Fact-checked by Grok 2 weeks ago

Chromothripsis

Chromothripsis is a mutational process characterized by the massive shattering of one or more chromosomes into hundreds of fragments during a single catastrophic cellular event, followed by their random and erroneous reassembly via pathways, leading to complex genomic rearrangements. This phenomenon, first described in , manifests as oscillating copy-number states (gains and losses) confined to localized chromosomal regions, often retaining heterozygosity in retained segments, distinguishing it from gradual accumulation of mutations. Key features of chromothripsis include its occurrence in a "one-off" crisis, typically involving or for fragment re-ligation, and association with double-strand breaks clustered in space and time. It predominantly affects cancer cells but has also been identified in contexts, contributing to congenital disorders through structural variations that disrupt multiple genes and regulatory elements. In tumors, chromothripsis drives oncogenesis by simultaneously inactivating tumor suppressors (e.g., TP53, ) and amplifying oncogenes (e.g., , ) in a single event, accelerating tumor evolution. Prevalence studies reveal chromothripsis in approximately 29–40% of human cancers across 2,658 analyzed whole-genome sequences, with higher rates in specific types such as 100% of liposarcomas, 77% of s, and 60–65% of pancreatic cancers. It often involves multiple chromosomes (up to ≥5 in 61% of cases) and co-occurs with or TP53 mutations, though it can arise independently. Beyond , germline chromothripsis underlies developmental syndromes, as seen in cases of severe congenital abnormalities from inherited shattered chromosomes affecting protein-coding genes. Mechanistically, chromothripsis is triggered by mitotic errors, including formation, bridges, or premature condensation, leading to rupture and DNA pulverization. Advances as of 2023 highlight mitotic clustering of pulverized fragments and tethering proteins like CIP2A-TOPBP1 that facilitate their inheritance, promoting ongoing genomic instability; more recent studies (2024–2025) have further elucidated ongoing chromothripsis in and multi-omic profiles in , reinforcing its role in tumor evolution. Clinically, it correlates with aggressive disease progression, therapy resistance, and poor prognosis in cancers like and , underscoring its role as a hallmark of punctuated .

Discovery and Definition

Initial Discovery

Chromothripsis was first identified in 2011 through whole-genome sequencing of a (CLL) sample, where researchers observed an unprecedented pattern of massive, localized genomic rearrangements confined to the long arm of (4q). This discovery, led by Philip J. Stephens and colleagues at the Wellcome Trust Sanger Institute, revealed 42 rearrangement breakpoints within a ~30-megabase region, suggesting a single catastrophic event rather than the incremental accumulation of mutations typically seen in cancer evolution. The phenomenon was termed "chromothripsis," derived from the Greek words for "color" (chromo) and "shattering" (thripsis), to describe the shattering of one or a few chromosomes into tens to hundreds of pieces, followed by an apparently random reassembly. In the , these rearrangements included deletions, inversions, and duplications that occurred in a localized manner, starkly contrasting with the progressive, genome-wide alterations observed in most tumors. The event was inferred to have happened in a single cellular crisis, as evidenced by the absence of progressive intermediate states in the sequencing data. Key genomic signatures distinguished this process: breakpoints were tightly clustered, with most occurring within the affected region and few elsewhere in the , indicating simultaneity rather than sequential events. Additionally, copy number profiles showed characteristic oscillations, alternating between two copies (diploid) and zero copies (homozygous deletion) in a 1:1 pattern along the chromosome arm, reflecting the rejoining of shattered fragments without distant inter-chromosomal exchanges. Initial analysis across a broader suggested chromothripsis occurs in 2–3% of all cancers, with a notably higher prevalence of approximately 25% in bone cancers such as . This finding, based on paired-end sequencing of 24 tumor genomes and validation in additional samples, highlighted chromothripsis as a widespread yet previously unrecognized driver of oncogenesis.

Core Definition and Characteristics

Chromothripsis is defined as a catastrophic genomic event in which one or a few chromosomes shatter into numerous fragments—typically 10 to 100 or more—which are subsequently reassembled in a randomized manner primarily through (NHEJ), resulting in complex structural variants such as inversions, deletions, duplications, and translocations. This process contrasts with gradual accumulation of , occurring instead as a singular, rapid phenomenon within a single , often leading to localized regions of extreme genomic instability. Key characteristics of chromothripsis include its confinement to one or a limited number of chromosomes, exhibiting high density with tens to hundreds of rearrangement junctions clustered in a confined genomic region, and patterns of oscillating copy number states (gains and losses) without evidence of progressive evolutionary steps. These events frequently produce amplifications or homozygous deletions that can drive oncogenesis, though they are not exclusively oncogenic and have been observed in non-cancerous contexts such as congenital disorders. First identified in , chromothripsis exemplifies how a single disruptive episode can profoundly alter the . Chromothripsis is distinct from related phenomena like chromoplexy, which involves chain-like rearrangements across multiple chromosomes with fewer breakpoints, and kataegis, which features hypermutation through clustered single-nucleotide variants at rearrangement sites rather than large-scale shattering. By 2025 estimates, chromothripsis occurs in approximately 30–50% of cancers across various types, underscoring its as a driver of genomic chaos in tumorigenesis.

