Fact-checked by Grok 2 weeks ago

Cancer cell

A cancer cell is an abnormal cell that proliferates uncontrollably, disregards normal regulatory signals, and can invade surrounding s or metastasize to distant sites in the body, forming the basis of malignant tumors. Unlike normal cells, which divide in a controlled manner to maintain function and repair, cancer cells acquire that disrupt this balance, leading to their hallmark behaviors of sustained , evasion of , and potential for widespread dissemination. The defining characteristics of cancer cells stem from genetic and epigenetic alterations that enable them to bypass the checkpoints governing normal cellular behavior. For instance, they often exhibit self-sufficiency in growth signals by producing their own stimulatory factors or overexpressing receptors, allowing proliferation without external cues. They also show insensitivity to antigrowth signals, ignoring inhibitors like tumor suppressor proteins such as or , which normally halt division in response to damage or overcrowding. Additionally, cancer cells resist through mechanisms like upregulation of anti-apoptotic proteins (e.g., members), enabling survival under stressful conditions that would eliminate normal cells. Beyond these core traits, cancer cells demonstrate unlimited replicative potential by maintaining telomere length via activation, avoiding the that limits normal cell divisions to roughly 50-70 cycles (the ). They further promote to secure nutrient supply by secreting factors like (VEGF), and acquire capabilities for tissue invasion and metastasis, often through epithelial-to-mesenchymal transition (EMT) that enhances motility and extracellular matrix degradation. These acquired abilities, collectively termed the "hallmarks of cancer," evolve through multistep accumulation of mutations in oncogenes and tumor suppressor genes, driven by factors such as environmental carcinogens, inherited predispositions, or chronic inflammation. In the context of disease, cancer cells originate from any tissue type but share these dysfunctional traits, classifying malignancies by their cell of origin (e.g., carcinomas from epithelial cells, sarcomas from mesenchymal cells). Their uncontrolled expansion disrupts organ function and, if untreated, can lead to systemic effects like or organ failure, underscoring the need for therapies targeting these specific vulnerabilities, such as checkpoint inhibitors or targeted molecular agents.

Definition and Characteristics

Hallmarks of Cancer

The hallmarks of cancer delineate the essential functional capabilities that enable normal cells to undergo , acquiring traits that sustain uncontrolled and survival in diverse tissue environments. Originally proposed in , these hallmarks provide a for understanding the multistep of tumors, emphasizing how cancer cells subvert normal regulatory circuits to promote their own growth and dissemination. This framework has since been refined to include enabling characteristics that facilitate hallmark acquisition and emerging hallmarks reflecting additional adaptive strategies observed in advanced cancers.00127-9) In 2022, the framework was further updated to incorporate new dimensions, including unlocking as a new hallmark capability, nonmutational epigenetic reprogramming and polymorphic microbiomes as additional enabling characteristics, and the influence of senescent cells in the . Collectively, these traits distinguish malignant cells from their normal counterparts by conferring selective advantages that drive tumor progression, such as sustained through activation or resistance to programmed death signals. The six original hallmarks represent the foundational biological capabilities acquired during tumorigenesis. Self-sufficiency in growth signals allows cancer cells to proliferate independently of exogenous mitogenic stimuli, often through autocrine loops where cells produce their own growth factors like (PDGF) in glioblastomas or overexpression of receptors such as HER2 in , bypassing the need for stromal-derived signals. Insensitivity to antigrowth signals enables evasion of tumor suppressor pathways, for instance via disruption of the (pRb) pathway through mutations in TGF-β receptors or loss of pRb function, as seen in human papillomavirus (HPV)-associated cervical cancers where the viral E7 protein inactivates pRb. Evasion of apoptosis confers resistance to , a critical barrier to tumor expansion, achieved through inactivation of (altered in over 50% of cancers) or overexpression of anti-apoptotic proteins like , which inhibits mitochondrial outer membrane permeabilization. Limitless replicative potential overcomes the of normal cell division by reactivating in 85-90% of tumors, maintaining length to prevent replicative senescence, or via alternative lengthening of telomeres (ALT) mechanisms in some sarcomas. Sustained ensures nutrient and oxygen supply for expanding tumors beyond the diffusion limit of 100-200 μm, primarily through upregulation of (VEGF) and fibroblast growth factors (FGFs), often triggered by hypoxia-inducible factors or loss. , responsible for approximately 90% of cancer mortality, involves alterations in (e.g., downregulation of E-cadherin) and remodeling via proteases like matrix metalloproteinases (MMPs), enabling local invasion and distant colonization. Two enabling characteristics underpin the acquisition and maintenance of these hallmarks by fostering an environment conducive to genetic and epigenetic changes. accelerates the mutation rate, generating the diverse variants needed for tumor evolution; this arises from defects in pathways, such as mismatch repair deficiencies in , or centrosome amplification leading to chromosomal instability.00127-9) Tumor-promoting inflammation recruits immune cells that release cytokines and growth factors, akin to but chronically, enhancing and ; for example, interleukin-6 (IL-6) and (TNF) from infiltrating macrophages support oncogene-driven growth in various carcinomas.00127-9) Two emerging hallmarks highlight adaptive reprogramming in cancer cells. Reprogramming energy metabolism, often termed the Warburg effect, shifts cells toward aerobic even in oxygen-rich environments, supporting rapid ; this is driven by oncogenes like or PI3K/AKT activation, which upregulate glucose transporters and glycolytic enzymes to fuel and for proliferation.00127-9) Evading immune destruction allows tumors to dodge recognition and elimination by the adaptive and innate immune systems, through mechanisms such as expression on cancer cells to inhibit T-cell activity via PD-1 blockade or recruitment of regulatory T cells to suppress anti-tumor responses.00127-9) The 2022 update expands the framework by recognizing unlocking as a hallmark, enabling cells to undergo , blocked , or , which facilitates adaptation to therapeutic pressures and . It also introduces nonmutational epigenetic as an enabling characteristic, where heritable changes in without DNA alterations drive tumor heterogeneity. Polymorphic microbiomes are highlighted as another enabler, with microbial communities influencing , , and immune evasion in the . Additionally, senescent cells—from cancer cells, fibroblasts, or endothelial cells—are noted as a key component, promoting tumorigenesis through secretion of pro-inflammatory factors despite their non-proliferative state. These additions reflect evolving insights into cancer's complexity and inform novel therapeutic strategies.

Differences from Normal Cells

Cancer cells exhibit distinct morphological differences from normal cells, primarily characterized by irregular shapes, increased size variation known as , and enlarged nuclei that appear hyperchromatic due to increased DNA content and prominent nucleoli. These alterations arise from uncontrolled proliferation and genetic instability, contrasting with the uniform, organized of normal cells that maintain structural integrity within s. For instance, normal cells typically display consistent nuclear-to-cytoplasmic ratios and symmetrical contours, while cancer cells often have scarce and pleomorphic features that disrupt tissue architecture. In terms of behavior, cancer cells lose contact inhibition, allowing them to beyond the formation seen in normal cells, and gain anchorage independence, enabling growth without attachment to a or . This anchorage independence facilitates survival in low-adhesion environments, such as during , unlike normal cells that require adhesion for and undergo anoikis—a form of —upon detachment. Consequently, cancer cells can form disorganized masses, invading surrounding tissues, in stark contrast to the orderly layers formed by normal epithelial cells that respect boundary signals. Regulatory differences further distinguish cancer cells, including disrupted that permit unchecked division despite DNA damage, reduced leading to immature phenotypes, and high intratumoral heterogeneity where subpopulations vary in growth rates and responses to stimuli. Normal cells adhere to stringent checkpoints, such as those at G1/S and G2/M phases, ensuring fidelity in replication and into specialized functions, whereas cancer cells bypass these via mutations in regulators like or cyclins. This heterogeneity, driven by non-genetic factors like epigenetic variations, allows cancer cell populations to adapt dynamically, evading therapies that target uniform traits in normal cells. These regulatory shifts align with broader hallmarks, such as sustained proliferative signaling, but manifest as observable deviations in behavior and organization.

