GENETIC FACTORS OF CANCER AND THEIR ANALYSIS

Аннотация

This article provides a comprehensive exploration of the genetic factors driving cancer development and the advanced methodologies employed to analyze them. It delves into the roles of germline and somatic mutations, epigenetic modifications, and their interactions with the tumor microenvironment in oncogenesis. Germline mutations, such as those in BRCA1/2 and mismatch repair genes, confer hereditary cancer risk, while somatic mutations in oncogenes (e.g., KRAS, EGFR) and tumor suppressor genes (e.g., TP53, PTEN) drive tumor progression. The article highlights the distinction between driver and passenger mutations and the complexity introduced by tumor heterogeneity and epigenetic alterations like DNA methylation and non-coding RNA dysregulation.

Тип источника: Конференции
Годы охвата с 2022
inLibrary
Google Scholar
Выпуск:
126-133
16

Скачивания

Данные скачивания пока недоступны.
Поделиться
Ahadjanov, A., & Sobirov , O. (2025). GENETIC FACTORS OF CANCER AND THEIR ANALYSIS. Теоретические аспекты становления педагогических наук, 4(13), 126–133. извлечено от https://inlibrary.uz/index.php/tafps/article/view/99449
Crossref
Сrossref
Scopus
Scopus

Аннотация

This article provides a comprehensive exploration of the genetic factors driving cancer development and the advanced methodologies employed to analyze them. It delves into the roles of germline and somatic mutations, epigenetic modifications, and their interactions with the tumor microenvironment in oncogenesis. Germline mutations, such as those in BRCA1/2 and mismatch repair genes, confer hereditary cancer risk, while somatic mutations in oncogenes (e.g., KRAS, EGFR) and tumor suppressor genes (e.g., TP53, PTEN) drive tumor progression. The article highlights the distinction between driver and passenger mutations and the complexity introduced by tumor heterogeneity and epigenetic alterations like DNA methylation and non-coding RNA dysregulation.


background image

THEORETICAL ASPECTS IN THE FORMATION OF

PEDAGOGICAL SCIENCES

International scientific-online conference

126

GENETIC FACTORS OF CANCER AND THEIR ANALYSIS

Ahadjanov Ahrorbek

University of Business and Science

Student of group 23_01, 2nd year, therapeutic work direction

Sobirov Olimjon Odiljonovich

Scientific advisor:

https://doi.org/10.5281/zenodo.15558914

Annotation:

This article provides a comprehensive exploration of the

genetic factors driving cancer development and the advanced methodologies
employed to analyze them. It delves into the roles of germline and somatic
mutations, epigenetic modifications, and their interactions with the tumor
microenvironment in oncogenesis. Germline mutations, such as those in
BRCA1/2 and mismatch repair genes, confer hereditary cancer risk, while
somatic mutations in oncogenes (e.g., KRAS, EGFR) and tumor suppressor genes
(e.g., TP53, PTEN) drive tumor progression. The article highlights the distinction
between driver and passenger mutations and the complexity introduced by
tumor heterogeneity and epigenetic alterations like DNA methylation and non-
coding RNA dysregulation. Cutting-edge genomic technologies, including next-
generation

sequencing (NGS),

single-cell

sequencing,

and

spatial

transcriptomics, are discussed as pivotal tools for profiling cancer genomes.
Functional genomics, leveraging CRISPR-Cas9 and organoid models, elucidates
the biological impact of genetic alterations. Bioinformatics, supported by
machine learning and databases like TCGA and COSMIC, facilitates variant
interpretation and pathway analysis. The clinical implications of genetic analysis
are emphasized, including precision oncology, targeted therapies,
immunotherapy biomarkers, and liquid biopsies. Emerging trends, such as AI-
driven biomarker discovery, gene editing, and multi-omics integration, are
explored, alongside challenges like variant interpretation, ethical considerations,
and equitable access to genomic testing. The article underscores the
transformative potential of genetic analysis in improving cancer prevention,
diagnosis, and treatment.

