Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
World Academy of Sciences Journal
Join Editorial Board Propose a Special Issue
Print ISSN: 2632-2900 Online ISSN: 2632-2919
Journal Cover
November-December 2025 Volume 7 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
November-December 2025 Volume 7 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Role of genetic polymorphisms within different genes and associated risks of cancer development (Review)

  • Authors:
    • Runjhun Mathur
    • Noopur Khare
    • Gaurav Saini
    • Sheo Prasad Shukla
    • Abhimanyu Kumar Jha
  • View Affiliations / Copyright

    Affiliations: Department of Environmental Science and Engineering, Dr. A.P.J Abdul Kalam Technical University, Lucknow, Uttar Pradesh 226031, India, Department of Biotechnology Engineering and Food Technology, Chandigarh University, Chandigarh, Punjab 140413, India, Department of Civil Engineering, Netaji Subhas University of Technology, Delhi 110078, India, Department of Civil Engineering, Rajkiya Engineering College, Atarra, Uttar Pradesh 210201, India, Department of Biotechnology and Bioengineering, School of Biosciences and Technology, Galgotias University, Greater Noida, Uttar Pradesh 203201, India
    Copyright: © Mathur et al. This is an open access article distributed under the terms of Creative Commons Attribution License [CC BY 4.0].
  • Article Number: 118
    |
    Published online on: October 13, 2025
       https://doi.org/10.3892/wasj.2025.406
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Cancer susceptibility varies widely among individuals due to complex interactions between genetic and environmental factors. Among genetic determinants, polymorphisms within key genes involved in carcinogen metabolism, DNA repair, cell cycle regulation, immune response and epigenetic modification significantly influence cancer risk. The present review comprehensively discusses the role of genetic polymorphisms across diverse gene families, including cytochrome P450 enzymes (notably CYP1A1 and CYP2E1), tumor suppressors (PTEN, TP53), DNA repair genes (XPD) and immune checkpoint regulators (PD‑L1), highlighting their contributions to cancer development. Additionally, the interplay between genetic variants and epigenetic mechanisms, such as DNA methylation is discussed, emphasizing their combined effects on gene expression modulation in carcinogenesis. Advances in genotyping technologies and genome‑wide association studies have expanded the understanding of these polymorphisms, providing insight into population‑specific susceptibilities and pharmacogenomic applications. The integration of genetic and epigenetic profiling holds promise for personalized cancer prevention and therapeutic strategies, tailored to individual genetic makeup and environmental exposures. The present review underscores the importance of continued research into genetic polymorphisms as biomarkers for cancer risk assessment, providing a foundation for targeted interventions and improved public health outcomes.

1. Introduction

The investigation of the genetic factors influencing cancer susceptibility has been increasing in recent years. The International Human Genome Sequencing Project and the International HapMap Project have both conducted substantial research into the location, amount, type and frequency of genetic variants in the human genome (1-4). Numerous observational studies examining the association among polymorphisms in genetic variations and the risk of cancer development have led to ongoing technology advancements that enable more rapid and more economical genotyping results (5). A recent genome-wide association study identified several novel susceptibility loci for prostate cancer risk, providing new insight into the genetic architecture of this disease across diverse populations (6). A large cohort study involving >113,000 women emphasized the contribution of both common and rare genetic variants to breast cancer susceptibility, demonstrating the complex genetic architecture underlying breast cancer risk (7).

Knowledge of the genetic propensity to cancer has generally improved with the increasing amount of research that has been performed. The lack of replication has been a major critique of genetic epidemiology. There have been many ‘false positive’ claims, as evidenced by the failure of numerous research that attempted to reproduce a statistically significant outcome for a genetic condition that had already been published (8,9). Meta- and pooled analyses have been employed to incorporate both statistically meaningful and non-significant data from several studies and grade these results according to their precision. Another significant methodological issue is the scale of these studies of genetic associations (a proportion of sample size) (10-12).

The programmed death-ligand 1 (PD-L1), encoded by the CD274 gene located on chromosome 9p24.1, plays a critical role in immune regulation by mediating immune checkpoint pathways that allow tumors to evade immune surveillance. PD-L1 expression is detected in various cells, including antigen-presenting cells, lymphocytes and epithelial cells, where its regulated activity helps maintain immune homeostasis during inflammation (13,14). Toll-like receptors (TLRs), expressed in immune cells, have been shown to modulate PD-L1 expression by responding to pathogen-associated molecular patterns, thus influencing the immune microenvironment.

Notably, the aberrant expression of PD-L1 has been implicated in the pathogenesis of several types of cancer. The overexpression of PD-L1 in tumors, such as non-small cell lung cancer is associated with oncogenic mutations, such as those in the epidermal growth factor receptor (EGFR), highlighting the interplay between oncogenic signaling pathways and immune evasion mechanisms. Genetic and epigenetic alterations, including polymorphisms and mutations in PD-L1 regulatory regions or associated signaling genes, such as phosphatase and tensin homolog (PTEN) and anaplastic lymphoma kinase (ALK), may contribute to a variable PD-L1 expression and consequent differences in tumor immune escape and patient prognosis (15).

Despite the recognized importance of PD-L1, comprehensive studies focusing on genetic polymorphisms within the CD274 gene and their associations with cancer susceptibility remain limited. Integrating the mechanistic understanding of PD-L1 regulation with genetic variation analyses could provide valuable insight into individual susceptibility and response to immune checkpoint therapies.

TLRs, a subtype of non-catalytic receptor that is widely expressed in APCs and activated by epitope molecular patterns, have a notable impact on PD-L1 expression (16). The driving oncogenic events in carcinogenesis may be the cause of PD-L1 overexpression. For instance, in lung cancer, PD-L1 expression is positively associated with EGFR mutations in the EGFR (17). Through the unregulated stimulation of the protein kinase pathway, PD-L1 overexpression is maintained in enzymatic activity and in PTEN-mutant tumors (18,19). Through constitutive STAT3 activation, the recombinant genes for the protein nucleophosmin (NPM) and ALK enhance the expression of PD-L1 in T-cell lymphoma (20). Lin et al (21) explored the genomic and transcriptomic characteristics regulating PD-L1 expression, highlighting the impact of genetic polymorphisms on PD-L1 and its role across various types of cancer.

Cytochrome P450 (CYP450) enzymes, which include those from the CYP1, CYP2 and CYP3 families, as well as additional exogenous and endogenous compounds, metabolize drugs. The key CYP enzymes involved in the metabolic activation of procarcinogens are CYP1A1, CYP1A2, CYP1B1, CYP2E1, CYP3A4 and CYP3A5(22).

The majority of CYPP450 enzymes that belong to the CYP 1, 2, or 3 groups are polymorphic due to gene deletion, single-nucleotide polymorphisms that occur all alone in combination, or gene duplications, where mutant alleles result in discontinued, reduced, modified, or enhanced enzyme activity. Genetic polymorphisms in CYP enzymes, such as CYP2D6 and CYP2E1 significantly affect enzyme activity and have been linked to altered cancer susceptibility across diverse populations (23,24). Phenotyping experiments were utilized to investigate the potential link between CYP polymorphisms and the risk of developing cancer before genotyping methods were developed. The delivery of CYP enzyme substrate and the analysis of the products in plasma and urine were common procedures used in research. Following the analysis of the associations among CYP polymorphisms and the risk of cancer development over the period of a decade in numerous studies encompassing thousands of patients, the association between CYP polymorphisms and cancer susceptibility is now understood; the mechanisms involved include simultaneous genes that code for phase 1 and phase 2 enzymes (25). Recent studies have shed light on CYP polymorphisms influencing the risk of cancer development and treatment outcomes. Tan et al (26) reported on the impact of CYP2D6 genotypes on breast cancer outcomes and pharmacogenomics, emphasizing the clinical relevance of CYP2D6 polymorphisms. Additionally, Jiang et al (27) conducted an updated meta-analysis linking CYP2E1 polymorphisms with an increased risk of developing various types of cancer, supporting the role of CYP2E1 genetic variations in cancer susceptibility.

Numerous biological mechanisms, including bone metabolism, inborn resistance, proliferation and differentiation are regulated by the vitamin-D endocrine system (28). Numerous common diseases, such as cancers, diabetes, cardiac disease, autoimmune disorders, rickets, as well as other bone diseases, have been firmly related to vitamin D deficiency by epidemiological and laboratory research (29-32). The biologically highly active naturally occurring metabolite of vitamin D, known as 1,25-dihydroxyvitamin D3 (1,25(OH)2D3, calcitriol), has been found to control the proliferation and differentiation of a range of cell types, including cancer cells (33-35). Research has also demonstrated that angiogenesis, cancer growth and cell death are all regulated in cancer (36-38). Previous research has revealed the existence of intracellular enzyme 25(OH) D3-1-hydroxylase (CYP27B1) activity in a wide range of cell types, including macrophages, keratinocytes, prostate and colon cancer cells (39,40). It has been shown that a number of tissues locally synthesize 1, 25(OH) 2D3. Recent research has uncovered that vitamin D can be activated via an alternative pathway by the steroidogenic enzyme CYP11A1. This pathway complements the classical activation involving CYP27B1 and expands the understanding of vitamin D metabolism beyond traditional mechanisms. The CYP11A1-mediated conversion produces novel secosteroids with potential biological activities relevant to cellular proliferation, differentiation, and immune modulation in cancer and other diseases. This alternative pathway adds complexity to vitamin D regulation and may have implications for cancer susceptibility and therapy (41,42).