Genomic Alterations

Chromosome Breakage Patterns

Chromothripsis involves the formation of massive, clustered double-strand breaks (DSBs) that shatter one or a few chromosomes into numerous fragments, typically tens to hundreds of breaks occurring in a single catastrophic event. These breaks are spatially confined and generate fragments of random sizes ranging from kilobases (kb) to megabases (Mb), with examples including segments as small as 150 kb and as large as 27 Mb. The breakage often manifests in the G1 or G2 phases of the cell cycle, where DSB processing leads to localized genomic instability. Notably, the resulting junctions from these breaks are predominantly tailless or blunt-ended, lacking microhomology, which distinguishes chromothripsis from other repair pathways involving templated insertions. A defining feature of chromothripsis in copy number profiles is the characteristic sawtooth pattern, marked by multiple rapid oscillations between two or three copy number states, such as alternating gains and losses along the affected . This oscillatory profile arises from the random reassembly of shattered fragments and is often confined to a single or subregion, sparing distant parts of the genome. In non-rearranged regions adjacent to the shattered area, retention of heterozygosity is typically preserved, contrasting with the loss of heterozygosity (LOH) commonly observed within the chromothripsis-affected segments. The structural rearrangements resulting from chromothripsis breakage include a variety of local alterations, such as inversions, deletions, duplications, and translocations, primarily within the affected arm. Inversions often appear as head-to-head or tail-to-tail orientations, while deletions and duplications contribute to the copy number oscillations, and translocations can link distant fragments from the same chromosome without widespread interchromosomal exchanges. These rearrangements are stitched together via error-prone mechanisms like (NHEJ), leading to complex but localized genomic architecture. Recent insights from 2025 highlight a distinct subtype of chromothripsis known as loss-translocation-amplification (LTA), particularly prevalent in , where it drives sequential genomic events including initial loss of tumor suppressor regions, followed by translocation and subsequent amplification of oncogenes.01418-1) This LTA pattern exemplifies how breakage can propagate specific structural outcomes, contributing to aggressive tumor evolution in high-grade osteosarcomas.

Repair and Reassembly Processes

Following the massive chromosome breakage in chromothripsis, the primary cellular repair mechanism is the error-prone non-homologous end joining (NHEJ) pathway, which directly ligates double-strand break ends without requiring homologous templates, frequently producing blunt-end junctions. This process is mediated by the Ku70/Ku80 heterodimer, which rapidly binds to DNA ends and recruits DNA-dependent protein kinase catalytic subunit (DNA-PKcs) to form a synaptic complex that stabilizes the breaks. Subsequent recruitment of XRCC4 and DNA ligase IV (LIG4) enables the final ligation, often with minimal or no nucleotide loss, though small insertions or deletions can occur due to the pathway's inherent inaccuracy. Experimental evidence from NHEJ-deficient cells demonstrates that inactivation of core components like DNA-PKcs or LIG4 drastically impairs the formation of complex, clustered rearrangements, shifting outcomes toward simpler translocations or unresolved damage. In certain cases, alternative end-joining pathways such as (MMEJ) contribute to repair, particularly when short stretches of microhomology (typically 2–15 base pairs) at break sites allow annealing and deletion of intervening sequences. MMEJ involves polymerases like Polθ and ligase III, operating as a to NHEJ during replication , and is implicated in a subset of chromothripsis breakpoints exhibiting microhomology signatures. However, genomic analyses reveal that MMEJ plays a limited role overall, as the majority of chromothripsis junctions are blunt-ended without microhomology, underscoring NHEJ's dominance in this context. Reassembly of the fragmented occurs through randomized of pieces during the S/G2 phases, enabling rapid but chaotic restructuring within a single . This process may be facilitated by localized disruptions, including potential rupture, which compromises integrity and promotes error-prone repair in confined subnuclear spaces like micronuclei. The resultant derivative chromosomes consist of fused segments from the original , often rearranged in inverted, duplicated, or translocated configurations that reflect the haphazard joining. These repair outcomes drive oncogenic potential by juxtaposing proto-oncogenes with active regulatory elements, such as enhancers, leading to their aberrant activation and contributing to tumor initiation or progression. For instance, chromothripsis-induced fusions can amplify oncogenes like or create novel drivers like EML4-ALK, amplifying selective pressures in cancer evolution.

Underlying Mechanisms

Micronuclei and Mitotic Errors

One prominent mechanism underlying chromothripsis involves the formation of micronuclei during , where mis-segregated chromosomes are encapsulated in separate, aberrant nuclear structures outside the primary nucleus. These micronuclei often exhibit defective nuclear envelopes due to asynchronous assembly, leading to premature disassembly of the and subsequent exposure of to cytoplasmic factors, which triggers extensive DNA double-strand breaks (DSBs) and chromosome pulverization.00709-5) Mitotic errors, such as chromosome missegregation, frequently initiate this process by generating lagging chromosomes that fail to align properly on the mitotic , resulting in their exclusion into micronuclei. Overexpression of family member 18A (KIF18A), a involved in chromosome congression, or topoisomerase II alpha (TOP2A), which resolves DNA catenations during segregation, can exacerbate these errors by disrupting spindle dynamics and increasing the incidence of lagging chromosomes, thereby promoting micronuclei formation and downstream chromothripsis events. Once formed, micronuclear rupture exposes pulverized to cytoplasmic exonucleases like three-prime repair exonuclease 1 (TREX1), which further degrades DNA ends and amplifies breakage.00709-5) Experimental evidence for this micronuclei model was first robustly demonstrated in 2012, when Crasta et al. showed that inducing micronuclei in human cells via mitotic errors led to shattering confined to the micronucleated , with reassembly patterns mimicking chromothripsis observed in cancers. Subsequent studies confirmed that these events occur independently of or arrest, relying instead on instability. More recent 2025 research has linked this pathway to TP53-deficient cells, where loss impairs checkpoints that eliminate cells with segregation errors, allowing micronuclei-driven chromothripsis to propagate and contribute to ongoing genomic instability. In tumor evolution, chromothripsis events originating from micronuclei often occur subclonally, driving intratumor heterogeneity and progression. For instance, multi-region whole-genome sequencing of osteosarcomas in 2025 revealed ongoing, subclonal chromothripsis in 74% of cases, frequently tied to mitotic dysfunction and micronuclei formation, highlighting its role in sustaining cancer adaptability without immediate cell death. Repair of these breaks primarily involves (NHEJ), as noted in broader genomic studies, but the initial damage stems from mitotic entrapment.01418-1)