Classification

By Tissue Origin

Cancer cells are classified by the tissue or from which they originate, a that provides a foundational histological framework for and . This histological distinguishes tumors based on their embryonic or adult tissue lineage, such as epithelial, mesenchymal, or hematopoietic origins, and has evolved through standardized international systems to ensure consistency in . The (WHO) Classification of Tumours, initiated by a 1956 resolution of the WHO Executive Board, represents the authoritative standard, with its first edition published in the 1960s and subsequent updates incorporating advances in and to refine tissue-based categorizations. The predominant category is carcinomas, which arise from epithelial tissues lining organs and glands, accounting for 80-90% of all human cancers due to the widespread distribution of epithelial cells throughout the body. Within carcinomas, malignant transformations often progress from benign precursors, such as adenomas (benign glandular tumors) to (malignant glandular cancers), the latter characterized by invasive growth and potential for distant spread; common examples include lung and colorectal . Squamous cell carcinomas, another major subtype, originate from squamous epithelium in areas like , lungs, and , frequently developing from premalignant lesions like . Sarcomas derive from mesenchymal tissues, including connective, supportive, or soft tissues such as , , , and , and represent a rarer comprising about 1% of adult cancers but more common in children and young adults. Benign counterparts, like osteomas (benign tumors), contrast with malignant forms such as , which exhibits aggressive destruction and a higher incidence in adolescents. Other sarcomas include from and from , highlighting the diverse mesenchymal origins and the malignant shift involving loss of tissue-specific differentiation. Leukemias and lymphomas originate from hematopoietic and lymphoid tissues, respectively, and are classified separately from solid tumors due to their liquid or systemic nature. Leukemias involve malignant proliferation of immature blood-forming cells in the , leading to overproduction of abnormal , with subtypes including and ; these differ from benign hematologic disorders by their uncontrolled clonal expansion. Lymphomas, solid malignancies of lymphocytes, include (characterized by Reed-Sternberg cells) and , often arising in lymph nodes or extranodal sites like the , where disrupts normal immune function. Other categories encompass germ cell tumors, which develop from primordial germ cells in the gonads or extragonadal sites like the , and can be benign (e.g., mature teratomas) or malignant (e.g., seminomas or tumors), with the latter showing pluripotential differentiation mimicking embryonic tissues. Mixed tumors, such as carcinosarcomas combining epithelial and mesenchymal elements, further illustrate the histological diversity within tissue origins, though molecular subtyping refines these groupings for precision .

By Molecular Subtypes

Cancer cells are increasingly classified by molecular subtypes, which delineate distinct genetic, proteomic, and transcriptomic profiles that refine categorization beyond tissue origin and . This approach evolved from early 20th-century histological systems to modern genomic paradigms, driven by initiatives like (TCGA), which sequenced over 11,000 primary tumors to reveal subtype-specific alterations across cancers. The transition emphasizes multi-omics integration, combining , expression, and protein data to capture tumor heterogeneity and improve prognostic accuracy over purely morphological methods. Recent editions of the WHO Classification of Tumours (5th edition, 2020-2025) further incorporate molecular features into histological categories for enhanced precision. Key techniques for molecular subtyping include next-generation genomic sequencing to identify driver mutations, (IHC) to assess protein biomarkers, and to cluster tumors based on transcriptomic signatures. For instance, the PAM50 assay uses quantitative reverse transcription on 50 genes to classify into luminal A, luminal B, HER2-enriched, basal-like, and normal-like subtypes, with luminal A tumors showing the highest stability. is pivotal for detecting HER2 overexpression in cells, where membrane staining intensity scores guide subtype assignment, while whole-exome sequencing uncovers somatic variants like BRAF in cells, present in approximately 50% of cutaneous melanomas. In lung adenocarcinoma, mutations—often G12C or G12D—define a major subtype, frequently co-occurring with /LKB1 alterations in 15-25% of KRAS-mutant cases, as identified through integrative genomic analyses. These molecular subtypes carry significant prognostic implications, influencing survival outcomes and therapeutic decision-making in precision as of the early . For example, early-stage HER2-positive has a 5-year overall survival rate exceeding 90% with targeted therapies like , while luminal A subtypes exceed 95%. BRAF-mutated subtypes show variable prognosis, with V600E variants linked to younger onset but poorer response to standard therapies compared to wild-type tumors. For metastatic KRAS-driven lung adenocarcinoma, median overall survival is approximately 12 months, with co-mutations like associated with resistance and worse outcomes. Emerging multi-omics frameworks, such as those using on TCGA datasets, further refine these subtypes by incorporating epigenomic and proteomic layers, enabling predictions of tumor evolution and personalized risk assessment.

Microscopic Features

Nuclear Alterations

Cancer cells exhibit distinct nuclear alterations that contribute to their identification under and reflect underlying genomic . One prominent feature is nuclear enlargement, often accompanied by anisonucleosis—a variation in nuclear size—and pleomorphism, characterized by irregular nuclear shapes. These changes arise primarily from increased DNA content due to or , which disrupts normal nuclear architecture. Hyperchromasia, or intensified nuclear staining, further accentuates these alterations, resulting from condensed and elevated density that enhances basophilic appearance during histological examination. Prominent nucleoli are another hallmark of cancer cell nuclei, appearing enlarged and multiple in number compared to normal cells. This prominence stems from heightened ribosomal RNA (rRNA) synthesis within the , supporting accelerated to meet the demands of rapid protein production in proliferating tumor cells. Such nucleolar correlates with upregulated transcription of rRNA genes, a process essential for the metabolic reprogramming observed in malignancies. Chromatin within cancer cell nuclei often displays margination, where it adheres to the periphery, and clumping into dense aggregates, deviating from the evenly dispersed in healthy cells. These structural shifts indicate altered patterns, as influences accessibility to transcriptional machinery and epigenetic modifications. Margination and clumping can impair normal regulatory functions, promoting the dysregulation of oncogenes and tumor suppressors. Micronuclei formation represents a critical alteration linked to chromosomal in cancer cells, where whole chromosomes or fragments are excluded from the main during , forming small extranuclear bodies. This phenomenon arises from errors such as lagging chromosomes or acentric fragments, serving as a marker of genomic that drives tumor evolution. The quantifies these structures by scoring their frequency in cells, providing a standardized method to assess and chromosomal aberrations in cancer diagnostics and research.

Cytoplasmic and Structural Changes

Cancer cells often exhibit increased cytoplasmic , a bluish of the observed under light microscopy, primarily due to elevated levels of ribosomal and associated with heightened protein synthesis demands. This basophilia reflects the accumulation of free ribosomes and polysomes in the , which supports the rapid and metabolic activity characteristic of . In histological sections of tumors such as , this feature manifests as intensified cytoplasmic alongside reduced cell volume. The of cancer cells frequently shows an abundance of mitochondria, adapted to fulfill the elevated bioenergetic and biosynthetic requirements despite reliance on aerobic . These organelles increase in number through enhanced biogenesis pathways, enabling the production of intermediates for synthesis and signaling that promote tumor progression. For instance, in aggressive cancers like and carcinomas, mitochondrial mass expansion correlates with metastatic potential and resistance to . Disruption of the is a hallmark structural change in , leading to irregular cell shapes, loss of , and enhanced motility essential for . Alterations in filaments, including disorganized and abnormal bundling mediated by actin-binding proteins, contribute to these morphological irregularities and facilitate pseudopod formation for migratory behavior. In mesenchymal-type , such as those in pancreatic ductal , stabilized or destabilized networks impair normal while promoting amoeboid-like movement through extracellular matrices. Organelle abnormalities further underscore the dysfunctional architecture of cancer cell cytoplasm. The Golgi apparatus often appears enlarged or fragmented, supporting increased secretory demands for remodeling and release during tumor progression. In stressed cancer cells, such as those under nutrient deprivation or , autophagic vacuoles—double-membrane-bound structures known as autophagosomes—accumulate to degrade damaged cytoplasmic components and maintain . These vacuoles, formed via ATG protein-mediated processes, enable survival in hypoxic tumor microenvironments but can also contribute to therapeutic resistance in cancers like . Examples of severe structural derangements include binucleation or multinucleation, arising from failed where cells complete karyokinesis but fail to divide the . This results in tetraploid or polyploid cells with multiple nuclei sharing a common , observed in anaplastic tumors such as those driven by mutations or ECT2 overexpression. In ovarian and colorectal cancers, such multinucleated giant cells promote genomic instability and , fostering aggressive phenotypes.