The nucleotide excision repair (NER) system is one of the main mechanisms by which cells defend themselves from genotoxic damage, such as that caused by UV radiation and exposure to chemical carcinogens. Human syndromes, such as trichotillomania, Cockayne syndrome and xeroderma pigmentosum (XP) are caused by NER system anomalies (43,44). An exceptionally high sensitivity to UV light and a comparatively high chance of developing skin cancer are two features of the heritable human condition known as XP. The UV sensitivity of patients with XP led to the initial connection between the illness and DNA repair (45). Based on the ability of distinct cell types to complement UV sensitivity, eight XP-complementation groups were identified (46,47), and the genes encoding the various complementation groups have also been identified (48-50). UV radiation plays a dual role in human homeostasis, with both harmful and beneficial effects that extend beyond simple DNA damage. Beyond its capacity to induce genotoxic lesions repaired by the nucleotide excision repair system, UV radiation also plays a central homeostatic role in neuro-immuno-endocrine regulation. Recent insights highlight how UV exposure modulates systemic physiology by affecting neural, immune and endocrine pathways, thus contributing positively to body regulation and immune surveillance. This dual nature of UV radiation underscores the complexity of its impact on health and carcinogenesis, balancing its carcinogenic potential against vital homeostatic functions (51).

In addition to genetic polymorphisms, epigenetic modifications, such as DNA methylation play a crucial role in regulating gene expression and influencing cancer susceptibility. DNA methylation involves the addition of a methyl group to cytosine residues, typically at CpG dinucleotides, leading to changes in chromatin structure and gene activity without altering the DNA sequence itself. Aberrant DNA methylation patterns can result in gene silencing or activation that contributes to carcinogenesis, affecting tumor suppressor genes, oncogenes and DNA repair genes. The interplay between genetic polymorphisms and epigenetic alterations is increasingly recognized as fundamental to understanding the risk of cancer development, progression and the therapeutic response. Therefore, integrating both genetic and epigenetic perspectives provides more comprehensive insight into cancer susceptibility and personalized medicine approaches.

Taken together, the diversity of biological systems, such as immune surveillance (antigen-presenting cells and checkpoint proteins), metabolic detoxification (cytochrome P450 enzymes), cellular signaling (vitamin D pathways), DNA repair (nucleotide excision repair system), and epigenetic regulation (DNA methylation) forms an intricate network maintaining cell homeostasis and genomic integrity. Genetic polymorphisms within the genes governing these systems can significantly alter their functions, leading to variability in how individuals respond to environmental exposures, carcinogens and intrinsic cellular stresses. These variations may influence cancer susceptibility by affecting immune evasion, the activation or detoxification of carcinogens, DNA damage repair efficiency, and the epigenetic regulation of oncogenes and tumor suppressors. Thus, understanding the complex interplay among these elements is essential for elucidating the multifactorial nature of cancer risk and identifying genetic and epigenetic markers for cancer prevention, diagnosis and personalized therapy.

2. Various genes and genetic polymorphisms

The following chapter provides an overview of genes involved in key cellular pathways; it is important to emphasize that the relevance of these genes to the risk of cancer development is principally determined by the specific genetic polymorphisms they harbor. A detailed understanding of the normal function of each gene provides the necessary foundation to appreciate how alterations in the genetic sequence, such as single nucleotide polymorphisms (SNPs), insertions, deletions and copy number variations, can influence an individual's susceptibility to cancer. Throughout this section, gene variants with established or potential links to cancer risk will be highlighted. It should be noted, however, that some gene polymorphisms discussed in the subsequent sections affect human health through mechanisms that may extend beyond cancer alone; these examples are included to underscore both the complexity and the far-reaching consequences of genetic variation in human populations. This integrative approach aims to provide a nuanced perspective, facilitating a comprehensive understanding of the multifactorial risk factors contributing to cancer and related diseases.

Within the broad family of CYP enzymes, several members such as CYP1A1, CYP1A2, CYP1B1, CYP2E1, CYP2D6, CYP3A4 and CYP3A5 participate in the metabolism of carcinogens and drugs. Among these, CYP1A1 and CYP2E1 have emerged as particularly critical, due to their significant roles in activating procarcinogens commonly found in tobacco smoke and other environmental toxins. Genetic polymorphisms in these enzymes influence enzymatic activity and expression levels, thereby modulating individual susceptibility to various cancers. While all CYP enzymes contribute to xenobiotic metabolism, the extent of evidence linking CYP1A1 and CYP2E1 variations to the risk of cancer development is more substantial, warranting focused research attention.

CYP1A1

On human chromosome no. 15, the CYP1A1 gene is only activated in organs other than the liver, such as the lungs. The enzyme CYP1A1 converts polycyclic aromatic compounds, which are present in cigarette smoke, into hazardous arene oxide that can result in DNA mutation and cancer (47,48). Since CYP1A1 is not expressed in the human liver, it is dubious whether human studies on animals have any application (49). There are several polymorphic CYP1A1 alleles known. A point mutation causes an MspI restriction fragment-length polymorphism in the 3' non-coding region (RFLP). The heme-binding domain of CYP1A1 contains the exon 7 polymorphism, which increases the inducibility of the enzyme. Recent research suggests that the MspI, as well as exon 7 mutation may increase the risk of developing lung cancer, although Asians are more likely to be affected. In the 3' non-coding region of CYP1A1, a second MspI RFLP is only present in individuals of African origin. Its connection to the risk of developing lung cancer remains unclear. According to recent studies, nicotine can boost pulmonary CYP1A1 activity (50). This research has important implications for how sensitive humans are to acquiring cancer, even if it is currently only applicable to rats. First, nicotine most definitely contributes to the induction of CYP1A1 by cigarette smoke. Second, the emergence of smoking cessation aids raises serious concerns about the possibility for nicotine replacement medications to increase CYP1A1 activity and hence trigger the bio-activation of carcinogens (51). Last but not least, it remains unknown how CYP1A1 functions in the presence of nicotine and an allele variation.

CYP2D6

CYP2D6 (debrisoquine hydroxylase) is considered to be involved in the metabolism of 25% of all prescribed medications (52). At least 29 allelic variations of CYP2D6 have been identified, and this has been linked to significant inter-individual variability in drug metabolism (53).

In ~6% of Caucasians, deletions, abnormal splicing and gene duplication result in the absence of functional CYP2D6. It is interesting that CYP2D6 cannot be induced. It is understood that CYP2D6 activates metabolism (54).

Smoke from cigarettes contains 4-(methylnitrosamine)-1- (3-pyridyl)-1-butanone, which is considered to cause cancer and is a factor in the development of human lung adenomas. Extensive metabolizers may be more susceptible to lung cancer (54).

E-Cadherin

The CDH1 gene, that is found on chromosome 16q22.1, is responsible for producing the 120 kD single transmembrane glycoprotein known as E-cadherin. One intracellular and five external domains are present. It converses with catenins. The development and maintenance of tissue architecture, cell polarity, intracellular signaling and intercellular adhesion depend on this protein. It plays a critical role in the formation of sticky junctions in epithelial cells. The lack of E-cadherin directly affects essential cellular processes including motility. Additionally, it has been demonstrated that its expression decreases typically happen during tissue metastasis. An association has been found between an increased aggressive behavior and the decreased expression of E-cadherin. The frequency of OSCC E-cadherin gene hypermethylation varies between 7 and 46% (55).

PTEN

The tumor-suppressor gene, PTEN, is located on chromosome 10q23.3. It is anticipated that essential cellular functions such as survivability, differentiating, proliferating, apoptosis and invasion are affected by the lack of expression Ras/phosphoinositide 3-kinase (PI3K)/Akt, which lack control over the signal transduction that control apoptosis and migration, and also play a crucial role in the survival, proliferation and metastasis of tumor cells. Due to mutations or epigenetic modifications, PTEN is frequently lacking in a variety of cancer types. Furthermore, it has been demonstrated that endometrial cancer, gastric cancer, non-small cell lung carcinoma and cervical cancer all exhibit the methylation of the PTEN promoter CpG islands (56). The functions, regulation and implications of PTEN polymorphisms in cancer were comprehensively reviewed by Song et al (57), detailing the role of PTEN as a key tumor suppressor. Han et al (58) further performed a meta-analysis of PTEN mutations, associating genetic alterations with prognosis across multiple cancer types.

p53

Cell cycle progression, cellular differentiation, DNA repair and apoptosis are some of the key cell activities that the TP53 gene, also known as p53, is involved in. It is located on chromosome 17p13.1. p53 levels increase in response to endogenous or exogenous stress, which stops the cell cycle and enables DNA repair. Genomic instability results from p53 loss of function, which affects how cells react to stress or damage. With a frequency ranging from 25 to 69%, p53 is mutated in the majority of human malignancies, including oral tumors (59,60). In addition to this, p53 frequently exhibits a decrease of function brought on by epigenetic rather than genetic processes. Comprehensive analyses of TP53 variations across multiple human cancers were provided by Bouaoun et al (61), utilizing database and genomic data to reveal new insight into TP53 mutation patterns. A further mechanistic understanding of mutant p53 roles in cancer pathogenesis was provided by Mantovani et al (62), elucidating its function as a cancer cell guardian.

