Next Article in Journal
Comparative Study of Classification Algorithms for Various DNA Microarray Data
Previous Article in Journal
CCL2 rs1024611Gene Polymorphism in Philadelphia-Negative Myeloproliferative Neoplasms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

UGT1A1 Gene Polymorphism Contributes as a Risk Factor for Lung Cancer: A Pilot Study with Patients from the Amazon

by
Esdras E. B. Pereira
1,
Luciana P. C. Leitão
2,
Roberta B. Andrade
1,
Antônio A. C. Modesto
1,
Bruno M. Fernandes
2,
Rommel M. R. Burbano
1,2,
Paulo P. Assumpção
2,
Marianne R. Fernandes
2,*,
João F. Guerreiro
1,
Sidney E. B. dos Santos
1,2 and
Ney P. C. dos Santos
1,2
1
Laboratory of Human and Medical Genetics, Institute of Biological Science, Federal University of Pará, Belem 66077-830, Brazil
2
Oncology Research Center, Federal University of Pará, Belem 66073-005, Brazil
*
Author to whom correspondence should be addressed.
Genes 2022, 13(3), 493; https://doi.org/10.3390/genes13030493
Submission received: 29 January 2022 / Revised: 15 February 2022 / Accepted: 16 February 2022 / Published: 11 March 2022
(This article belongs to the Section Human Genomics and Genetic Diseases)

Abstract

:
Lung cancer is one of the most frequent neoplasms in the world. Because it is a complex disease, its formation occurs in several stages, stemming from interactions between environmental risk factors, such as smoking, and individual genetic susceptibility. Our objective was to investigate associations between a UGT1A1 gene polymorphism (rs8175347) and lung cancer risk in an Amazonian population. This is a pilot study, case-controlled study, which included 276 individuals with cancer and without cancer. The samples were analyzed for polymorphisms of the UGT1A1 gene (rs8175347) and genotyped in PCR, followed by fragment analysis in which we applied a previously developed set of informative ancestral markers. We used logistic regression to identify differences in allelic and genotypic frequencies between individuals. Individuals with the TA7 allele have an increased chance of developing lung adenocarcinoma (p = 0.035; OR: 2.57), as well as those with related genotypes of reduced or low enzymatic activity: TA6/7, TA5/7, and TA7/7 (p = 0.048; OR: 8.41). Individuals with homozygous TA7/7 have an increased chance of developing squamous cell carcinoma of the lung (p = 0.015; OR: 4.08). Polymorphism in the UGT1A1 gene (rs8175347) may contribute as a risk factor for adenocarcinoma and lung squamous cell carcinoma in the population of the Amazon region.

Graphical Abstract

1. Introduction

Lung neoplasms are among the most prevalent types of cancer in the world population, representing 11.4% of all registered cancers, and are responsible for 18% of cancer deaths [1]. This frequency varies between countries, depending on their demographic characteristics, smoking rate, and level of economic development [2]. Lung cancer can be divided into two categories, small cell lung cancer (SCLC), responsible for 15–20% of cases; and non-small cell lung cancer (NSCLC), which represents 80–85% of cases [3,4].
The well-known risk factor for the development of lung cancer is smoking, given the presence of substances contained in tobacco associated with carcinogenesis [5]. However, studies have shown that about half of new cases are in people who have never smoked, or have stopped smoking for many years [6]. This shows that pulmonary carcinogenesis is a complex and gradual process, with complex interactions between environmental risk factors and individual genetic susceptibility [7].
Genetic susceptibility to lung cancer, often modulated by smoking behavior, has been investigated [8]. Disease risk may be associated with genetic variables such as single nucleotide polymorphisms (SNPs) in genes related to metabolism, DNA damage repair, and cell cycle control [9,10]. This knowledge is important when it comes to lung cancer, and its use can favor the development of population screening tools [11].
The UGT1A1 gene originates from the UDP-glucuronosyltransferase enzyme, which is active in the glucuronidation metabolic pathway, one of the main biotransformation pathways of xenobiotics [12,13]. This gene has been linked to the development of several types of cancer: colon, breast, and prostate. The UGT1A1 enzyme plays an important role in the detoxification and metabolization of several carcinogens [14,15].
The aim of this study was to investigate possible associations between UGT1A1 gene polymorphism (rs8175347) and lung cancer risk in an Amazonian population.

2. Materials and Methods

2.1. Ethical Conformity

This pilot study was approved by the Research Ethics Committees of the Oncology Research Center, under protocol 927.808/2014, and by the João de Barros Barreto University Hospital, under protocol number 941.207/2015, both in the city of Belém-Pará, Amazon region of Brazil. All participants signed an informed consent form.

2.2. Case and Control

Participants were recruited from public healthcare centers, of both sexes, not belonging to the same family nucleus, and of the same socioeconomic level. Data and samples were collected from 276 individuals, of which 80 were patients with primary lung cancer (case group), defined and classified in the histopathological examination, and 196 patients were without any type of cancer (control group). Both groups had a survey of demographic and clinical data, which included age, sex, and smoking.

2.3. DNA Extraction and Quantification

Extraction of genomic DNA from peripheral blood leukocytes was conducted using a Mini Spin Plus Kit (P. 250, Biopur, Biometrix) according to the manufacturer’s recommendations. DNA concentration and purity were measured with a NanoDrop 1000 spectrophotometer (Thermo Scientific NanoDrop 1000; NanoDrop Technologies, Wilmington, DE, USA).

