The interplay of genetics and cancer
Cancer and genetics
Authors
Abstract
Cancer is fundamentally a genetic disease driven by alterations in the genome that disrupt normal cellular functions and promote malignant transformation. Genetic mutations, including point mutations, chromosomal rearrangements, gene amplifications, and epigenetic changes, play critical roles in initiating and sustaining tumorigenesis. These alterations affect key pathways regulating cell proliferation, apoptosis, DNA repair, and angiogenesis, leading to uncontrolled growth and metastasis. The discovery of oncogenes, such as KRAS and MYC, and tumor suppressor genes, such as TP53 and RB1, has provided critical insights into cancer biology and facilitated the development of targeted therapies that inhibit specific molecular drivers. Advances in genomics, particularly next- generation sequencing, have revolutionized cancer research and clinical practice by enabling comprehensive tumor profiling. This approach underpins precision oncology, where treatments are tailored to the unique genetic makeup of a patient’s tumor, improving outcomes and minimizing adverse effects. Despite these advances, challenges remain due to tumor heterogeneity, the emergence of drug resistance, and the influence of the tumor microenvironment on genetic evolution. Liquid biopsy technologies and germline genetic screening offer promise for early cancer detection and personalized prevention strategies. While the integration of genetics into cancer research has transformed our understanding of the disease, ongoing efforts are required to address its complexity. Future research into tumor evolution, resistance mechanisms, and the interplay between genetics and the immune system will further enhance therapeutic strategies. By unraveling the intricate relationship between cancer and genetics, the potential for curative treatments and improved patient survival continues to grow.
Keywords
Introduction
Cancer is one of the leading causes of morbidity and mortality worldwide, representing a major public health challenge despite significant advances in early detection, diagnosis, and treatment [1]. At its core, cancer is a disease of genetic alterations. These alterations disrupt the normal regulation of cell growth, differentiation, and survival, leading to the development of malignant tumors [2]. Understanding the intricate interplay between genetics and cancer has transformed modern oncology, providing insights into the mechanisms of tumorigenesis, therapeutic resistance, and metastasis [3].
The link between cancer and genetics was first suggested in the mid-20th century, with the identification of chromosomal abnormalities in cancer cells [4]. The discovery of the Philadelphia chromosome in chronic myelogenous leukemia (CML) marked a watershed moment, as it provided the first direct evidence of a genetic basis for cancer [5]. Subsequent research has revealed that cancer arises from the accumulation of genetic mutations in somatic cells, as well as inherited genetic predispositions that increase the risk of specific cancer types. Genetic alterations can be broadly categorized into two types: somatic mutations, which are acquired during a person’s lifetime and are restricted to tumor cells, and germline mutations, which are inherited and present in every cell of the body [6].
The genetic landscape of cancer is complex, involving a wide array of molecular alterations that contribute to tumor initiation and progression. Oncogenes, for instance, are genes that, when mutated or overexpressed, promote cell proliferation and survival [7]. Prominent examples include KRAS, MYC, and BRAF. Tumor suppressor genes, on the other hand, act as cellular safeguards by regulating cell cycle progression and repairing DNA damage. Mutations in genes such as TP53, BRCA1, and RB1 disable these protective mechanisms, leading to genomic instability and uncontrolled cell growth [8]. Additionally, alterations in DNA repair pathways, such as those associated with mismatch repair genes, contribute to an accumulation of mutations, further driving tumor evolution [9].
Advances in high-throughput genomic technologies, particularly next-generation sequencing (NGS), have provided unprecedented insights into the genetic basis of cancer [9, 10]. These tools allow for the comprehensive profiling of tumor genomes, uncovering the full spectrum of genetic mutations, structural variants, and epigenetic changes. The advent of large-scale genomic initiatives, such as The Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC), has further expanded our understanding of the genetic diversity within and between cancer types [11]. Such efforts have identified key driver mutations, common pathways implicated in tumorigenesis, and molecular subtypes that have prognostic and therapeutic implications. The integration of genetics into oncology has revolutionized clinical practice, giving rise to the field of precision oncology. By leveraging genomic information, clinicians tailor treatments to the specific molecular profile of a patient’s tumor. Targeted therapies, such as tyrosine kinase inhibitors and monoclonal antibodies, exemplify the success of this approach. Furthermore, genetic insights have informed the development of immunotherapies, including immune checkpoint inhibitors and personalized cancer vaccines, which exploit the genetic and immunological vulnerabilities of tumors [12].