XPD

TFIIH phosphorylates a wide range of substrates, including nuclear hormone receptors such as RAR or ER, transcription activators and RNA polymerase II (63). Additionally, TFIIH is present as a nine-subunit complex, a transcriptionally active core TFIIH, and a CDK-activating kinase (CAK) complex (Fig. 1). The XPD protein is a component of all three of these complexes (64). The activity of these complexes can be decreased by mutations in the XPD gene, which can cause issues with transcription, the apoptotic response, repair, or, most likely, hormonal function. All of these deficiencies result in syndromes that are associated with immature sexual development, skeletal abnormalities, mental impairment, and, in the case of the majority of patients with XPD mutations, a high propensity for cancer. Only a limited amount of research has been performed into the connection between XPD mutations and DNA repair capacity, as determined by the biological tests mentioned in the literature. The Lys/Lys codon 751 XPD genotype was previously linked to a decreased repair of X-ray-induced DNA damage in a brief study on 31 women at risk of developing breast cancer (65). More chromatid aberrations are present in individuals with the wildtype Lys allele than in those carrying one or more Gln alleles (65).

Diagram illustrating the associations
between the XPD protein and potential clinical diseases caused by
XPD gene mutations.

Figure 1

Diagram illustrating the associations between the XPD protein and potential clinical diseases caused by XPD gene mutations.

3. Methods for analyzing genetic polymorphisms

Association studies are most frequently used to determine whether polymorphisms contribute to genetic susceptibility or progression. As a result, the focus is on variables that determine the effectiveness of associations. If a specific allele exhibits a greater frequency in cases compared to controls, that polymorphism is then considered to be linked to the disease (66). Researchers compare individuals with extreme phenotypes when analyzing polymorphisms as contributing factors to the course of disease, rather than diseased individuals with unaffected controls. There are three possibilities if a substantial association is found: Either the polymorphism is at the locus of interest, it is in genetic linkage [namely linkage disequilibrium (LD)] with the locus, or confounding factors are involved.

When alleles from two different genetic loci co-occur more frequently than would be predicted according to their respective allelic frequencies, a population is considered to exhibit LD. Possible sources of LD include early mutation, founder effects and selection. Population admixture, which occurs when groups that have been separated for a long time unite to form a hybrid population, is another possible source of LD (67). The resulting LD can be prolonged beyond distances typically observed in populations with greater stability, depending on the sort of mixing. The most precise estimates of LD in an outbred population indicate that LD is unlikely to span distances >1-2 centimorgans (cM), or ~1-2 million base pairs, whereas LD may occur across 10-fold that distance in an inbred group, such as the Hutterites (68).

Confounding variables need to be taken into account, particularly when polymorphisms reported in one study are absent in another ethnic group. One perplexing aspect is population stratification. This could be brought on by unequal ethnic admixture, such as the presence of Caucasians in the gene pool of an African-American community. Fortunately, this issue can be resolved by taking great precautions during the analysis or research plan phases. Association studies based on families are specifically created to take into account the genetic background that may add confounding variables, and they include genotyping of affected people, their parents, and/or their unaffected siblings (69). These types of investigations frequently employ the transmissions disparity test (TDT), as well as the haplo-type relative risk test statistics (HRR). The TDT compares how frequently each allele is transferred from a heterozygous parent to a child who has the disease. Similar to the TDT, the HRR analyses genetic transmission (haplotype) as compared to allele transmission (70).

The decision of which phenotype to explore is crucial in genetic polymorphism investigations. In fact, the presence of numerous phenotypes in the case sample has hampered much research that analyzing genetic polymorphisms. For instance, research on asthma has used patient samples from patients with mild, severe, adult-onset, intrinsic and extrinsic asthma (71). Studying a more precisely defined intermediate phenotype can significantly enhance studies that aim to ascertain whether there is a link between a polymorphism and disease. Total IgE and bronchial hyper responsiveness are examples of intermediate phenotypes in the case of asthma. Another factor to take into account is the possibility that the genes involved in illness progression may not be the same genes responsible for disease susceptibility. It may be helpful to restrict the sample to those with a particular stage or severity of disease. In a previous study, to evaluate potential genes for the disease, the researchers focused on patients with severe early-onset chronic obstructive pulmonary disease (71).

The evaluation of DNA polymorphisms close to or within putative genes is the only application of association studies. Linkage analysis employing families or affected siblings is necessary to carry out a genome screen to look for candidate genes. Although linkage analysis is rigorous and uncovers genes that significantly affect disease susceptibility, it has very limited power and will miss genes that merely increase the risk of mild to moderate disease. For instance, hundreds to thousands of families would need to be typed if a disease susceptibility allele increased disease risk by 2-fold relative to the wild-type allele, which may not be a realistic sample size (72). Although linkage analysis detects connections over considerably larger genomic regions (thousands of base pairs), association studies have a stronger power (millions of base pairs). Including current technology, a genome scan with association studies would need tens of thousands of markers. The majority of lung diseases need some type of environmental trigger before they may become visible. Given that genetic susceptibility only accounts for a small part of illness variation, failure to consider environmental factors can drastically degrade gene-finding research for the majority of complex disorders. Furthermore, genome screening or association studies carried out on populations that were neither chosen nor stratified according to their environmental exposure could only be able to find genes whose environmental exposure is common in that group. For instance, investigating a randomly chosen sample of asthmatics from the Midwest region of the USA would be able to discover genes crucial for regulating the response to house dust mites, although possibly not with absolute certainty (73).

Gene-environment interactions can take on many different shapes, such as distinct exposure risk implications based on the genotype of an individual or diverse gene risk implications based on the exposure of an individual. Biological and statistical interactions are the two main interactions. The coefficient of the product term of the genetic and environmental risk variables represents a statistical risk factor interaction, and the interaction is quantified as a divergence from a multiplicative model (gene and environment). This approach is arbitrary, relies on models, and may overlook biological synergy or interaction. The biologic interaction paradigm states that when two factors cause a disease to start, they interact. Occasionally, this co-participation could stand out as a divergence from an additive model (74).

4. SNPs in the human population

SNPs are single nucleotide variations in the genomic DNA that occur at different positions in different individuals in a community (75). Genome-wide datasets are more frequently used in the drug development process, as well as to identify molecular pathways and networks underlying complex disorders (Table I) (76-79). In particular, functional pathway analysis of genomic data provides the possibility of greater capability for discovery and organic links to biological phenomena. SNPs are the consequence of single base-pair variations (substitutions or deletions) caused by point mutations in chromosome sequences, and account for a large portion of the genetic variation found in the human genome. Finding SNPs in a genome can be achieved in a variety of laboratories and via computational mechanisms; however, they all involve comparing the same DNA segment from various individuals or haplotypes.

Table I

Methods of selecting SNPs.

Table I

Methods of selecting SNPs.

MethodsAdvantagesLimitations(Refs.)
Pathway gene methodIt is simple to study a subset of SNP based on a description of the pathways relating to the drug's pharmacokinetics and mechanism of action. Clinically, general vulnerability is observed in complex genetic disorders.The pathway gene approach will result in fewer false-positive findings than the genome wide approach because of the disadvantage of multiple testing.(76,77)
Candidate gene methodFunctional SNPs are those whose genetic variant affects how a protein functions. This technique has resulted in the identification of a sizable number of pertinent SNPs in pharmacogenetics.As the majority of complex traits are not considered to be monogenetic, selecting SNPs using this strategy will frequently result in a limited explanation of variation in medication response.(78,79)
Genome-wide methodThis strategy may identify unexpected SNPs linked to drug response. New associations between SNPs and medication response have been found in genome-wide association investigations, and complicated features can be studied, while taking into account polygenetic variation.In identifying a related SNP, the discrepancy between type I errors (false positive findings) and eventually type II faults (false negative results) may create some issues.(78,79)

[i] SNP, single nucleotide polymorphism.

SNPs can be located using expressive sequence tags, which are created by single-run sequencing of cDNAs obtained from various individuals and the assembly of overlapping sequences for the same region. This allows for the discovery of novel SNPs. Depending on whether they are located in regions of the genome that regulate genes, non-coding SNPs can be categorized. A number of complex disorders may be caused by quantitative discrepancies in gene products rather than qualitative differences. Based on whether they alter the amino acid sequences of the protein that the altered gene encodes, coding SNPs can be categorized. By their impact on protein structure, modifications that change protein sequences can be categorized (80).

Genome-wide association studies have emerged as a crucial method for identifying genes that predispose to complicated disorders. With the aid of population-based information like as allele frequency, LD and recombination rates, researchers may perform genome-wide association analysis on millions of SNP markers. Some of the discrepancies in association results between populations for particular traits of interest can be explained by HapMap data, such as population-specific common variants and LD blocks (81).