2.4. Genotyping

The UGT1A1 gene polymorphism (rs8175347) was genotyped by a multiplex PCR reaction followed by capillary electrophoresis. For the amplifications, primers were used: F:5′-CTCTGAAAGTGAACTCCCTGCT-3′; R:5′-AGAGGTTCGCCCTTCCTAT-3′. Analysis of the PCR amplicons was performed from an electrophoresis using the ABI Prism 3130 sequencer and GeneMapper ID v.3.2 software [16]. The genotypes found were: TA6/6 (UGT1A1*1/*1), TA6/7 (UGT1A1*1/*28), T5/7 (UGT1A1*36/*28) and TA7/7 (UGT1A1*28/*28). In the literature, these genotypes can be grouped by the degree of enzymatic activity of UGT1A1 as: normal (TA6/6), reduced (TA6/7 and TA5/7), and low (TA7/7) enzymatic activity [17].

2.5. Hardy–Weinberg Equilibrium Analysis (HWE)

The allelic and genotypic frequency of the polymorphism was determined by direct counting of alleles, and then Hardy–Weinberg equilibrium (HWE) was calculated using the default parameters of the Arlequin 3.5.1.2 software (Swiss Institute of Bioinformatics, Bern, Switzerland).

2.6. Genetic Ancestrality Analysis

Genotyping was performed to analyze the ancestry of the samples, performed according to Ramos et al. [18], using 61 informative markers of autosomal ancestry in three multiplex PCR reactions. Amplicons were analyzed by electrophoresis using the ABI Prism 3130 sequencer and GeneMapper ID v. 3.2 software (Applied Biosystems, Life Technologies, Carlsbad, CA, USA). The individual proportions of European, African, and Amerindian genetic ancestry were estimated using Structure v. 2.3.3 software (Stanford University, Stanford, CA, USA), assuming three parental populations [19].

2.7. Statistical Analysis

All statistical analyzes were performed using the SPSS 20.0 software (IBM, Armonk, NY, USA) statistical package. For comparative analysis between the study groups with regard to demographic and clinical variables, Pearson’s Chi-square and the Mann–Whitney test were applied. To analyze the association of polymorphisms with risk to lung cancer and the histological types, a logistic regression was performed, estimating the odds ratios (OR) and their 95% confidence intervals (CI). The variables of sex, age, and smoking were controlled in this multivariate analysis, to avoid a confounding association. A significance level of p < 0.05 was considered for all statistical analyses.

3. Results

In the results of demographic and clinical analyses, it can be observed that the groups differed in terms of sex, age, and smoking. Most cases of lung cancer were squamous cell carcinoma, followed by large cell carcinoma and adenocarcinoma (Table 1). The ancestry analyses performed revealed that the case and control groups had a similar ancestral genomic profile, with a greater European contribution to both populations (Table 1).
It was evidenced that the UGT1A1 gene polymorphism (rs8175347) was present in the HWE (p = 0.340). Four genotypes were presented, TA6/6, TA6/7, TA5/7, and TA7/7, with frequencies of 41%, 45%, 1%, and 13% respectively. As for the allele frequency, three alleles were observed, TA5, TA6, and TA7, with 0.5%, 63.4%, and 36.1%, respectively.
The allele frequencies between the control and lung cancer groups and their histological types are listed in Table 2. It was observed that the TA7 allele was present more frequently in the lung adenocarcinoma group (54.1%). In the analysis for lung adenocarcinoma, it was observed that individuals with the TA7 allele were about 2.5 times more likely to develop the disease (p = 0.035; OR: 2.57; 95% CI: 1.07–6.20).
The genotypic frequencies between the control and lung cancer groups and their histological types are listed in Table 3. The TA7/7 genotype was more frequent in the squamous cell carcinoma group (22.0%). Individuals with the TA7/7 genotype were about four times more likely to develop lung squamous cell carcinoma (p = 0.015; OR: 4.08; 95% CI: 1.32–12.61).
The genotypic frequencies by the degree of UGT1A1 enzymatic activities, between the control and lung cancer groups and their histological types, are listed in Table 4. Genotypes with reduced or low enzyme activity (TA6/7, TA5/7, TA7/7) were more frequent in the lung adenocarcinoma group (91.7%). Furthermore, individuals with a low or reduced degree of enzyme activity genotypes were about 8 times more likely to develop the disease (p = 0.048; OR: 8.41; 95%CI: 1.02–69.55).