The Genetic Basis of Cancer
Oncogenes and Tumor Suppressor Genes
The development and progression of cancer are fundamentally linked to the dysregulation of two critical classes of genes: oncogenes and tumor suppressor genes [13]. These genes are integral to the regulation of cellular processes, such as proliferation, differentiation, and apoptosis. Their alterations disrupt the delicate balance of cell cycle control, leading to uncontrolled cellular growth and, ultimately, tumor formation. Understanding the molecular mechanisms underlying their function provides significant insights into the biology of cancer and serves as the foundation for the development of targeted therapies [14].
Oncogenes are mutated or overexpressed derivatives of normal cellular genes, known as proto-oncogenes, which typically encode proteins involved in signaling pathways that regulate cell growth and survival. Under normal conditions, proto-oncogenes play essential roles in cellular functions, including growth factor signaling, transcription regulation, and cell cycle progression [15]. However, genetic alterations such as point mutations, chromosomal rearrangements, or gene amplifications can convert these proto-oncogenes into oncogenes, leading to their constitutive activation. For instance, the RAS family of genes, frequently mutated in cancers such as pancreatic, colorectal, and lung cancers, produces proteins that remain persistently active, driving excessive cell proliferation and inhibiting apoptosis [16]. Similarly, chromosomal translocations such as the formation of the BCR-ABL fusion gene in chronic myeloid leukemia (CML) result in the production of a constitutively active tyrosine kinase, promoting unregulated cell division [17]. Oncogene activation not only accelerates tumor growth but also contributes to processes such as angiogenesis and metastasis, further enhancing the aggressiveness of the malignancy.
In contrast, tumor suppressor genes serve as a counterbalance to oncogenes, acting to maintain genomic stability and prevent malignant transformation. These genes encode proteins that regulate the cell cycle, facilitate DNA repair, and promote apoptosis in response to cellular stress or damage. The loss of tumor suppressor gene function is often a critical step in carcinogenesis, as it removes essential barriers to unchecked cellular growth. Genetic alterations such as deletions, loss of heterozygosity, or inactivating point mutations frequently target these genes [18]. One of the most well-studied tumor suppressor genes is TP53, which encodes the p53 protein. Dubbed the “guardian of the genome,” p53 plays a pivotal role in preserving genomic integrity by halting the cell cycle in response to DNA damage and initiating repair or apoptosis if the damage is irreparable [19]. Mutations in TP53 are found in more than 50% of human cancers and are strongly associated with poor prognosis and resistance to therapy. Another key tumor suppressor gene, RB1, regulates the G1/S transition of the cell cycle, and its loss is implicated in cancers such as retinoblastoma and osteosarcoma.
The interplay between oncogenes and tumor suppressor genes underscores the complexity of cancer biology [20]. Oncogene activation often works in tandem with the inactivation of tumor suppressor genes to drive tumor progression. For example, in colorectal cancer, the activation of the KRAS oncogene frequently coincides with the loss of the APC tumor suppressor gene, creating a synergistic effect that accelerates tumorigenesis. Additionally, hereditary cancer syndromes provide compelling evidence of the significance of these genes. Germline mutations in tumor suppressor genes, such as BRCA1 and BRCA2 in hereditary breast and ovarian cancer, significantly increase an individual’s cancer risk. These genes are critical for homologous recombination repair of DNA double-strand breaks, and their loss predisposes cells to genomic instability and malignant transformation [21].