Etiology

Genetic Causes

Cancer cells arise from intrinsic genetic alterations that disrupt normal cellular regulation, primarily through mutations affecting and tumor suppressor genes. , derived from proto-oncogenes, promote cell growth and division when activated by gain-of-function mutations, such as point mutations or gene amplifications. For instance, mutations in the family of oncogenes, occurring in approximately 30% of human cancers, lead to constitutive activation of signaling pathways that drive uncontrolled proliferation. Similarly, amplification of the oncogene, a overexpressed in over 70% of cancers, enhances progression and metabolic reprogramming, facilitating tumorigenesis. In contrast, tumor suppressor genes inhibit cell growth, and their loss-of-function mutations or deletions allow . The TP53 gene, mutated in more than 50% of cancers, encodes the protein, which normally induces arrest, , or in response to damage; its inactivation impairs these safeguards, enabling survival of genetically unstable cells. These genetic changes can be inherited as mutations, conferring predisposition to hereditary cancers. For example, pathogenic variants in and genes, which repair DNA double-strand breaks, increase lifetime risk to 55-72% for BRCA1 carriers and 45-69% for BRCA2 carriers by age 70-80, with risks of 39-44% and 17-31%, respectively; these mutations account for 5-10% of breast cancers overall. Most cancer-initiating genetic alterations, however, are acquired during a lifetime through multistep , where sequential accumulation of transforms normal cells into malignant ones. In this process, driver mutations confer selective growth advantages to cells, such as enhanced or , while passenger mutations arise incidentally without functional impact but hitchhike with drivers during clonal expansion; sequencing reveals tumors often harbor 2-8 driver mutations alongside thousands of passengers. Chromosomal abnormalities further contribute to cancer development by altering or creating fusion genes. , an imbalance in chromosome number affecting nearly all solid tumors, disrupts proteostasis and promotes genomic instability, acting as both a cause and consequence of . Balanced translocations, such as the t(9;22) in chronic myeloid leukemia (CML), fuse BCR and ABL1 genes to produce a hyperactive that drives leukemogenesis in over 95% of CML cases.

Environmental and Lifestyle Factors

Environmental and lifestyle factors play a significant role in the initiation of cancer by inducing DNA damage that can lead to oncogenic mutations in cells. These extrinsic influences interact with cellular processes, often requiring chronic exposure to accumulate sufficient genetic alterations for . While individual genetic susceptibilities can modulate risk, the primary impact stems from exposure to specific agents that directly or indirectly promote . Chemical carcinogens, such as those in tobacco smoke, are among the most well-established environmental contributors to cancer development. Tobacco smoke contains polycyclic aromatic hydrocarbons (PAHs) like benzopyrene, which form DNA adducts that, if unrepaired, lead to mutations in critical genes; epidemiological studies show a strong dose-response relationship, where the risk of lung cancer increases linearly with pack-years smoked. Similarly, asbestos fibers, particularly chrysotile and amphibole types, cause chronic inflammation and oxidative stress in mesothelial cells, resulting in malignant mesothelioma; the International Agency for Research on Cancer (IARC) classifies asbestos as a Group 1 carcinogen based on consistent evidence from occupational cohorts demonstrating risk proportional to cumulative exposure duration and intensity. Physical agents, including ultraviolet (UV) radiation and ionizing radiation, exert genotoxic effects by directly damaging DNA structure. UV radiation, primarily UVB wavelengths from sunlight, induces cyclobutane pyrimidine dimers (CPDs) and 6-4 photoproducts between adjacent thymine or cytosine bases, impairing replication and transcription; this is a key mechanism in skin cancers like melanoma, with fair-skinned individuals showing higher incidence due to reduced melanin protection. Ionizing radiation, from sources such as radon gas or medical imaging, generates reactive oxygen species and causes double-strand breaks in DNA, leading to chromosomal aberrations; atomic bomb survivor studies have established a linear no-threshold dose-response, where even low doses elevate leukemia risk. Biological agents, notably oncogenic viruses, contribute to cancer by hijacking host cellular machinery to promote uncontrolled growth. Human papillomavirus (HPV), particularly high-risk types 16 and 18, encodes E6 and E7 oncoproteins that bind and degrade and tumor suppressors, respectively, disabling and ; this is central to pathogenesis, with over 99% of cases linked to persistent HPV as per global surveillance data. Other viruses like hepatitis B virus (HBV) induce through chronic inflammation and integration of viral DNA into host genomes, while Epstein-Barr virus (EBV) is associated with lymphomas via latent gene expression that activates signaling. Lifestyle factors amplify cancer through modifiable behaviors that foster a pro-carcinogenic . Diets high in processed meats introduce N-nitroso compounds from nitrates and nitrites, which can form DNA-alkylating agents during , elevating ; meta-analyses indicate a 18% increased per 50g daily intake. consumption generates , a mutagenic that forms DNA adducts, particularly in the upper aerodigestive tract, with rising dose-dependently—over 3 drinks per day triples esophageal cancer odds. promotes systemic inflammation via adipokine dysregulation and elevated insulin/IGF-1 levels, creating a microenvironment conducive to and endometrial cancers; cohort studies link a 5-unit increase to a 20-50% higher for these sites.

Molecular Mechanisms

DNA Repair and Mutations

Defects in mechanisms are a fundamental driver of genomic instability in cancer cells, allowing the accumulation of that promote tumorigenesis. These defects impair the cell's ability to correct DNA damage from endogenous or exogenous sources, leading to a higher frequency of genetic alterations compared to normal cells. As a result, cancer cells exhibit elevated mutation rates, which fuel the selection of advantageous clones and contribute to tumor progression. The major DNA repair pathways include (BER), which removes damaged bases such as those caused by ; (NER), which excises bulky lesions like UV-induced adducts; mismatch repair (MMR), which corrects replication errors and small insertions/deletions; (HR), which accurately repairs double-strand breaks (DSBs) using a sister chromatid ; and (NHEJ), an error-prone pathway that ligates DSB ends directly. Each pathway operates through specific proteins: for instance, BER involves PARP-1 and APE1, NER relies on XPA-XPG factors, MMR uses MLH1/MSH2 complexes, HR depends on /2 and RAD51, and NHEJ requires Ku70/80 and . Defects in these pathways compromise repair fidelity, resulting in persistent DNA damage and chromosomal aberrations that characterize cancer genomes. Notable deficiencies include MMR defects associated with Lynch syndrome, an autosomal dominant condition caused by germline mutations in MLH1, MSH2, MSH6, or PMS2 genes, which lead to (MSI)—a hypermutator phenotype marked by expansions or contractions in repetitive DNA sequences. This instability arises because unrepaired replication errors accumulate, particularly in microsatellites, driving frameshift mutations in target genes and increasing cancer risk for colorectal, endometrial, and other tumors. Similarly, HR defects in BRCA1- or BRCA2-mutated cells impair DSB repair, causing genomic scars such as and structural variants, which predispose individuals to , ovarian, and cancers by allowing unrepaired breaks to propagate lethal or oncogenic changes. Mutation rates in cancer cells are substantially elevated due to these repair deficiencies; while normal cells maintain a rate of approximately 10^{-9} mutations per per , cancer cells with defects like MMR deficiency can exhibit 100- to 1,000-fold increases, reaching 10^{-6} to 10^{-7} per per division in hypermutated tumors. This hypermutation, often quantified as MSI-high status, amplifies the within the tumor. The consequences of unrepaired DNA damage include accelerated clonal and intratumor heterogeneity, where subpopulations of cancer cells acquire diverse that confer survival advantages, such as resistance to or enhanced invasiveness. This evolutionary process, driven by repair pathway failures, enables the tumor to adapt dynamically, with dominant clones emerging from a heterogeneous pool shaped by selective pressures.30066-1)