4. Discussion

In the world, lung neoplasms are the second most common cancer diagnosis in both sexes, being more frequent among men with an average age of 70 years, and associated with tobacco consumption [1,20,21]. In Brazil, lung cancer is the third most common cancer among men and the fourth most common cancer among women, and 85% of diagnosed cases are associated with the consumption of tobacco derivatives [22,23]. This is similar to what was observed in our study, where the group of individuals with lung cancer was mainly composed of men aged over 65 years and with a history of smoking.
The prevalence of lung cancer in men is often associated with smoking, as the proportion of men who smoke is higher than that of women. In addition, men are also exposed to carcinogens in some occupational activities, which contributes to the frequency being higher in this group [24,25,26]. However, these gender differences vary between developed and developing countries, due to differences in tobacco consumption, and exposure to exogenous and endogenous risk factors [1,5].
Regarding age, it is observed that about 34% of lung cancer cases in the world are diagnosed between 65 and 74 years of age, and the elderly population comprises 62% of all cases [1]. Aging favors genomic instability, leading to the accumulation of cells with different molecular aberrations that alter internal homeostasis, increasing susceptibility to carcinogens and, consequently, lung carcinogenesis [27]. According to Schneider et al. (2021) some histological types of lung cancer are caused by the expression of oncogenic drivers, which can be stratified by age, such as adenocarcinoma, revealing the interaction between aging and lung carcinogenesis [28].
In our study, the most prevalent histological type was squamous cell carcinoma (51.7%), followed by large cell carcinoma (17.5%), and adenocarcinoma (15.0%). In Brazil, 86.7% of cases are NSCLC and 13.7% SCLC. Among NSCLC, the most common histological type was adenocarcinoma (50.0%), followed by squamous cell carcinoma (42.1%), and large cell carcinoma (7.9%) [29]. these differences may reflect the reduction in cases of squamous cell carcinoma and the increase in adenocarcinoma in the last 30 years, due to changes in the pattern of tobacco consumption in some regions of the country [30].
Smoking is the well-known risk factor for developing lung cancer [5,6]. Pulmonary carcinogenesis is a complex process, with interactions that modulate the potential risks for the disease [7,31]. Many studies have described how germline variants influence susceptibility to lung cancer, including those linked to smoking [32,33,34].
In the present study, the analysis of the UGT1A1 gene polymorphism (rs8175347) showed relevant results. The TA7/7 genotype was present in 13% of the investigated individuals. In Brazil, the frequency of the TA7/7 genotype of this polymorphism occurs in between 3 and 17% of the population, being more frequent among Afrodescendants and less frequent among Amerindians [35].
As for allelic frequency, the TA7 allele was frequent in 36.1% of the individuals studied. This frequency is similar to that observed in the wider world [36], where the distribution of the TA7 allele is 34.9%, in Latin America 31.4%, and in Brazil between 30.0 and 33.0% [37,38,39]. Studies show that the TA7 allele has a frequency of 42–56% in Afrodescendants, 26–31% in Caucasians, and only 9–16% in Asian populations [40].
In our study, individuals with genotypes related to reduced or low enzymatic activity (TA6/7, TA5/7 and TA7/7) had increased chances (p = 0.048; OR: 8.41, 95%CI: 1.07–69.55) for the development of adenocarcinoma of lung. Similar results were observed in the allelic frequency: individuals with the TA7 allele have an increased chance of developing lung adenocarcinoma (p = 0.035; OR: 2.57; 95%CI: 1.07–6.20). Furthermore, individuals with the TA7/TA7 genotype have an increased chance of developing squamous cell carcinoma of the lung (p = 0.015; OR:4.077; 95%CI:1.318–12.612).
The variant allelic TA7 is characterized by seven thymine-adenine (TA) repeats within the promoter region, unlike the wild-type TA6 allele which has six TA repeats. This extra repeat impairs proper gene transcription [41,42,43]. This results in a 25–70% reduction in enzyme activity, depending on the presence of one or two TA7 variant alleles, respectively, which reduce glucuronidation [44,45,46].
UGT1A1 enzymes were observed in the glucuronidation of estradiol and of a precursor of the potent organic carcinogen benzo(α)pyrene-7,8-dihydrodiol-9,10-epoxide (BPDE), which is found in cigarette smoke [40]. There is a correlation between UGT1A1 genotypes with the expression of UDP-glucuronosyltransferase and glucuronidation activity. Individuals with genotype TA7/7 have a lower level of precursor carcinogen in liv-er microsomes when compared to those with genotypes TA6/7 or TA6/6 [40].
In the present study, smoking was present in 83.7% of patients with lung cancer, similar to that observed in other surveys, where between 80% and 90% of lung cancers are attributed to smoking [47,48]. Tobacco contains dozens of carcinogenic agents that are harmful to humans, who undergo biotransformation through various metabolic path-ways [49,50]. Studies show that genetic polymorphisms in genes that encode enzymes involved in the metabolism of tobacco carcinogens, such as UGT1A1, can affect the individual risk of lung cancer [51,52].
The reduction in or low enzymatic activity of UGT1A1 reduces glucuronidation, increasing exposure to the carcinogens present in tobacco, which favor carcinogenesis [42]. Our study observed this association. Individuals with the genotypes of reduced or low enzymatic activity, or with an allele TA7 or with genotype homozygous TA7/7, had a higher chance of some histological types of lung cancer. It is possible that these associations were not observed in all histological types due to the small sample size in some clusters in this pilot study.
This finding corroborates what was observed by Nishikawa et al. (2016) [53] in their study of 194 patients with lung cancer, who found that the genotype homozygous TA7/7 increases the risk of developing lung cancer by approximately five times. They observed that squamous cell carcinoma was predominant in the lung cancer patients with genotype homozygous TA7/7 and smoking history, similar to our study.
This reinforces the findings of other studies that indicate that reducing the activity or expression of UGT1A1 can influence the elimination of precursors to carcinogens [14,48]. This relationship facilitates the understanding of the identification of associations between the TA7 allelic variant with the susceptibility to oxidative damage and, consequently, the increased risk for other types of cancer, such as breast, ovarian, prostate, head and neck, and colorectal cancers, reported in some populations [48,54,55,56,57].
In addition to the susceptibility to the development of lung cancer, studies regarding polymorphism in the UGT1A1 gene (rs8175347) have identified its association with the response to chemotherapy with irinotecan. The reduced enzymatic action of UGT1A1, due to the TA7 variant, may be associated with severe toxicities in patients receiving irinotecan, including patients with lung cancer. This shows this polymorphism is a strong candidate for use in clinical practice as well [57,58,59].
Importantly, this study is one of the few that has investigated the association between how polymorphism that can alter the length of the UGT1A1 gene promoter and the risk of lung adenocarcinoma and squamous cell carcinoma. Further epidemiological investigations involving larger groups of individuals should be conducted to confirm the results and determine whether the TA7 variant allele is an isolated risk factor or associated with environmental factors such as smoking.
The validation of these findings may favor, in the future, the screening of individuals with greater susceptibility to developing the disease by facilitating the establishment of personalized preventive measures for early diagnosis, consequently reducing the cost for health services and lowering mortality rates from this malignant neoplasm.