5. VNTR polymorphisms in the human population

CYP2E1 is a key enzyme found in the microsomal ethanol oxidation system. It belongs to the CYP superfamily and is primarily located in the membranes of the endoplasmic reticulum. CYP2E1 plays a crucial role in the metabolism of various hydrophobic toxic compounds (82-84). Additionally, it contributes to the conversion of certain pro-carcinogens and drugs into highly reactive metabolites.

The activation of N-nitrosamines, which are present in tobacco smoke, foodstuffs, and certain industrial and endogenous carcinogens, is facilitated by CYP2E1. Furthermore, this enzyme can generate highly reactive compounds, such as superoxide anion radical (O2-), singlet oxygen (O2), hydrogen peroxide (H2O2) and hydroxyl radical (OH·) by reducing molecular oxygen. These reactive species are capable of causing DNA damage and promoting carcinogenesis (85-87). The human CYP2E1 gene is situated on chromosome 10q26.3 and consists of 9 exons and 8 introns. The expression of the CYP2E1 gene can be regulated at multiple levels, including transcription, translation, mRNA stability and protein degradation. As with other CYP genes, CYP2E1 exhibits various polymorphic sites in its 5'-flanking region, introns, and transcribed gene regions. One notable polymorphism is a variable number tandem repeat (VNTR) sequence located ~2.0 kilo base pairs upstream of the transcription start site (88-90).

The activity of the CYP2E1 enzyme exhibits significant variability among different ethnic groups, influenced by both environmental and genetic factors, such as polymorphisms. Polymorphic CYP genes can lead to differences in the ability to metabolize, detoxify, or activate various substances. Several studies have indicated that certain polymorphic genes, in conjunction with alcohol consumption, play a role in the development of specific types of cancer. Although alcohol itself is not a carcinogen, it can function as a co-carcinogen by amplifying the effects of other chemicals that are activated by enzymes, such as CYP2E1. Research has demonstrated that CYP2E1 is highly expressed in the liver and pancreas following ethanol ingestion. The production of acetaldehyde during ethanol oxidation may directly cause cell damage through the generation of reactive oxygen species (91-95).

Furthermore, it is widely known that certain tumors have a hereditary component, and environmental factors associated with specific habits can increase the risk of tumor occurrence.

6. Using a different approach to identify human gene polymorphisms

The function of biotransformation enzymes with reference to occupational health exposure is to provide effective purification of endogenous or exogenous substances by particular biochemical pathways. These turn harmful molecules into inactive compounds, which are then eliminated in urine, preventing the buildup of metabolites and injury to the human body (96). Although the scanning of specific gene polymorphisms by molecular biology laboratories is the optimal approach to determine each the susceptibility of each study participant, the willingness of study participants to provide the biosample is essential to move forward with the genetic analysis. There are some issues, such as the unwillingness of study participants to consent to venipuncture or, more generally, to the collection of biopsies, either as they are simply unaccustomed to the procedure as they consider it to be an invasive and painful technique, or because they are afraid of the possible outcomes of the analysis (97). However, the evaluation of gene polymorphisms does not provide any diagnostic information regarding the propensity of an individual to develop a specific disease. In comparison to collecting urine samples, collecting blood samples may be more difficult. The lack of language proficiency, the difficulties in communicating, and the study participants varied cultures, customs, dietary preferences, and religious beliefs could all play a role in this. To combat this critical issue and obtain ethnic-specific genotype information without using laboratory analysis, a publicly available online library (http://grch37.ensembl.org/Homosapiens/Variation) appears to contain a tentative catalogue of the majority of genotype and allele frequencies of several ethnic groups. It is possible to collect the genetic profiles of many ethnic groups using this resource, which aids in the prediction and identification of population-specific susceptibilities in silico study. This model was created to assess the risk level of the homozygous variation and heterozygous genotype in relation to the world population in four macro-groups that includes Africans, Eastern Asians, South Asians and Europeans. The principal component analysis statistical technique serves as the foundation of the model (98). It is intended to identify the crucial vulnerabilities in the polymorphisms of genes linked to three main functional biochemical processes, including detoxification, oxidative stress and DNA repair, following exposure to the harmful compounds. The SNPs were selected based on their exposure to harmful and cancer-causing chemicals that are frequently present in manufacturing plants and shipyards (99).

7. Benefits of including polymorphisms in studies on health-related effects

Incorporating polymorphisms opens up a wide range of intriguing options for investigating the health effects of exposure to toxins and toxicants in the environment. It is possible to identify various risk ranges in subgroups of individuals who were exposed by stratifying a health result or biomarker in accordance with the relevant genotype (or phenotype) (100), as illustrated in Fig. 2. An implication that corresponds to the average risk for both fast and slow acetylators is shown by research evaluating the probability of bladder cancer linked to exposure to aromatic amines (100). This estimate does not imply that aromatic amines are as significant etiological factors for sub-populations or as powerful carcinogens as a stratified study might. For routine exposures, to food ingredients or carbon emission, for example, whose relation to a disease consequence is normally small, effect dilution may be especially crucial. Second, proof of impact change by genotype provides insight into the fundamental biologic mechanism of cytotoxicity or carcinogenicity when substrates or targets of possible genetic variants are identified as likely causal agents (101). Lipopolysaccharide (LPS), a constituent of particulates in rural regions and sometimes known as an endotoxin, may have a negative impact on lung function metrics. Kelada et al (101) demonstrated that the response to LPS varied by TLR4 genotype. The TLR4, which is encoded by TLR4, binds LPS and begins a signaling pathway that causes lung inflammation. According to their findings, although individuals with the mutated TLR4 genotype may be more susceptible to an inflammatory process, they may not be as susceptible to an inflammation of the lungs brought on by LPS. These results may contribute to answering the difficult question of whether particulates matter component(s) is/are accountable for the multitude of documented health consequences, particularly in rural areas where LPS levels are substantial. The development of drugs or dietary interventions that postpone the start or progression of disease may be made possible by the increased understanding of pathologic pathways gained via combined epidemiological and toxicological investigations. Oltipraz [OPZ; 5-(2-pyrazinyl)-4-methyl-1, 2-dithiole-3-thione] is an example of a medication that activates phase II XMEs, specifically the GSTs (102). Aflatoxin B1 can cause liver cancer in rats, according to early research. Research has also revealed that administering OPZ to patients significantly improved the clearance of a phase II substance known as aflatoxin-mercapturic acids (103). Research suggests that OPZ may function by competitively inhibiting CYP1A2, preventing the activation of aflatoxin. Ultimately, hypothesis-based epidemiological research and the knowledge of aflatoxin biotransformation routes from investigations on human tissue grown in vitro and animal models has helped to establish a chemoprevention method for aflatoxin-induced hepatocellular carcinoma. Studies on the consequences of exposure to controlled environmental pollutants that take genetic sensitivities into consideration will increase our understanding of the range of human genetic variation in response to these pollutants. Genes that may be associated with susceptibility should be included in studies designed to examine the effects of these substances. By replacing the standard default assumptions (i.e., uncertainty factor of 10) with more accurate estimates of human variability, the risk assessment may be improved. As a result, acceptable exposure levels may be redefined, improving overall public health protection and industry regulation. Although this benefit has been promoted for some time, there is still no concrete illustration of how it may be achieved, particularly in light of the myriad social, legal, or ethical issues that surround use of genetic data (104). Preventive efforts on those who are genetically sensitive to disease has started in the environmental health field, with a focus on the intrinsically complicated ethical, legal and social issues (105).

Schematic depiction of (A) various
risk factors associated with genetic polymorphism-induced cancer,
and (B) various health promotion approaches for the management of
genetic polymorphism-induced cancer.

Figure 2

Schematic depiction of (A) various risk factors associated with genetic polymorphism-induced cancer, and (B) various health promotion approaches for the management of genetic polymorphism-induced cancer.

8. Synopsis

Polymorphisms and DNA methylation both play critical roles in the development of cancer. Polymorphisms are DNA sequence variations that occur when a single nucleotide in the DNA sequence is altered. These variations can lead to changes in the function of the gene or the expression of the gene product, which can result in cancerous changes to cells. Epigenetic changes, such as DNA methylation are biochemical processes wherein enzymes add a methyl group to the DNA strand at 5th position of the cytosine to be precise. These methylation sites can control gene expression by either silencing a gene or increasing its activity depending upon the number of methylation sites present. DNA methylation can also alter splicing patterns, downregulate or ‘turn off’ gene expression, which can lead to cancerous changes in cells. Overall, both polymorphisms and DNA methylation function as key regulators of gene expression and can therefore exert significant effects on the development of cancer (106). However, they do differ in their mechanisms of action.

The role of genetic polymorphisms within different genes in the development and risk of cancer development is a complex and multifactorial process. While genetic polymorphisms can contribute to the susceptibility of an individual to cancer, they are not the sole determinant. Environmental factors, lifestyle choices and other non-genetic factors also play a crucial role. Numerous studies have identified specific genetic polymorphisms that are associated with an increased risk of developing cancer. For example, variations in the BRCA1 and BRCA2 genes have been linked to an increased risk of developing breast and ovarian cancer, while variations in the TP53 gene have been linked to an increased risk of developing several types of cancer, including breast, ovarian and colorectal cancer (107,108). However, it is important to note that the presence of a genetic polymorphism does not necessarily mean that an individual will develop cancer. A number of individuals with these genetic variations never develop cancer, and numerous individuals without these genetic variations do develop cancer.