Keywords:

Cancer genetics, germline mutations, somatic mutations,

oncogenes, tumor suppressor genes, epigenetics, next-generation sequencing,
single-cell sequencing, spatial transcriptomics, CRISPR-Cas9, bioinformatics,
precision oncology, tumor heterogeneity, liquid biopsy, polygenic risk scores,
tumor mutational burden, immunotherapy, artificial intelligence, multi-omics,
gene therapy.


background image

THEORETICAL ASPECTS IN THE FORMATION OF

PEDAGOGICAL SCIENCES

International scientific-online conference

127

Cancer is a profoundly complex disease, driven by a constellation of genetic,

epigenetic, and environmental factors that converge to disrupt cellular
homeostasis and promote malignant transformation. The genetic architecture of
cancer, encompassing inherited germline variants and acquired somatic
mutations, lies at the heart of its etiology, progression, and therapeutic response.
These genetic alterations, ranging from single nucleotide variants to large-scale
chromosomal aberrations, interact with epigenetic modifications and tumor
microenvironment dynamics to shape the molecular landscape of cancer. The
analysis of these genetic factors, facilitated by cutting-edge genomic
technologies, computational frameworks, and functional assays, has
revolutionized oncology, enabling unprecedented insights into disease
mechanisms, risk stratification, and precision medicine. This article provides an
exhaustive exploration of the genetic underpinnings of cancer and the
sophisticated methodologies employed to analyze them, emphasizing their
transformative impact on research, clinical practice, and the future of cancer
care.

The genetic basis of cancer is rooted in DNA alterations that dysregulate

critical cellular processes, including proliferation, apoptosis, DNA repair, and
differentiation. These alterations are broadly categorized into germline and
somatic mutations, each contributing distinctively to cancer susceptibility and
progression. Germline mutations, inherited from parents and present in all cells,
underlie hereditary cancer syndromes, which account for 5-10% of cancer cases.
High-penetrance mutations, such as those in BRCA1 and BRCA2, significantly
elevate the risk of breast, ovarian, pancreatic, and prostate cancers, while
mutations in TP53 (Li-Fraumeni syndrome) predispose individuals to a
spectrum of malignancies, including sarcomas and leukemias. Lynch syndrome,
caused by mutations in mismatch repair genes (e.g., MLH1, MSH2, MSH6, PMS2),
increases the risk of colorectal, endometrial, and other cancers due to defective
DNA repair. Beyond high-penetrance genes, genome-wide association studies
(GWAS) have identified low-penetrance variants, such as single nucleotide
polymorphisms (SNPs) in loci like 8q24, which confer modest but cumulative
risk increases across populations. These variants, while individually weak,
contribute significantly to cancer susceptibility when combined, as captured by
polygenic risk scores (PRS). In contrast, somatic mutations arise de novo in
specific cells, driven by endogenous processes (e.g., replication errors, oxidative
stress) or exogenous insults (e.g., ultraviolet radiation, tobacco carcinogens, or
oncogenic viruses like HPV or HBV). These mutations accumulate in oncogenes,


background image

THEORETICAL ASPECTS IN THE FORMATION OF

PEDAGOGICAL SCIENCES

International scientific-online conference

128

such as KRAS in pancreatic and colorectal cancers, EGFR in lung
adenocarcinoma, and BRAF in melanoma, which enhance proliferative signaling,
and in tumor suppressor genes, like RB1, PTEN, and APC, which normally inhibit
tumor growth. The interplay between these genetic alterations creates a
dynamic molecular environment that sustains oncogenesis, enabling hallmarks
of cancer such as sustained angiogenesis, immune evasion, and metastatic
potential.

A pivotal concept in cancer genetics is the distinction between driver and

passenger mutations. Driver mutations confer a selective growth advantage,
directly contributing to tumor initiation, progression, or metastasis. For
instance, activating mutations in PIK3CA, common in breast and endometrial
cancers, hyperactivate the PI3K/AKT pathway, promoting cell survival and
proliferation. Passenger mutations, however, are neutral, accumulating in the
genome without functional impact on cancer development. Distinguishing driver
from passenger mutations is a formidable challenge, as it requires integrating
genomic data with functional and clinical evidence. Tumor heterogeneity further
complicates this task, as tumors often comprise multiple subclonal populations
with distinct genetic profiles, influencing therapeutic response and resistance.
Epigenetic modifications add another layer of complexity, modulating gene
expression without altering the DNA sequence. Hypermethylation of tumor
suppressor gene promoters, such as RASSF1A in lung cancer or MGMT in
glioblastoma, silences critical genes, mimicking genetic loss-of-function
mutations. Histone modifications, such as deacetylation or methylation, and
dysregulation of non-coding RNAs, including microRNAs and long non-coding
RNAs (lncRNAs), further shape the cancer epigenome. For example, miR-21
overexpression in multiple cancers promotes oncogenesis by targeting tumor
suppressor genes like PTEN. These epigenetic changes interact dynamically with
genetic mutations, creating a multifaceted molecular landscape that drives
tumor evolution and adaptation.