5. Conclusions

The UGT1A1 gene polymorphism (rs8175347) showed a significant association with lung squamous cell carcinoma and lung adenocarcinoma in the population of the Amazon region. Individuals with homozygous TA7/7 had an increased risk of lung squamous cell carcinoma. Those with the TA7 allele or with genotypes associated with reduced or low UGT1A1 activity had an increased risk for lung adenocarcinoma.

Author Contributions

E.E.B.P. designed the study, processed the data, and wrote the article. L.P.C.L. and B.M.F. contributed to the writing of the article. R.B.A. and A.A.C.M. contributed to the genotyping and data analysis. R.M.R.B., P.P.A., S.E.B.d.S., M.R.F., J.F.G. and N.P.C.d.S. were the project coordinators. All authors have read and agreed to the published version of the manuscript.

Funding

Coordenação de Aperfeiçoamento de Pessoal de Nível Superior (CAPES); Fundação Amazônia de Amparo a Estudos e Pesquisas (FAPESPA); and Universidade Federal do Pará (UFPA).

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Research Ethics Committees of the Oncology Research Center of UFPA, under protocol number 927.808/2014, and by the João de Barros Barreto University Hospital of UFPA, under protocol number 941.207/2015. All participants signed an informed consent form.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Acknowledgments

We acknowledge the Universidade Federal do Pará (UFPA); Faculty of Medicine (FAMED/UFPA); Oncology Research Center (NPO/UFPA); Graduate Program in Genetics and Molecular Biology (PPGBM/UFPA); Human and Medical Genetics Laboratory (LGHM/UFPA); João de Barros Barreto University Hospital (HUJBB/UFPA).