Radiation and Cellular Stress Models

Ionizing radiation exposure, particularly during , can induce chromothripsis by generating clusters of DNA double-strand breaks (DSBs) that overwhelm repair mechanisms and lead to catastrophic shattering. This process is amplified in vulnerable cell cycle phases, where even low doses of —such as low-linear energy transfer (LET) types—produce persistent DSB clusters of 2–3 breaks, promoting complex rearrangements rather than simple . Experimental models using γ-irradiation at doses like 2 demonstrate that these clustered DSBs destabilize , resulting in chromothripsis-like patterns confined to specific chromosomal regions. Another mechanism involves aborted apoptosis, where partial activation of caspases triggers DNA fragmentation without completing cell death, allowing fragmented chromosomes to persist and reassemble erroneously. This process is often linked to TP53 mutations that disrupt full apoptotic execution, enabling survival of cells with extensive DNA damage. The fragmentation exhibits DNase II-like activity, resembling lysosomal nuclease-mediated cleavage seen in early apoptosis stages, which generates high-density breaks amenable to chromothripsis. Telomeric dysfunction contributes to chromothripsis through iterative breakage-fusion-bridge (BFB) cycles, where short or eroded s lead to end-to-end fusions, forming dicentric chromosomes that break during mitosis.01573-1) These cycles amplify local genomic , culminating in widespread shattering and reassembly of fragments, as observed in telomere crisis models bypassing via and loss. Such events produce characteristic oscillating copy number patterns, distinguishing them from gradual mutations. Recent 2025 research highlights chromothripsis in radiotherapy-resistant cancers, where irradiation elevates nuclear p62 levels, impairing ESCRT-III-mediated nuclear envelope repair and exacerbating chromatin bridge resolution failures. This leads to persistent fragmentation and chromothripsis, driving tumor evolution and resistance to treatments like ionizing radiation. These findings underscore how cellular stress from radiation sustains chromothripsis in clinical contexts, particularly in p53-deficient tumors.

Alternative Pathways

In rare instances, activity has been implicated in triggering chromothripsis, especially in the . For example, LINE-1 mediated retrotransposition has been shown to drive chromothripsis by facilitating Alu/Alu and subsequent DNA shattering. poisoning represents another auxiliary route, where chemotherapeutic agents like stabilize topoisomerase II-DNA cleavage complexes, generating clusters of double-strand breaks (DSBs) during that mimic chromothripsis patterns through erroneous repair. These DSB clusters, if unresolved, lead to extensive chromosomal rearrangements confined to specific loci, highlighting the role of replication-associated stress in non-mitotic initiation. Recent 2025 studies have expanded chromothripsis beyond , revealing its involvement in non-cancer contexts such as developmental disorders, including chromothripsis on chromosomes 3q and 5q causing 5q14.3 microdeletion-associated developmental epileptic encephalopathy. Additionally, hybrid models integrating abortive with dysfunction propose that incomplete apoptotic fragmentation combined with erosion during crisis can precipitate chromothripsis-like events, where unprotected chromosome ends fuse and shatter iteratively. Emerging evidence from 2024 also implicates the pathway in inducing chromothripsis and formation, contributing to genomic instability in both and cells. These frameworks suggest multifaceted triggers that blend pathways with replicative instability, offering new insights into and origins.

