Skip to main content
Erschienen in: Cancer and Metastasis Reviews 3-4/2013

Open Access 01.12.2013

The detection and implication of genome instability in cancer

verfasst von: Larissa Pikor, Kelsie Thu, Emily Vucic, Wan Lam

Erschienen in: Cancer and Metastasis Reviews | Ausgabe 3-4/2013

Abstract

Genomic instability is a hallmark of cancer that leads to an increase in genetic alterations, thus enabling the acquisition of additional capabilities required for tumorigenesis and progression. Substantial heterogeneity in the amount and type of instability (nucleotide, microsatellite, or chromosomal) exists both within and between cancer types, with epithelial tumors typically displaying a greater degree of instability than hematological cancers. While high-throughput sequencing studies offer a comprehensive record of the genetic alterations within a tumor, detecting the rate of instability or cell-to-cell viability using this and most other available methods remains a challenge. Here, we discuss the different levels of genomic instability occurring in human cancers and touch on the current methods and limitations of detecting instability. We have applied one such approach to the surveying of public tumor data to provide a cursory view of genome instability across numerous tumor types.

1 Introduction

Cancer is a disease characterized and fuelled by dynamic genomic changes. The vast number of structural abnormalities present in cancer genomes is largely attributed to genomic instability, a transient or persistent state that increases the spontaneous mutation rate, leading to gross genetic alterations such as rearrangements and changes in chromosome number (aneuploidy). Genomic instability is therefore a driving force of tumorigenesis in that continuous modification of tumor cell genomes promotes the acquisition of further DNA alterations, clonal evolution, and tumor heterogeneity [1]. It is a feature of almost all cancers and has been observed in a range of malignant stages, from pre-neoplastic lesions prior to acquired TP53 mutations to advanced cases [24]. Numerous theories regarding the source of genome instability have been proposed. These theories, which include the mutator phenotype, DNA damage-induced replication stress, telomere dysfunction, and mitotic checkpoint failure [511], vary principally in their supposition of how early in tumorigenesis instability occurs, mechanisms leading to sequence level alteration, and whether instability initiates tumorigenesis or is merely a consequence of malignant transformation. While these mechanisms may all contribute to instability phenotypes to some extent in cancer in general, their prevalence varies across tumors derived from distinct cell types or in response to different carcinogens or selective pressures.
Genomic instability refers to a variety of DNA alterations, encompassing single nucleotide to whole chromosome changes, and is typically subdivided into three categories based on the level of genetic disruption. Nucleotide instability (NIN) is characterized by an increased frequency of base substitutions, deletions, and insertions of one or a few nucleotides; microsatellite instability (MIN or MSI) is the result of defects in mismatch repair genes which leads to the expansion and contraction of short nucleotide repeats called microsatellites; chromosomal instability (CIN) is the most prevalent form of genomic instability and leads to changes in both chromosome number and structure [12]. While instability is a characteristic of almost all human cancers, cancer genomes vary considerably in both the amount and type of genomic instability they harbor. Importantly, the instability phenotype has implications in patient prognosis as well as patient management, specifically with the choice of therapeutic agents [1315].
Currently, detection of genome instability can be achieved using a variety of technologies, ranging from single-cell approaches to high-throughput multicellular techniques, each capable of detecting different levels of genomic changes. However, at present, no assay is capable of reliably measuring the rate (cell-to-cell variability) of small chromosomal changes such as deletions, amplification, and inversions within a population of cells. There is therefore a great need for sensitive, high-resolution techniques capable of detecting genomic instability over time as this would afford critical insights into the mechanisms that underlie genomic instability and the role of instability in tumorigenesis. In this review, we discuss the different levels of genomic instability and various methods of and limitations to detecting instability and describe global trends in genome instability across numerous tumor types.

2 Levels of genomic instability

2.1 Nucleotide instability

NIN typically develops due to replication errors and impairment of the base excision repair and nucleotide excision repair pathways, leading to subtle sequence changes involving only one or a few nucleotides (substitutions, deletions, insertions, etc.) which can affect gene structure and/or expression (Fig. 1a). While less common than the other forms of genomic instability, when present, single nucleotide alterations can cause dramatic phenotypes. For example, inherited defects in these repair pathways (germline mutations in XPC, ERCC2, DDB2, and MYH) lead to disorders such as xeroderma pigmentosum and MYH-associated polyposis, which result in genomic instability and the accumulation of DNA mutations, consequently predisposing these individuals to skin and colon cancers, respectively [16, 17]. Similar to the nuclear genome, the mitochondrial genome also displays NIN, and coupling of the high rate of reactive oxygen species generation with inefficient DNA repair can result in a rate of mtDNA mutations that is substantially higher than that of nuclear DNA [18, 19].