Genomic Instability and DNA Repair Mechanisms
Genomic instability is a hallmark of cancer and a driving force behind tumorigenesis, characterized by the accumulation of mutations, chromosomal rearrangements, and aneuploidy [22]. This instability arises from a combination of endogenous factors, such as errors during DNA replication, and exogenous factors, including ultraviolet (UV) radiation, ionizing radiation, and chemical carcinogens. The integrity of the genome is critical for normal cellular function, and cells have evolved highly sophisticated DNA repair mechanisms to maintain genomic stability [23]. When these repair pathways are compromised, the resulting accumulation of DNA damage can lead to the activation of oncogenes, the inactivation of tumor suppressor genes, and the promotion of cancer development.
The primary sources of genomic instability include DNA replication errors, oxidative damage, and double-strand breaks (DSBs). Replication errors occur when DNA polymerases introduce mismatched bases or slippage events during replication. Although these errors are rare, they are efficiently corrected by the mismatch repair (MMR) system. Oxidative damage, caused by reactive oxygen species (ROS), results in the formation of mutagenic lesions such as 8-oxo guanine [24]. Base excision repair (BER) is the primary pathway responsible for correcting oxidative lesions, involving glycosylases that recognize and excise damaged bases, followed by DNA polymerase and ligase activities to restore the DNA sequence. DNA double-strand breaks (DSBs) represent one of the most deleterious forms of DNA damage, as they disrupt the continuity of the chromosome and can lead to translocations or deletions if improperly repaired. Other important DNA repair mechanisms include nucleotide excision repair (NER) and the Fanconi anemia (FA) pathway. NER is responsible for removing bulky DNA adducts and helix-distorting lesions caused by UV light and chemical agents [25]. The pathway involves the recognition of DNA damage, unwinding of the DNA helix by TFIIH, excision of the damaged strand, and synthesis of a new strand using the undamaged strand as a template.
The dysregulation of DNA repair mechanisms contributes to cancer susceptibility and influences therapeutic responses. Tumors with defective DNA repair pathways, such as those with BRCA mutations, exhibit hypersensitivity to DNA-damaging agents like platinum-based chemotherapy and poly (ADP- ribose) polymerase (PARP) inhibitors. PARP inhibitors exploit synthetic lethality by targeting the base excision repair pathway in cells already deficient in homologous recombination. While this therapeutic approach has shown promise, resistance mechanisms, including the restoration of BRCA function or upregulation of alternative repair pathways, remain significant challenges [26].
In addition to cancer, genomic instability is implicated in aging and neurodegenerative diseases. Accumulation of unrepaired DNA damage over time contributes to cellular senescence and the decline of tissue homeostasis. Moreover, defects in DNA repair pathways are associated with conditions such as ataxia- telangiectasia, Cockayne syndrome, and Werner syndrome, all of which exhibit features of premature aging and genomic instability.
Inherited Genetic Predispositions
Hereditary Cancer Syndromes
Hereditary cancer syndromes are genetic conditions characterized by an increased risk of developing specific types of cancer due to inherited mutations in cancer susceptibility genes. These mutations are typically germline, meaning they are present in every cell of the body and passed down through families in an autosomal dominant or recessive manner. While hereditary cancer syndromes account for only 5-10% of all cancers, their identification is crucial for early detection, targeted prevention, and personalized treatment strategies. Understanding the molecular and genetic basis of these syndromes provides valuable insights into tumorigenesis and offers opportunities for intervention.
One of the most well-known hereditary cancer syndromes is hereditary breast and ovarian cancer (HBOC) syndrome, primarily associated with mutations in the BRCA1 and BRCA2 genes. These genes play essential roles in homologous recombination repair, a high-fidelity DNA repair pathway. Germline mutations in BRCA1 or BRCA2 compromise DNA repair, leading to genomic instability and an elevated lifetime risk of breast, ovarian, prostate, and pancreatic cancers. Women with BRCA mutations have up to a 70% lifetime risk of breast cancer and a significantly increased risk of ovarian cancer. Genetic testing for BRCA mutations has become a cornerstone of risk assessment, enabling the implementation of preventive measures such as enhanced surveillance, prophylactic surgeries (e.g., mastectomy and salpingo-oophorectomy), and chemoprevention. Moreover, tumors in BRCA mutation carriers are often sensitive to platinum-based chemotherapies and PARP inhibitors, which exploit the DNA repair deficiency in cancer cells.