Telomerase and Immortality

Telomeres are structures at the ends of linear chromosomes, consisting of tandem repeats of the hexanucleotide sequence TTAGGG in humans, typically spanning 5–15 kilobases in length. In normal somatic cells, telomeres progressively shorten with each due to the end-replication problem, where cannot fully replicate the 3' ends of chromosomes, leading to a loss of 50–200 base pairs per replication cycle. This telomere attrition eventually triggers a DNA damage response, culminating in or , thereby limiting the replicative lifespan of cells and serving as a tumor-suppressive . Cancer cells evade this replicative senescence by reactivating , a that extends using an template to add TTAGGG repeats to ends. comprises two main components: the catalytic subunit encoded by the TERT gene (), which synthesizes the telomeric DNA, and the TERC component (), which provides the template sequence for repeat addition. In most cancers, is upregulated through mechanisms such as TERT promoter mutations (e.g., C228T or C250T hotspots), which create binding sites for transcription factors, or TERT , leading to sustained activity and telomere maintenance. Telomerase activity is detected in approximately 90% of human cancers, including carcinomas, but is largely absent in normal somatic tissues, where it is restricted to cells, cells, and activated lymphocytes. This high prevalence underscores 's role in conferring replicative , a hallmark enabling indefinite proliferation. In the remaining 10–15% of telomerase-negative cancers, s are maintained via alternative lengthening of s (ALT), a recombination-mediated mechanism involving break-induced replication between s, often characterized by heterogeneous lengths and extrachromosomal telomeric DNA. ALT relies on proteins and is prevalent in certain sarcomas, gliomas, and neuroendocrine tumors.

Pathological Behavior

Uncontrolled Proliferation

Uncontrolled proliferation in cancer cells arises from dysregulated cell cycle control, enabling these cells to divide rapidly and indefinitely without external signals that normally restrict growth in healthy tissues. This hallmark behavior is driven by alterations in key regulatory pathways, allowing cancer cells to bypass physiological checkpoints and maintain a proliferative state even in nutrient-poor or stressful environments. Such deregulation contrasts with normal cells, which exit the into quiescence () under unfavorable conditions, highlighting how cancer cells exploit these mechanisms for survival and expansion. A primary mechanism of uncontrolled involves at the of the , often due to overexpression of and its associated kinases CDK4/6. In normal cells, /CDK4/6 complexes phosphorylate the (Rb), partially releasing E2F transcription factors to initiate ; however, in cancer, amplified or hyperactive CDK4/6 leads to excessive Rb phosphorylation, fully derepressing E2F and driving unchecked progression into . This overexpression is common in various malignancies, such as and cancers, where it sustains independent of growth factors. Cancer cells also achieve growth factor independence through autocrine signaling loops, exemplified by (PDGF) in s. In these tumors, cells produce PDGF ligands that bind and activate their own PDGF receptors (PDGFR), forming a self-stimulatory loop that activates downstream pathways like PI3K/Akt to promote continuous without paracrine support from the . Blocking this autocrine PDGF signaling inhibits Akt and suppresses cell growth, underscoring its role in maintaining proliferative autonomy. Failures in cell cycle checkpoints further exacerbate proliferation, particularly through defects in the mitotic spindle assembly checkpoint (SAC), which normally halts until chromosomes align properly. In cancer cells, weakened SAC function—often from mutations in genes like MAD2 or BUB1—allows improper spindle attachments, leading to chromosome missegregation and , a state of abnormal numbers that paradoxically enhances tumorigenic potential by increasing genetic instability and adaptive proliferation. This checkpoint evasion enables cells to complete despite errors, perpetuating cycles of division. Evasion of quiescence is facilitated by reduced levels of CDK inhibitors, such as p16INK4a, which normally binds CDK4/6 to prevent Rb and induce G0 arrest. Alterations in the p16INK4a pathway, observed in up to 77% of bladder carcinomas, free CDK4/6 to drive proliferation by releasing other inhibitors (e.g., p27KIP1) for alternative uses and bypassing stress-induced quiescence. This deregulation not only sustains active cycling but also links to broader survival strategies, such as limited evasion of through overlapping Rb pathway alterations.

Invasion and Metastasis

Invasion and metastasis are defining features of malignant cancer cells, enabling them to escape the primary tumor, enter circulation, and establish secondary tumors at distant sites, which accounts for over 90% of cancer-related deaths. This process begins with the acquisition of migratory and invasive capabilities, allowing cells to breach membranes and tissues, followed by and . A pivotal in initiating is the epithelial-mesenchymal transition (), where cancer cells lose epithelial polarity and adhesion while gaining mesenchymal traits that promote motility. During EMT, E-cadherin expression is downregulated, disrupting cell-cell junctions, while and N-cadherin are upregulated, enhancing cytoskeletal reorganization and invasive potential. This transition is orchestrated by transcription factors such as and , induced by signals from the like TGF-β, and partial EMT states allow collective migration of cell clusters. To penetrate the (), cancer cells form specialized protrusions called invadopodia, which concentrate proteolytic activity for localized degradation. Matrix metalloproteinases (MMPs), particularly MMP-2 and MMP-9, are secreted or membrane-bound at invadopodia tips, cleaving ECM components like and to create paths for advancement. Invadopodia assembly involves polymerization driven by proteins like cortactin and Arp2/3, regulated by tyrosine kinases such as , enabling efficient tissue remodeling without widespread proteolysis. Intravasation occurs when invasive cells enter vascular or lymphatic systems, a rate-limiting step influenced by tumor-associated macrophages that facilitate vessel permeability. Circulating tumor cells (CTCs) must then evade anoikis, detachment-induced , through resistance conferred by integrin-mediated signaling; for instance, β1 and β3 engage remnants or platelets to activate PI3K/Akt pathways, promoting survival during transit. This anoikis resistance, often amplified by EMT-induced changes, ensures a subset of CTCs remains viable despite circulatory stresses like shear forces and immune surveillance. Colonization at secondary sites requires the establishment of a supportive metastatic niche, where CTCs adapt to foreign microenvironments. Primary tumors precondition distant organs via exosomes, small extracellular vesicles carrying miRNAs and proteins that mobilize bone marrow-derived cells, induce , and remodel to form pre-metastatic niches. For example, exosomal S100A8/A9 recruits myeloid cells to or liver sites, creating an inflammatory milieu that aids and outgrowth. This niche fosters dormancy escape and proliferation, transforming micrometastases into clinically detectable tumors.

Historical Development

Early Observations

The earliest documented observations of what we now recognize as cancerous growths date back to ancient civilizations, long before the advent of allowed for cellular examination. In , the , dating to approximately 2500 BCE and transcribed around 1600 BCE, provides the oldest known surgical treatise describing tumors, including eight cases of breast tumors characterized as bulging masses that could not be treated surgically due to their inoperable nature. These descriptions focused on macroscopic swellings and symptoms, such as ulceration and discharge, without any reference to cellular structures, reflecting the limitations of pre-microscopic . Similarly, the from the same era mentions treatments for various tumors using ointments and incantations, underscoring an early recognition of malignant-like growths as distinct pathological entities. The development of in the marked a pivotal shift, enabling the first direct visualizations of cancer at the cellular level. In 1838, German pathologist Johannes Müller published a seminal monograph on tumors, in which he systematically analyzed microscopic sections of human neoplasms and identified distinctive "cancer cells" in breast tumors through detailed illustrations and descriptions of their granular, irregular . Müller's work challenged prevailing humoral theories, such as the idea that cancer arose from , by demonstrating that tumors consisted of proliferated cells rather than fluid accumulations, based on scrapings and thin slices observed under early microscopes. This observation laid the groundwork for understanding cancer as a cellular , though the full implications would emerge later. Building on these insights, Rudolf Virchow advanced the field in 1858 with his lectures on cellular pathology, introducing the principle "omnis cellula e cellula" (every cell from a cell), which posited that all diseases, including cancer, originate from abnormal proliferation of preexisting cells rather than spontaneous generation. Virchow's theory linked cancerous growths to dysregulated cellular division, emphasizing histological examination of tissues to identify pathological changes, such as excessive cell multiplication in organs like the liver and uterus. His approach transformed pathology from a descriptive to an explanatory science, directly influencing the study of cancer as a disorder of cellular behavior. Concurrent with these advancements, early techniques enhanced the visibility of cancer cells' abnormal features under the . The hematoxylin-eosin (H&E) method, developed in the mid-to-late , became instrumental in highlighting pleomorphic (variably shaped) nuclei and cytoplasmic details in tumor tissues; hematoxylin, introduced for nuclear by Friedrich Böhmer in 1865, was combined with eosin's counterstain by around 1875 to differentiate cellular components effectively. This technique allowed pathologists to discern the irregular, hyperchromatic nuclei characteristic of cancer cells, facilitating more precise diagnoses of in biopsies.