Emerging evidence demonstrates that tumors possess the capacity not only to modulate their local microenvironment, but also to dysregulate systemic body homeostasis through neuroendocrine pathways. This tumor-driven autoregulation interferes with normal physiological processes, such as metabolism, immune function and stress response, effectively hijacking the neuroendocrine system of the body to support cancer progression and evade host defenses. Understanding this intricate interplay reveals cancer as a systemic disease with extensive body-wide impacts, highlighting potential therapeutic targets to restore homeostatic balance and counter tumor-driven systemic disruption (109).

The study of genetic polymorphisms and cancer risk is an active area of research, and new discoveries are continuously being made. As the understanding of the genetic basis of cancer increases, it is likely that the identification of novel genetic variations that are associated with an increased risk of cancer development will be achieved, and the development of novel strategies for the prevention and treatment of cancer may be possible.

9. Limitations and future challenges

While the present review comprehensively covers the role of genetic polymorphisms across various genes associated with susceptibility to cancer, several limitations should be acknowledged. First, the heterogeneity of study designs, sample sizes and populations in the original research contributes to variability and potential inconsistency in reported associations. A number of genetic association studies face challenges, such as population stratification, limited replication and publication bias, which affect the strength and generalizability of conclusions. Second, the complex interplay between genetic polymorphisms, epigenetic modifications and environmental factors remains incompletely understood, limiting the ability to fully elucidate causal mechanisms. Third, the functional characterization of numerous polymorphisms is still lacking, impeding translation to clinical applications. Finally, rapidly evolving genomic technologies and the emergence of multi-omics approaches necessitate continuous updates to maintain a current and holistic perspective on genetic risks in cancer. Future studies incorporating large, well-characterized cohorts with integrated genomic, epigenetic, and environmental data are essential to overcome these limitations and advance personalized cancer prevention and therapy.

10 Conclusion

In conclusion, DNA methylation and genetic polymorphisms both have a major impact on the occurrence and risk of developing cancer. Genetic polymorphisms are differences in DNA sequence that may have an impact on gene expression and function, possibly causing malignant cell alterations. Contrarily, DNA methylation is an epigenetic change that can influence gene expression by activating or silencing genes, having an effect on the onset of cancer. New findings are continuously being made as a result of ongoing research into genetic variants and the risk of developing cancer. Additional genetic variants linked to the risk of developing cancer will probably be discovered as the knowledge of the genetic basis of cancer increases. This knowledge could potentially lead to the development of personalized strategies for cancer prevention and treatment based on the unique genetic makeup of an individual.

Acknowledgements

The authors gratefully acknowledge the Galgotias University (Greater Noida, India) for providing the academic resources and valuable technical assistance during the preparation of the present review.

Funding

Funding: No funding was received.

Availability of data and materials

Not applicable.

Author's contributions

RM and NK conducted a significant portion of the literature search and drafted the manuscript. GS contributed to editing specific sections of the manuscript. SPS edited and proofread the manuscript. AKJ contributed to the initial conception and scope of the review, provided critical review and feedback on the manuscript. All authors have read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Lander ES, Linton LM, Birren B, Nusbaum C, Zody MC, Baldwin J, Devon K, Dewar K, Doyle M, FitzHugh W, et al: Initial sequencing and analysis of the human genome. Nature. 409:860–921. 2001.PubMed/NCBI View Article : Google Scholar

2 

Venter JC, Adams MD, Myers EW, Li PW, Mural RJ, Sutton GG, Smith HO, Yandell M, Evans CA, Holt RA, et al: The sequence of the human genome. Science. 291:1304–1351. 2001.PubMed/NCBI View Article : Google Scholar

3 

International Human Genome Sequencing Consortium: Finishing the euchromatic sequence of the human genome. Nature. 431:931–945. 2004.PubMed/NCBI View Article : Google Scholar

4 

International HapMap Consortium: The international HapMap project. Nature. 426:789–796. 2003.PubMed/NCBI View Article : Google Scholar

5 

Lin BK, Clyne M, Walsh M, Gomez O, Yu W, Gwinn M and Khoury MJ: Tracking the epidemiology of human genes in the literature: The HuGE published literature database. Am J Epidemiol. 164:1–4. 2006.PubMed/NCBI View Article : Google Scholar

6 

Schumacher FR, Al Olama AA, Berndt SI, Benlloch S, Ahmed M, Saunders EJ, Dadaev T, Leongamornlert D, Anokian E, Cieza-Borrella C, et al: Association analyses of more than 140,000 men identify 63 new prostate cancer susceptibility loci. Nat Genet. 50:928–936. 2018.PubMed/NCBI View Article : Google Scholar

7 

Breast Cancer Association Consortium. Dorling L, Carvalho S, Allen J, González-Neira A, Luccarini C, Wahlström C, Pooley KA, Parsons MT, Fortuno C, et al: Breast cancer risk genes-association analysis in more than 113,000 women. N Engl J Med. 384:428–439. 2021.PubMed/NCBI View Article : Google Scholar

8 

Ioannidis JP, Ntzani EE, Trikalinos TA and Contopoulos-Ioannidis DG: Replication validity of genetic association studies. Nat Genet. 29:306–309. 2001.PubMed/NCBI View Article : Google Scholar

9 

Morgan TM, Krumholz HM, Lifton RP and Spertus JA: Nonvalidation of reported genetic risk factors for acute coronary syndrome in a large-scale replication study. JAMA. 297:1551–1561. 2007.PubMed/NCBI View Article : Google Scholar

10 

Dickersin K and Berlin JA: Meta-analysis: State-of-the-science. Epidemiol Rev. 14:154–176. 1992.PubMed/NCBI View Article : Google Scholar

11 

Mosteller F and Colditz GA: Understanding research synthesis (meta-analysis). Annu Rev Public Health. 17:1–23. 1996.PubMed/NCBI View Article : Google Scholar

12 

Morris RD: Meta-analysis in cancer epidemiology. Environ Health Perspect. 102 (Suppl 8):S61–S66. 1994.PubMed/NCBI View Article : Google Scholar

13 

Chen J, Jiang CC, Jin L and Zhang XD: Regulation of PD-L1: A novel role of pro-survival signalling in cancer Ann. Oncol. 27:409–416. 2016.PubMed/NCBI View Article : Google Scholar

14 

Bardhan K, Anagnostou T and Boussiotis VA: The PD1:PD-L1/2 pathway from discovery to clinical implementation. Front Immunol. 7(550)2016.PubMed/NCBI View Article : Google Scholar

15 

Wang Q, Lin W, Tang X, Li S, Guo L, Lin Y and Kwok HF: The roles of microRNAs in regulating the expression of PD-1/PD-L1 immune checkpoint. Int J Mol Sci. 18(2540)2017.PubMed/NCBI View Article : Google Scholar

16 

Keir ME, Butte MJ, Freeman GJ and Sharpe AH: PD-1 and its ligands in tolerance and immunity. Annu Rev Immunol. 26:677–704. 2008.PubMed/NCBI View Article : Google Scholar

17 

Tang Y, Fang W, Zhang Y, Hong S, Kang S, Yan Y, Chen N, Zhan J, He X, Qin T, et al: The association between PD-L1 and EGFR status and the prognostic value of PD-L1 in advanced non-small cell lung cancer patients treated with EGFR-TKIs. Oncotarget. 6:14209–14219. 2015.PubMed/NCBI View Article : Google Scholar

18 

Cretella D, Digiacomo G, Giovannetti E and Cavazzoni A: PTEN Alterations as a Potential Mechanism for Tumor Cell Escape from PD-1/PD-L1 Inhibition. Cancers (Basel). 11(1318)2019.PubMed/NCBI View Article : Google Scholar

19 

Suzuki A, Nakano T, Mak TW and Sasaki T: Portrait of PTEN: Messages from mutant mice. Cancer Sci. 99:209–213. 2008.PubMed/NCBI View Article : Google Scholar

20 

Marzec M, Zhang Q, Goradia A, Raghunath PN, Liu X, Paessler M, Wang HY, Wysocka M, Cheng M, Ruggeri BA and Wasik MA: Oncogenic kinase NPM/ALK induces through STAT3 expression of immunosuppressive protein CD274 (PD-L1, B7-H1). Proc Natl Acad Sci USA. 105:20852–20857. 2008.PubMed/NCBI View Article : Google Scholar

21 

Lin H, Wei S, Hurt EM, Green MD, Zhao L, Vatan L, Szeliga W, Herbst R, Harms PW, Fecher LA, et al: Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade-mediated tumor regression. J Clin Invest. 128:805–815. 2018.PubMed/NCBI View Article : Google Scholar : Erratum in: J Clin Invest 128: 1708, 2018.