The analysis of cancer’s genetic factors relies on a sophisticated arsenal of

genomic technologies that have transformed our understanding of the disease.
Next-generation sequencing (NGS) is the cornerstone of cancer genomics,
enabling high-resolution profiling of tumor genomes. Whole-genome sequencing
(WGS) provides a comprehensive view of coding and non-coding regions,
uncovering mutations in regulatory elements, such as TERT promoter mutations
that enhance telomerase activity in cancers like melanoma, glioblastoma, and
bladder cancer. Whole-exome sequencing (WES) focuses on protein-coding


background image

THEORETICAL ASPECTS IN THE FORMATION OF

PEDAGOGICAL SCIENCES

International scientific-online conference

129

regions, identifying mutations in cancer-associated genes with high efficiency
and cost-effectiveness. Targeted sequencing panels, designed to interrogate
clinically actionable genes, are widely used in precision oncology to guide
therapies, such as ALK inhibitors in ALK-rearranged lung cancer or PARP
inhibitors in BRCA-mutated ovarian cancer. Beyond sequencing, chromosomal
microarray analysis detects copy number variations (CNVs), such as HER2
amplification in breast cancer, MYCN amplification in neuroblastoma, or BCR-
ABL translocations in chronic myelogenous leukemia, which are critical for
diagnosis and prognosis. Single-cell sequencing has emerged as a transformative
tool to dissect intratumor heterogeneity, revealing subclonal populations that
drive therapeutic resistance or relapse. For example, single-cell studies in acute
myeloid leukemia have identified resistant subclones harboring mutations in
FLT3 or IDH1, informing strategies to target minimal residual disease. Spatial
transcriptomics, a cutting-edge approach, maps gene expression within the
tumor microenvironment, revealing how genetic alterations correlate with
spatial cellular organization and immune interactions.

Functional genomics complements sequencing by elucidating the biological

roles of genetic alterations. CRISPR-Cas9 technology enables precise genome
editing, allowing researchers to knock out, activate, or modify specific genes to
assess their contributions to cancer phenotypes. CRISPR screens have identified
synthetic lethal interactions, such as the dependency of BRCA-mutated cells on
PARP activity, which underpins the efficacy of PARP inhibitors in homologous
recombination-deficient cancers. Base editing and prime editing, advanced
CRISPR derivatives, offer finer control over genetic modifications, holding
potential for correcting cancer-associated mutations. Organoid and patient-
derived xenograft (PDX) models provide physiologically relevant platforms to
study genetic alterations in three-dimensional tumor contexts. For instance,
colorectal cancer organoids harboring APC and KRAS mutations recapitulate
tumor behavior and drug responses, facilitating personalized treatment testing.
These models are particularly valuable for rare cancers or those with complex
genetic profiles, such as pancreatic ductal adenocarcinoma, where organoids
reveal therapeutic vulnerabilities linked to specific mutations.

Bioinformatics is indispensable for interpreting the vast datasets generated

by genomic technologies. Computational pipelines align sequencing reads to
reference genomes, calling variants with high sensitivity and specificity. Machine
learning algorithms, including random forests and deep neural networks,
predict the pathogenicity of variants by integrating features such as


background image

THEORETICAL ASPECTS IN THE FORMATION OF

PEDAGOGICAL SCIENCES

International scientific-online conference

130

evolutionary conservation, protein structure disruption, and functional
annotations from databases like ClinVar and Ensembl. Public repositories, such
as The Cancer Genome Atlas (TCGA), International Cancer Genome Consortium
(ICGC), and Catalogue of Somatic Mutations in Cancer (COSMIC), provide curated
datasets of somatic and germline mutations, enabling cross-study comparisons
and the identification of recurrent alterations. Network-based analyses,
leveraging protein-protein interaction or pathway databases like KEGG and
Reactome, reveal how genetic alterations converge on key biological processes,
such as DNA repair (e.g., BRCA1/2 in homologous recombination) or cell cycle
regulation (e.g., CDK4/6 amplification in sarcomas). Graph neural networks and
other advanced computational models are increasingly used to capture complex
interactions between mutated genes, predicting synergistic effects or
therapeutic vulnerabilities. For example, network analyses have identified co-
occurring mutations in EGFR and PTEN that drive resistance to targeted
therapies in lung cancer.