Familial adenomatous polyposis (FAP) is another inherited syndrome, caused by mutations in the APC gene. This condition is characterized by the development of hundreds to thousands of colorectal adenomas, often beginning in adolescence or early adulthood. Without intervention, nearly all individuals with FAP develop colorectal cancer by the age of 40. Prophylactic colectomy is a standard preventive measure for individuals diagnosed with FAP. Variants of FAP, such as attenuated FAP and Gardner syndrome, may present with fewer polyps or extracolonic manifestations, including desmoid tumors, osteomas, and thyroid cancers. Advances in genetic testing and family counseling have improved the management and prevention of FAP-related cancers [27, 28].
Li-Fraumeni syndrome (LFS) is another hereditary cancer syndrome caused by germline mutations in the TP53 gene, which encodes the p53 tumor suppressor protein. TP53 is a critical regulator of the cell cycle, DNA repair, and apoptosis, and its inactivation predisposes individuals to a wide range of cancers, including sarcomas, breast cancer, brain tumors, and adrenocortical carcinomas. Individuals with LFS often develop cancer at an early age, and the syndrome is associated with a high lifetime cancer risk exceeding 90% in some cases. Given the spectrum of cancers in LFS, surveillance protocols using whole-body MRI have been developed to enable early detection of malignancies [29].
Hereditary cancer syndromes represent a critical area of cancer research and clinical practice. The identification of germline mutations in cancer susceptibility genes has not only improved our understanding of cancer biology but also enabled the development of personalized risk-reduction strategies and therapeutic interventions. Ongoing advancements in genetic testing technologies and the expansion of hereditary cancer screening programs hold the potential to further transform the landscape of cancer prevention and treatment.
Advances in Next-Generation Sequencing
Next-generation sequencing (NGS) has revolutionized the field of genomics by enabling high-throughput, cost-effective, and comprehensive analysis of DNA and RNA sequences. Unlike traditional Sanger sequencing, which relies on capillary electrophoresis to determine nucleotide sequences one fragment at a time, NGS allows for the simultaneous sequencing of millions of fragments in a massively parallel fashion. This technological advancement has transformed basic research, clinical diagnostics, and personalized medicine, providing unprecedented insights into the genetic basis of health and disease [30].
The core principle of NGS involves fragmenting DNA or RNA into smaller pieces, attaching adapters, and amplifying the fragments to create a library. This library is then loaded onto a sequencing platform, where individual fragments are sequenced using a variety of methods, such as sequencing-by-synthesis, sequencing-by-ligation, or ion semiconductor sequencing [31]. In oncology, NGS has become an indispensable tool for precision medicine. Targeted gene panels, which focus on clinically relevant cancer-associated genes, enable the detection of actionable mutations, guiding the selection of targeted therapies. For instance, mutations in EGFR, ALK, and BRAF can be identified through NGS, informing the use of tyrosine kinase inhibitors or other targeted agents. Furthermore, NGS has been instrumental in the identification of tumor mutational burden (TMB) and microsatellite instability (MSI), biomarkers that predict responsiveness to immunotherapy.
Despite its transformative potential, NGS faces challenges that require ongoing innovation. The vast amount of data generated necessitates robust bioinformatics tools and computational infrastructure for data analysis, storage, and interpretation. The accuracy of NGS can be affected by biases introduced during library preparation, sequencing, or alignment, necessitating quality control measures [32].
The Role of Epigenetics in Cancer
Epigenetics refers to heritable changes in gene expression that occur without alterations in the underlying DNA sequence. These changes are mediated by mechanisms such as DNA methylation, histone modification, and non-coding RNAs, which collectively regulate chromatin structure and gene activity [33].