Predisposing Factors

Genetic Vulnerabilities

Mutations in the TP53 gene, which encodes the tumor suppressor protein, are strongly associated with chromothripsis across various cancers, as plays a critical role in maintaining the G1 and preventing cells with DNA damage from progressing through . In sonic hedgehog-driven medulloblastomas, for instance, all cases with TP53 mutations (10 out of 10) exhibited chromothripsis, highlighting a near-complete linkage in this context. Pan-cancer analyses further confirm that TP53 mutations predispose individuals to chromothripsis by impairing DNA damage response pathways. Similarly, deficiencies in or , key players in repair of double-strand breaks, heighten susceptibility to chromothripsis by inducing replication stress and mitotic errors that lead to chromosome shattering. In models with BRCA1/2 loss, such defects promote the formation of micronuclei and chromatin bridges, structures vulnerable to catastrophic rearrangements characteristic of chromothripsis. Abnormal ploidy states, such as tetraploidy or , increase the risk of chromosome mis-segregation during , thereby predisposing cells to chromothripsis events. Experimental models demonstrate that hyperploid cells are significantly more likely to undergo chromothripsis than diploid counterparts; for example, studies found chromothripsis in 9 out of 58 hyperploid transformants compared to none in 40 diploid ones. validation in sonic hedgehog medulloblastomas similarly showed higher rates in hyperploid tumors (5 out of 11) versus diploid ones (2 out of 34). Tetraploidy, often arising from failure, exacerbates this vulnerability by complicating centrosome clustering and promoting multipolar spindles, which facilitate the isolation of chromosome fragments into micronuclei prone to shattering. Germline predispositions, particularly in rare syndromes like Li-Fraumeni syndrome (LFS) caused by inherited TP53 mutations, are linked to elevated chromothripsis rates in affected tumors. In LFS-associated medulloblastomas, 6 out of 6 cases with germline TP53 mutations displayed chromothripsis, suggesting that constitutional TP53 inactivation precedes and promotes these catastrophic events. This predisposition extends to other LFS-related cancers, where TP53 often accompanies chromothripsis, amplifying mutant allele gains and driving oncogenesis.

Environmental and Acquired Risks

Ionizing radiation exposure, including from therapeutic sources and diagnostic procedures like CT scans, elevates the risk of chromothripsis by generating localized DNA double-strand breaks in mitotic cells, which can trigger complex chromosomal rearrangements resembling chromothripsis patterns. Studies using proton microbeam irradiation have demonstrated that targeted nuclear exposure induces multiple copy number alterations and translocations across chromosomes, such as chromosomes 7, 11, and 12, mimicking the clustered breakpoints characteristic of chromothripsis. High-linear energy transfer radiation, as seen in particle therapy, further promotes irreparable breaks that may culminate in chromothripsis-like genome shattering during cell division. Chemotherapeutic agents, particularly II inhibitors like and spindle poisons such as or , contribute to acquired chromothripsis risk by disrupting mitotic processes and inducing micronuclei formation. These poisons trap DNA- complexes or impair spindle attachment, leading to missegregation, ultrafine bridges, and micronuclei that serve as sites for DNA fragmentation and erroneous reassembly, thereby facilitating chromothripsis. Viral infections, including human papillomavirus (HPV) and Epstein-Barr virus (EBV), promote chromothripsis through genome integration that destabilizes host chromosomes and induces breakage-fusion-bridge cycles. HPV integration often occurs at fragile sites, triggering local chromothripsis-like events with chromosome arm translocations and massive rearrangements in infected cells. Similarly, EBV integration disrupts DNA replication junctions, causing dose-dependent chromosomal breakage that can evolve into complex structural variants akin to chromothripsis. As of 2025, epidemiological analyses indicate higher chromothripsis incidence in populations exposed to or , linked to that compromises DNA integrity and repair in cells. Tobacco smoke drives complex mutagenesis, including chromothripsis signatures, through that induce double-strand breaks in head and neck tissues. Asbestos exposure in cohorts correlates with frequent chromothripsis, as fiber-induced inflammation and persistent DNA damage in mesothelial cells heighten shattering events. In aging cells, accumulated acquired mutations and declining repair efficiency further predispose to chromothripsis, often amplified by vulnerabilities like TP53 alterations.

Role in Disease

Contribution to Carcinogenesis

Chromothripsis drives carcinogenesis primarily through the creation of extensive chromosomal rearrangements that disrupt genomic stability, leading to the amplification of key oncogenes and the inactivation of tumor suppressor genes. For instance, in various solid tumors, chromothripsis events have been linked to the amplification of oncogenes such as and , which promote uncontrolled cell proliferation and survival signaling. Similarly, deletions resulting from these catastrophic events often eliminate tumor suppressors like and PTEN, removing critical barriers to tumor initiation and progression. These alterations can occur in a single cellular event, accelerating the transformation of normal cells into malignant ones by concentrating multiple driver mutations within localized genomic regions. The prevalence of chromothripsis underscores its significant role in cancer development, with events detected in up to 50% of certain pediatric cancers, including high frequencies in bone sarcomas. In , a common pediatric solid tumor, subclonal chromothripsis occurs in 74% of cases, often involving loss-translocation-amplification patterns that inactivate TP53 and amplify nearby oncogenes. Across solid tumors more broadly, chromothripsis is associated with poor patient survival, as it correlates with aggressive disease courses in entities like and . Beyond direct oncogenic effects, chromothripsis confers an evolutionary advantage to tumors by fostering rapid genomic adaptation and intratumoral heterogeneity. Subclonal chromothripsis events generate diverse populations, enabling tumors to evade therapies and metastasize more effectively through ongoing instability. This dynamic process fuels tumor progression, as seen in sarcomas where repeated chromothripsis shapes clonal . While predominantly oncogenic, chromothripsis is rare in non-cancer contexts but has been implicated in congenital disorders, including reports of neurodevelopmental anomalies arising from events. Such cases highlight its potential to cause severe developmental disruptions when occurring outside of cancer cells.