22 

McDonnell AM and Dang CH: . Basic review of the cytochrome p450 system. J Adv Pract Oncol. 4:263–268. 2013.PubMed/NCBI View Article : Google Scholar

23 

Chan CWH, Li C, Xiao EJ, Li M, Phiri PGM, Yan T and Chan JYW: Association between genetic polymorphisms in cytochrome P450 enzymes and survivals in women with breast cancer receiving adjuvant endocrine therapy: a systematic review and meta-analysis. Expert Rev Mol Med. 24(e1)2022.PubMed/NCBI View Article : Google Scholar

24 

Datkhile KD, Durgawale PP, Gudur RA, Gudur AK and Patil SR: CYP2D6 and CYP2E1 gene polymorphisms and their association with cervical cancer susceptibility: A hospital based case-control study from South-Western Maharashtra. Asian Pac J Cancer Prev. 23:2591–2597. 2022.PubMed/NCBI View Article : Google Scholar

25 

Agundez JAG: Cytochrome P450 gene polymorphism and cancer. Curr Drug Metab. 5:211–224. 2004.PubMed/NCBI View Article : Google Scholar

26 

Tan EY, Bharwani L, Chia YH, Soong RCT, Lee SSY, Chen JJC and Chan PMY: . Impact of cytochrome P450 2D6 polymorphisms on decision-making and clinical outcomes in adjuvant hormonal therapy for breast cancer. World J Clin Oncol. 13:712–724. 2022.PubMed/NCBI View Article : Google Scholar

27 

Jiang O, Zhou R, Wu D, Liu Y, Wu W and Cheng N: CYP2E1 polymorphisms and colorectal cancer risk: a HuGE systematic review and meta-analysis. Tumor Biol. 34:1215–1224. 2013.PubMed/NCBI View Article : Google Scholar

28 

Holick MF: Sunlight and vitamin D for bone health and prevention of autoimmune diseases, cancers, and cardiovascular disease. Am J Clin Nutr. 80 (6 Suppl):1678S–1688S. 2004.PubMed/NCBI View Article : Google Scholar

29 

Mathieu C and Badenhoop K: Vitamin D and type 1 diabetes mellitus: State of the art. Trends Endocrinol Metab. 16:261–266. 2005.PubMed/NCBI View Article : Google Scholar

30 

Misra M, Pacaud D, Petryk A, Collett-Solberg PF and Kappy M: Drug and Therapeutics Committee of the Lawson Wilkins Pediatric Endocrine Society. Vitamin D deficiency in children and its management: Review of current knowledge and recommendations. Pediatrics. 122:398–417. 2008.PubMed/NCBI View Article : Google Scholar

31 

Handono K, Sidarta YO, Pradana BA, Nugroho RA, Hartono IA, Kalim H and Endharti AT: Vitamin D prevents endothelial damage induced by increased neutrophil extracellular traps formation in patients with systemic lupus erythematosus. Acta Med Indones. 46:189–198. 2014.PubMed/NCBI

32 

Van Belle TL, Vanherwegen AS, Feyaerts D, De Clercq P, Verstuyf A, Korf H, Gysemans C and Mathieu C: 1,25-Dihydroxyvitamin D3 and its analog TX527 promote a stable regulatory T cell phenotype in T cells from type 1 diabetes patients. PLoS One. 9(e109194)2014.PubMed/NCBI View Article : Google Scholar

33 

Reddy KK: Reply to Glossmann: Vitamin D compounds and oral supplementation methods. J Invest Dermatol. 133(2649)2013.PubMed/NCBI View Article : Google Scholar

34 

Takahashi H, Hatta Y, Iriyama N, Hasegawa Y, Uchida H, Nakagawa M, Makishima M, Takeuchi J and Takei M: Induced differentiation of human myeloid leukemia cells into M2 macrophages by combined treatment with retinoic acid and 1α,25-dihydroxyvitamin D3. PLoS One. 9(e113722)2014.PubMed/NCBI View Article : Google Scholar

35 

Zhang Z, Zhang H, Hu Z, Wang P, Wan J and Li B: Synergy of 1,25-dihydroxyvitamin D3 and carboplatin in growth suppression of SKOV-3 cells. Oncol Lett. 8:1348–1354. 2014.PubMed/NCBI View Article : Google Scholar

36 

Henry HL: Regulation of vitamin D metabolism. Best Pract Res Clin Endocrinol Metab. 25:531–541. 2011.PubMed/NCBI View Article : Google Scholar

37 

Hossein-nezhad A and Holick MF: Vitamin D for health: A global perspective. Mayo Clin Proc. 88:720–755. 2013.PubMed/NCBI View Article : Google Scholar

38 

Cantorna MT, Zhu Y, Froicu M and Wittke A: Vitamin D status, 1,25-dihydroxyvitamin D3, and the immune system. Am J Clin Nutr. 80 (6 Suppl):1717S–1720S. 2004.PubMed/NCBI View Article : Google Scholar

39 

Weinstein SJ, Purdue MP, Smith-Warner SA, Mondul AM, Black A, Ahn J, Huang WY, Horst RL, Kopp W, Rager H, et al: Serum 25-hydroxyvitamin D, vitamin D binding protein and risk of colorectal cancer in the prostate, lung, colorectal and ovarian cancer screening trial. Int J Cancer. 136:E654–E664. 2015.PubMed/NCBI View Article : Google Scholar

40 

Slominski AT, Kim TK, Janjetovic Z, Slominski RM, Li W, Jetten AM, Indra AK, Mason RS and Tuckey RC: Biological effects of CYP11A1-derived vitamin D and lumisterol metabolites in the skin. J Invest Dermatol. 144:2145–2161. 2024.PubMed/NCBI View Article : Google Scholar

41 

Slominski AT, Tuckey RC, Jenkinson C, Li W and Jetten AM: Alternative pathways for vitamin D metabolism. In: Hewison M, Bouillon R, Giovanucci E, Goltzman D, Meyer M and Welsh J (eds.), Feldman and Pike's Vitamin D: Volume One: Biochemistry, Physiology and Diagnostics. 5th edition. Academic Press, pp85-109, 2024.

42 

Hoeijmakers JH: Genome maintenance mechanisms for preventing cancer. Nature. 411:366–374. 2001.PubMed/NCBI View Article : Google Scholar

43 

Stary A and Sarasin A: The genetics of the hereditary xeroderma pigmentosum syndrome. Biochimie. 84:49–60. 2002.PubMed/NCBI View Article : Google Scholar

44 

Cleaver JE: Defective repair replication of DNA in xeroderma pigmentosum. Nature. 218:652–656. 1968.PubMed/NCBI View Article : Google Scholar

45 

Kraemer KH, Levy DD, Parris CN, Gozukara EM, Moriwaki S, Adelberg S and Seidman MM: Xeroderma pigmentosum and related disorders: Examining the linkage between defective DNA repair and cancer. J Invest Dermatol. 103 (5 Suppl):96S–101S. 1994.PubMed/NCBI View Article : Google Scholar

46 

Wood RD: DNA damage recognition during nucleotide excision repair in mammalian cells. Biochimie. 81:39–44. 1999.PubMed/NCBI View Article : Google Scholar

47 

de Boer J and Hoeijmakers JH: Nucleotide excision repair and human syndromes. Carcinogenesis. 21:453–460. 2000.PubMed/NCBI View Article : Google Scholar

48 

Friedberg EC, Feaver WJ and Gerlach VL: The many faces of DNA polymerases: Strategies for mutagenesis and for mutational avoidance. Proc Natl Acad Sci USA. 97:5681–5683. 2000.PubMed/NCBI View Article : Google Scholar

49 

Whitlock JP Jr: Induction of cytochrome P4501A1. Annu Rev Pharmacol Toxicol. 39:103–125. 1999.PubMed/NCBI View Article : Google Scholar

50 

Iba MM, Scholl H, Fung J, Thomas PE and Alam J: Induction of pulmonary CYP1A1 by nicotine. Xenobiotica. 28:827–843. 1998.PubMed/NCBI View Article : Google Scholar

51 

San Jose C, Cabanillas A, Benitez J, Carrillo JA, Jimenez M and Gervasini G: CYP1A1 gene polymorphisms increase lung cancer risk in a high-incidence region of Spain: A case control study. BMC Cancer. 10(463)2010.PubMed/NCBI View Article : Google Scholar

52 

Johansson I, Xanthopoulou EM, Zhou Y, Sanchez-Spitman A, van der Lee M, Wollmann BM, Størset E, Swen JJ, Guchelaar HJ, Molden E, et al: Improved prediction of CYP2D6 catalyzed drug metabolism by taking variant substrate specificities and novel polymorphic haplotypes into account. Clin Pharmacol Ther. 118:218–231. 2025.PubMed/NCBI View Article : Google Scholar

53 

Wolf CR and Smith G: Cytochrome P450 CYP2D6. In: Metabolic Polymorphisms and Susceptibility to Cancer. Vol 148. International Agency for Research on Cancer, Lyon, pp209-229, 1999.