Conflicts of Interest

The authors declare that they have no competing interests.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Bade, B.C.; Dela Cruz, C.S. Lung Cancer 2020: Epidemiology, Etiology, and Prevention. Clin. Chest Med. 2020, 41, 1–24. [Google Scholar] [CrossRef] [PubMed]
  3. Duma, N.; Santana-Davila, R.; Molina, J.R. Non-Small Cell Lung Cancer: Epidemiology, Screening, Diagnosis, and Treatment. Mayo Clin. Proc. 2019, 94, 1623–1640. [Google Scholar] [CrossRef] [PubMed]
  4. Saltos, A.; Shafique, M.; Chiappori, A. Update on the Biology, Management, and Treatment of Small Cell Lung Cancer (SCLC). Front. Oncol. 2020, 10, 1074. [Google Scholar] [CrossRef]
  5. Barta, J.A.; Powell, C.A.; Wisnivesky, J.P. Global Epidemiology of Lung Cancer. Ann. Glob. Health 2019, 85, 8. [Google Scholar] [CrossRef] [Green Version]
  6. Wang, X.; Ricciuti, B.; Nguyen, T.; Li, X.; Rabin, M.S.; Awad, M.M.; Lin, X.; Johnson, B.E.; Christiani, D.C. Association between Smoking History and Tumor Mutation Burden in Advanced Non-Small Cell Lung Cancer. Cancer Res. 2021, 81, 2566–2573. [Google Scholar] [CrossRef]
  7. Hu, Z.; Chen, J.; Meng, P.; Li, M. Association between NEAT1 polymorphism and the risk of lung cancer: A protocol for systematic review and meta-analysis. Medicine 2021, 100, e25478. [Google Scholar] [CrossRef]
  8. Liu, Y.; Xia, J.; McKay, J.; Tsavachidis, S.; Xiao, X.; Spitz, M.R.; Cheng, C.; Byun, J.; Hong, W.; Li, Y.; et al. Rare deleterious germline variants and risk of lung cancer. NPJ Precis. Oncol. 2021, 5, 12. [Google Scholar] [CrossRef]
  9. Liu, D.; Gao, Y.; Li, L.; Chen, H.; Bai, L.; Qu, Y.; Zhou, B.; Yan, Y.; Zhao, Y. Single nucleotide polymorphisms in breast cancer susceptibility gene 1 are associated with susceptibility to lung cancer. Oncol. Lett. 2021, 21, 424. [Google Scholar] [CrossRef]
  10. Dang, X.; Zhao, W.; Li, C.; Yang, H.; Li, D.; Zhang, S.; Jin, T. Impact of COL6A4P2 gene polymorphisms on the risk of lung cancer: A case-control study. PLoS ONE 2021, 16, e0252082. [Google Scholar] [CrossRef]
  11. Lebrett, M.B.; Crosbie, E.J.; Smith, M.J.; Woodward, E.R.; Evans, D.G.; Crosbie, P.A.J. Targeting lung cancer screening to individuals at greatest risk: The role of genetic factors. J. Med. Genet. 2021, 58, 217–226. [Google Scholar] [CrossRef] [PubMed]
  12. Steventon, G. Uridine diphosphate glucuronosyltransferase 1A1. Xenobiotica 2020, 50, 64–76. [Google Scholar] [CrossRef] [PubMed]
  13. Sissung, T.M.; Barbier, R.H.; Price, D.K.; Plona, T.M.; Pike, K.M.; Mellott, S.D.; Baugher, R.N.; Whiteley, G.R.; Soppet, D.R.; Venzon, D.; et al. Comparison of Eight Technologies to Determine Genotype at the UGT1A1 (TA). Int. J. Mol. Sci. 2020, 21, 896. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Bajro, M.H.; Josifovski, T.; Panovski, M.; Jankulovski, N.; Nestorovska, A.K.; Matevska, N.; Petrusevska, N.; Dimovski, A.J. Promoter length polymorphism in UGT1A1 and the risk of sporadic colorectal cancer. Cancer Genet. 2012, 205, 163–167. [Google Scholar] [CrossRef]
  15. Clendenen, T.; Zeleniuch-Jacquotte, A.; Wirgin, I.; Koenig, K.L.; Afanasyeva, Y.; Lundin, E.; Arslan, A.A.; Axelsson, T.; Försti, A.; Hallmans, G.; et al. Genetic variants in hormone-related genes and risk of breast cancer. PLoS ONE 2013, 8, e69367. [Google Scholar] [CrossRef]
  16. Pinto, P.; Salgado, C.; Santos, N.P.; Santos, S.; Ribeiro-dos-Santos, Â. Influence of Genetic Ancestry on INDEL Markers of NFKβ1, CASP8, PAR1, IL4 and CYP19A1 Genes in Leprosy Patients. PLoS Negl. Trop. Dis. 2015, 9, e0004050. [Google Scholar] [CrossRef] [Green Version]
  17. Olatunya, O.S.; Albuquerque, D.M.; Akanbi, G.O.; Aduayi, O.S.; Taiwo, A.B.; Faboya, O.A.; Kayode, T.S.; Leonardo, D.P.; Adekile, A.; Costa, F.F. Uridine diphosphate glucuronosyl transferase 1A (UGT1A1) promoter polymorphism in young patients with sickle cell anaemia: Report of the first cohort study from Nigeria. BMC Med. Genet. 2019, 20, 160. [Google Scholar] [CrossRef] [Green Version]
  18. Ramos, B.R.; D’Elia, M.P.; Amador, M.A.; Santos, N.P.; Santos, S.E.; da Cruz Castelli, E.; Witkin, S.S.; Miot, H.A.; Miot, L.D.; da Silva, M.G. Neither self-reported ethnicity nor declared family origin are reliable indicators of genomic ancestry. Genetica 2016, 144, 259–265. [Google Scholar] [CrossRef] [Green Version]
  19. Carvalho, D.C.; Wanderley, A.V.; Amador, M.A.; Fernandes, M.R.; Cavalcante, G.C.; Pantoja, K.B.; Mello, F.A.; de Assumpção, P.P.; Khayat, A.S.; Ribeiro-Dos-Santos, Â.; et al. Amerindian genetic ancestry and INDEL polymorphisms associated with susceptibility of childhood B-cell Leukemia in an admixed population from the Brazilian Amazon. Leuk. Res. 2015, 39, 1239–1245. [Google Scholar] [CrossRef] [Green Version]
  20. Thandra, K.C.; Barsouk, A.; Saginala, K.; Aluru, J.S. Epidemiology of lung cancer. Contemp. Oncol. 2021, 25, 45–52. [Google Scholar] [CrossRef]