Prognostic and Diagnostic Relevance

The presence of chromothripsis in tumors is strongly associated with aggressive disease behavior and poor patient outcomes across multiple cancer types. In , chromothripsis events are detected in over 60% of metastatic cases, significantly higher than in primary tumors, and contribute to tumor progression by generating oncogenic fusions and disrupting key genes such as and ERBB2. Similarly, chromothripsis has been detected in approximately 30–45% of cancers across various grades, including both clinically insignificant and high-grade tumors. Recent analyses, including those from 2024-2025 studies, indicate that chromothripsis-positive cancers exhibit shorter overall and higher risk compared to those without, with ongoing subclonal chromothripsis events further driving therapy resistance through heterogeneous genomic evolution. Detection of chromothripsis relies primarily on whole-genome sequencing (WGS) to identify characteristic patterns of clustered rearrangements, copy number oscillations, and structural variants confined to one or few chromosomes. Computational algorithms enhance this process by automating signature recognition; for instance, ShatterSeek, an R-based tool, analyzes NGS data to quantify chromothripsis likelihood through metrics like oscillation scores and breakpoint clustering, enabling reliable identification in large cohorts. These tools have been validated in pan-cancer datasets, improving diagnostic accuracy for complex genomic events that may otherwise be overlooked in targeted sequencing approaches. Therapeutically, chromothripsis often co-occurs with DNA repair deficiencies, making affected tumors vulnerable to , which exploit in homologous recombination-impaired cells. Clinical studies in pediatric cancers with chromothripsis have shown promising responses to , particularly when combined with HDAC inhibitors to target residual repair pathways and reduce toxicity. Additionally, monitoring subclonal chromothripsis via serial sequencing can reveal emerging resistant populations, guiding adaptive therapies to counteract evolution-driven relapse in cancers like and .

Evidence and Debates

Key Experimental Findings

One of the earliest experimental validations of chromothripsis came from in vitro models using human cell lines, where researchers induced micronuclei formation through mitotic errors and observed chromosome shattering. In a seminal 2015 study, Zhang et al. developed a technique combining live-cell imaging with single-cell whole-genome sequencing to track mis-segregated chromosomes forming micronuclei in RPE-1 hTERT-immortalized retinal pigment epithelial cells; they demonstrated that DNA damage within these micronuclei leads to tens to hundreds of double-strand breaks on a single chromosome, followed by random reassembly characteristic of chromothripsis. This work provided direct visual and genomic evidence that chromothripsis can occur in a single catastrophic event during or shortly after mitosis, with fragmented chromatids reincorporated into the main nucleus. Animal models have further corroborated these findings by revealing spontaneous chromothripsis in contexts of genomic instability. In (TP53) knockout mice, which mimic Li-Fraumeni syndrome, whole-genome sequencing of thymic lymphomas showed frequent chromothripsis events, often involving massive rearrangements on one or few chromosomes, occurring as early drivers of tumorigenesis. More recent studies using human tumor samples have demonstrated ongoing chromothripsis as a subclonal process, with multi-region sequencing identifying loss-translocation-amplification (LTA) patterns in 74% of tumors, where chromothripsis continues to fuel clonal evolution even after initial tumor formation. These models highlight how TP53 deficiency predisposes cells to repeated shattering events . Large-scale human cohort analyses using (TCGA) data have expanded these observations, quantifying chromothripsis prevalence and timing across cancers. Expansions of TCGA whole-genome sequencing datasets to over 2,600 tumors revealed chromothripsis in 29% of cases, often as clustered rearrangements confined to one arm, validating its role in rapid genome evolution. Multi-region sequencing in cohorts like clear cell and has shown that chromothripsis events can be early clonal (truncal, present in all tumor regions) or late subclonal (branching, driving heterogeneity), with phylogenetic reconstruction indicating they precede or coincide with key oncogenic amplifications. Recent 2025 studies have provided high-resolution evidence using advanced sequencing and genetic screens. Single-cell whole-genome sequencing in (CLL) cohorts confirmed chromothripsis as a single-cycle event, with amplifications occurring late in leukemic evolution, as tracked by variant frequencies and epigenetic timing analyses. Complementing this, CRISPR-Cas9 knockout screens in chromosomally unstable cell lines identified KIF18A, a mitotic , as a key trigger; its depletion increased micronuclei formation and chromothripsis-like rearrangements by disrupting chromosome alignment, underscoring the mitotic origins of these catastrophes.

Criticisms and Open Questions

One major criticism of chromothripsis detection involves the potential for over-diagnosis due to artifacts in sequencing data, where progressive rearrangements can produce patterns mimicking the characteristic clustered breakpoints and oscillating copy number states. Simulations demonstrate that chromosomes with 50–55 breakpoints from gradual accumulation exhibit 2–3 copy number states in 3.9% of cases, leading to false positives when detection criteria are relaxed to include fewer than breakpoints. A central concerns whether chromothripsis truly represents a singular catastrophic event or if it often results from the gradual accumulation of rearrangements that simulate shattering. inversions and deletions can generate up to 237 s with only two copy number states on a single , as seen in cell line analyses, undermining the exclusivity of the one-event model without additional mechanistic proof. Limitations in chromothripsis research include its extreme rarity outside cancer contexts, where it is primarily documented in a handful of constitutional cases linked to congenital malformations or reproductive issues, with 24 reported instances as of 2019 showing varied phenotypes like subtle dysmorphic features; subsequent studies have identified additional cases. Distinguishing chromothripsis from other complex rearrangements, such as chromoanasynthesis or chromoplexy, poses significant challenges due to overlapping features like clustering and random joins, compounded by the absence of a defined minimum threshold and the ability of sequential events to replicate its signature. Open questions persist regarding the frequency of chromothripsis in normal aging, as current studies lack comprehensive genomic surveys of healthy cells, leaving its potential role in age-related genomic instability unresolved. Preventive strategies remain unexplored, with no established interventions to mitigate the underlying mitotic errors or micronuclei formation that trigger shattering, though emerging research on disrupting fragment suggests possible therapeutic avenues. The role of chromothripsis in resistance has gained attention, where it correlates with reduced cytotoxic T-cell infiltration and poorer responses to checkpoint blockade, as shown in a 2023 pan-cancer analysis, potentially via immune evasion phenotypes in chromothripsis-affected tumors. Evolving views in critique the underestimation of chromothripsis as an ongoing process rather than a one-time event, with multi-region sequencing revealing subclonal occurrences in 74% of osteosarcomas that drive continuous genomic instability and clonal evolution through unstable derivative chromosomes.