54 

Crespi CL, Penman BW, Gelboin HV and Gonzalez FJ: A tobacco smoke-derived nitrosamine, 4-(methylnitrosamino)-1-(3- pyridyl)-1-butanone, is activated by multiple human cytochrome P450s including the polymorphic human cytochrome P4502D6,. Carcinogenesis. 12:1197–1201. 1991.PubMed/NCBI View Article : Google Scholar

55 

Lorenzo-Pouso AI, Silva FFE, Pérez-Jardón A, Chamorro-Petronacci CM, Oliveira-Alves MG, Álvarez-Calderón-Iglesias Ó, Caponio VCA, Pinti M, Perrotti V and Pérez-Sayáns M: Overexpression of E-cadherin is a favorable prognostic biomarker in oral squamous cell carcinoma: A systematic review and meta-analysis. Biology (Basel). 12(239)2023.PubMed/NCBI View Article : Google Scholar

56 

Chen CY, Chen J, He L and Stiles BL: PTEN: Tumor Suppressor and Metabolic Regulator. Front Endocrinol (Lausanne). 9(338)2018.PubMed/NCBI View Article : Google Scholar

57 

Song MS, Salmena L and Pandolfi PP: The functions and regulation of the PTEN tumour suppressor. Nat Rev Mol Cell Biol. 13:283–296. 2012.PubMed/NCBI View Article : Google Scholar

58 

Han F, Hu R, Yang H, Liu J, Sui J, Xiang X, Wang F, Chu L and Song S: PTEN gene mutations correlate to poor prognosis in glioma patients: a meta-analysis. Onco Targets Ther. 9:3485–3492. 2016.PubMed/NCBI View Article : Google Scholar

59 

Aubrey BJ, Kelly GL, Janic A, Herold MJ and Strasser A: How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 25:104–113. 2018.PubMed/NCBI View Article : Google Scholar

60 

Vaddavalli P and Schumacher B: The p53 network: Cellular and systemic DNA damage responses in cancer and aging. Trends Genet. 38:598–612. 2022.PubMed/NCBI View Article : Google Scholar

61 

Bouaoun L, Sonkin D, Ardin M, Hollstein M, Byrnes G, Zavadil J and Olivier M: TP53 variations in human cancers: New lessons from the IARC TP53 database and genomics data. Hum Mutat. 37:865–876. 2016.PubMed/NCBI View Article : Google Scholar

62 

Mantovani F, Collavin L and Del Sal G: Mutant p53 as a guardian of the cancer cell. Cell Death Differ. 26:199–212. 2019.PubMed/NCBI View Article : Google Scholar

63 

Keriel A, Stary A, Sarasin A, Rochette-Egly C and Egly JM: XPD mutations prevent TFIIH-dependent transactivation by nuclear receptors and phosphorylation of RARalpha. Cell. 109:125–135. 2002.PubMed/NCBI View Article : Google Scholar

64 

Coin F, Marinoni JC, Rodolfo C, Fribourg S, Pedrini AM and Egly JM: Mutations in the XPD helicase gene result in XP and TTD phenotypes, preventing interaction between XPD and the p44 subunit of TFIIH. Nat Genet. 20:184–188. 1998.PubMed/NCBI View Article : Google Scholar

65 

Lunn RM, Helzlsouer KJ, Parshad R, Umbach DM, Harris EL, Sanford KK and Bell DA: XPD polymorphisms: Effects on DNA repair proficiency. Carcinogenesis. 21:551–555. 2000.PubMed/NCBI View Article : Google Scholar

66 

Mohamed NS, Ali Albsheer MM, Abdelbagi H, Siddig EE, Mohamed MA, Ahmed AE, Omer RA, Muneer MS, Ahmed A, Osman HA, et al: Genetic polymorphism of the N-terminal region in circumsporozoite surface protein of Plasmodium falciparum field isolates from Sudan. Malar J. 18(333)2019.PubMed/NCBI View Article : Google Scholar

67 

Ju HL, Kang JM, Moon SU, Kim JY, Lee HW, Lin K, Sohn WM, Lee JS, Kim TS and Na BK: Genetic polymorphism and natural selection of Duffy binding protein of Plasmodium vivax Myanmar isolates. Malar J. 11:1–110. 2012.PubMed/NCBI View Article : Google Scholar

68 

Thompson EE, Sun Y, Nicolae D and Ober C: Shades of gray: A comparison of linkage disequilibrium between Hutterites and Europeans. Genet Epidemiol. 34:133–139. 2010.PubMed/NCBI View Article : Google Scholar

69 

Tibayrenc M: Human genetic diversity and the epidemiology of parasitic and other transmissible diseases. Adv Parasitol. 64:377–422. 2007.PubMed/NCBI View Article : Google Scholar

70 

Guo CY, DeStefano AL, Lunetta KL, Dupuis J and Cupples LA: Expectation maximization algorithm based haplotype relative risk (EM-HRR): Test of linkage disequilibrium using incomplete case-parents trios. Hum Hered. 59:125–135. 2005.PubMed/NCBI View Article : Google Scholar

71 

Silverman EK, Chapman HA, Drazen JM, Weiss ST, Rosner B, Campbell EJ, O'Donnell WJ, Reilly JJ, Ginns L, Mentzer S, et al: Genetic epidemiology of severe, early-onset chronic obstructive pulmonary disease. Risk to relatives for airflow obstruction and chronic bronchitis. Am J Respir Crit Care Med. 157:1770–1778. 1998.PubMed/NCBI View Article : Google Scholar

72 

Karahalil B, Bohr VA and Wilson DM III: Impact of DNA polymorphisms in key DNA base excision repair proteins on cancer risk. Hum Exp Toxicol. 31:981–1005. 2012.PubMed/NCBI View Article : Google Scholar

73 

Bookman EB, McAllister K, Gillanders E, Wanke K, Balshaw D, Rutter J, Reedy J, Shaughnessy D, Agurs-Collins T, Paltoo D, et al: Gene-environment interplay in common complex diseases: Forging an integrative model-recommendations from an NIH workshop. Genet Epidemiol. 35:217–225. 2011.PubMed/NCBI View Article : Google Scholar

74 

Daly AK: Pharmacogenetics and human genetic polymorphisms. Biochem J. 429:435–449. 2010.PubMed/NCBI View Article : Google Scholar

75 

Mathur R, Rana BS and Jha AK: Single nucleotide polymorphism (SNP). In: Vonk J, Shackelford T (eds). Encyclopedia of Animal Cognition and Behavior. Springer, Cham, pp1-4, 2018.

76 

Ando Y, Saka H, Ando M, Sawa T, Muro K, Ueoka H, Yokoyama A, Saitoh S, Shimokata K and Hasegawa Y: Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan toxicity: A pharmacogenetic analysis. Cancer Res. 60:6921–6926. 2000.PubMed/NCBI

77 

Mallal S, Phillips E, Carosi G, Molina JM, Workman C, Tomazic J, Jägel-Guedes E, Rugina S, Kozyrev O, Cid JF, et al: HLA-B*5701 screening for hypersensitivity to abacavir. N Engl J Med. 358:568–579. 2008.PubMed/NCBI View Article : Google Scholar

78 

Halder I and Shriver MD: Measuring and using admixture to study the genetics of complex diseases. Hum Genomics. 1:52–62. 2003.PubMed/NCBI View Article : Google Scholar

79 

Hu D and Ziv E: Confounding in genetic association studies and its solutions. Methods Mol Biol. 448:31–39. 2008.PubMed/NCBI View Article : Google Scholar

80 

Hettiarachchi G and Komar AA: GWAS to identify SNPs associated with common diseases and individual risk: Genome wide association studies (GWAS) to identify SNPs associated with common diseases and individual risk. In: Sauna ZE, Kimchi-Sarfaty C (eds). Single Nucleotide Polymorphisms. Springer, Cham, pp51-76, 2022.

81 

Uffelmann E, Huang QQ, Munung NS, De Vries J, Okada Y, Martin AR, Martin HC, Lappalainen T and Posthuma D: Genome-wide association studies. Nat Rev Methods Primers. 1(59)2021.

82 

Umeno M, McBride OW, Yang CS, Gelboin HV and Gonzalez FJ: Human ethanol-inducible P450IIE1: Complete gene sequence, promoter characterization, chromosome mapping, and cDNA-directed expression. Biochemistry. 27:9006–9013. 1988.PubMed/NCBI View Article : Google Scholar

83 

Guengerich FP, Kim DH and Iwasaki M: Role of human cytochrome P-450 IIE1 in the oxidation of many low molecular weight cancer suspects. Chem Res Toxicol. 4:168–179. 1991.PubMed/NCBI View Article : Google Scholar

84 

Nakajima T and Aoyama T: Polymorphism of drug-metabolizing enzymes in relation to individual susceptibility to industrial chemicals. Ind Health. 38:143–152. 2000.PubMed/NCBI View Article : Google Scholar

85 

Wang AH, Sun CS, Li LS, Huang JY and Chen QS: Relationship of tobacco smoking CYP1A1 GSTM1 gene polymorphism and esophageal cancer in Xi'an. World J Gastroenterol. 8:49–53. 2002.PubMed/NCBI View Article : Google Scholar

86 

Bartsch H, Nair U, Risch A, Rojas M, Wikman H and Alexandrov K: Genetic polymorphism of CYP genes, alone or in combination, as a risk modifier of tobacco-related cancers. Cancer Epidemiol Biomarkers Prev. 9:3–28. 2000.PubMed/NCBI