In cancer, epigenetic dysregulation plays a pivotal role in tumor initiation, progression, and resistance to therapy. Unlike genetic mutations, epigenetic alterations are reversible, making them attractive targets for therapeutic intervention. Understanding the role of epigenetics in cancer biology provides critical insights into tumor heterogeneity, the tumor microenvironment, and potential strategies for precision medicine.
DNA methylation, the addition of a methyl group to the cytosine residues in CpG islands, is one of the most extensively studied epigenetic mechanisms in cancer. In normal cells, CpG islands in gene promoter regions are typically unmethylated, allowing for active transcription. However, in cancer cells, hypermethylation of tumor suppressor gene promoters, such as CDKN2A (encoding p16), leads to gene silencing and loss of cell cycle control. Conversely, global hypomethylation of the genome contributes to genomic instability and the activation of oncogenes. Aberrant DNA methylation patterns serve as biomarkers for cancer detection and prognosis, exemplified by the FDA-approved methylation-based test for MGMT promoter methylation in glioblastoma.
Histone modifications, including acetylation, methylation, phosphorylation, and ubiquitination, regulate chromatin accessibility and gene expression by altering the interaction between histones and DNA. Histone acetylation, catalyzed by histone acetyltransferases (HATs), generally promotes transcription by loosening chromatin structure, while histone deacetylation, mediated by histone deacetylases (HDACs), leads to transcriptional repression [34]. Dysregulated histone modification enzymes, such as overexpression of HDACs or mutations in histone methyltransferases, are frequently observed in cancer and contribute to the dysregulation of oncogenes and tumor suppressor genes. HDAC inhibitors, such as vorinostat and romidepsin, have shown efficacy in treating hematological malignancies, underscoring the therapeutic potential of targeting histone-modifying enzymes [35].
Non-coding RNAs, particularly microRNAs (miRNAs) and long non-coding RNAs (lncRNAs), also play crucial roles in the epigenetic regulation of cancer [36]. miRNAs are small, non-coding RNAs that post-transcriptionally regulate gene expression by binding to target mRNAs. Dysregulated miRNA expression in cancer can result in the suppression of tumor suppressor genes or the activation of oncogenes [37]. Epigenetic modifications are not only intrinsic to cancer cells but also influenced by the tumor microenvironment. Hypoxia, inflammation, and metabolic stress within the tumor microenvironment can induce epigenetic changes that promote tumor progression and immune evasion.
The Tumor Microenvironment and Genetics
The tumor microenvironment (TME) plays a crucial role in the development, progression, and therapeutic resistance of cancer. It is composed of various cell types, extracellular matrix (ECM) components, soluble factors, and blood vessels that interact dynamically with tumor cells. Understanding the intricate relationship between the TME and genetic alterations in tumor cells is essential for advancing cancer therapies and improving clinical outcomes [38]. The TME consists of multiple cellular components, including tumor-associated fibroblasts (CAFs), immune cells (such as macrophages, T cells, and dendritic cells), endothelial cells, adipocytes, and extracellular matrix components. These components interact with each other and the tumor cells in a highly complex and evolving manner, contributing to tumor growth, immune evasion, metastasis, and resistance to therapy. The tumor microenvironment and the genetic landscape of tumor cells are inextricably linked, with reciprocal interactions driving cancer progression, metastasis, and therapeutic resistance. Tumor genetic alterations not only enhance the malignant potential of tumor cells but also shape the composition and function of the TME, creating a supportive environment for tumor growth and immune evasion. Conversely, the TME exerts selective pressure on tumor cells, promoting genetic diversity and clonal evolution. Advances in understanding these complex interactions hold the potential for the development of novel therapeutic strategies that target both the tumor and its microenvironment, ultimately improving patient outcomes in cancer therapy.