2.2 Microsatellite instability

Microsatellites are repetitive DNA sequences comprising 1–6 bp located throughout the genome [2022]. Within the population, microsatellite size is highly variable; however, each individual possesses unique microsatellites of a set length. MSI results from defects in DNA mismatch repair (MMR), specifically alterations of the MLH1, MSH2, MSH6, and PMS2 genes, which causes deletions or random insertion and expansion of microsatellites and a hypermutable phenotype (Fig. 1b). MSI is a characteristic feature of a number of cancers, including gastric, endometrial, ovarian, lung, and colorectal cancer (CRC), where it was first described and has been studied most extensively [2328]. MSI occurs in approximately 15 % of CRC, which typically arise in the proximal colon, posses a normal karyotype, and are associated with a better prognosis than non-MSI tumors.
MSI occurs in both hereditary (Lynch syndrome) and sporadic forms of colon cancer, although via distinct mechanisms [13]. Hereditary non-polyposis colorectal cancer (Lynch syndrome) is characterized by inactivating germline mutations to MSH2, MSH6, PMS2, or MLH1, whereas sporadic CRC with MSI is associated with hypermethylation and loss of expression of MLH1 [2732]. The majority of sporadic CRC with MSI arise in a background of extensive aberrant promoter methylation—referred to as the CpG island methylator phenotype (CIMP) [3335]. CIMP tumors develop and progress by methylating the promoters of tumor suppressor genes such as p16, IGF-2, and MLH1 and possess clinical features distinct from non-CIMP tumors [33, 3638].

2.3 Chromosomal instability

CIN is an increase in the rate of gain or loss of segmental and whole chromosomes during cell division and is the most prominent form of genomic instability in solid tumors, with roughly 90 % of human cancers exhibiting chromosomal abnormalities and aneuploidy [3, 39]. CIN tumors are characterized by global aneuploidy, amplifications, deletions, loss of heterozygosity (LOH), homozygous deletions, translocations, and inversions (Fig. 2). These alterations lead to karyotypic instability and the simultaneous growth of diverse tumor subpopulations, resulting in genomic inter- and intra-tumor heterogeneity [39]. CIN develops early in tumorigenesis (detectable in premalignant lesions) and is associated with intrinsic multidrug resistance [40] and poor prognosis [15, 41], making its detection clinically relevant.
Despite the prominence and fundamental importance of CIN to cancer biology, the molecular mechanisms underlying CIN in sporadic cancers remain poorly understood. This is due primarily to the fact that disruption of countless genes can give rise to CIN, including, but not limited to, those involved in chromosome condensation and segregation (STAG2) [42], telomere dysfunction (TRF1 and Tankyrase) [43], as well as DNA damage (ATM) [44, 45] and spindle checkpoint genes (BUB1, Mad2) [4648], highlighting the heterogeneous nature of CIN in sporadic cancers. Attempts to explain the presence and molecular basis of CIN in sporadic cancers have led to the development of three prevailing theories: the mutator hypothesis, the oncogene-induced DNA damage model, and instability due to telomere erosion, which are reviewed in [5, 10, 49].
The advent of sequencing technologies has led to the recent discovery of an intriguing form of genome chaos and CIN, whereby only one or a few distinct chromosomes in a cancer cell are characterized by the presence of upwards of hundreds of complex genomic rearrangements [11]. These distinct chromosomal rearrangements were proposed by Stephens et al. to have developed through chromosome shattering (“thripsis” in Greek) or incomplete fragmentation and the inaccurate stitching together of chromosomes in a single stochastic event in a process termed “chromothripsis,” an event in contrast to the widely accepted notion of gradual accumulation of cancer genome rearrangements. Chromothripsis has been proposed to occur in ∼2–3 % of a wide spectrum of cancers (with a higher incidence in bone cancers), where chromosome-specific massive rearrangements have been described [11, 50]. The mechanisms underlying chromothripsis, and its clinical implications, have been recently reviewed by Forment et al. [51].

2.4 Interplay between instability types

While all levels of instability can co-occur within the same cell, and work in concert to disrupt a single gene, protein complex, or pathway, in colorectal and endometrial cancers, an inverse relationship between CIN and MIN has been observed [52, 53]. Although both types of instability appear to occur early in tumor development and increase with tumor progression, cancers with an MMR deficiency tend to be diploid and exhibit normal rates of gross chromosomal changes, whereas MMR-proficient tumors are typically aneuploid and display increased rates of chromosomal alterations [12]. Moreover, the fusion of MIN and CIN cells results in CIN, but not MIN, suggesting that CIN is a dominant phenotype that may result from gain-of-function alterations rather than gene inactivation [3, 46].