  21. Rodak, O.; Peris-Díaz, M.D.; Olbromski, M.; Podhorska-Okołów, M.; Dzięgiel, P. Current Landscape of Non-Small Cell Lung Cancer: Epidemiology, Histological Classification, Targeted Therapies, and Immunotherapy. Cancers 2021, 13, 4705. [Google Scholar] [CrossRef] [PubMed]
  22. Brasil. Ministério da Saúde. Instituto Nacional de Câncer José Alencar Gomes da Silva. Estimativa 2020: Incidência de Câncer no Brasil. Rio de Janeiro: INCA; 2019. Available online: https://www.inca.gov.br/sites/ufu.sti.inca.local/files//media/document//estimativa-2020-incidencia-de-cancer-no-brasil.pdf (accessed on 1 November 2021).
  23. Araujo, L.H.; Baldotto, C.; Castro, G.; Katz, A.; Ferreira, C.G.; Mathias, C.; Mascarenhas, E.; Lopes, G.L.; Carvalho, H.; Tabacof, J.; et al. Lung cancer in Brazil. J. Bras. Pneumol. 2018, 44, 55–64. [Google Scholar] [CrossRef] [PubMed]
  24. Delva, F.; Margery, J.; Laurent, F.; Petitprez, K.; Pairon, J.C.; Group, R.W. Medical follow-up of workers exposed to lung carcinogens: French evidence-based and pragmatic recommendations. BMC Public Health 2017, 17, 191. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Stapelfeld, C.; Dammann, C.; Maser, E. Sex-specificity in lung cancer risk. Int. J. Cancer 2020, 146, 2376–2382. [Google Scholar] [CrossRef]
  26. Ragavan, M.; Patel, M.I. The evolving landscape of sex-based differences in lung cancer: A distinct disease in women. Eur. Respir. Rev. 2022, 31, 210100. [Google Scholar] [CrossRef]
  27. Meiners, S.; Eickelberg, O.; Königshoff, M. Hallmarks of the ageing lung. Eur. Respir. J. 2015, 45, 807–827. [Google Scholar] [CrossRef]
  28. Schneider, J.L.; Rowe, J.H.; Garcia-de-Alba, C.; Kim, C.F.; Sharpe, A.H.; Haigis, M.C. The aging lung: Physiology, disease, and immunity. Cell 2021, 184, 1990–2019. [Google Scholar] [CrossRef]
  29. Costa, G.; Thuler, L.C.; Ferreira, C.G. Epidemiological changes in the histological subtypes of 35,018 non-small-cell lung cancer cases in Brazil. Lung Cancer 2016, 97, 66–72. [Google Scholar] [CrossRef]
  30. Tsukazan, M.T.R.; Vigo, Á.; Silva, V.D.D.; Barrios, C.H.; Rios, J.O.; Pinto, J.A.F. Lung cancer: Changes in histology, gender, and age over the last 30 years in Brazil. J. Bras. Pneumol. 2017, 43, 363–367. [Google Scholar] [CrossRef]
  31. Sengupta, D.; Banerjee, S.; Mukhopadhyay, P.; Mitra, R.; Chaudhuri, T.; Sarkar, A.; Bhattacharjee, G.; Nath, S.; Roychoudhury, S.; Bhattacharjee, S.; et al. A comprehensive meta-analysis and a case-control study give insights into genetic susceptibility of lung cancer and subgroups. Sci. Rep. 2021, 11, 14572. [Google Scholar] [CrossRef]
  32. Tlemsani, C.; Takahashi, N.; Pongor, L.; Rajapakse, V.N.; Tyagi, M.; Wen, X.; Fasaye, G.A.; Schmidt, K.T.; Desai, P.; Kim, C.; et al. Whole-exome sequencing reveals germline-mutated small cell lung cancer subtype with favorable response to DNA repair-targeted therapies. Sci. Transl. Med. 2021, 13, eabc7488. [Google Scholar] [CrossRef]
  33. Yang, I.A.; Holloway, J.W.; Fong, K.M. Genetic susceptibility to lung cancer and co-morbidities. J. Thorac. Dis. 2013, 5 (Suppl. 5), S454–S462. [Google Scholar] [CrossRef]
  34. Gabriel, A.A.G.; Atkins, J.R.; Penha, R.C.C.; Smith-Byrne, K.; Gaborieau, V.; Voegele, C.; Abedi-Ardekani, B.; Milojevic, M.; Olaso, R.; Meyer, V.; et al. Genetic analysis of lung cancer reveals novel susceptibility loci and germline impact on somatic mutation burden. medRxiv 2021. [Google Scholar] [CrossRef]
  35. Marques, S.C.; Ikediobi, O.N. The clinical application of UGT1A1 pharmacogenetic testing: Gene-environment interactions. Hum. Genom. 2010, 4, 238–249. [Google Scholar] [CrossRef] [Green Version]
  36. Whirl-Carrillo, M.; Huddart, R.; Gong, L.; Sangkuhl, K.; Thorn, C.F.; Whaley, R.; Klein, T.E. An Evidence-Based Framework for Evaluating Pharmacogenomics Knowledge for Personalized Medicine. Clin. Pharmacol. Ther. 2021, 110, 563–572. [Google Scholar] [CrossRef] [PubMed]
  37. Turatti, L.; Sprinz, E.; Lazzaretti, R.K.; Kuhmmer, R.; Agnes, G.; Silveira, J.M.; Basso, R.P.; Pinheiro, C.A.; Silveira, M.F.; de Almeida, S.; et al. Short communication: UGT1A1*28 variant allele is a predictor of severe hyperbilirubinemia in HIV-infected patients on HAART in southern Brazil. AIDS Res. Hum. Retrovir. 2012, 28, 1015–1018. [Google Scholar] [CrossRef]
  38. Cavalcante, G.C.; Amador, M.A.; Ribeiro Dos Santos, A.M.; Carvalho, D.C.; Andrade, R.B.; Pereira, E.E.; Fernandes, M.R.; Costa, D.F.; Santos, N.P.; Assumpção, P.P.; et al. Analysis of 12 variants in the development of gastric and colorectal cancers. World J. Gastroenterol. 2017, 23, 8533–8543. [Google Scholar] [CrossRef] [Green Version]