Key Scientific Advances

In 1911, Peyton Rous demonstrated that a filterable agent from a could transmit the tumor to healthy birds, marking the first identification of an oncogenic virus and laying the groundwork for understanding viral contributions to cancer cell transformation. During the 1970s, the discovery of cellular oncogenes revolutionized cancer biology, with J. Michael Bishop and showing that the src gene in originated from normal cellular DNA, which could be activated to drive uncontrolled proliferation in cancer cells; this work, building on earlier retroviral studies, earned them the 1989 Nobel Prize in Physiology or Medicine. Their 1976 findings revealed that DNA sequences related to viral oncogenes, such as src kinase, are present in normal and mammalian genomes, suggesting that mutations in these proto-oncogenes underlie tumorigenesis. In the and , advances in pathways highlighted how cancer cells evade , exemplified by the identification of the gene in 1985 as a proto-oncogene involved in t(14;18) translocations in , where its overexpression inhibits and promotes cell survival. Concurrently, the discovery of as key regulators of the , first reported by in the early through studies on embryos showing periodic synthesis and degradation of cyclin proteins that activate cyclin-dependent kinases, provided mechanistic insights into how dysregulated arises in cancer cells; this contribution shared the 2001 Nobel Prize in Physiology or Medicine. From the 2000s onward, Douglas Hanahan and Robert A. Weinberg synthesized these insights in their seminal 2000 paper, outlining six "" that define the acquired capabilities of cancer cells, including self-sufficiency in growth signals, insensitivity to anti-growth signals, evasion of , limitless replicative potential, sustained , and tissue invasion and , which framed cancer as a of disrupted cellular . Their 2011 update expanded this framework to include two emerging hallmarks—reprogramming of energy metabolism and evading immune destruction—along with enabling characteristics like and tumor-promoting , integrating genomic and systems-level views of cancer cell behavior. Parallel to these conceptual advances, , adapted for mammalian cells in the early , enabled high-throughput in , allowing precise screens to identify essential genes and synthetic lethal interactions in tumor cells; a landmark 2014 study demonstrated genome-scale CRISPR screens in human cancer cell lines, revealing vulnerabilities like dependencies on oncogenic pathways that guide targeted therapies. These tools have since accelerated the dissection of cancer cell heterogeneity and resistance mechanisms, transforming experimental .

Therapeutic Approaches

Conventional Treatments

Conventional treatments for cancer primarily include , , and , which aim to eliminate or control tumors by exploiting the rapid of cancer cells while often affecting normal tissues as well. These approaches have been the of for decades, offering curative potential in many cases but limited by their non-specific nature and associated toxicities. Surgery involves the physical removal of tumor masses, providing definitive local control when feasible. Chemotherapy and radiation target cellular processes essential for division, such as DNA integrity, but can induce significant side effects due to their impact on healthy proliferating cells. Surgical excision is a primary intervention for localized tumors, where the goal is to remove the entire cancerous mass along with a margin of surrounding healthy to prevent recurrence. This procedure is particularly effective for solid tumors that are accessible and have not metastasized, achieving high cure rates in early-stage cancers like or . By directly excising the tumor, addresses the pathological accumulation of cancer cells without relying on systemic agents, though it may not eradicate microscopic distant spread. Chemotherapy employs cytotoxic drugs to interfere with and in rapidly proliferating cancer cells. Alkylating agents, such as , exert their effects by forming cross-linkages between DNA strands at the guanine N-7 position via their phosphoramide mustard, thereby preventing DNA unwinding and transcription essential for cell survival. Antimetabolites like 5-fluorouracil (5-FU) inhibit , an enzyme critical for synthesizing needed for , leading to nucleotide depletion and arrest primarily in the . Many chemotherapeutic agents exhibit phase specificity, targeting cancer cells during vulnerable stages of proliferation while sparing quiescent normal cells to varying degrees. Radiation therapy utilizes to induce lethal DNA double-strand breaks (DSBs) in cancer cells, a process that disrupts genomic integrity and triggers if unrepaired. Cancer cells often display heightened due to inherent deficiencies in DSB repair pathways, such as or , making them less able to recover from this damage compared to most normal tissues. This therapy is typically localized to the tumor site using external beam techniques, minimizing exposure to distant organs while exploiting the uncontrolled of cancer cells. A major limitation of these conventional treatments is their non-selective , which affects normal rapidly dividing cells, particularly in the , leading to myelosuppression. Myelosuppression manifests as reduced production of red blood cells, , and platelets, increasing risks of , infections, and , and often necessitates dose adjustments or supportive care like growth factors. This shared vulnerability with normal proliferative tissues underscores the need for adjunctive strategies to mitigate toxicities while enhancing antitumor efficacy.

Targeted and Emerging Therapies

Targeted therapies represent a in by selectively interfering with molecular pathways dysregulated in cancer cells, minimizing damage to healthy tissues. These approaches leverage the unique genetic and proteomic alterations in malignant cells, such as activated kinases or overexpressed receptors, to achieve precision in therapeutic intervention. Unlike conventional , which indiscriminately targets rapidly dividing cells, targeted therapies focus on cancer-specific vulnerabilities identified through molecular . Small-molecule inhibitors are orally bioavailable compounds designed to bind and inhibit specific enzymes critical for cancer cell survival. A landmark example is , which targets the BCR-ABL fusion resulting from the translocation in chronic myeloid leukemia (CML) cells. By competitively binding the ATP-binding site of BCR-ABL, imatinib blocks downstream signaling pathways like PI3K/AKT and MAPK, halting uncontrolled proliferation in CML cells. In a phase 1 trial, imatinib achieved complete hematologic responses in 98% of chronic-phase CML patients and major cytogenetic responses in 31%, establishing it as the first to dramatically improve survival in this disease. Broader classes of inhibitors (TKIs), such as those targeting in non-small cell or ALK in anaplastic large cell , similarly exploit kinase addictions in cancer cells, with response rates often exceeding 70% in biomarker-selected patients. Monoclonal antibodies offer extracellular targeting by binding cell surface antigens on cancer cells, triggering immune-mediated destruction or signaling inhibition. , a humanized IgG1 antibody, specifically binds the extracellular domain of the HER2 receptor overexpressed in approximately 15-20% of breast cancers, preventing HER2 dimerization and downstream activation of proliferative pathways. Additionally, recruits natural killer cells to induce (ADCC), enhancing tumor cell lysis. In a pivotal phase 3 trial, adding to first-line in HER2-positive patients doubled the median time to progression to 7.4 months compared to chemotherapy alone. Other antibodies, like rituximab targeting on B-cell lymphomas, similarly promote ADCC and , achieving complete response rates of up to 80% in combination regimens. Emerging immunotherapies, including chimeric antigen receptor (CAR) T-cell therapies, engineer patient-derived T cells to express synthetic receptors that recognize tumor s. CAR-T cells targeting , a surface marker on B-cell malignancies, have shown transformative efficacy in relapsed/refractory hematologic cancers. For instance, , a CD19-directed CAR-T product, reprograms T cells to release perforin and granzymes upon binding, leading to targeted of malignant B cells. In the ZUMA-1 trial, achieved an objective response rate of 83% in refractory large patients, with 58% achieving complete remission at a follow-up of 15.4 months. inhibitors, such as imetelstat, address the immortality conferred by reactivation in cancer cells by competitively binding the RNA component, shortening telomeres and inducing or . In the phase 3 IMerge trial, imetelstat achieved 8-week red blood cell transfusion independence in 39.1% of lower-risk patients refractory to erythropoiesis-stimulating agents, compared to 15% with placebo, leading to its FDA approval in June 2024. Gene therapies harness molecular tools to directly modify or destroy cancer cells. CRISPR-Cas9-based editing enables precise correction of oncogenic mutations in cancer cells, such as restoring tumor suppressor function or disrupting driver genes like . Preclinical studies have demonstrated CRISPR-mediated of PD-1 in T cells to enhance anti-tumor immunity, with early-phase trials showing feasibility in editing hematopoietic stem cells for . Oncolytic viruses, engineered to selectively replicate in and lyse cancer cells with defective antiviral responses, represent another modality. (T-VEC), a modified expressing GM-CSF, infects cells, replicates intracellularly, and causes oncolysis while stimulating systemic anti-tumor immunity. In the OPTiM phase 3 trial, intratumoral T-VEC improved durable response rates to 16.3% in advanced patients versus 2.1% with GM-CSF alone, leading to its FDA approval in 2015. These approaches continue to evolve, with ongoing trials combining them for synergistic effects against heterogeneous cancer cell populations.