References

  1. [1]
  2. [2]
    Molecular dissection of germline chromothripsis in a developmental ...
    Jan 26, 2017 · Germline chromothripsis causes complex genomic rearrangements that are likely to affect multiple genes and their regulatory contexts.Rna Extraction And... · Analysis Of Rna Sequencing... · Hi-C Data Generation And...
  3. [3]
    Comprehensive analysis of chromothripsis in 2,658 human cancers ...
    Feb 5, 2020 · Chromothripsis is a mutational phenomenon characterized by massive, clustered genomic rearrangements that occurs in cancer and other diseases. ...
  4. [4]
  5. [5]
    Chromothripsis is a common mechanism driving ... - Genome Biology
    Oct 19, 2011 · Local chromosome shattering, termed chromothripsis, is a mechanism proposed to be the cause of clustered chromosomal rearrangements and was ...
  6. [6]
    Patterns and mechanisms of structural variations in human cancer
    Aug 7, 2018 · Like chromothripsis, chromoplexy is readily explained by the presence of a catastrophic hit that produces multiple DNA double-strand breaks ( ...
  7. [7]
    Chromothripsis in cancer - PubMed
    Nov 15, 2024 · This extreme form of genome instability has been detected in 30-50% of cancers. Studies conducted in the past few years have uncovered insights ...
  8. [8]
    The landscape of chromothripsis across adult cancer types - Nature
    May 8, 2020 · We show that chromothripsis affects a substantial proportion of human cancers, with a prevalence of 49% across all cases.
  9. [9]
    Chromothripsis in Healthy Individuals Affects Multiple Protein ... - NIH
    ... size from 150 kb to 27 Mb and involved 2 or 3 chromosomes per child (Figure S1). Giemsa (G)-banded chromosome analysis revealed the presence of CCRs ...
  10. [10]
    Chromothripsis and genomic instability from mitotic errors
    Jan 4, 2024 · Within these MN bodies, fragmented DSB ends activate the DNA damage response during G1 phase for processing by the NHEJ machinery (Figure 4) ...
  11. [11]
    Non-homologous end joining shapes the genomic rearrangement ...
    In cancers genomes and germline disorders with chromothripsis, the majority of rearrangement breakpoints harbor blunt-ended junctions without homology; however, ...
  12. [12]
    Chromothripsis and DNA Repair Disorders - PMC - NIH
    Chromothripsis is characterized by local “shattering” or the generation of clustered DNA double-strand breaks (DSBs) involving one or multiple chromosomes and ...
  13. [13]
    Of Simple and Complex Genome Rearrangements, Chromothripsis ...
    Jan 14, 2017 · Chromothripsis is currently defined as a process in which one or several chromosomes are broken into many segments with narrowly spaced breakpoints.
  14. [14]
    Chromothripsis - an overview | ScienceDirect Topics
    Repaired chromosomes contain chromosomal rearrangements such as deletions, head-to-head and tail-to-tail inversions, and tandem duplications. With mitosis exit, ...
  15. [15]
    Non-homologous end joining shapes the genomic rearrangement ...
    Jul 4, 2024 · Chromothripsis frequently drives complex and localized genomic rearrangements owing to the error-prone reassembly of the fragmented chromosome.
  16. [16]
    Ongoing chromothripsis underpins osteosarcoma genome ...
    Jan 23, 2025 · Using multi-region whole-genome sequencing, we find that chromothripsis is an ongoing mutational process, occurring subclonally in 74% of osteosarcomas.
  17. [17]
    Rebuilding Chromosomes After Catastrophe - NIH
    The resultant chromosomal fragments are re-ligated in random order by DNA double-strand break repair during the subsequent interphase. Chromothripsis scars the ...
  18. [18]
    Chromothripsis-like chromosomal rearrangements induced by ...
    Mar 1, 2016 · Chromothripsis is the massive but highly localized chromosomal rearrangement in response to a one-step catastrophic event, rather than an ...
  19. [19]
    Chromatin and the Cellular Response to Particle Radiation-Induced ...
    Even with exposure to low-LET radiation, persistent clusters of 2–3 DSBs (4–6 pKu70 foci) were observed, and it was postulated that these formed in ...
  20. [20]
    Mitotic clustering of pulverized chromosomes from micronuclei | Nature
    May 10, 2023 · The prevalence of canonical chromothripsis in cancer genomes probably reflects strong positive selection pressure owing to the increased risk of ...
  21. [21]
    Chromothripsis—Explosion in Genetic Science - PMC
    Chromothripsis has been defined as complex patterns of alternating genes copy number changes (normal, gain or loss) along the length of a chromosome or ...
  22. [22]
    Apoptosis – Fueling the oncogenic fire - Castillo Ferrer - FEBS Press
    Nov 11, 2020 · Sustained DNA damage can also lead to a process known as chromothripsis, a mechanism of chromosome fragmentation that can promote the ...
  23. [23]
    Degradation of chromosomal DNA during apoptosis - Nature
    Mar 25, 2003 · DNase II is a lysosomal enzyme, and apoptotic DNA degradation occurs at the early stage of apoptosis when lysosomes have not yet been destroyed, ...Missing: chromothripsis | Show results with:chromothripsis