87 

Itoga S, Nomura F, Makino Y, Tomonaga T, Shimada H, Ochiai T, Iizasa T, Baba M, Fujisawa T and Harada S: Tandem repeat polymorphism of the CYP2E1 gene: An association study with esophageal cancer and lung cancer. Alcohol Clin Exp Res. 26 (8 Suppl):15S–19S. 2002.PubMed/NCBI View Article : Google Scholar

88 

Danko IM and Chaschin NA: Association of CYP2E1 gene polymorphism with predisposition to cancer development. Exp Oncol. 27:248–256. 2005.PubMed/NCBI

89 

Hayashi S, Watanabe J and Kawajiri K: Genetic polymorphisms in the 5'-flanking region change transcriptional regulation of the human cytochrome P450IIE1 gene. J Biochem. 110:559–565. 1991.PubMed/NCBI View Article : Google Scholar

90 

Watanabe M: Polymorphic CYP genes and disease predisposition-what have the studies shown so far? Toxicol Lett. 102-103:167–171. 1998.PubMed/NCBI View Article : Google Scholar

91 

Yang B, O'Reilly DA, Demaine AG and Kingsnorth AN: Study of polymorphisms in the CYP2E1 gene in patients with alcoholic pancreatitis. Alcohol. 23:91–97. 2001.PubMed/NCBI View Article : Google Scholar

92 

Yu SZ, Huang XE, Koide T, Cheng G, Chen GC, Harada K, Ueno Y, Sueoka E, Oda H, Tashiro F, et al: Hepatitis B and C viruses infection, lifestyle and genetic polymorphisms as risk factors for hepatocellular carcinoma in Haimen, China. Jpn J Cancer Res. 93:1287–1292. 2002.PubMed/NCBI View Article : Google Scholar

93 

Munaka M, Kohshi K, Kawamoto T, Takasawa S, Nagata N, Itoh H, Oda S and Katoh T: Genetic polymorphisms of tobacco- and alcohol-related metabolizing enzymes and the risk of hepatocellular carcinoma. J Cancer Res Clin Oncol. 129:355–360. 2003.PubMed/NCBI View Article : Google Scholar

94 

Norton ID, Apte MV, Haber PS, McCaughan GW, Pirola RC and Wilson JS: Cytochrome P4502E1 is present in rat pancreas and is induced by chronic ethanol administration. Gut. 42:426–430. 1998.PubMed/NCBI View Article : Google Scholar

95 

Catanzaro I, Naselli F, Saverini M, Giacalone A, Montalto G and Caradonna F: Cytochrome P450 2E1 variable number tandem repeat polymorphisms and health risks: A genotype-phenotype study in cancers associated with drinking and/or smoking. Mol Med Rep. 6:416–420. 2012.PubMed/NCBI View Article : Google Scholar

96 

Christiani DC, Mehta AJ and Yu CL: Genetic susceptibility to occupational exposures. Occup Environ Med. 65:430–436, 436, 397. 2008.PubMed/NCBI View Article : Google Scholar

97 

Pizzino G, Bitto A, Interdonato M, Galfo F, Irrera N, Mecchio A, Pallio G, Ramistella V, De Luca F, Minutoli L, et al: Oxidative stress and DNA repair and detoxification gene expression in adolescents exposed to heavy metals living in the Milazzo-Valle del Mela area (Sicily, Italy). Redox Biol. 2:686–693. 2014.PubMed/NCBI View Article : Google Scholar

98 

Kringel D, Lippmann C, Parnham MJ, Kalso E, Ultsch A and Lötsch J: A machine-learned analysis of human gene polymorphisms modulating persisting pain points to major roles of neuroimmune processes. Eur J Pain. 22:1735–1756. 2018.PubMed/NCBI View Article : Google Scholar

99 

Pandiyan A, Lari S, Vanka J, Kumar BS, Ghosh S, Jee B and Jonnalagadda PR: Genetic polymorphism in xenobiotic metabolising genes and increased oxidative stress among pesticides exposed agricultural workers diagnosed with cancers. Asian Pac J Cancer Prev. 24:3795–3804. 2023.PubMed/NCBI View Article : Google Scholar

100 

Rothman N, Wacholder S, Caporaso NE, Garcia-Closas M, Buetow K and Fraumeni JF Jr: The use of common genetic polymorphisms to enhance the epidemiologic study of environmental carcinogens. Biochim Biophys Acta. 1471:C1–C10. 2001.PubMed/NCBI View Article : Google Scholar

101 

Kelada SN, Eaton DL, Wang SS, Rothman NR and Khoury MJ: The role of genetic polymorphisms in environmental health. Environ Health Perspect. 111:1055–1064. 2003.PubMed/NCBI View Article : Google Scholar

102 

Carr BA and Franklin MR: Drug-metabolizing enzyme induction by 2,2'-dipyridyl, 1,7-phenanthroline, 7,8-benzoquinoline and oltipraz in mouse. Xenobiotica. 28:949–956. 1998.PubMed/NCBI View Article : Google Scholar

103 

Langouët S, Coles B, Morel F, Becquemont L, Beaune P, Guengerich FP, Ketterer B and Guillouzo A: Inhibition of CYP1A2 and CYP3A4 by oltipraz results in reduction of aflatoxin B1 metabolism in human hepatocytes in primary culture. Cancer Res. 55:5574–5579. 1995.PubMed/NCBI

104 

Kensler TW, Egner PA, Wang JB, Zhu YR, Zhang BC, Lu PX, Chen JG, Qian GS, Kuang SY, Jackson PE, et al: Chemoprevention of hepatocellular carcinoma in aflatoxin endemic areas. Gastroenterology. 127 (5 Suppl 1):S310–S318. 2004.PubMed/NCBI View Article : Google Scholar

105 

Schulte P and Howard J: Genetic susceptibility and the setting of occupational health standards. Annu Rev Public Health. 32:149–159. 2011.PubMed/NCBI View Article : Google Scholar

106 

Naselli F, Catanzaro I, Bellavia D, Perez A, Sposito L and Caradonna F: Role and importance of polymorphisms with respect to DNA methylation for the expression of CYP2E1 enzyme. Gene. 536:29–39. 2014.PubMed/NCBI View Article : Google Scholar

107 

Sharma S, Sambyal V, Guleria K, Manjari M, Sudan M, Uppal MS, Singh NR, Bansal D and Gupta A: TP53 polymorphisms in sporadic North Indian breast cancer patients. Asian Pac J Cancer Prev. 15:6871–6879. 2014.PubMed/NCBI View Article : Google Scholar

108 

Petrucelli N, Daly MB and Pal T: BRCA1- and BRCA2-associated hereditary breast and ovarian cancer. 1998 Sep 4 [Updated 2025 Mar 20]. In: Adam MP, Feldman J, Mirzaa GM, Pagon RA, Wallace SE and Amemiya A (eds). GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle, 1993.

109 

Slominski RM, Raman C, Chen JY and Slominski AT: How cancer hijacks the body's homeostasis through the neuroendocrine system. Trends Neurosci. 46:263–275. 2023.PubMed/NCBI View Article : Google Scholar

Related Articles

  • Abstract
  • View
  • Download
Copy and paste a formatted citation
Spandidos Publications style
Mathur R, Khare N, Saini G, Shukla SP and Jha AK: Role of genetic polymorphisms within different genes and associated risks of cancer development (Review). World Acad Sci J 7: 118, 2025.
APA
Mathur, R., Khare, N., Saini, G., Shukla, S.P., & Jha, A.K. (2025). Role of genetic polymorphisms within different genes and associated risks of cancer development (Review). World Academy of Sciences Journal, 7, 118. https://doi.org/10.3892/wasj.2025.406
MLA
Mathur, R., Khare, N., Saini, G., Shukla, S. P., Jha, A. K."Role of genetic polymorphisms within different genes and associated risks of cancer development (Review)". World Academy of Sciences Journal 7.6 (2025): 118.
Chicago
Mathur, R., Khare, N., Saini, G., Shukla, S. P., Jha, A. K."Role of genetic polymorphisms within different genes and associated risks of cancer development (Review)". World Academy of Sciences Journal 7, no. 6 (2025): 118. https://doi.org/10.3892/wasj.2025.406
Copy and paste a formatted citation
x
Spandidos Publications style
Mathur R, Khare N, Saini G, Shukla SP and Jha AK: Role of genetic polymorphisms within different genes and associated risks of cancer development (Review). World Acad Sci J 7: 118, 2025.
APA
Mathur, R., Khare, N., Saini, G., Shukla, S.P., & Jha, A.K. (2025). Role of genetic polymorphisms within different genes and associated risks of cancer development (Review). World Academy of Sciences Journal, 7, 118. https://doi.org/10.3892/wasj.2025.406
MLA
Mathur, R., Khare, N., Saini, G., Shukla, S. P., Jha, A. K."Role of genetic polymorphisms within different genes and associated risks of cancer development (Review)". World Academy of Sciences Journal 7.6 (2025): 118.
Chicago
Mathur, R., Khare, N., Saini, G., Shukla, S. P., Jha, A. K."Role of genetic polymorphisms within different genes and associated risks of cancer development (Review)". World Academy of Sciences Journal 7, no. 6 (2025): 118. https://doi.org/10.3892/wasj.2025.406
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team