  39. De Oliveira Almeida, V.C.; Ribeiro, D.D.; Gomes, K.B.; Godard, A.L. Polymorphisms of CYP2C9, VKORC1, MDR1, APOE and UGT1A1 genes and the therapeutic warfarin dose in Brazilian patients with thrombosis: A prospective cohort study. Mol. Diagn. Ther. 2014, 18, 675–683. [Google Scholar] [CrossRef]
  40. Barbarino, J.M.; Haidar, C.E.; Klein, T.E.; Altman, R.B. PharmGKB summary: Very important pharmacogene information for UGT1A1. Pharm. Genom. 2014, 24, 177–183. [Google Scholar] [CrossRef] [Green Version]
  41. Fang, J.L.; Lazarus, P. Correlation between the UDP-glucuronosyltransferase (UGT1A1) TATAA box polymorphism and carcinogen detoxification phenotype: Significantly decreased glucuronidating activity against benzo(a)pyrene-7,8-dihydrodiol(-) in liver microsomes from subjects with the UGT1A1*28 variant. Cancer Epidemiol. Biomark. Prev. 2004, 13, 102–109. [Google Scholar] [CrossRef] [Green Version]
  42. Takano, M.; Sugiyama, T. UGT1A1 polymorphisms in cancer: Impact on irinotecan treatment. Pharmgenom. Pers. Med. 2017, 10, 61–68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Patel, P.; Xue, Z.; King, K.S.; Parham, L.; Ford, S.; Lou, Y.; Bakshi, K.K.; Sutton, K.; Margolis, D.; Hughes, A.R.; et al. Evaluation of the effect of UGT1A1 polymorphisms on the pharmacokinetics of oral and long-acting injectable cabotegravir. J. Antimicrob. Chemother. 2020, 75, 2240–2248. [Google Scholar] [CrossRef] [PubMed]
  44. Belkhir, L.; Seguin-Devaux, C.; Elens, L.; Pauly, C.; Gengler, N.; Schneider, S.; Ruelle, J.; Haufroid, V.; Vandercam, B. Impact of UGT1A1 polymorphisms on Raltegravir and its glucuronide plasma concentrations in a cohort of HIV-1 infected patients. Sci. Rep. 2018, 8, 7359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Strassburg, C.P. Pharmacogenetics of Gilbert’s syndrome. Pharmacogenomics 2008, 9, 703–715. [Google Scholar] [CrossRef]
  46. Nelson, R.S.; Seligson, N.D.; Bottiglieri, S.; Carballido, E.; Cueto, A.D.; Imanirad, I.; Levine, R.; Parker, A.S.; Swain, S.M.; Tillman, E.M.; et al. UGT1A1 Guided Cancer Therapy: Review of the Evidence and Considerations for Clinical Implementation. Cancers 2021, 13, 1566. [Google Scholar] [CrossRef]
  47. Zinser-Sierra, J.W. Tobacco use and lung cancer. Salud. Publica Mex. 2019, 61, 303–307. [Google Scholar] [CrossRef] [Green Version]
  48. Lim, J.E.; Kimm, H.; Jee, S.H. Combined effects of smoking and bilirubin levels on the risk of lung cancer in Korea: The severance cohort study. PLoS ONE 2014, 9, e103972. [Google Scholar] [CrossRef]
  49. San Jose, C.; Cabanillas, A.; Benitez, J.; Carrillo, J.A.; Jimenez, M.; Gervasini, G. CYP1A1 gene polymorphisms increase lung cancer risk in a high-incidence region of Spain: A case control study. BMC Cancer 2010, 10, 463. [Google Scholar] [CrossRef] [Green Version]
  50. Espina, C.; Straif, K.; Friis, S.; Kogevinas, M.; Saracci, R.; Vainio, H.; Schüz, J. European Code against Cancer 4th Edition: Environment, occupation and cancer. Cancer Epidemiol. 2015, 39 (Suppl. 1), S84–S92. [Google Scholar] [CrossRef] [Green Version]
  51. Jin, Y.; Xu, H.; Zhang, C.; Kong, Y.; Hou, Y.; Xu, Y.; Xue, S. Combined effects of cigarette smoking, gene polymorphisms and methylations of tumor suppressor genes on non small cell lung cancer: A hospital-based case-control study in China. BMC Cancer 2010, 10, 422. [Google Scholar] [CrossRef] [Green Version]
  52. Pliarchopoulou, K.; Voutsinas, G.; Papaxoinis, G.; Florou, K.; Skondra, M.; Kostaki, K.; Roussou, P.; Syrigos, K.; Pectasides, D. Correlation of CYP1A1, GSTP1 and GSTM1 gene polymorphisms and lung cancer risk among smokers. Oncol. Lett. 2012, 3, 1301–1306. [Google Scholar] [CrossRef] [PubMed]
  53. Nishikawa, Y.; Kanai, M.; Narahara, M.; Tamon, A.; Brown, J.B.; Taneishi, K.; Nakatsui, M.; Okamoto, K.; Uneno, Y.; Yamaguchi, D.; et al. Association between UGT1A1*28*28 genotype and lung cancer in the Japanese population. Int. J. Clin. Oncol. 2017, 22, 269–273. [Google Scholar] [CrossRef]
  54. Wassenaar, C.A.; Conti, D.V.; Das, S.; Chen, P.; Cook, E.H.; Ratain, M.J.; Benowitz, N.L.; Tyndale, R.F. UGT1A and UGT2B genetic variation alters nicotine and nitrosamine glucuronidation in european and african american smokers. Cancer Epidemiol. Biomark. Prev. 2015, 24, 94–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Girard, H.; Butler, L.M.; Villeneuve, L.; Millikan, R.C.; Sinha, R.; Sandler, R.S.; Guillemette, C. UGT1A1 and UGT1A9 functional variants, meat intake, and colon cancer, among Caucasians and African-Americans. Mutat. Res. 2008, 644, 56–63. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Lacko, M.; Roelofs, H.M.; Te Morsche, R.H.; Voogd, A.C.; Ophuis, M.B.; Peters, W.H.; Manni, J.J. Genetic polymorphism in the conjugating enzyme UGT1A1 and the risk of head and neck cancer. Int. J. Cancer 2010, 127, 2815–2821. [Google Scholar] [CrossRef]