  24. [24]
    Chromothripsis and kataegis induced by telomere crisis - PMC - NIH
    Telomere fusions are infrequent in senescence, most likely because of the low frequency of dysfunctional telomeres. Upon by-pass of senescence due to loss of ...
  25. [25]
    Chromothripsis: Breakage-fusion-bridge over and over again - PMC
    Keywords: chromothripsis, breakage-fusion-bridge, chromosomal instability, centromere fission, mitosis, telomere ... Telomere dysfunction, genome instability and ...
  26. [26]
    Irradiation-induced increase in nuclear p62 levels contributes to ...
    Aug 1, 2025 · Our findings suggest that irradiation-induced p62 accumulation is crucial for chromothripsis and may affect nuclear repair processes, impacting chromosomal ...<|control11|><|separator|>
  27. [27]
    Homologous recombination promotes non-immunogenic mitotic cell ...
    Jan 13, 2025 · Here, we use single-cell analysis of long-duration live imaging to investigate mitotic catastrophe in human cells exposed to ionizing radiation ...<|separator|>
  28. [28]
    Mutational signatures in tumours induced by high and low energy ...
    Jan 20, 2020 · Chromothripsis is particularly associated with tumour development in patients with Li-Fraumeni syndrome as they bear germline TP53 mutations, ...
  29. [29]
    Chromosome Inequality: Causes and Consequences of Non ...
    Chromosome Inequality: Causes and Consequences of Non-Random Segregation Errors in Mitosis and Meiosis ... reverse segregation of acrocentric or smaller ...
  30. [30]
    Chromothripsis: potential origin in gametogenesis and ...
    Endogenous retroviruses and transposable elements have been found to be activated in both blastomere-stage embryos and PGCCs [115,165], demonstrating an ...
  31. [31]
    Germline Chromothripsis Driven by L1-Mediated Retrotransposition ...
    Chromothripsis is a phenomenon where multiple localized double-stranded DNA breaks result in complex genomic rearrangements. Although the DNA-repair ...<|separator|>
  32. [32]
    Long-term fate of etoposide-induced micronuclei and ... - NIH
    Jul 17, 2020 · In addition, micronuclei are discussed to be involved in chromothripsis, which could explain the local occurrence of massive chromosomal damages ...
  33. [33]
    TDP2 suppresses chromosomal translocations induced by DNA ...
    Aug 10, 2017 · In this study we have addressed the impact of TDP2-dependent DSB repair on chromosome rearrangements mediated by the abortive activity of TOP2.
  34. [34]
    De Novo Chromosomes 3q and 5q Chromothripsis Leads to a 5q14 ...
    Jan 30, 2025 · This is the first reported case of chromosomes 3q and 5q chromothripsis leading to a developmental epileptic encephalopathy (DEE) due to disruption of 5q14.3.
  35. [35]
    Chromothripsis: an emerging crossroad from aberrant mitosis to ...
    Chromothripsis involves the rapid acquisition of tens to hundreds of structural rearrangements within a short period, leading to complex alterations in one or ...
  36. [36]
    Chromothripsis-like chromosomal rearrangements induced by ...
    Mar 1, 2016 · These findings suggested that localized ionizing irradiation within the nucleus may induce chromothripsis-like complex chromosomal alterations ...Missing: environmental CT scans
  37. [37]
  38. [38]
    Chromosomal Rearrangements and Chromothripsis: The Alternative ...
    Jan 2, 2023 · Chromothripsis defines a genetic phenomenon where up to hundreds of clustered chromosomal rearrangements can arise in a single catastrophic event.
  39. [39]
    [PDF] Genomic structures and regulation patterns at HPV integration sites ...
    Nov 5, 2023 · HPV integration is associated with full chromosome arm translocations ... chromothripsis event28. Akagi et al.29 described this patterning ...
  40. [40]
    Chromosomal fragile site breakage by EBV-encoded EBNA1 at ...
    We demonstrate that increasing levels of EBNA1 binding trigger dose-dependent breakage at 11q23, producing a fusogenic centromere-containing fragment and an ...Missing: chromothripsis | Show results with:chromothripsis
  41. [41]
    (PDF) The complexity of tobacco smoke-induced mutagenesis in ...
    Mar 31, 2025 · Our results demonstrate the multiple pathways by which tobacco smoke can influence the evolution of cancer cell clones.
  42. [42]
    Does chromothripsis make mesothelioma an immunogenic cancer?
    During the interphase, the micronuclei membrane disrupts leading to chromosome shattering because the DNA is fragmented by exposure to cytoplasmic nucleases.
  43. [43]
    Mutational game changer: Chromothripsis and its emerging ...
    The seminal discovery of chromothripsis in cancer genomes in 2011 not only challenged the long-standing concept of progressive genetic events driving ...
  44. [44]
    Scrambling the genome in cancer: causes and consequences of ...
    Three chromothripsis-associated mechanisms are thought to be capable of driving tumorigenesis: 1) disruption or loss of tumor suppressors, 2) generation of ...
  45. [45]
  46. [46]
    Chromothripsis and focal copy number alterations determine poor ...
    Chromothripsis describes a single catastrophic cellular event, in which one or a few chromosomes, chromosome arms or chromosomal subregions are shattered into ...Missing: sawtooth | Show results with:sawtooth
  47. [47]
    Long-read sequencing reveals chromothripsis in a molecularly ...
    Mar 12, 2024 · Most junctions display blunt ends (70%) and some microhomology of 1–5 nucleotides (23%), suggesting the occurrence of non-homologous end joining ...
  48. [48]
    Constitutional Chromothripsis on Chromosome 2: A Rare Case with ...
    Jan 30, 2024 · Constitutional chromothripsis is a rare occurrence, reported in children presenting with a wide range of birth defects.
  49. [49]
    Chromothripsis in Human Breast Cancer - AACR Journals
    Chromothripsis is a form of genome instability, whereby one or a few chromosomes are affected by tens to hundreds of clustered DNA rearrangements (1–4).<|separator|>
  50. [50]
    Chromosomal catastrophe is a frequent event in clinically ... - NIH
    In a few cancer types, chromothripsis was found to be associated with poor prognosis. ... Poly-gene fusion transcripts and chromothripsis in prostate cancer.
  51. [51]
    Ongoing chromothripsis underpins osteosarcoma genome ...
    Jan 23, 2025 · Using multi-region whole-genome sequencing, we find that chromothripsis is an ongoing mutational process, occurring subclonally in 74% of osteosarcomas.
  52. [52]
    Multi-omic and single-cell profiling of chromothriptic ... - Nature
    Nov 23, 2024 · We characterise subclonal variation of chromothripsis and its effects on extrachromosomal circular DNA, cancer drivers and putatively druggable ...
  53. [53]
    parklab/ShatterSeek - GitHub
    ShatterSeek is an R package that provides utilities to detect chromothripsis events from next-generation sequencing (NGS) data.
  54. [54]
    Comprehensive analysis of chromothripsis in 2,658 human cancers ...
    Chromothripsis is a mutational phenomenon characterized by massive, clustered genomic rearrangements that occurs in cancer and other diseases.
  55. [55]
    Chromothripsis, DNA repair and checkpoints defects - ScienceDirect
    Chromothripsis is a unique form of genome instability characterized by tens to hundreds of DNA double-strand breaks on one or very few chromosomes, followed by ...
  56. [56]
    and PARP inhibitors in childhood tumors with chromothripsis
    Aug 15, 2022 · Rearrangements associated with chromothripsis are detectable in numerous tumor entities and linked with poor prognosis in some of these, such as ...Missing: deficient | Show results with:deficient
  57. [57]
    Detection of Genomic Copy Number Variations in Ovarian Cancer in ...
    Chromothripsis leads to a sudden separation of the genome in the chromatids during anaphase [12,13,14,15]. Chromoanasynthesis results from errors in chromosome ...
  58. [58]
    Cell‐free DNA aneuploidy score as a dynamic early response ... - NIH
    Mar 14, 2025 · Cell‐free circulating tumor DNA (ctDNA) has emerged as a promising biomarker for response evaluation in metastatic castration‐resistant prostate ...
  59. [59]
    Chromothripsis from DNA damage in micronuclei - Nature
    May 27, 2015 · We demonstrate that the mechanism for chromothripsis can involve the fragmentation and subsequent reassembly of a single chromatid from a micronucleus.
  60. [60]
    The evolution of thymic lymphomas in p53 knockout mice
    In this study, the p53 knockout mouse was employed as a model to study the mutational evolution of tumorigenesis. ... chromothripsis, a pattern observed in human ...
  61. [61]
    Chromothripsis-associated chromosome 21 amplification ... - Nature
    Jun 9, 2025 · Chromothripsis, the chaotic shattering and repair of chromosomes, is common in cancer. Whether chromothripsis generates actionable ...
  62. [62]
    Targeting chromosomally unstable tumors with a selective KIF18A ...
    Jan 2, 2025 · We identify and validate KIF18A, a mitotic kinesin, as a vulnerability of chromosomally unstable cancer cells. Knockdown of KIF18A leads to mitotic defects and ...
  63. [63]
    The elusive evidence for chromothripsis - PMC - NIH
    Jun 17, 2014 · The chromothripsis hypothesis suggests an extraordinary one-step catastrophic genomic event allowing a chromosome to 'shatter into many ...
  64. [64]
    On the Complexity of Mechanisms and Consequences of ...
    Apr 30, 2019 · We discuss the challenges of chromothripsis detection and its distinction from other chromoanagenesis events. Along with already known ...
  65. [65]
    Tethering of Shattered Chromosomal Fragments Paves Way for New ...
    Jun 15, 2023 · By destroying the tether, they might prevent the rearranged chromosomes from forming, thereby reducing the number of cells potentially carrying ...
  66. [66]
    Chromothripsis is correlated with reduced cytotoxic immune ... - NIH
    Feb 27, 2023 · Chromothripsis can be used as an independent predictor, and patients with low-CPSs experienced longer overall survival (OS) after immunotherapy ...