Case Presentation
The intricate relationship between cancer and genetics underscores the profound impact of hereditary and acquired genetic variations in driving tumorigenesis. Advances in molecular genetics have revolutionized our understanding of the genetic basis of cancer, identifying key mutations, epigenetic changes, and pathways that fuel malignant transformation and progression. The identification of genetic predispositions, such as BRCA mutations in breast and ovarian cancer, has also paved the way for precision medicine, enabling targeted therapies and personalized treatment strategies. Despite these advancements, the genetic landscape of cancer remains vast and complex, with emerging challenges such as tumor heterogeneity and resistance to therapies highlighting the need for continuous exploration. Integrating genomic data with other fields, such as proteomics and epigenomics, holds promise for more comprehensive insights into cancer biology. Ultimately, a deeper understanding of the interplay between genetic factors and environmental influences will be crucial to unraveling the complexities of cancer. By translating these insights into clinical applications, we move closer to a future where cancer diagnosis, prevention, and treatment are guided by the unique genetic makeup of each patient, enhancing outcomes and improving quality of life.
References
-
Schwartz SM. Epidemiology of Cancer. Clin Chem. 2024;70(1):140-9.
-
Brothwell MRS, Barnett GC. Cancer Genetics and Genomics - Part 1. Clin Oncol (R Coll Radiol). 2022;34(6):e254-e61.
-
Yin W, Wang J, Jiang L, James Kang Y. Cancer and stem cells. Exp Biol Med (Maywood). 2021;246(16):1791-801.
-
Tran KA, Kondrashova O, Bradley A, Williams ED, Pearson JV, Waddell N. Deep learning in cancer diagnosis, prognosis and treatment selection. Genome Med. 2021;13(1):152.
-
Minciacchi VR, Kumar R, Krause DS. Chronic Myeloid Leukemia: A Model Disease of the Past, Present and Future. Cells. 2021;10(1):117.
-
Recillas-Targa F. Cancer Epigenetics: An Overview. Arch Med Res. 2022;53(8):732-40.
-
Baltanas FC, Santos E. Advances in Molecular Research of Oncogenes. Int J Mol Sci. 2023;24(8):7222.
-
Aryanti C, Uwuratuw JA, Labeda I, et al. The Mutation Portraits of Oncogenes and Tumor Supressor Genes in Predicting the Overall Survival in Pancreatic Cancer: A Bayesian Network Meta-Analysis. Asian Pac J Cancer Prev. 2023;24(8):2895-902.
-
Tufail M. DNA repair pathways in breast cancer: from mechanisms to clinical applications. Breast Cancer Res Treat. 2023;200(3):305-21.
-
Satam H, Joshi K, Mangrolia U, et al. Next-Generation Sequencing Technology: Current Trends and Advancements. Biology (Basel). 2023;12(7):997.
-
Johnson JA, Moore BJ, Syrnioti G, Eden CM, Wright D, Newman LA. Landmark Series: The Cancer Genome Atlas and the Study of Breast Cancer Disparities. Ann Surg Oncol. 2023;30(11):6427-40.
-
Ebrahimi N, Fardi E, Ghaderi H, et al. Receptor tyrosine kinase inhibitors in cancer. Cell Mol Life Sci. 2023;80(4):104.
-
Weinberg RA. Oncogenes and tumor suppressor genes. CA Cancer J Clin. 1994;44(3):160-70.
-
Aunoble B, Sanches R, Didier E, Bignon YJ. Major oncogenes and tumor suppressor genes involved in epithelial ovarian cancer (review). Int J Oncol. 2000;16(3):567-76.
-
Torry DS, Cooper GM. Proto-oncogenes in development and cancer. Am J Reprod Immunol. 1991;25(3):129-32.
-
Weiss RA. A perspective on the early days of RAS research. Cancer Metastasis Rev. 2020;39(4):1023-8.
-
Melo JV. The diversity of BCR-ABL fusion proteins and their relationship to leukemia phenotype. Blood. 1996;88(7):2375-84.
-
Liu Y, Hu X, Han C, et al. Targeting tumor suppressor genes for cancer therapy. Bioessays. 2015;37(12):1277-86.
-
Aubrey BJ, Strasser A, Kelly GL. Tumor-Suppressor Functions of the TP53 Pathway. Cold Spring Harb Perspect Med. 2016;6(5):026062.
-
Kontomanolis EN, Koutras A, Syllaios A, et al. Role of Oncogenes and Tumor- suppressor Genes in Carcinogenesis: A Review. Anticancer Res. 2020;40(11):6009- 15.