References

  1. [1]
    What Is Cancer? - NCI - National Cancer Institute
    Oct 11, 2021 · Cancer cells differ from normal cells in many ways. For instance, cancer cells: grow in the absence of signals telling them to grow. Normal ...Common Cancer Myths and · Cancer Statistics · Defining Cancer · Tumor Grade
  2. [2]
    Cell Biology of Cancer - SEER Training Modules
    A cancer cell is a cell that grows out of control. Unlike normal cells, cancer cells ignore signals to stop dividing, to specialize, or to die and be shed.
  3. [3]
    Hallmarks of Cancer: New Dimensions | Cancer Discovery
    Jan 12, 2022 · The hallmarks of cancer were proposed as a set of functional capabilities acquired by human cells as they make their way from normalcy to neoplastic growth ...Unlocking Phenotypic Plasticity · Polymorphic Microbiomes · Senescent Cells
  4. [4]
    Cancer Classification - SEER Training Modules
    Cancers are classified in two ways: by the type of tissue in which the cancer originates (histological type) and by primary site, or the location in the body ...
  5. [5]
  6. [6]
    TUMOR CELL MORPHOLOGY - Comparative Oncology - NCBI - NIH
    Morphologically, the cancerous cell is characterized by a large nucleus, having an irregular size and shape, the nucleoli are prominent, the cytoplasm is scarce ...
  7. [7]
    A Tale of Two States: Normal and Transformed, With and ... - NIH
    For many years, major differences in morphology, motility, and mechanical characteristics have been observed between transformed cancer and normal cells.
  8. [8]
    Cell adhesion in cancer: Beyond the migration of single cells - PMC
    Two major hallmarks of cancer, loss of cell-to-cell adhesion and anchorage-independent growth, are both dependent on cell adhesion molecules.Missing: cycle | Show results with:cycle
  9. [9]
    Interdependence of cell attachment and cell cycle signaling - PMC
    After cell transformation, transformed cells become anchorage-independent and cease to be contact ... disrupted apical junction complexes and show loss of cell ...
  10. [10]
    Epithelial cell polarity and tumorigenesis - PubMed Central - NIH
    A strong correlation between malignancy and loss of epithelial organization has been histologically documented for almost types of tumor deriving from ...
  11. [11]
    Clinical and Biological Implications of the Tumor Microenvironment
    Malignant tumors are disorganized masses of living material composed of different tissues in which cancerous and normal components are intimately intermingled ...
  12. [12]
    What Makes a Cancer Cell a Cancer Cell? - NCBI - NIH
    Loss of Cell-Cycle Control and Resistance to Apoptosis. Normal cells respond to a variety of suboptimal growth conditions by entering a quiescent phase in ...
  13. [13]
    Non‐genetic heterogeneity, altered cell fate and differentiation therapy
    Feb 8, 2021 · Altered capacity for self‐renewal and differentiation is a hallmark of cancer, and many tumors are composed of cells with a developmentally immature phenotype.Missing: cycle | Show results with:cycle
  14. [14]
    The Cancer Genome Atlas Program (TCGA) - NCI
    The Cancer Genome Atlas (TCGA) is a landmark cancer genomics program that sequenced and molecularly characterized over 11000 cases of primary cancer samples ...GDC Data Portal · Access OCG Data · Using TCGA · Program History<|separator|>
  15. [15]
    Tumor classification: molecular analysis meets Aristotle - BMC Cancer
    Mar 17, 2004 · This challenge is intended to lay the groundwork for changing the basis of tumor classification from morphological to molecular characteristics.
  16. [16]
    DeepCC: a novel deep learning-based framework for cancer ...
    Aug 16, 2019 · In PAM50, five distinct (intrinsic) molecular subtypes are defined: Basal-like, Her2, Luminal A, Luminal B, and Normal-like. Using TCGA RNA-Seq ...
  17. [17]
    Molecular Characterization and Classification of HER2-Positive ...
    Nov 4, 2024 · HER2-positive breast cancer represents a distinct aggressive subtype, accounting for 15% to 20% of all breast cancers, and features the ...
  18. [18]
    Co-occurring Genomic Alterations Define Major Subsets of KRAS ...
    Integrative analysis identified three major clusters of KRAS-mutant lung adenocarcinoma characterized by co-occurring genetic events in STK11/LKB1, TP53,
  19. [19]
    Molecular Features and Survival Outcomes of the Intrinsic Subtypes ...
    Aug 19, 2014 · Here, we evaluated the molecular features and survival outcomes of the intrinsic subtypes within HER2+ breast cancer.
  20. [20]
    A machine learning toolkit for subtyping cancer in existing and new ...
    Mar 5, 2025 · Tumour subtypes based on molecular characteristics can be valuable to care for patients with cancer and informative to the prognosis and ...
  21. [21]
    Update on Oral Epithelial Dysplasia and Progression to Cancer - PMC
    Abnormal variation in nuclear size and shape (anisonucleosis and pleomorphism) ... changes such as hyperchromatism, and prominent cell and nuclear pleomorphism may ...
  22. [22]
    CANCER DIAGNOSIS - Comparative Oncology - NCBI Bookshelf - NIH
    The most important nuclear changes are: different sizes and shapes, alterations in the structure of chromatin and nucleoli. There are also changes in the ...<|separator|>
  23. [23]
    Diagnostic cellular abnormalities in neoplastic and non ... - NIH
    Nuclear irregularity, increased N/C ratio, conspicuous nucleoli, crowding and hyperchromasia are the most useful indicators of malignancy.
  24. [24]
    Clinicopathological Characteristics of Pleomorphic High-Grade ...
    Aug 15, 2020 · PHSILs were histologically characterized by significant nuclear enlargement, marked pleomorphism, hyperchromasia, increased mitotic activity, and frequent ...
  25. [25]
    Nucleolus, Ribosomes, and Cancer - PMC - PubMed Central - NIH
    The available evidence indicates that the morphological and functional changes in the nucleolus, widely observed in cancer tissues, are a consequence of both ...
  26. [26]
    The relationship between the nucleolus and cancer
    Increased number and size of nucleoli, which most likely reflect the increased rate of ribosome biogenesis, are frequently observed in malignant tumors such as ...
  27. [27]
    Impaired ribosome biogenesis: mechanisms and relevance to ...
    Importantly, increased size of nucleoli usually reflects intense ribosome biogenesis and has been recognized by physicians for a long time as a hallmark of many ...Impaired Rrna Synthesis · Impaired Rrna Maturation · Aberrant Ribosome Biogenesis...
  28. [28]
    Isotropic 3D Nuclear Morphometry of Normal, Fibrocystic and ... - NIH
    Jan 5, 2012 · Margination of high-density clumps in the abnormal and especially in metastatic cell nuclei suggests likely functional significance. It could ...
  29. [29]
    Micronuclei and Cancer - PMC - PubMed Central - NIH
    Micronuclei arise from chromosome mis-segregation and characterize tumors with elevated rates of chromosomal instability. Although their association with cancer ...
  30. [30]
    Micronuclei as biomarkers of DNA damage, aneuploidy, inducers of ...
    Micronuclei (MNi) are among the most widely studied biomarkers of DNA damage and chromosomal instability in humans. They originate from chromosome fragments ...
  31. [31]
    A new assay for measuring chromosome instability (CIN) and ...
    May 22, 2013 · Micronucleus assays (MNi) are widely used to detect broken or lagging chromosomes, but fail to detect non-balanced chromosome segregation [7].
  32. [32]
    micronuclAI enables automated quantification of micronuclei for ...
    Mar 4, 2025 · An automated, single cell quantitative imaging microscopy approach to assess micronucleus formation, genotoxicity and chromosome instability.
  33. [33]
    Pathologic and molecular features of hepatocellular carcinoma
    Small cell change is characterized by decreased cell volume, increased nuclear-cytoplasmic ratio, cytoplasmic basophilia, mild nuclear pleomorphism, and ...
  34. [34]
    Mitochondria and Cancer - PMC - PubMed Central - NIH
    Mitochondria play a central and multi-functional role in malignant tumor progression, and targeting mitochondria provides therapeutic opportunities.
  35. [35]
    Targeting mitochondria metabolism for cancer therapy - PMC
    The high levels of mitochondrial ROS activate signaling pathways proximal to the mitochondria to promote cancer cell proliferation and tumorigenesis. However, ...
  36. [36]
    A Review of Advances in Mitochondrial Research in Cancer - PMC
    Nov 2, 2024 · This review provides a comprehensive analysis of the various aspects of mitochondrial aberrations and addresses drugs that target mitochondrial therapy.
  37. [37]
    Involvement of Actin and Actin-Binding Proteins in Carcinogenesis
    In the case of cancer cells, actin and ABPs (actin-binding proteins) are involved in all stages of carcinogenesis.
  38. [38]
    Scared stiff: Stabilizing the actin cytoskeleton to stop invading ...
    We discovered that stabilizing the actin cytoskeleton via elevated expression of Tm5NM1 specifically inhibits mesenchymal-type cancer cell invasion.
  39. [39]
    Adaptation of the Golgi Apparatus in Cancer Cell Invasion and ...
    Taken together, Golgi disorganization, ranging from mild enlargement to vesicular dispersal, has been linked to the orchestration of molecular pathways related ...
  40. [40]
    Autophagy and autophagy-related proteins in cancer - PMC
    Jan 22, 2020 · Autophagy plays a dynamic tumor-suppressive or tumor-promoting role in different contexts and stages of cancer development.
  41. [41]
    Understanding Cytokinesis Failure - PMC - PubMed Central - NIH
    Cytokinesis failure leads to both centrosome amplification and production of tetraploid cells, which may set the stage for the development of tumor cells.
  42. [42]
    A non-genetic route to aneuploidy in human cancers - PMC - NIH
    Aneuploidy can result from cytokinesis failure, which produces binucleate cells that generate aneuploid offspring with subsequent divisions. In cancers, ...
  43. [43]
    RAS oncogenes: weaving a tumorigenic web - PMC - PubMed Central
    In this Review, we describe how RAS oncogenes exploit their extensive signalling reach to affect multiple cellular processes that drive tumorigenesis.
  44. [44]
    The MYC oncogene — the grand orchestrator of cancer growth ... - NIH
    Abstract. The MYC proto-oncogenes encode a family of transcription factors that are among the most commonly activated oncoproteins in human neoplasias.
  45. [45]
    Tumor-Suppressor Functions of the TP53 Pathway - PMC
    The TP53 protein is a critical tumor suppressor that plays a fundamental and multifaceted role in the development of cancer and cancer therapy.
  46. [46]
    BRCA Gene Changes: Cancer Risk and Genetic Testing Fact Sheet
    Jul 19, 2024 · About 30%–40% of breast cancer survivors with inherited BRCA1 changes and 25% of those with inherited BRCA2 changes will develop contralateral ...
  47. [47]
    Penetrance estimates for BRCA1 and BRCA2 based on genetic ...
    Breast cancer penetrance to 70 and to 80 years was 68% (95%CI 64.7–71.3%) and 79.5% (95%CI 75.5–83.5%) respectively for BRCA1 and 75% (95%CI 71.7–78.3%) and 88% ...
  48. [48]
    Accumulation of driver and passenger mutations during tumor ...
    Passenger mutations are defined as those which do not alter fitness but occurred in a cell that coincidentally or subsequently acquired a driver mutation, and ...Abstract · Sign Up For Pnas Alerts · Oncogenes And Tumor...
  49. [49]
    Distinguishing between driver and passenger mutations in ...
    In somatic cancer genomes, delineating genuine driver mutations against a background of multiple passenger events is a challenging task.
  50. [50]
    Losing balance: the origin and impact of aneuploidy in cancer - PMC
    It is important to recognize that aneuploidy and CIN are not synonymous: aneuploidy defines the 'state' of having abnormal chromosome content, whereas CIN ...
  51. [51]
    The Philadelphia chromosome in leukemogenesis - PMC
    Common gene abnormalities found in Ph-positive leukemia are discussed in Table 1. Table 1. Recurrent genomic abnormalities in Philadelphia chromosome (Ph)- ...
  52. [52]
    DNA damage repair: historical perspectives, mechanistic pathways ...
    Jul 9, 2021 · This review highlights the roles of DNA damage, response and repair in cancer development and cancer therapy from perspectives of the ...
  53. [53]
    Deficient mismatch repair: Read all about it (Review) - PMC - NIH
    When the MMR system develops a functional error or defect, this results in a particular phenotype called microsatellite instability (MSI). This is characterised ...
  54. [54]
    Homologous Recombination Deficiency: Cancer Predispositions ...
    Jun 2, 2021 · Alteration in HR genes is prevalent among many cancer types (13%–17%), especially breast, ovarian, and pancreatic cancers [1]. BRCA1 and BRCA2 ...
  55. [55]
    calculating how cancers may arise with normal mutation rates - PMC
    Jan 5, 2010 · Normal mutation rates are low and around one mutation per billion bases per division ... With a mutation rate of 10-9 per base per division ...
  56. [56]
    Structure and Maintenance of Telomeric DNA Repeats and ...
    Telomeric repeats may be regular (such as TTAGGG in humans and most vertebrates or TTGGGG in Tetrahymena) or irregular (such as G1–3T in yeast Saccharomyces ...
  57. [57]
    Telomerase and cancer | Human Molecular Genetics
    With each cell division, telomeres shorten by ∼50–200 bp (23), primarily because the lagging strand of DNA synthesis is unable to replicate the extreme 3′ end ...
  58. [58]
    Role of Telomeres and Telomerase in Aging and Cancer - PMC - NIH
    When a few telomeres become critically shortened there is a growth arrest state, at which time a DNA damage signal and cellular senescence is normally triggered ...
  59. [59]
    Telomere and telomerase in oncology | Cell Research - Nature
    Mar 1, 2002 · In this review we discuss the structure and function of telomere and telomerase and their roles in cell immortalization and oncogenesis.
  60. [60]
    TERT gene: its function and dysregulation in cancer
    In this review, we summarise the function and structure of telomerase reverse transcriptase (TERT) in humans, including its regulation.
  61. [61]
    The regulations of telomerase reverse transcriptase (TERT) in cancer
    Jan 26, 2024 · It is indicated that TERT overexpression, caused by promoter mutation, is attributed to telomerase abnormal activation in cancer, although only ...
  62. [62]
    Prevalence of telomerase activity in human cancer - PubMed
    Telomerase is activated in 80-90% of human carcinomas, but not in normal somatic cells, therefore, its detection holds promise as a diagnostic marker for cancer ...
  63. [63]
    Roles of telomeres and telomerase in cancer, and advances in ...
    Jun 20, 2016 · In the large majority of cancer cells, telomere length is maintained by telomerase. Thus, telomere length and telomerase activity are crucial ...
  64. [64]
    ALTernative Telomere Maintenance and Cancer - PubMed Central
    Alternative lengthening of telomeres (ALT) is a recombination-based telomere maintenance mechanism (TMM) utilized by 10-15% of human cancers. ALT is ...
  65. [65]
    The alternative lengthening of telomeres mechanism ... - PNAS
    Sep 19, 2022 · Alternative lengthening of telomeres (ALT) cancers achieve immortality by reelongating their telomeres in the G2 and M phases of the cell cycle ...
  66. [66]
  67. [67]
  68. [68]
    Inhibition of platelet-derived growth factor signalling induces ...
    2 мар. 2004 г. · Malignant gliomas highly coexpress platelet-derived growth factor (PDGF) and its receptor, suggesting the presence of an autocrine loop.
  69. [69]
    The Molecular Balancing Act of p16INK4a in Cancer and Aging - NIH
    Several lines of evidence suggest that p16INK4a may function both through CDK4/6-dependent and -independent mechanisms to regulate the cell cycle. CYCLIN D-CDK4 ...
  70. [70]
  71. [71]
    Invasion and metastasis in cancer: molecular insights and ... - Nature
    The local tumor microenvironment (TME) supports EMT and ECM breakdown, enabling intravasation into the bloodstream or lymphatic system (Fig. 3).
  72. [72]
    The gate to metastasis: key players in cancer cell intravasation
    Jun 2, 2021 · Here, we discuss the role of intrinsic properties of cancer cells, tumor microenvironment, and mechanical cues in the intravasation process.Abstract · Introduction · Intrinsic properties of cancer... · Metabolism of cancer cells
  73. [73]
    Epithelial-mesenchymal transition - Cell Press
    Oct 2, 2025 · Epithelial-mesenchymal transition (EMT) is a fundamental mechanism involved in the morphogenesis of metazoans.Epithelial-Mesenchymal... · Emt In Gastrulation... · Emt And Metabolism
  74. [74]
    Epithelial-Mesenchymal Transition: A Fundamental Cellular and ...
    Feb 11, 2024 · EMT is a complex process that involves many transcription factors and genes that enable the tumor cell to leave the primary location, invade the basement ...3. Emt Induction And... · 4. Emt Proteome And Genome... · Figure 2
  75. [75]
    Two distinct epithelial-to-mesenchymal transition programs control ...
    Oct 16, 2024 · Epithelial-to-mesenchymal transition (EMT) triggers cell plasticity in embryonic development, adult injured tissues and cancer.
  76. [76]
    Invadopodia: clearing the way for cancer cell invasion - PMC
    Although invadopodia are very complex and dynamic structures, it is expected that their key regulators would be good targets for anticancer therapy.
  77. [77]
    Matrix Metalloproteinases in Cancer Cell Invasion - NCBI - NIH
    In this chapter, we discuss the role of MMPs and their inhibitors in tumor cell invasion as a basis for prognostication and targeted therapeutic intervention.Missing: paper | Show results with:paper
  78. [78]
    Invadopodia in cancer metastasis: dynamics, regulation, and ...
    May 16, 2025 · Growing evidence suggests that invadopodia plays a pivotal role in cancer invasion and metastasis. This highlights the potential of targeting ...
  79. [79]
    Tumor cell intravasation - PMC - NIH
    Although this review is on intravasation by cancer cells, intravasation is a naturally occurring process that is important during development, as well as for a ...
  80. [80]
    Anoikis molecular pathways and its role in cancer progression
    Cancer cells can achieve resistance to anoikis through: i) a specific switch in their integrins, thereby adapting to the metastatic site, ii) undergoing to EMT, ...Missing: paper | Show results with:paper
  81. [81]
    Pre-metastatic niche: formation, characteristics and therapeutic ...
    Sep 25, 2024 · This review delves into the intricate landscape of the pre-metastatic niche, focusing on the roles of tumor-derived secreted factors, ...Missing: paper | Show results with:paper
  82. [82]
    Effects of exosomes on pre-metastatic niche formation in tumors - PMC
    Mar 11, 2019 · In this review, we analyzed the effects of exosomes on pre-metastatic niches from various perspectives, including inflammation, immune response, angiogenesis, ...
  83. [83]
    Exosomes: Key mediators of metastasis and pre-metastatic niche ...
    The roles exosomes have in pre-metastatic niche formation are diverse, and range from metabolic reprogramming, to recruitment of numerous immune and non-immune ...Missing: paper | Show results with:paper
  84. [84]
    The Key Role of Exosomes on the Pre-metastatic Niche Formation in ...
    Evidence shows that tumor-derived exosomes mediate tumor pre-metastatic niche (PMN) remodeling to establish a supportive and receptive niche.Missing: paper | Show results with:paper
  85. [85]
    Cancer: A Historic Perspective - SEER Training Modules
    Two of them, known as the "Edwin Smith" and "George Ebers" papyri, contain descriptions of cancer written around 1600 B.C., and are believed to date from ...
  86. [86]
    The past and future of breast cancer treatment—from the papyrus to ...
    Jun 8, 2017 · The papyrus describes 48 generic clinical cases where rudimentary surgical procedures were used to treat wounds, fractures, and cancer, ...
  87. [87]
    A note from history: Landmarks in history of cancer, part 3
    Jul 12, 2011 · In 1838, Johannes Muller illustrated for the first time the microscopic appearance of cancer. (Top) Muller's Figure 14 depicts breast cancer ...
  88. [88]
    Foundation of Diagnostic Cytology
    Johannes Müller (1801–1858), a pathologist in Berlin (Fig. 1⇓), was the first, in 1838, to show cancer cells as they appeared in the microscope on scrapings ...
  89. [89]
    [PDF] Rudolf Virchow (1821-1902): Founder of Cellular Pathology and ...
    Virchow's Cellular Pathology is based on the cellular theory, formulated by the adage “Omnis cel- lula a cellula”, which opposed completely the notion of ...
  90. [90]
    Rudolf Virchow - PMC - NIH
    He stated that all diseases involve changes in normal cells, that is, all pathology ultimately is cellular pathology. This insight led to major progress in the ...
  91. [91]
    From silks to science: The history of hematoxylin and eosin staining
    Jan 11, 2023 · These dyes possess a profound and alluring history, which includes stories of the fabric industry, pirates, fine art, and eventually histology.
  92. [92]
    Hematoxylin and eosin staining of tissue and cell sections - PubMed
    May 1, 2008 · INTRODUCTIONHematoxylin and eosin (H&E) stains have been used for at least a century and are still essential for recognizing various tissue ...
  93. [93]
    CRISPR/Cas9 somatic multiplex-mutagenesis for high-throughput ...
    Oct 27, 2015 · Our studies show that multiplexed CRISPR/Cas9 can be used for recessive genetic screening or high-throughput cancer gene validation in mice.
  94. [94]
    Targeting HER2-positive breast cancer: advances and future ...
    Nov 7, 2022 · Trastuzumab binds to the extracellular domain (ECD) of HER2, suppresses intracellular HER2 signalling pathways, inhibits cell cycle arrest and ...
  95. [95]
    Efficacy and Safety of a Specific Inhibitor of the BCR-ABL Tyrosine ...
    Apr 5, 2001 · Efficacy and Safety of a Specific Inhibitor of the BCR-ABL Tyrosine Kinase in Chronic Myeloid Leukemia. Authors: Brian J. Druker, M.D., Moshe ...
  96. [96]
    Chronic myeloid leukemia: the paradigm of targeting oncogenic ...
    Feb 19, 2018 · This review summarizes the main biological acquisitions about BCR-ABL1 as a therapeutically druggable oncogenic tyrosine kinase and provides an update on drug ...Missing: paper | Show results with:paper
  97. [97]
    Use of Chemotherapy plus a Monoclonal Antibody against HER2 for ...
    Mar 15, 2001 · Trastuzumab increases the clinical benefit of first-line chemotherapy in metastatic breast cancer that overexpresses HER2.
  98. [98]
    Axicabtagene Ciloleucel CAR T-Cell Therapy in Refractory Large B ...
    Drs. Neelapu and Locke contributed equally to this article. This article was published on December 10, 2017, at NEJM.org. Supported by Kite Pharma and the ...
  99. [99]
    Comprehensive review of CRISPR-based gene editing
    Jan 9, 2024 · The CRISPR system is a revolutionary genome editing tool that has the potential to revolutionize the field of cancer research and therapy.
  100. [100]
    Talimogene Laherparepvec: Moving From First-In-Class to Best-In ...
    Feb 21, 2022 · While T-VEC was approved for the treatment of melanoma, the virus was able to demonstrate activity against tumor cells derived from other ...Abstract · Introduction · The Development of... · Real World Experience With T...