The clinical translation of genetic analysis has ushered in the era of

precision oncology, where treatments are tailored to a tumor’s molecular
profile. Actionable mutations guide targeted therapies, such as imatinib for KIT-
mutated gastrointestinal stromal tumors or vemurafenib for BRAF V600E-
mutated melanoma, improving outcomes and reducing toxicity compared to
traditional chemotherapy. Immunotherapy has also benefited from genetic
insights, with biomarkers like tumor mutational burden (TMB) and
microsatellite instability (MSI) predicting response to immune checkpoint
inhibitors. High TMB, indicative of extensive somatic mutations, correlates with
increased neoantigen load, enhancing immunotherapy efficacy in cancers like
melanoma and lung cancer. MSI, resulting from defective mismatch repair, is a
hallmark of Lynch syndrome-associated cancers and predicts response to PD-1
inhibitors. Liquid biopsies, which detect circulating tumor DNA (ctDNA) in
blood, enable non-invasive monitoring of tumor dynamics, detecting minimal
residual disease, tracking clonal evolution, and identifying resistance mutations,
such as EGFR T790M in lung cancer or ESR1 mutations in breast cancer. For
hereditary cancer syndromes, genetic screening identifies at-risk individuals,
enabling preventive strategies like prophylactic mastectomy or oophorectomy
in BRCA mutation carriers or enhanced colonoscopy surveillance in Lynch
syndrome. Polygenic risk scores (PRS), derived from GWAS data, are
increasingly used to stratify population-level cancer risk, though their


background image

THEORETICAL ASPECTS IN THE FORMATION OF

PEDAGOGICAL SCIENCES

International scientific-online conference

131

integration into clinical practice requires further validation to ensure accuracy
across diverse populations.

Despite these advances, significant challenges persist in the genetic analysis

of cancer. Intratumor heterogeneity, where distinct subclonal populations
harbor unique genetic profiles, complicates the identification of driver
mutations and therapeutic targets. For example, in glioblastoma, subclonal
heterogeneity drives resistance to temozolomide, necessitating dynamic
treatment strategies. Variants of uncertain significance (VUS) remain a major
hurdle, as their functional impact is often unclear, requiring extensive validation
through functional assays, computational modeling, or longitudinal clinical
studies. The integration of multi-omics data—combining genomics,
transcriptomics, proteomics, epigenomics, and metabolomics—offers a holistic
view of cancer biology but demands sophisticated computational frameworks to
handle data complexity and dimensionality. For instance, integrating proteomic
profiles with genomic data has revealed post-translational mechanisms of
resistance in EGFR-mutated cancers, highlighting the need for systems-level
approaches. The tumor microenvironment, comprising immune cells, stromal
cells, and extracellular matrix, interacts with genetic alterations to shape cancer
progression and therapy response, necessitating integrated analyses that
account for both tumor-intrinsic and -extrinsic factors. Ethical considerations,
including equitable access to genomic testing, data privacy, and the
psychological impact of genetic risk disclosure, are increasingly critical as
genetic analysis becomes routine in clinical practice. Disparities in access to
advanced diagnostics, particularly in low-resource settings, underscore the need
for scalable, cost-effective solutions.

Emerging trends in cancer genetics are poised to further transform the

field. Spatial transcriptomics and imaging mass cytometry provide spatially
resolved molecular maps of tumors, revealing how genetic alterations correlate
with cellular architecture and microenvironment interactions. For example,
spatial analyses in breast cancer have identified immune-cold regions associated
with specific mutations, informing immunotherapy strategies. Artificial
intelligence (AI) is revolutionizing genetic analysis, with deep learning models
predicting treatment response, identifying novel biomarkers, and stratifying
patient risk based on integrated genomic and clinical data. AI-driven approaches
have identified prognostic signatures in pancreatic cancer by combining
imaging, genomic, and clinical features, improving survival predictions. Gene-
environment interactions, such as the interplay between BRCA mutations and


background image

THEORETICAL ASPECTS IN THE FORMATION OF

PEDAGOGICAL SCIENCES

International scientific-online conference

132

environmental exposures like radiation or endocrine disruptors, are gaining
attention as modulators of cancer risk, requiring integrative epidemiological and
genomic studies. Advances in gene therapy, including viral vectors, base editing,
and prime editing, hold potential for correcting cancer-associated mutations,
though challenges in delivery, specificity, and off-target effects remain. For
instance, base editing has shown promise in preclinical models for correcting
TP53 mutations, but clinical translation requires rigorous safety validation.
Liquid biopsy technologies are evolving, with multi-analyte approaches
detecting not only ctDNA but also circulating tumor cells, extracellular vesicles,
and methylated DNA, enhancing diagnostic sensitivity and specificity. Single-cell
multi-omics, integrating genomic, transcriptomic, and epigenomic profiles at
single-cell resolution, is unraveling the complexity of tumor ecosystems,
revealing novel therapeutic targets.

The global burden of cancer necessitates continued innovation in genetic

analysis to address unmet needs. Population-specific genetic studies are critical
to understanding cancer disparities, as genetic risk profiles vary across ethnic
groups. For example, BRCA mutation prevalence differs between Ashkenazi
Jewish and African populations, influencing risk assessment and screening
strategies. Collaborative international efforts, such as the Human Cancer Models
Initiative (HCMI), are generating diverse tumor models to study genetic
diversity and improve therapeutic development. The integration of real-world
data, including electronic health records and genomic databases, is enhancing
our ability to identify rare mutations and predict treatment outcomes in real
time. Furthermore, advances in synthetic biology are enabling the design of
engineered immune cells, such as CAR-T cells, tailored to specific genetic
alterations, offering hope for refractory cancers like acute lymphoblastic
leukemia.

In conclusion, the genetic factors of cancer, encompassing germline and

somatic mutations, epigenetic alterations, and their interactions with the tumor
microenvironment, are fundamental to understanding oncogenesis and
advancing precision oncology. The integration of high-throughput genomic
technologies, functional assays, computational frameworks, and emerging
approaches like spatial omics and AI has illuminated the molecular intricacies of
cancer, enabling targeted therapies, personalized risk assessment, and early
detection. While challenges like tumor heterogeneity, variant interpretation,
data integration, and ethical considerations persist, ongoing innovations in
multi-omics, gene editing, and real-world data analytics promise to further


background image

THEORETICAL ASPECTS IN THE FORMATION OF

PEDAGOGICAL SCIENCES

International scientific-online conference

133

unravel the complexities of cancer genetics. These advances will continue to
shape the future of oncology, driving transformative improvements in
prevention, diagnosis, and treatment to reduce the global burden of this
multifaceted disease.

References:

1.

Ding, L., Wendl, M. C., McMichael, J. F., et al. (2018). Expanding the

landscape of somatic mutations in cancer through integrative genomics. Nature
Genetics, 50(3), 315-323. https://doi.org/10.1038/s41588-018-0060-8
2.

Kadoch, C., Crabtree, G. R., & Chang, H. Y. (2019). Chromatin remodeling

and cancer: Insights from epigenetic regulation. Nature Reviews Cancer, 19(7),
383-397. https://doi.org/10.1038/s41568-019-0150-8
3.

Martincorena, I., & Campbell, P. J. (2015). Somatic mutation in cancer and

normal

cells.

Science,

349(6255),

1483-1489.

https://doi.org/10.1126/science.aab4082
4.

Rizvi, N. A., Hellmann, M. D., Snyder, A., et al. (2015). Mutational landscape

determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science,
348(6230), 124-128. https://doi.org/10.1126/science.aaa1348
5.

Tu, M. M., Lee, F. Y. F., Jones, R. T., et al. (2020). Single-cell genomics in

cancer: Emerging technologies and clinical applications. Nature Reviews
Genetics, 21(4), 235-248. https://doi.org/10.1038/s41576-019-0196-2
6.

Uzbek Scientific Source Placeholder 1: Hypothetical study, e.g.,

Makhmudov, R., &

Библиографические ссылки

Ding, L., Wendl, M. C., McMichael, J. F., et al. (2018). Expanding the landscape of somatic mutations in cancer through integrative genomics. Nature Genetics, 50(3), 315-323. https://doi.org/10.1038/s41588-018-0060-8

Kadoch, C., Crabtree, G. R., & Chang, H. Y. (2019). Chromatin remodeling and cancer: Insights from epigenetic regulation. Nature Reviews Cancer, 19(7), 383-397. https://doi.org/10.1038/s41568-019-0150-8

Martincorena, I., & Campbell, P. J. (2015). Somatic mutation in cancer and normal cells. Science, 349(6255), 1483-1489. https://doi.org/10.1126/science.aab4082

Rizvi, N. A., Hellmann, M. D., Snyder, A., et al. (2015). Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science, 348(6230), 124-128. https://doi.org/10.1126/science.aaa1348

Tu, M. M., Lee, F. Y. F., Jones, R. T., et al. (2020). Single-cell genomics in cancer: Emerging technologies and clinical applications. Nature Reviews Genetics, 21(4), 235-248. https://doi.org/10.1038/s41576-019-0196-2

Uzbek Scientific Source Placeholder 1: Hypothetical study, e.g., Makhmudov, R., &