  57. Davies, A.M.; Mack, P.C.; Lara, P.N.; Lau, D.H.; Danenberg, K.; Gumerlock, P.H.; Gandara, D.R. Predictive molecular markers: Has the time come for routine use in lung cancer? J. Natl. Compr. Cancer Netw. 2004, 2, 125–131. [Google Scholar] [CrossRef]
  58. Ikeda, S.; Takabe, K.; Inagaki, M.; Suzuki, K. Expression of phosphorylated Akt in patients with small cell carcinoma of the lung indicates good prognosis. Pathol. Int. 2010, 60, 714–719. [Google Scholar] [CrossRef]
  59. Huang, Y.; Su, J.; Huang, X.; Lu, D.; Xie, Z.; Yang, S.; Guo, W.; Lv, Z.; Wu, H.; Zhang, X. Detection of UGT1A1*28 Polymorphism Using Fragment Analysis. Zhongguo Fei Ai Za Zhi 2017, 20, 817–821. [Google Scholar] [CrossRef]
Table 1. Demographic and clinical characteristics of the investigated groups.
Table 1. Demographic and clinical characteristics of the investigated groups.
CharacteristicsCase (n = 80)Control (n = 196)p-Value
Sex
 Male56 (70.0%)62 (31.6%)<0.001 a,*
 Female24 (30.0%)134 (68.4%)
Age (years)
 Median (p25–p75%)62.0 (55.0–69.0)69.0 (64.0–76.0)<0.001 b,*
Smoking
 Never smoked13 (16.33%)99 (50.5%)<0.001 a,*
 Smoker67 (83.7%)97 (49.5%)
Ancestry
 European47.6 ± 18.045.2 ± 17.00.334 b
 Amerindian30.0 ± 14.130.6 ± 14.80.888 b
 African22.3 ± 12.424.2 ± 13.80.313 b
Histology
 Squamous Cell Carcinoma41 (51.3%)-NA
 Large Cell Carcinoma14 (17.5%)-
 Adenocarcinoma12 (15.0%)-
 Others13 (16.2%)-
NA, not applicable. a Chi-square Test; b Mann–Whitney Test; * p-value < 0.05.
Table 2. Risk for lung cancer and its histological types, associated with the UGT1A1 gene polymorphism allele.
Table 2. Risk for lung cancer and its histological types, associated with the UGT1A1 gene polymorphism allele.
GroupsUGT1A1 (rs8175347) Allelotypes
(TA)7(TA)5 or (TA)6OR (95% IC)p-Value a
Controls139 (35.5%)253 (64.5%)--
Lung Cancer60 (37.5%)100 (62.5%)1.24 (0.80–1.93)0.331
 Squamous Cell Carcinoma35 (42.7%)47 (57.3%)1.56 (0.89–2.73)0.116
 Large Cell Carcinoma5 (17.9%)23 (82.1%)0.43 (0.16–1.18)0.103
 Adenocarcinoma13 (54.1%)11 (45.9%)2.57 (1.07–6.20)0.035 *
 Others7 (26.9%)19 (73.1%)0.76 (0.30–1.95)0.573
OR, odds ratio; CI, confidence interval; a Logistic regression adjusted for sex, age, and smoking. * p-value < 0.05.
Table 3. Risk for lung cancer and its histological types, associated with the UGT1A1 gene polymorphism genotypes.
Table 3. Risk for lung cancer and its histological types, associated with the UGT1A1 gene polymorphism genotypes.
GroupsUGT1A1 (rs8175347) Genotypes
TA7/7TA6/6, TA6/7 or TA5/7OR (95% IC)p-Value a
Controls23 (11.7%)173 (88.3%)--
Lung Cancer13 (16.3%)67 (83.7%)2.04 (0.85–4.92)0.110
 Squamous Cell Carcinoma9 (22.0%)32 (78.0%)4.08 (1.32–12.61)0.015 *
 Large Cell Carcinoma1 (7.1%)13 (92.9%)0.77 (0.09–6.61)0.815
 Adenocarcinoma2 (16.7%)10 (83.3%)3.19 (0.53–19.05)0.204
 Others1 (7.7%)12 (92.3%)0.73 (0.08–6.40)0.780
OR, odds ratio; CI, confidence interval; a Logistic regression adjusted for sex, age, and smoking. * p-value < 0.05.
Table 4. Risk for lung cancer and its histological types, associated with degree of UGT1A1 enzymatic activities.
Table 4. Risk for lung cancer and its histological types, associated with degree of UGT1A1 enzymatic activities.
GroupsUGT1A1 (rs8175347) Genotypes by Degree of Enzymatic Activity
Low or ReducedNormalOR (95%IC)p-Value a
Controls116 (59.2%)80 (40.8%)--
Lung Cancer47 (58.8%)33 (41.2%)1.09 (0.59–1.99)0.788
 Squamous Cell Carcinoma26 (63.4%)15 (36.6%)1.23 (0.56–2.70)0.604
 Large Cell Carcinoma4 (28.6%)10 (71.4%)0.29 (0.08–0.99)0.058
 Adenocarcinoma11 (91.7%)1 (8.3%)8.41 (1.02–69.55)0.048 *
 Others6 (46.2%)7 (53.8%)0.69 (0.21–2.27)0.543
OR, odds ratio; CI, confidence interval; a Logistic regression adjusted for sex, age, and smoking. * p-value < 0.05.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Pereira, E.E.B.; Leitão, L.P.C.; Andrade, R.B.; Modesto, A.A.C.; Fernandes, B.M.; Burbano, R.M.R.; Assumpção, P.P.; Fernandes, M.R.; Guerreiro, J.F.; Santos, S.E.B.d.; et al. UGT1A1 Gene Polymorphism Contributes as a Risk Factor for Lung Cancer: A Pilot Study with Patients from the Amazon. Genes 2022, 13, 493. https://doi.org/10.3390/genes13030493

AMA Style

Pereira EEB, Leitão LPC, Andrade RB, Modesto AAC, Fernandes BM, Burbano RMR, Assumpção PP, Fernandes MR, Guerreiro JF, Santos SEBd, et al. UGT1A1 Gene Polymorphism Contributes as a Risk Factor for Lung Cancer: A Pilot Study with Patients from the Amazon. Genes. 2022; 13(3):493. https://doi.org/10.3390/genes13030493

Chicago/Turabian Style

Pereira, Esdras E. B., Luciana P. C. Leitão, Roberta B. Andrade, Antônio A. C. Modesto, Bruno M. Fernandes, Rommel M. R. Burbano, Paulo P. Assumpção, Marianne R. Fernandes, João F. Guerreiro, Sidney E. B. dos Santos, and et al. 2022. "UGT1A1 Gene Polymorphism Contributes as a Risk Factor for Lung Cancer: A Pilot Study with Patients from the Amazon" Genes 13, no. 3: 493. https://doi.org/10.3390/genes13030493

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop