Fact-checked by Grok 2 weeks ago

Oncogene

An oncogene is a mutated or altered form of a normal cellular , known as a proto-oncogene, that drives uncontrolled and contributes to the development of cancer by promoting . Proto-oncogenes typically encode proteins involved in essential cellular processes such as growth signaling, progression, and differentiation, but when activated abnormally, they function like a "stuck accelerator" in the cell's regulatory machinery. This activation often results from genetic changes including point , chromosomal translocations, gene amplifications, or insertions, leading to overexpression or constitutive activity of the encoded protein. The concept of oncogenes emerged from studies of tumor-causing viruses in the 1970s, with key discoveries revealing that viral oncogenes like in originated from captured cellular proto-oncogenes. Pioneering work by researchers such as Harold Varmus and J. Michael Bishop demonstrated that these proto-oncogenes exist in normal cells and can be converted to oncogenes through mutations, earning them the 1989 Nobel Prize in Physiology or Medicine. In human cancers, oncogenes play a central role in tumorigenesis by disrupting signaling pathways—such as the RAS/MAPK pathway—that regulate cell division, survival, and inhibition. Unlike tumor suppressor genes, which act as "brakes" on cell growth and require loss-of-function mutations to contribute to cancer, oncogenes exhibit gain-of-function alterations that actively promote tumor initiation, progression, and . Notable examples include the family of oncogenes, which harbor point mutations in approximately 20-30% of human cancers, particularly in pancreatic, colorectal, and lung malignancies, leading to persistent activation of downstream growth signals. The oncogene, often amplified or translocated as in Burkitt's lymphoma, regulates transcription to accelerate entry and inhibit . Similarly, amplification of HER2 (also known as ERBB2) occurs in about 20% of cancers, enhancing signaling for tumor growth. The BCR-ABL fusion oncogene, resulting from the translocation, drives by constitutively activating activity. Oncogenes frequently cooperate with inactivated tumor suppressors like or RB1 to fully unleash carcinogenic potential, underscoring their role in the multi-hit model of cancer development.

Fundamentals

Definition

An oncogene is a mutated or overexpressed form of a proto-oncogene (a normal that regulates and division), resulting in uncontrolled and tumor formation. These genes arise from alterations in normal cellular genes known as proto-oncogenes, which typically regulate and division; when mutated, oncogenes drive uncontrolled cellular activity that disrupts normal tissue . At the cellular level, oncogenes act in a dominant manner, meaning that alteration of only one is sufficient to promote oncogenic effects, in contrast to tumor suppressor genes, which require inactivation of both alleles to lose their protective function. This dominant behavior stems from the enhanced or hyperactive protein products they encode, which override normal regulatory signals to stimulate proliferation even in the presence of a wild-type allele. Well-known examples include the oncogene, which encodes a involved in pathways, and the oncogene, a that regulates progression and ; both are frequently mutated or amplified across various cancers. Oncogenes were first identified as viral oncogenes (v-onc) in retroviruses capable of inducing tumors in animals, such as the src gene in .

Relation to Proto-oncogenes

Proto-oncogenes, often denoted as c-onc genes, are normal cellular genes that encode proteins essential for regulating , , division, and in eukaryotic cells. These genes are ubiquitous across and as positive regulators of cellular processes under physiological conditions. When altered—typically through gain-of-function mutations, amplification, or dysregulation—they convert into oncogenes, which promote uncontrolled and contribute to tumorigenesis. In their unmutated form, proto-oncogenes are integral to key cellular pathways, including , where they relay extracellular signals to intracellular responses; transcriptional regulation, influencing that drives progression; and apoptosis control, balancing cell survival and death to maintain tissue . For instance, the proto-oncogene EGFR encodes the , a transmembrane that, upon binding, activates downstream cascades like the RAS-MAPK pathway to stimulate and survival in response to growth factors. Similarly, c-MYC regulates transcription of genes involved in and , while BCL2 modulates to prevent unnecessary cell loss during development or stress. These roles ensure coordinated cellular responses to environmental cues, preventing aberrant growth in healthy tissues. Proto-oncogenes are highly conserved across diverse species, from to mammals, reflecting their essential roles in eukaryotic over hundreds of millions of years. Their viral counterparts (v-onc) originated from ancient retroviral capture and of these cellular genes into viral genomes, a process that occurred millions of years ago and facilitated the discovery of proto-oncogenes through studies of tumor-inducing viruses. The high degree of sequence and functional conservation, as seen in homologs like c-src in vertebrates derived from viral v-src, highlights their indispensable contributions to development and . The transformation of a proto-oncogene into an oncogene requires only a single genetic alteration—the "single-hit" model—due to the gain-of-function nature of these mutations, which can dominantly override normal regulatory controls. This contrasts with tumor suppressor genes, as even heterozygous mutations in proto-oncogenes can lead to or hyperactivity, providing a selective advantage for clonal expansion in early . For example, a in KRAS can lock the protein in an active GTP-bound state, sufficient to drive pancreatic tumorigenesis without needing a second hit.

Historical Development

Early Discoveries

The discovery of oncogenes began with the pioneering work of Peyton Rous in 1911, when he identified a filterable agent capable of transmitting sarcomas in chickens, later named the (RSV). This finding provided the first evidence that viruses could cause cancer, challenging prevailing views that tumors arose solely from cellular mutations or environmental factors. Rous's experiments demonstrated that cell-free extracts from the tumors could induce malignancy in healthy birds, establishing the concept of viral oncogenesis. For this breakthrough, Rous was awarded the in or in 1966. In the 1970s, research on led to the identification of the gene as the first recognized oncogene, marking a pivotal advancement in understanding cancer's genetic basis. J. Michael and demonstrated that the viral (v-src) gene was responsible for 's transforming ability and originated from a cellular gene in the host . Their work revealed that acute transforming retroviruses, such as , acquire oncogenes (v-onc genes) through from host cellular sequences during viral replication, enabling rapid tumor induction. This discovery earned and Varmus the in Physiology or Medicine in 1989. The realization that v-onc genes derived from cellular counterparts, termed proto-oncogenes or c-onc genes, shifted the paradigm from purely viral causation to the role of endogenous genes in oncogenesis. Studies showed that these normal cellular genes, present in all multicellular organisms, regulate and but can become oncogenic when altered, as seen in viral . This insight laid the groundwork for molecular by highlighting how viruses exploit host to drive cancer.

Key Experiments

In the late 1970s, building briefly on earlier insights from viral oncogene research, key experiments shifted focus to non-viral contexts by demonstrating that DNA from transformed mammalian cells could transfer oncogenic properties to normal cells via . A seminal study in by Robert Weinberg and colleagues used calcium phosphate-mediated DNA to introduce genomic DNA from chemically transformed mouse and rat cells into NIH 3T3 mouse fibroblasts, resulting in the formation of transformed foci—piles of multilayered, anchorage-independent cells indicative of oncogenic activity. This assay confirmed that transforming sequences were present in the DNA of non-virally transformed cells and could be propagated through multiple rounds of , establishing a functional test for oncogenes without relying on viral vectors. This transfection approach was rapidly extended to human tumor DNA in the early 1980s, providing direct evidence of human oncogenes. In 1981, Weinberg's group transfected DNA from the human bladder carcinoma cell line EJ into NIH 3T3 cells, observing focus formation and tumorigenicity in nude mice, which indicated the presence of an activated transforming gene distinct from known viral oncogenes. Similar results were reported independently by groups led by Michael Wigler and Mariano Barbacid using DNA from various human carcinomas, including lung and bladder tumors, further validating the method's sensitivity in detecting rare transforming sequences (occurring at frequencies of about 10^{-5} to 10^{-6}). These experiments not only isolated the first human oncogene—a mutated RAS homolog—but also highlighted the assay's utility in pinpointing dominant, gain-of-function genetic alterations driving cellular transformation. Throughout the , focus-forming assays in fibroblasts became central to identifying and characterizing specific oncogenes, particularly the family. of tumor DNA into NIH 3T3 or Rat-1 fibroblasts produced quantifiable foci of transformed cells, allowing researchers to map transforming activity to specific genomic fragments; for instance, Barbacid's group used this to clone the activated oncogene from human tumors, demonstrating its homology to sarcoma genes while confirming its role in morphological and soft . These assays revealed that activation conferred a advantage in low-serum conditions, with transformed foci exhibiting significantly enhanced proliferation compared to untransfected controls, underscoring RAS's potent oncogenic potential in mammalian systems. Hybridization studies using Southern blotting provided complementary evidence for the existence of cellular oncogene (c-onc) homologs in normal genomes, bridging viral and cellular discoveries. In the early , probes derived from viral oncogenes (e.g., v-ras or v-myc) were hybridized to restriction-digested DNA, revealing conserved sequences in non-tumorigenic cells; for example, Southern blots showed single-copy c-ras loci on chromosomes, with tumor samples displaying altered restriction patterns indicative of mutations or amplifications. This technique, applied to diverse tissues, confirmed that proto-oncogenes were ubiquitous in the , present at one or two copies per haploid , and laid the groundwork for detecting structural changes in cancer. Key milestones from these efforts included the 1982 cloning of the human c-myc proto-oncogene from cells, where Southern blotting and library screening identified its translocation to the locus on chromosome 14, juxtaposing it with enhancer elements to drive overexpression. Concurrently, evidence emerged for point mutations as a mechanism of oncogene activation; sequencing of the gene from the EJ bladder carcinoma revealed a G-to-A transition at codon 12, substituting with and locking the protein in its GTP-bound, active state, which was sufficient to transform NIH at efficiencies comparable to viral . These findings solidified point mutations as a common activation route for proto-oncogenes in sporadic human cancers.

Activation Mechanisms

Genetic Mutations

Genetic mutations in oncogenes primarily involve point mutations, which are single nucleotide alterations in the DNA sequence of proto-oncogenes that result in their conversion to constitutively active oncogenes. These mutations often occur at critical codons, leading to proteins that promote uncontrolled cell proliferation by disrupting normal regulatory mechanisms. A common type is the missense mutation, where a single amino acid substitution alters the protein's function, frequently locking enzymes in an active state. For instance, the G12V mutation in the KRAS proto-oncogene replaces glycine with valine at codon 12, impairing the GTPase activity required for signal termination and causing persistent activation of downstream pathways like MAPK/ERK. This exemplifies how such mutations in GTPases lead to constitutive signaling, a hallmark of oncogenic transformation. Similarly, in the BRAF proto-oncogene, the substitutes with at codon 600, resulting in domain activation independent of upstream signals and driving melanomagenesis through hyperactive RAF-MEK-ERK signaling. This is particularly prevalent in , underscoring its role in tissue-specific oncogenesis. Mutations in the RAS family (KRAS, NRAS, HRAS) are found in approximately 19% of all human cancers, with higher frequencies in solid tumors such as pancreatic (over 90% KRAS), colorectal (about 40% KRAS), and lung adenocarcinomas (around 30% KRAS). These alterations typically impair GTP hydrolysis, maintaining the protein in its GTP-bound active form and facilitating tumor initiation and progression. Detection of these point mutations relies on next-generation sequencing (NGS) techniques, which enable high-throughput identification of single nucleotide variants in tumor DNA, often through targeted panels focusing on hotspot regions in known oncogenes. This approach has become standard for precise diagnosis and guiding targeted therapies, such as inhibitors for specific mutants like BRAF V600E.

Amplification and Rearrangement

Gene amplification represents a key mechanism for oncogene , wherein multiple tandem copies of a proto-oncogene are generated within the , leading to elevated expression levels of the encoded protein and heightened signaling activity without any alteration to the gene's coding sequence. This process increases the effective , often resulting in aggressive tumor phenotypes. A well-characterized instance is the amplification of the HER2 (ERBB2) gene in , which occurs in 20–30% of cases and drives overexpression of the HER2 , promoting and while correlating with poorer and shorter relapse-free . Another critical example involves the MYCN oncogene in , where amplification is detected in approximately 20–25% of tumors, particularly those classified as high-risk, and serves as an independent predictor of adverse outcomes by enhancing MYCN-mediated transcription of genes involved in and survival. This amplification fosters rapid tumor progression and resistance to therapy, underscoring its role as a driver of aggressive pediatric malignancies. Chromosomal rearrangements, including translocations, activate oncogenes by juxtaposing proto-oncogene sequences with potent regulatory elements or by generating chimeric proteins with deregulated function. The paradigmatic case is the t(9;22) translocation, known as the , which fuses the BCR and ABL1 genes to produce the BCR-ABL oncoprotein in over 95% of chronic (CML) patients. This fusion imparts constitutive kinase activity to ABL1, disrupting normal cellular signaling, inhibiting , and enabling uncontrolled , thereby initiating and sustaining the leukemic state. Insertional mutagenesis provides another route for oncogene activation, particularly through viral integration, where the viral genome inserts adjacent to a proto-oncogene, co-opting viral (LTR) promoters or enhancers to drive . Retroviruses exemplify this mechanism; for instance, avian leukosis virus integration near the c-myc proto-oncogene in chickens leads to its overexpression, transforming B-lymphocytes and inducing lymphomas by amplifying MYC-dependent proliferative signals. Such events highlight how structural genomic disruptions can enhance oncogene signaling independently of sequence mutations, contributing to oncogenesis across diverse contexts.

Regulatory Changes

Regulatory changes in oncogene activation encompass epigenetic and transcriptional mechanisms that dysregulate gene expression without altering the underlying DNA sequence, thereby promoting overexpression or aberrant activity in cancer cells. These alterations often involve modifications to chromatin structure, enhancer-promoter interactions, and post-transcriptional processing, which can silence repressive elements or enhance activating signals to drive tumorigenesis. Promoter hypomethylation represents a key epigenetic mechanism where reduced at CpG islands in oncogene promoters leads to their transcriptional derepression and overexpression. For instance, hypomethylation of in B-cell (CLL) results in elevated levels, contributing to apoptotic resistance and disease progression. Hypermethylation in specific regulatory regions can also activate oncogenes by relieving repression. For instance, hypermethylation of the (TERT) promoter in the telomerase hypermethylated oncological region (THOR) activates TERT expression, contributing to replicative immortality observed in approximately 90% of human cancers overall, including those of the , , and . Enhancer hijacking occurs when chromosomal translocations reposition oncogenes adjacent to potent enhancers, thereby placing them under the control of constitutively active regulatory elements and causing their inappropriate overexpression. A prominent example is the t(8;14) translocation in , which juxtaposes the oncogene with (IGH) enhancers, leading to MYC deregulation and lymphomagenesis through enhanced transcriptional activation. This mechanism exemplifies how structural rearrangements can exploit distant regulatory landscapes to fuel oncogene activation in hematopoietic malignancies. MicroRNA (miRNA) dysregulation contributes to oncogene activation by downregulating miRNAs that normally repress target oncogenes at the post-transcriptional level, resulting in unchecked protein expression. In CLL, deletion or downregulation of the miR-15/16 cluster, located at 13q14, relieves inhibition of BCL2 translation, leading to BCL2 overexpression and impaired , which is a hallmark of the disease. This miRNA-oncogene axis highlights the role of non-coding RNAs in fine-tuning oncogene activity during cancer initiation. Post-transcriptional changes, particularly altered mRNA splicing, generate oncogenic isoforms that promote tumor progression by evading normal regulatory controls. For example, in various cancers including and colon, alternative splicing of the PKM gene favors the isoform over PKM1, enhancing aerobic (the effect) and supporting rapid through regulation by splicing factors like hnRNPA1 and PTBP1. Likewise, splicing of BCL2L1 to produce the anti-apoptotic isoform predominates in many tumors, inhibiting and driven by elevated PTBP1 expression. These isoform switches underscore the splicing machinery's vulnerability in oncogenic transformation.

Classification

By Protein Function

Oncogenes are classified by the biochemical functions of their encoded proteins, which typically correspond to key steps in cellular growth and pathways. This functional categorization highlights how dysregulated proteins drive oncogenesis by mimicking or amplifying normal signaling. Common groups include , receptors, intracellular signal transducers, nuclear transcription factors, and regulators, with additional roles in anti-apoptotic processes. Growth factor oncogenes encode proteins that stimulate by binding to and activating their cognate receptors. A representative example is PDGFB, which encodes beta and promotes in gliomas, leading to uncontrolled glial . Activation of such oncogenes often occurs through or overexpression, enabling persistent mitogenic stimulation independent of external cues. Growth factor receptor oncogenes primarily involve receptor tyrosine kinases (RTKs) that transduce extracellular signals into intracellular responses. (epidermal growth factor receptor), for instance, is frequently activated in various carcinomas through mutations or amplifications, resulting in ligand-independent dimerization and downstream signaling that enhances cell survival and motility. Similarly, ERBB2 (also known as HER2) functions as an RTK that amplifies signaling from other family members; its overexpression, often due to , potently drives proliferation in breast and ovarian cancers by forming constitutive heterodimers. Intracellular signaling oncogenes mediate the relay of signals from receptors to downstream effectors, often through kinase cascades or lipid second messengers. Non-receptor tyrosine kinases like propagate signals by phosphorylating multiple substrates, thereby integrating inputs with cytoskeletal reorganization and metabolic changes; activation, typically via or , contributes to invasive phenotypes. G-proteins such as exemplify oncogenes that lock in an active state due to point mutations, constitutively activating pathways like MAPK and sustaining proliferation signals. The PI3K/AKT pathway represents another critical transducer group, where oncogenic PIK3CA s hyperactivate kinase activity, leading to AKT-mediated promotion of survival and nutrient uptake. Transcription factor oncogenes directly regulate gene expression to enforce proliferative programs. MYC, a basic helix-loop-helix leucine zipper protein, drives the transcription of genes involved in biomass accumulation and cell cycle entry; its deregulation through translocation or amplification broadly reprograms cellular metabolism toward growth. JUN, part of the AP-1 complex, similarly activates promoters for proliferation and angiogenesis genes; oncogenic forms, often from amplification, enhance survival under stress by modulating immediate early response elements. Cell cycle regulator oncogenes, such as (CCND1), facilitate progression through G1/S checkpoints by activating cyclin-dependent kinases (CDKs). Overexpression of CCND1 sequesters CDK inhibitors, promoting hyperphosphorylation of and E2F release, which commits cells to ; this is commonly seen in endocrine-responsive tumors via chromosomal rearrangements. Anti-apoptotic oncogenes counteract , allowing survival of damaged cells. The , particularly itself, inhibits mitochondrial outer membrane permeabilization by sequestering pro-apoptotic members like BAX and BAK; translocation-induced overexpression, as in , confers resistance to and clonal expansion. Other family members, such as , similarly tilt the balance toward survival in response to oncogenic stress.

By Cellular Location

Oncogenes can be classified based on the subcellular location where their protein products primarily function, as this localization influences their role in oncogenic signaling and cellular transformation. This spatial organization determines how oncogenes interact with cellular components, such as membranes for receptor-mediated signaling or the nucleus for transcriptional regulation, often overlapping with functional categories like receptors or transcription factors. Membrane-bound oncogenes typically encode s or membrane-associated proteins that initiate signaling cascades upon ligand binding or mutation, driving uncontrolled cell proliferation. For instance, the MET proto-oncogene product is a transmembrane overexpressed in , where it promotes invasive growth through hepatocyte growth factor stimulation. Other examples include RAS family members like H-RAS and K-RAS, which are anchored to the inner plasma membrane and act as in . Cytoplasmic oncogenes often produce signaling proteins that relay messages from the membrane to downstream effectors, amplifying mitogenic signals within the cell. RAF kinases, such as BRAF, are serine/threonine kinases located in the that activate the MAPK/ERK pathway in response to RAS activation, with mutations frequently observed in melanomas and other cancers. Additional cytoplasmic examples include and ABL tyrosine kinases, which phosphorylate targets to enhance cell motility and survival. Nuclear oncogenes encode transcription factors that directly regulate gene expression, leading to the promotion of cell cycle progression and inhibition of differentiation. The FOS proto-oncogene product forms part of the AP-1 transcription factor complex in the nucleus, where it heterodimerizes with JUN to activate genes involved in proliferation, as seen in osteosarcomas induced by v-fos. MYC and MYB are other nuclear examples, binding DNA to drive oncogenic transcription programs. Mitochondrial oncogenes are less common but play critical roles in modulating and cellular metabolism at the organelle's membranes. , an anti-apoptotic proto-oncogene, localizes to the mitochondrial outer membrane, where it prevents release and inhibits , contributing to survival. Variants or related proteins like AKT can also localize mitochondrially to suppress through calcium regulation. Secreted oncogenes produce growth factors that act in an autocrine or paracrine manner to stimulate nearby cells, fostering support. FGF3, a member of the family, is secreted and binds FGFRs to promote and , with amplification observed in breast cancers. The v-sis product, homologous to PDGF-B, exemplifies this by enabling ligand-independent receptor activation.

Role in Cancer

Oncogenic Pathways

Oncogenic pathways represent critical signaling cascades that, when dysregulated by oncogenes, drive uncontrolled , survival, and other hallmarks of tumorigenesis. These pathways often originate from developmental or signaling networks that become aberrantly activated through genetic alterations in oncogenes such as mutated or amplified receptor kinases. Central to this process is the -RAF-MEK-ERK pathway, a (MAPK) cascade that transduces signals from extracellular stimuli to the nucleus, promoting progression and proliferation. Upon activation by oncogenic mutations, which occur in approximately 30% of human cancers but lead to pathway hyperactivity in up to 50% through various mechanisms including upstream receptor amplification, RAF kinases phosphorylate and activate MEK1/2, which in turn phosphorylate ERK1/2; this culminates in the transcription of genes like c-MYC and that sustain tumor growth. Another major oncogenic pathway is the PI3K-AKT-mTOR axis, which regulates , , and protein synthesis, often hijacked by oncogenes like PIK3CA mutations or loss of the tumor suppressor PTEN. Activation begins with PI3K phosphorylating PIP2 to PIP3, recruiting and activating AKT, which then inhibits pro-apoptotic proteins like FOXO and BAD while stimulating to enhance anabolic processes; PTEN loss, observed in 40-60% of endometrial cancers and 15-25% of cancers, removes negative regulation, leading to persistent signaling that confers resistance to and metabolic reprogramming favoring tumor expansion. The Wnt/β-catenin pathway further contributes to oncogenesis by maintaining stemness and promoting , particularly through stabilizing β-catenin via loss-of-function mutations in the tumor suppressor or gain-of-function mutations in CTNNB1 (β-catenin). In the pathway, Wnt ligand binding inhibits the destruction complex (including and GSK3β), allowing β-catenin to accumulate, translocate to the , and activate transcription factors like TCF/LEF that upregulate genes such as AXIN2 and LEF1, fostering cancer properties and epithelial-mesenchymal transition () for metastatic potential. Developmental pathways like and are also frequently co-opted by oncogenes in cancer, amplifying proliferative and differentiative signals. The pathway, activated by ligands such as or Delta-like, leads to proteolytic cleavage and release of the Notch intracellular domain (NICD), which translocates to the nucleus to co-activate transcription factors like RBPJ, promoting genes involved in fate decisions; oncogenic mutations or amplifications, as seen in T-ALL, hijack this for sustained proliferation and inhibition of differentiation. Similarly, the (Hh) pathway, driven by ligands like Sonic binding to Patched, relieves inhibition of , activating transcription factors that induce targets like PTCH1 and ; aberrant activation via loss or SMO mutations in exemplifies how this developmental cascade supports tumor initiation and maintenance. These pathways do not operate in isolation; extensive cross-talk amplifies oncogenic effects, such as signaling enhancing activity or PI3K-AKT intersecting with Wnt to boost β-catenin stability, creating robust networks that sustain tumorigenesis across diverse cancer types.

Multi-step Carcinogenesis

The development of cancer follows a multi-step process involving the sequential accumulation of genetic alterations, in which oncogenes contribute gain-of-function that drive tumor initiation and progression. This framework adapts Knudson's , originally proposed for tumor suppressor genes requiring biallelic inactivation, to incorporate oncogenes as dominant, single-hit events that provide initiating "gain" alterations complemented by suppressor losses. In this extended model, a single activating in an oncogene can confer proliferative advantages, but full tumorigenesis demands cooperation with additional hits, such as suppressor gene disruptions, to bypass cellular checkpoints. A seminal illustration of this sequential cooperation is the Vogelstein model of colorectal carcinogenesis, which delineates the orderly progression from normal colonic epithelium to and then to through specific genetic events. Inactivation of the represents an early hit that initiates small adenomas; this is followed by activating mutations in the oncogene, which enlarge adenomas and confer growth autonomy; subsequent loss of TP53 function then propels the transition to malignant by impairing DNA damage responses. Here, the oncogene exemplifies how a gain-of-function alteration integrates with suppressor losses to advance multistage tumor , with mutations accumulating in a temporal sequence that correlates with histopathological progression. Within this multi-hit paradigm, clonal evolution further amplifies oncogene roles by enabling Darwinian selection of aggressive tumor subpopulations. Tumors arise from a single and progress through rounds of genetic diversification and subclonal expansion, where oncogene activations—such as those enhancing or survival—impart selective advantages, favoring the dominance of fitter clones in the . This process, driven by oncogene-induced genetic , generates intratumor heterogeneity and promotes the emergence of variants with heightened , underscoring how oncogenes shape adaptive landscapes during . Achieving full often involves threshold effects, wherein multiple oncogene activations must accumulate alongside other genetic hits to surpass critical barriers to tumorigenesis. Computational analyses of somatic reveal that cancer typically requires 2–8 hits, with oncogenic combinations synergistically enabling by overwhelming regulatory networks; for example, single oncogene alone rarely suffice, but paired with complementary alterations, they achieve high toward tumor formation. These thresholds highlight the cooperative necessity of diverse oncogene engagements in sustaining the multistep trajectory to cancer.

Clinical Relevance

Detection and Diagnosis

Detection and diagnosis of oncogene alterations involve a range of molecular and histopathological techniques applied to tumor samples or circulating biomarkers to identify activating , amplifications, fusions, or overexpression that drive cancer progression. These methods enable precise identification of oncogenic drivers, facilitating cancer subtyping and personalized management. Common approaches include genomic sequencing for point and structural variants, cytogenetic assays for gene copy number changes, and protein-based assays for expression levels, often integrated into clinical workflows for solid tumors like , , and colorectal cancers. According to the (NCCN) guidelines as of 2025, comprehensive genomic profiling is recommended for advanced non-small cell (NSCLC), , and to detect actionable oncogene alterations. Next-generation sequencing (NGS) has become a cornerstone for detecting oncogene mutations, offering high-throughput analysis of tumor DNA to identify somatic alterations such as point mutations, insertions, deletions, and fusions. In non-small cell lung cancer (NSCLC), NGS panels routinely screen for EGFR mutations, including exon 19 deletions, which occur in approximately 10-15% of cases in Western populations (higher in Asian cohorts) and are actionable targets; for instance, targeted NGS achieves over 95% sensitivity and specificity compared to single-gene assays like PCR. Studies validate NGS for comprehensive profiling, detecting not only EGFR but also co-occurring variants in genes like KRAS and BRAF, guiding subtype classification in advanced NSCLC. Fluorescence in situ hybridization (FISH) is widely used to detect oncogene amplifications by visualizing gene copy number gains directly on chromosomes, particularly for genes like HER2 in , where amplification is present in 15-20% of cases and correlates with aggressive disease. (IHC) complements FISH by assessing protein overexpression, scoring HER2 on a 0-3+ scale; scores of 3+ indicate high expression, though concordance with FISH is about 90%, with discrepancies resolved by reflex testing. In diagnostics, IHC serves as an initial screen due to its accessibility, while FISH confirms equivocal (2+) results, ensuring accurate identification of amplified oncogenes. Liquid biopsies, analyzing (ctDNA) from , provide a non-invasive for oncogene detection, capturing , amplifications, and fusions shed from tumors with sensitivities exceeding 80% in advanced stages. Techniques like targeted NGS on ctDNA identify EGFR in NSCLC with comparable accuracy to biopsies, enabling serial without invasive procedures. ctDNA assays detect fusions such as EML4-ALK in NSCLC, present in 3-7% of cases, supporting real-time subtyping in metastatic settings. As of 2025, NCCN guidelines endorse liquid biopsies for initial and in eligible patients with advanced NSCLC. The status of oncogenes holds significant prognostic value by informing cancer subtyping and risk stratification; for example, ALK fusions in NSCLC define a distinct subtype with unique clinical behavior, often associated with younger patients and never-smokers, influencing therapeutic decisions despite variable survival impacts across studies. Detection of such alterations via integrated diagnostics refines , as ALK-positive cases may exhibit better responses to specific inhibitors compared to wild-type tumors. Overall, oncogene profiling enhances diagnostic precision, with guidelines recommending upfront testing in eligible cancers to subtype and prognosticate effectively.

Targeted Therapies

Targeted therapies for oncogene-driven cancers focus on inhibiting the activity of oncogenic proteins or disrupting the signaling pathways they activate, offering precision-based interventions that improve outcomes compared to traditional . These approaches exploit oncogene addiction, where cancer cells become dependent on the continued activity of the mutated oncogene for survival, allowing selective targeting with minimal impact on normal cells. inhibitors (TKIs) represent a of this strategy, particularly for receptor and non-receptor frequently altered in cancers. Imatinib, a seminal TKI, specifically targets the BCR-ABL fusion oncoprotein in chronic (CML), binding to its ATP-binding site and inhibiting constitutive activity. In the phase III IRIS trial, imatinib achieved a complete cytogenetic response in 74% of newly diagnosed chronic-phase CML patients at 18 months, establishing it as first-line therapy and dramatically improving survival rates. However, acquired often arises through secondary point mutations in the BCR-ABL , such as T315I, which alter the drug-binding conformation and reduce efficacy in up to 20-30% of patients progressing on therapy. Monoclonal antibodies provide another key modality by blocking extracellular domains of oncogene products. , a targeting the HER2 oncogene (amplified in ~15-20% of s), inhibits HER2 dimerization and downstream signaling while also eliciting . In the pivotal trial, trastuzumab added to reduced the risk of recurrence by 46% and mortality by 33% in HER2-positive early patients, with benefits persisting beyond 10 years of follow-up. Small-molecule inhibitors targeting intracellular oncogenes have also transformed treatment landscapes. , approved for BRAF V600E-mutant (present in ~50% of cases), selectively inhibits the mutant BRAF , leading to tumor regression. In the phase III BRIM-3 trial, extended median to 5.3 months versus 1.6 months with in unresectable or metastatic BRAF V600E . To counter rapid resistance via MAPK pathway reactivation, combination with MEK inhibitors like has become standard; the coBRIM trial demonstrated that plus improved to 9.9 months compared to 6.2 months with alone, delaying resistance through dual blockade of the RAF-MEK-ERK cascade. For oncogenes, which are mutated in 20-30% of human cancers, targeted therapies have emerged more recently. G12C inhibitors, such as (approved by the FDA in 2021 for NSCLC) and (approved in 2022), covalently bind the mutant KRAS protein, locking it in an inactive state. In the CodeBreaK 100 trial, achieved an objective response rate of 37.1% and median of 6.8 months in previously treated KRAS G12C-mutant NSCLC patients. As of 2025, combinations with or other agents are under investigation to overcome resistance and expand applicability to other RAS mutations. Despite these advances, acquired resistance remains a major challenge, often driven by secondary mutations or pathway bypass. For instance, the EGFR T790M mutation emerges in ~50-60% of non-small cell lung cancer patients progressing on first-generation EGFR TKIs like , sterically hindering inhibitor binding while enhancing kinase activity. Third-generation TKIs such as overcome T790M but face their own resistance mechanisms, underscoring the need for sequential or combinatorial strategies. approaches offer promising alternatives by exploiting vulnerabilities in oncogene-addicted cells; for example, like induce lethal DNA damage in cancers with /2 alterations (which can co-occur with oncogenic drivers), achieving objective response rates of ~40-50% in platinum-sensitive ovarian cancers through selective to homologous recombination-deficient cells. Oncogene-driven tumors generally exhibit higher response rates to targeted therapies than non-oncogene-addicted cancers, highlighting the clinical value of . In ROS1 fusion-positive non-small cell (~1-2% of cases), TKIs like yield objective response rates of approximately 70%, with median exceeding 18 months, far surpassing historical benchmarks.

References

  1. [1]
    Oncogenes - The Cell - NCBI Bookshelf - NIH
    Specific genes (called oncogenes) are capable of inducing cell transformation, thereby providing the first insights into the molecular basis of cancer.
  2. [2]
    Cell Cycle Control, Oncogenes, Tumor Suppressors - Nature
    These are the proteins involved in cell cycle regulation that operate by stimulating cellular growth and division (Figure 3). A common analogy equates oncogenes ...<|control11|><|separator|>
  3. [3]
    Oncogenes and tumor suppressor genes: functions and roles in ...
    All cancers are rooted to mutations in oncogenes (OCGs). OCGs can be defined as a modified version of a proto‐OCG, a class of genes involved in normal cell ...
  4. [4]
    Definition of oncogene - NCI Dictionary of Cancer Terms
    When a proto-oncogene is changed so that too many copies are made or it becomes more active than normal, it is called an oncogene. Oncogenes may cause normal ...
  5. [5]
    Oncogene: What They Are & What They Do - Cleveland Clinic
    An oncogene is a gene that may cause cancer. It's a mutated (changed) form of a normal gene, called a proto-oncogene, that manages cell growth.
  6. [6]
    Proto-oncogenes to Oncogenes to Cancer | Learn Science at Scitable
    Mutations in proto-oncogenes are typically dominant in nature, and the mutated version of a proto-oncogene is called an oncogene.
  7. [7]
    Cell Oncogene - an overview | ScienceDirect Topics
    In contrast to tumour suppressor genes, which are dealt with next, oncogenes are generally dominant and mutation of only one allele is therefore sufficient to ...
  8. [8]
    MYC and KRAS cooperation: from historical challenges to ... - Nature
    Aug 21, 2024 · RAS and MYC rank amongst the most commonly altered oncogenes in cancer, with RAS being the most frequently mutated and MYC the most amplified.
  9. [9]
    Retroviral Oncogenes: A Historical Primer - PMC - PubMed Central
    Src was the first retroviral oncogene discovered. This was no accident. Preparations of Rous sarcoma virus (RSV), the avian sarcoma virus carrying the src gene, ...
  10. [10]
    Understanding Cancer - NIH Curriculum Supplement Series - NCBI
    The role of oncogenes​​ Most proto-oncogenes code for proteins that are involved in molecular pathways that receive and process growth-stimulating signals from ...
  11. [11]
    A History of Cancer Research: Retroviral Oncogenes - PMC - NIH
    The evolutionarily conserved cellular gene (c-src) from which the viral oncogene (v-src) had arisen became known as a proto-oncogene. Figure 9. Figure 9 ...Missing: across | Show results with:across
  12. [12]
    The RSV Oncogene and Its Progenitor - NCBI - NIH
    The viral oncogenes all had cellular progenitors (proto-oncogenes); and to knowing that some of the properties of oncogenic proteins were potentially useful ...
  13. [13]
    Peyton Rous – Facts - NobelPrize.org
    When research showed that viruses can operate by affecting the genetic material of normal germ cells, interest in Rous' discovery was reignited. To cite ...Missing: 1911 | Show results with:1911
  14. [14]
    J. Michael Bishop – Biographical - NobelPrize.org
    Together with Harold Varmus he directed the research that led to the discovery of proto-oncogenes – normal genes that can be converted to cancer genes by ...
  15. [15]
    Oncogenesis - Retroviruses - NCBI Bookshelf
    Retroviruses were first discovered in association with cancer around the turn of this century. An avian erythroblastosis virus (AEV) was isolated from a ...
  16. [16]
    Passage of phenotypes of chemically transformed cells via ... - PNAS
    Nov 15, 1979 · Passage of phenotypes of chemically transformed cells via transfection of DNA and chromatin. C Shih, B Z Shilo, M P Goldfarb ...
  17. [17]
    Classification of KRAS activating mutations and the implications for ...
    KRAS is a proto-oncogenic GTPase. The first RAS oncogenes were isolated from retroviruses that induced spontaneous soft-tissue tumors in mice (1,2).
  18. [18]
    Oncogenic KRAS: Signaling and Drug Resistance - PMC - NIH
    Nov 9, 2021 · According to the research, Ras activated either by point mutation (G12V) or by GTP-bound interacts with Raf-1 through guanylyl-imidodiphosphate ...
  19. [19]
    Article A first-in-class EGFR-directed KRAS G12V selective inhibitor
    Missense mutations in KRAS often result in its constitutive activation, leading to dysregulated downstream signaling cascades that drive oncogenic ...
  20. [20]
    BRAF Mutations in Melanoma: Biological Aspects, Therapeutic ...
    The most frequent oncogenic BRAF mutations consist of a single point mutation at codon 600 (mostly V600E) that leads to constitutive activation of the BRAF/MEK/ ...2. Braf Mutations As Key... · 3.2. Braf Mutations And... · 4. Targeted Therapy For...
  21. [21]
    The role of BRAF V600 mutation in melanoma
    May 3, 2012 · About 50 % of melanomas harbors activating BRAF mutations (over 90 % V600E). BRAFV600E has been implicated in different mechanisms underlying melanomagenesis.Vemurafenib: Phase I And Ii... · The Mutated-Braf Approach · B-Raf Inhibitors In...
  22. [22]
    Worldwide Prevalence and Clinical Characteristics of RAS ...
    Mutations in RAS family members are well-established drivers of cancer. Gain-of-function mutations in RAS genes are found in ∼19% of human cancers, most ...
  23. [23]
    Targeted next-generation sequencing detects point mutations ...
    Targeted next-generation sequencing detects point mutations, insertions, deletions and balanced chromosomal rearrangements as well as identifies novel leukemia- ...
  24. [24]
    High-Throughput Detection of Actionable Genomic Alterations in ...
    Targeted, sequencing-based profiling of archival tumor samples identifies genetic alterations that can direct personalized therapy.
  25. [25]
    Molecular landscape and efficacy of HER2-targeted therapy ... - Nature
    Oct 30, 2020 · Human epidermal growth factor receptor 2 (HER2, also known as ERBB2) amplification or overexpression is detected in 20–30% of patients with ...Results · Her2 Mutations In Patients... · Patients And Sample...
  26. [26]
    Human Breast Cancer: Correlation of Relapse and Survival with ...
    Amplification of the HER-2/neu gene was a significant predictor of both overall survival and time to relapse in patients with breast cancer.Missing: seminal | Show results with:seminal
  27. [27]
    MYCN-amplified neuroblastoma maintains an aggressive ... - PNAS
    Jan 26, 2018 · MYCN is frequently amplified in high-risk NB and is linked to an undifferentiated phenotype and poor prognosis. Estrogen and nerve growth factor ...
  28. [28]
    Neuroblastoma and MYCN - PMC - PubMed Central - NIH
    MYCN is amplified in ∼25% of neuroblastoma patients, and this correlates with poor prognosis. Thus, efforts continue to elucidate the roles for MYCN in ...
  29. [29]
    Discovery of the Philadelphia chromosome: a personal perspective
    Almost 50 years ago, David Hungerford and I noticed an abnormally small chromosome in cells from patients with chronic myelogenous leukemia (CML).
  30. [30]
    Chronic myeloid leukemia: the paradigm of targeting oncogenic ...
    Feb 19, 2018 · Two major mechanisms have been implicated in the malignant transformation by BCR-ABL1: a) altered adhesion to bone marrow stroma cells and ...Cml: The Disease · The Role Of Bcr-Abl1 In... · Bcr-Abl1 Inhibition...
  31. [31]
    Insertional Oncogenesis by Non-Acute Retroviruses - NIH
    Ab-MuLV is an acute transforming retrovirus whose replication-defective genome carries the v-abl oncogene. In order to efficiently infect mice and induce ...
  32. [32]
    [PDF] Retroviral insertional mutagenesis: past, present and future - MIT
    Overexpression of the c-Myc proto-oncogene is a frequently found oncogenic mutation in retrovirally induced tumours, and mainly induces haematopoietic tumours ...
  33. [33]
    Alternative splicing and cancer: a systematic review - Nature
    Feb 24, 2021 · The abnormal splicing events contributed to tumor progression as oncogenic drivers and/or bystander factors. The alterations in splicing factors ...
  34. [34]
    The role of aberrant DNA methylation in cancer initiation and clinical ...
    Jan 28, 2024 · Certain proto-oncogenes can also be activated by loss of promoter methylation, for example, BCL2 in B-cell CLL 6 and NFATC1 in B-cell CLL, 7 ...
  35. [35]
    Genomic and transcriptomic changes complement each other in the ...
    Mar 29, 2019 · ... enhancer hijacking through the IGH-MYC ... MINCR is a MYC-induced lncRNA able to modulate MYC's transcriptional network in Burkitt lymphoma cells.
  36. [36]
    Lineage-specific RUNX2 super-enhancer activates MYC ... - Nature
    Apr 10, 2019 · The deletion of the mutant-allele super-enhancer of RUNX2 significantly reduced the expression of MYC and impaired the proliferative capacity of ...
  37. [37]
    BCL2 and miR-15/16: from gene discovery to treatment - Nature
    Oct 6, 2017 · In this review, we discuss the discovery, functions and clinical relevance of miR-15/16 and BCL2.
  38. [38]
    Role of miR-15/16 in CLL | Cell Death & Differentiation - Nature
    Jun 27, 2014 · The fact that miR-15/16 targets BCL2 expression was a first link between major tumor-suppressor microRNA and major oncogene in CLL, and these ...
  39. [39]
    Discovery and identification of oncogenes - NCBI
    The demonstration of activated protooncogenes in human tumors was first shown by the DNA-mediated transformation technique. ... Robert Weinberg, which ...
  40. [40]
    PDGF in gliomas: more than just a growth factor? - PubMed Central
    Infusion of PDGF-A in the ventricular system for several days can cause expansion of cells expressing the PDGFRA and induction of some glioma characteristics.
  41. [41]
    EGFR in Cancer: Signaling Mechanisms, Drugs, and Acquired ...
    Jun 1, 2021 · Here we review the epidermal growth factor (EGF), which controls epithelial cells, the precursors of all carcinomas, and the cognate cell surface receptor, ...
  42. [42]
    The past, present, and future of HER2 (ERBB2) in cancer ...
    Apr 2, 2019 · Human epidermal growth factor receptor 2 (HER2) (also known as ERBB2) is located on chromosome 17p21 and encodes a 185-kDa receptor tyrosine ...
  43. [43]
    Historical retrospective of the SRC oncogene and new perspectives ...
    Jul 14, 2020 · In the present review, we present a flashback to the history of the Src research, while focusing on the most important milestones in the field.
  44. [44]
    RAS oncogenes: weaving a tumorigenic web | Nature Reviews Cancer
    Oct 13, 2011 · In this Review, we describe how RAS oncogenes exploit their extensive signalling reach to affect multiple cellular processes that drive tumorigenesis.
  45. [45]
    Oncogenic PI3K and its role in cancer - PubMed
    Purpose of review: The purpose of this review is to examine the contribution of the PI3K signaling pathway to the development of human tumors and to propose ...
  46. [46]
    MYC: a multipurpose oncogene with prognostic and therapeutic ...
    Aug 9, 2021 · MYC oncogene is a transcription factor with a wide array of functions affecting cellular activities such as cell cycle, apoptosis, DNA damage response, and ...
  47. [47]
    Selective antagonism of cJun for cancer therapy
    Sep 11, 2020 · This review will focus on cJun, an AP-1 family member which is found to be upregulated or overexpressed in a large number of cancers.
  48. [48]
    Functions of cyclin D1 as an oncogene and regulation of ... - PubMed
    In this review, we summarize the role of cyclin D1 on tumor development and malignant transformation. In addition, our chemical biology study to understand ...
  49. [49]
    The bcl-2 oncogene and apoptosis - PubMed
    Bcl-2 functions to inhibit apoptosis in a variety of in vitro and in vivo experiments, suggesting interference with a central mechanism of apoptosis.
  50. [50]
    The role of BCL-2 family proteins in regulating apoptosis and cancer ...
    Presently, the purpose of this review is to highlight the current findings on the role of BCL-2 family proteins in regulating apoptosis and the development of ...
  51. [51]
    Review Raf kinases: Function, regulation and role in human cancer
    The Ras-Raf-MAPK pathway regulates diverse physiological processes by transmitting signals from membrane based receptors to various nuclear, cytoplasmic and ...
  52. [52]
    The role of c-Met in prognosis and clinicopathology of renal cell ...
    The c-Met proto-oncogene pathway plays an important role in the progression of various cancers. However, the effect of the c-Met pathway on renal cell ...
  53. [53]
  54. [54]
    fos-associated cellular p39 is related to nuclear transcription factor ...
    We show that one of the fos-associated proteins, p39, is immunologically and structurally related to nuclear factor AP-1.
  55. [55]
    The proto-oncogene Bcl-2 and its role in regulating apoptosis - Nature
    Jun 1, 1997 · A novel death-inducing protein shares a distinct sequence motif with Bcl-2 family proteins and interacts with viral and cellular survival-promoting proteins.
  56. [56]
  57. [57]
    FGF3 - an overview | ScienceDirect Topics
    FGF3 and FGF7 are members of the fibroblast growth factor (FGF) family, which are cytokines involved in cell growth, development, and proliferation.
  58. [58]
    The MEK/ERK Network as a Therapeutic Target in Human Cancer
    The RAS–RAF–MEK–ERK pathway is the most well-studied of the MAPK cascades and is critical for cell proliferation, differentiation, and survival.
  59. [59]
    Targeting the RAS/RAF/MAPK pathway for cancer therapy - Nature
    Dec 18, 2023 · In this review, we delve deeper into the mechanistic insights surrounding RAF kinase signaling in tumorigenesis and RAFi-resistance.
  60. [60]
    PI3K/AKT/mTOR signaling transduction pathway and targeted ...
    Aug 18, 2023 · The PI3K/AKT/mTOR (PAM) signaling pathway is a highly conserved signal transduction network in eukaryotic cells that promotes cell survival, cell growth, and ...
  61. [61]
    PTEN Tumor Suppressor Network in PI3K-Akt Pathway Control - PMC
    The PI3K-Akt pathway is a major survival pathway activated in cancer. Efforts to develop targeted therapies have not been fully successful, mainly because ...
  62. [62]
    Wnt/β-catenin signaling in cancers and targeted therapies - Nature
    Aug 30, 2021 · Wnt/β-catenin signaling has been broadly implicated in human cancers and experimental cancer models of animals.
  63. [63]
    Targeting the Wnt/β-catenin signaling pathway in cancer
    Dec 4, 2020 · This scoping review aims at outlining the latest progress on the current approaches and perspectives of Wnt/β-catenin signaling pathway targeted therapy in ...
  64. [64]
    Context Matters: NOTCH Signatures and Pathway in Cancer ...
    The Notch signaling pathway is a critical player in embryogenesis but also plays various roles in tumorigenesis, with both tumor suppressor and oncogenic ...
  65. [65]
    Hedgehog Signal Transduction: Key Players, Oncogenic Drivers ...
    Aug 22, 2016 · The Hedgehog (Hh) signaling pathway governs complex developmental processes, including proliferation and patterning within diverse tissues.
  66. [66]
    The Role of Notch, Hedgehog, and Wnt Signaling Pathways in the ...
    The major signaling pathways involved in the developmental process, such as Notch, Hedgehog, and Wnt, play a vital role in development, tumorigenesis, and also ...
  67. [67]
    Mutation and Cancer: Statistical Study of Retinoblastoma | PNAS
    ### Summary of Knudson's Two-Hit Hypothesis and Related Concepts
  68. [68]
  69. [69]
    The two-hit theory hits 50 - PMC - PubMed Central
    Few ideas in cancer genetics have been as influential as the “two-hit” theory of tumor suppressors. This idea was introduced in 1971 by Al Knudson.
  70. [70]
  71. [71]
    Mutations in APC, Kirsten-ras, and p53—alternative genetic ...
    A genetic model for colorectal cancer has been proposed in which the sequential accumulation of mutations in specific genes, including adenomatous polyposis ...
  72. [72]
    The Clonal Evolution of Tumor Cell Populations
    ### Summary of Clonal Evolution in Tumor Cell Populations by Nowell
  73. [73]
    Clonal evolution in cancer - PubMed - NIH
    Jan 18, 2012 · Cancers evolve by a reiterative process of clonal expansion, genetic diversification and clonal selection within the adaptive landscapes of tissue ecosystems.
  74. [74]
    Identifying multi-hit carcinogenic gene combinations - Nature
    Feb 6, 2020 · The classic example for the multi-hit hypothesis is the study of retinoblastoma by Knudson9.
  75. [75]
    Evolutionary Determinants of Cancer - AACR Journals
    Our understanding of cancer is being transformed by exploring clonal diversity, drug resistance, and causation within an evolutionary framework.
  76. [76]
    Liquid biopsy: current technology and clinical applications
    Sep 12, 2022 · Liquid biopsies are increasingly used for cancer molecular profiling that enables a precision oncology approach.
  77. [77]
    Next-generation sequencing-based detection of EGFR, KRAS ... - NIH
    Molecular assays of EGFR, KRAS, BRAF, NRAS, PIK3CA and Her-2 are widely used to guide individualized treatment in NSCLC patients.Patients And Methods · The Patient Mutation Profile · Comparison Of Ngs, Sanger...
  78. [78]
    Comparison of Immunohistochemical Methods (IHC) and ... - NIH
    The present study is the first study to compare IHC and FISH methods in the diagnosis of HER2 gene overexpression in patients with breast cancer in western ...
  79. [79]
    Comparison of the IHC, FISH, SISH and qPCR methods for the ...
    May 17, 2012 · Her2 proto-oncogene amplification and protein overexpression is observed in 20-40% of patients with breast cancer and plays a crucial role ...
  80. [80]
    Evaluation of HER-2/neu Gene Amplification by Fluorescence In Situ ...
    This study tested the use of fluorescence in situ hybridization (FISH) and immunohistochemistry (IHC) to evaluate the HER-2/neu gene status and to ascertain the ...
  81. [81]
    Liquid biopsy in cancer: current status, challenges and future ...
    Dec 2, 2024 · Several molecular markers can be detected by liquid biopsy, such as circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), tumor-derived ...
  82. [82]
    Overview of the role of liquid biopsy in cancer management
    Nearly all targetable mutations could be detected by ctDNA analysis with high sensitivity and specificity like EGFR mutations [15], BRAF [16], KRAS [17], BRCA 1 ...
  83. [83]
    ALK fusions in the pan-cancer setting: another tumor-agnostic target?
    Sep 29, 2023 · In NSCLC, ALK gene alterations act as oncogenic drivers, and occur in ~3–7% of cases. One such molecular alteration—an activating ...
  84. [84]
    Prevalence and natural history of ALK positive non-small-cell lung ...
    Takeuchi et al63 found that the presence of a kinase fusion (ALK, ROS 1, and RET fusion) was an independent favorable prognostic factor after taking into ...
  85. [85]
    Mutation status analysis of 58 patients with advanced ALK fusion ...
    To analyze the characteristics and prognostic values of Anaplastic Lymphoma Kinase (ALK) fusion gene partner, gene subtype and abundance in tumor tissues
  86. [86]
    Bcr-Abl kinase domain mutations, drug resistance, and the road to a ...
    Mutations in the kinase domain (KD) of BCR-ABL are the most prevalent mechanism of acquired imatinib resistance in patients with chronic myeloid leukemia (CML).Introduction · How do point mutations cause... · Can mutations conferring...
  87. [87]
    Trastuzumab after Adjuvant Chemotherapy in HER2-Positive Breast ...
    Oct 20, 2005 · One year of treatment with trastuzumab after adjuvant chemotherapy significantly improves disease-free survival among women with HER2-positive breast cancer.Trastuzumab After Adjuvant... · Study Design · Eligibility Criteria
  88. [88]
    FDA Approval Summary: Vemurafenib for Treatment of Unresectable ...
    Vemurafenib represents an important new treatment option for patients with BRAFV600E mutation–positive unresectable or metastatic melanoma and is the first ...Abstract · Introduction · Clinical Trials · Discussion<|separator|>
  89. [89]
    Combined BRAF and MEK Inhibition versus BRAF Inhibition Alone ...
    Sep 29, 2014 · Combined BRAF and MEK inhibition, as compared with BRAF inhibition alone, delays the emergence of resistance and reduces toxic effects in ...
  90. [90]
    The T790M mutation in EGFR kinase causes drug resistance by ...
    We conclude that the T790M mutation is a “generic” resistance mutation that will reduce the potency of any ATP-competitive kinase inhibitor.
  91. [91]
    PARP Inhibitors: The First Synthetic Lethal Targeted Therapy - PMC
    Mar 16, 2017 · Tumors arising in patients who carry germline mutations in either BRCA1 or BRCA2 are sensitive to PARPi because they have a specific type of DNA ...
  92. [92]
    Efficacy of Neoadjuvant Therapy for ROS1-Positive Locally ... - NIH
    Dec 24, 2024 · In a trial cohort of 50 patients with ROS1-rearranged NSCLCs, the objective response rate (ORR) to crizotinib was 72%, the disease control rate ...