisms and lung cancer risk among a select group of Iranian people. Asian Pac J Cancer Prev APJCP 19(10):2921–2927

    PubMed  CAS  Google Scholar 

  17. Demir A, Altin S, Pehlivan D, Demir M, Yakar F, Seyhan EC et al (2007) The role of GSTM1 gene polymorphisms in lung cancer development in Turkish population. J Carcinog 6:13

    Article  PubMed  PubMed Central  Google Scholar 

  18. Honma HN, De Capitani EM, Perroud MW, Barbeiro AS, Toro IFC, Costa DB et al (2008) Influence of p53 codon 72 exon 4, GSTM1, GSTT1 and GSTP1*B polymorphisms in lung cancer risk in a Brazilian population. Lung Cancer Amst Neth 61(2):152–162

    Article  Google Scholar 

  19. Garte S, Gaspari L, Alexandrie AK, Ambrosone C, Autrup H, Autrup JL et al (2001) Metabolic gene polymorphism frequencies in control populations. Cancer Epidemiol Biomark Prev Publ Am Assoc Cancer Res Cosponsored Am Soc Prev Oncol 10(12):1239–1248

    CAS  Google Scholar 

  20. Oztürk O, Isbir T, Yaylim I, Kocatürk CI, Gürses A (2003) GST M1 and CYP1A1 gene polymorphism and daily fruit consumption in Turkish patients with non-small cell lung carcinomas. Vivo Athens Greece 17(6):625–632

    Google Scholar 

  21. Hemlata, Singh J, Bhardwaj A, Kumar A, Singh G, Priya K et al (2022) Comparative frequency distribution of glutathione S-transferase mu (GSTM1) and theta (GSTT1) allelic forms in Himachal Pradesh population. Egypt J Med Hum Genet 23(1):86

    Article  Google Scholar 

  22. Sharma A, Pandey A, Sharma S, Chatterjee I, Mehrotra R, Sehgal A et al (2014) Genetic polymorphism of glutathione S-transferase P1 (GSTP1) in Delhi population and comparison with other global populations. Meta Gene 2:134–142

    Article  PubMed  PubMed Central  Google Scholar 

  23. Kelsey KT, Spitz MR, Zuo ZF, Wiencke JK (1997) Polymorphisms in the glutathione S-transferase class mu and theta genes interact and increase susceptibility to lung cancer in minority populations (Texas, United States). Cancer Causes Control CCC 8(4):554–559

    Article  PubMed  CAS  Google Scholar 

  24. Roco A, Quiñones L, Agúndez JAG, García-Martín E, Squicciarini V, Miranda C et al (2012) Frequencies of 23 functionally significant variant alleles related with metabolism of antineoplastic drugs in the chilean population: comparison with caucasian and asian populations. Front Genet 3:229

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Uddin MMN, Ahmed MU, Islam MS, Islam MS, Sayeed MSB, Kabir Y et al (2014) Genetic polymorphisms of GSTM1, GSTP1 and GSTT1 genes and lung cancer susceptibility in the Bangladeshi population. Asian Pac J Trop Biomed 4(12):982–989

    Article  CAS  Google Scholar 

  26. Masood N, Taseer B, Yasmin A (2016) Association of GSTM1 and GSTT1 deletion with lung cancer development in Pakistani population. J Cancer Res Ther 12(2):731–734

    Article  PubMed  CAS  Google Scholar 

  27. Benhamou S, Lee WJ, Alexandrie AK, Boffetta P, Bouchardy C, Butkiewicz D et al (2002) Meta- and pooled analyses of the effects of glutathione S-transferase M1 polymorphisms and smoking on lung cancer risk. Carcinogenesis 23(8):1343–1350

    Article  PubMed  CAS  Google Scholar 

  28. Kalikaki A, Kanaki M, Vassalou H, Souglakos J, Voutsina A, Georgoulias V et al (2009) DNA repair gene polymorphisms predict favorable clinical outcome in advanced non-small-cell lung cancer. Clin Lung Cancer 10(2):118–123

    Article  PubMed  CAS  Google Scholar 

  29. Pinarbasi H, Silig Y, Cetinkaya O, Seyfikli Z, Pinarbasi E (2003) Strong association between the GSTM1-null genotype and lung cancer in a Turkish population. Cancer Genet Cytogenet 146(2):125–129

    Article  PubMed  CAS  Google Scholar 

  30. Langevin SM, Ioannidis JPA, Vineis P, Taioli E (2010) Genetic Susceptibility to Environmental Carcinogens group (GSEC). Assessment of cumulative evidence for the association between glutathione S-transferase polymorphisms and lung cancer: application of the Venice interim guidelines. Pharmacogenet Genomics 20(10):586–97

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Liu D, Wang F, Wang Q, Guo X, Xu H, Wang W et al (2012) Association of glutathione S-transferase M1 polymorphisms and lung cancer risk in a Chinese population. Clin Chim Acta Int J Clin Chem 414:188–190

    Article  CAS  Google Scholar 

  32. Liu X, Li Z, Zhang Z, Zhang W, Li W, Xiao Z et al (2014) Meta-analysis of GSTM1 null genotype and lung cancer risk in Asians. Med Sci Monit Int Med J Exp Clin Res 20:1239–1245

    Google Scholar 

  33. Sharma N, Singh A, Singh N, Behera D, Sharma S (2015) Genetic polymorphisms in GSTM1, GSTT1 and GSTP1 genes and risk of lung cancer in a North Indian population. Cancer Epidemiol 39(6):947–955

    Article  PubMed  Google Scholar 

  34. Zhao Y, Zeng J, Zhang Y, Lu S, Zhao E, Huang Z et al (2014) GSTM1 polymorphism and lung cancer risk among East Asian populations: a meta-analysis. Tumour Biol J Int Soc Oncodevelop Biol Med 35(7):6493–6500

    Article  CAS  Google Scholar 

  35. Bhara R, Tiwari S, Vijayaraghavalu S, Kumar M (2019) Genetic polymorphisms of xenobiotic metabolizing genes (GSTM1, GSTT1, GSTP1), gene-gene interaction with association to lung cancer risk in North India; a case control study. Asian Pac J Cancer Prev APJCP 20(9):2707–14

    Article  Google Scholar 

  36. Carlsten C, Sagoo GS, Frodsham AJ, Burke W, Higgins JPT (2008) Glutathione S-Transferase M1 (GSTM1) Polymorphisms and lung cancer: a literature-based systematic HuGE review and meta-analysis. Am J Epidemiol 167(7):759–774

    Article  PubMed  CAS  Google Scholar 

Download references

Funding

Not applicable.

Author information

Authors and Affiliations

Authors

Contributions

G.H.: Data collection, editing and revision. A.G.: Manuscript writing, review, editing and final revision. N.A.E.: Genetic analysis and PCR. E.A.: Genetic analysis and PCR. M.F.H: revision R.I.EK.: revision N.E.G.: Data collection, editing and revision. All authors have read and approved this manuscript.

Corresponding author

Correspondence to Gehan Hamdy.

Ethics declarations

Ethics approval and consent to participate

The study was approved by the ethical committee of the National Research Centre. Reference number: 10088. All subjects gave written informed consent.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s Note

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

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/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hamdy, G., Gharib, A., Emara, N.A.M. et al. Impact of the deletion glutathione S-transferase (class Mu) on lung cancer risk among smokers. Egypt J Bronchol 18, 54 (2024). https://doi.org/10.1186/s43168-024-00306-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43168-024-00306-w