-
Zhang J, Walsh MF, Wu G, et al. Germline Mutations in Predisposition Genes in Pediatric Cancer. N Engl J Med. 2015;373(24):2336-46.
-
Chen M, Linstra R, van Vugt M. Genomic instability, inflammatory signaling and response to cancer immunotherapy. Biochim Biophys Acta Rev Cancer. 2022;1877(1):188661.
-
Chatterjee N, Walker GC. Mechanisms of DNA damage, repair, and mutagenesis. Environ Mol Mutagen. 2017;58(5):235-63.
-
Özcan O, Erdal H, Çakırca G, Yönden Z. Oxidative stress and its impacts on intracellular lipids, proteins and DNA. J Clin Exp Invest. 2015;6(3):331-6.
-
Pasqui A, Boddi A, Campanacci DA, et al. Alteration of the Nucleotide Excision Repair (NER) Pathway in Soft Tissue Sarcoma. Int J Mol Sci. 2022;23(15):8360.
-
Yoshida K, Miki Y. Role of BRCA1 and BRCA2 as regulators of DNA repair, transcription, and cell cycle in response to DNA damage. Cancer Sci. 2004;95(11):866-71.
-
Half E, Bercovich D, Rozen P. Familial adenomatous polyposis. Orphanet J Rare Dis. 2009;4:22.
-
Lal G, Gallinger S. Familial adenomatous polyposis. Semin Surg Oncol. 2000;18(4):314-23.
-
Keymling M, Schlemmer HP, Kratz C, et al. [Li-Fraumeni syndrome]. Radiologie (Heidelb). 2022;62(12):1026-32.
-
Hu T, Chitnis N, Monos D, Dinh A. Next-generation sequencing technologies: An overview. Hum Immunol. 2021;82(11):801-11.
-
Levy SE, Boone BE. Next-Generation Sequencing Strategies. Cold Spring Harb Perspect Med. 2019;9(7):a025791.
-
Le Gallo M, Lozy F, Bell DW. Next-Generation Sequencing. Adv Exp Med Biol. 2017;943:119-48.
-
Dawson MA, Kouzarides T. Cancer epigenetics: from mechanism to therapy. Cell. 2012;150(1):12-27.
-
Zaib S, Rana N, Khan I. Histone Modifications and their Role in Epigenetics of Cancer. Curr Med Chem. 2022;29(14):2399-411.
-
Audia JE, Campbell RM. Histone Modifications and Cancer. Cold Spring Harb Perspect Biol. 2016;8(4):a019521.
-
Hombach S, Kretz M. Non-coding RNAs: Classification, Biology and Functioning. Adv Exp Med Biol. 2016;937:3-17.
-
Lu TX, Rothenberg ME. MicroRNA. J Allergy Clin Immunol. 2018;141(4):1202- 7.
-
Xiao Y, Yu D. Tumor microenvironment as a therapeutic target in cancer. Pharmacol Ther. 2021;221:107753.
Declarations
Scientific Responsibility Statement
The authors declare that they are responsible for the article’s scientific content, including study design, data collection, analysis and interpretation, writing, and some of the main line, or all of the preparation and scientific review of the contents, and approval of the final version of the article.
Animal and Human Rights Statement
All procedures performed in this study were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards.
Funding
None
Conflict of Interest
The authors declare that there is no conflict of interest.
Data Availability
The datasets used and/or analyzed during the current study are not publicly available due to patient privacy reasons but are available from the corresponding author on reasonable request.
Additional Information
Publisher’s Note
Bayrakol MP remains neutral with regard to jurisdictional and institutional claims.
Rights and Permissions
About This Article
How to Cite This Article
Muge Sonmez, Huseyin Erdal. The interplay of genetics and cancer. Ann Clin Anal Med 2025; DOI: 10.4328/ACAM.22599
Publication History
- Received:
- February 8, 2025
- Accepted:
- March 10, 2025
- Published Online:
- March 19, 2025