3 Methods for the detection and analysis of genome instability

A number of established strategies exist to detect genomic instability in cancer. However, it is important to keep in mind that genomic instability is a matter of rate of chromosomal alterations and is therefore a gauge of variability in chromosomal state between individual cells within a tumor [54]. To accurately assess instability, repeated measurements across cell populations throughout tumor evolution or, ideally, measurements in individual cancer cells are required to define the actual rate or variability in genomic changes for a particular tumor [54]. Although these measurements are more easily obtainable for cancer cell lines, measuring genome stability accurately in clinical tumor specimens where material is often limited and substantial cellular heterogeneity exists is considerably more difficult. As a result, few studies have determined the actual rate of chromosomal alterations in different cancer types and, thus, characterized true genomic instability [54]. Because of the difficulty in measuring actual genomic instability, various methods to calculate the frequency and extent of genomic changes for static tumor cell populations have been used as a surrogate to describe genomic instability. Therefore, caution must be taken when interpreting claims about instability in cancer. Since genomic instability occurs across multiple genetic levels, any method capable of detecting chromosomal, microsatellite, or nucleotide changes is adequate to measure a component of genomic instability. Such methods include, but are not limited to, karyotyping, flow cytometry, single nucleotide polymorphism (SNP) arrays, genome sequencing, and polymerase chain reaction (PCR), which are summarized in Table 1.
Table 1
Currently available methods of detecting genome instability
Method
Cellularity
Alterations detected
Rate and state
Karyotyping
Single cell
Whole and segmental CIN, aneuploidy
a
a
Single-cell sequencing
Single cell
Whole and segmental CIN, translocations, insertions, deletions, and mutations
a
c
Flow cytometry
Multi-cell
Cell ploidy/aneuploidy
b
b
Array-CGH
Multi-cell
Whole and segmental CIN
N/A
a
SNP arrays
Multi-cell
Whole and segmental CIN, SNP, UPD, LOH
N/A
a
Whole-genome sequencing
Multi-cell
Whole and segmental CIN, translocations, insertions, deletions, and mutations
N/A
c
PCR
Multi-cell
MSI, mitochondrial instability
b
a
SNP single nucleotide polymorphisms, UPD uniparental disomy, LOH loss of heterozygosity, CGH comparative genomic hybridization, N/A cannot detect
aBest approach for measuring rate and state
bUseful but not ideal at measuring rate and state
cNot very useful at measuring rate and state

3.1 Single-cell approaches

Karyotyping is the visualization of a cell’s entire complement of chromosomes, or karyotype. Assessment of a cell karyotype enables the identification of abnormalities in chromosome number (aneuploidy) and large structural rearrangements like inversions and translocations [55, 56]. Traditionally, metaphase chromosomes are stained with a DNA-binding dye, such as Giemsa stain, which is taken up readily by gene-poor A,T-rich genomic regions and results in a chromosome-specific banding pattern that can be used to differentiate chromosomes and identify abnormalities. The use of multicolored fluorescence in situ hybridization (FISH) probes has greatly facilitated the assessment of CIN and is referred to as spectral karyotyping (SKY). The SKY technique results in coloring, or painting, of each chromosome with a different colored fluorophore, readily enabling the identification of chromosomes and rearrangements [55, 57]. Although excellent for detecting global CIN changes, even the most advanced FISH strategies cannot accurately measure somatic mutations throughout the genome. While karyotyping is one of the few techniques available that enable the identification of alterations within a single cell, and the only one capable of profiling both clonal and non-clonal chromosomal alterations [58], like most other methods, it offers only a static picture of the state of chromosomal alterations with no information regarding the extent of variability between cells. Furthermore, it is labor-intensive and metaphase spreads from even short-term cultures can acquire culturing artifacts that induce additional genomic changes. Despite these limitations, karyotyping remains the most reliable method to detect non-clonal chromosomal aberrations and assess genomic variability among cells.
Advances in next-generation sequencing and whole-genome amplification technologies have enabled the advent of single-cell sequencing, which offers promising insight into understanding genomic instability as it provides not only a comprehensive look at the state of genomic alterations of a tumor cell but also cell-to-cell heterogeneity. Because single-cell sequencing relies on gene amplification, sequence bias and adequate genome coverage remain major challenges. However, new amplification methodologies such as multiple annealing and looping-based amplification cycles, which enable over 90 % genome coverage and can accurately detect mutations and copy number variations [59], are in development and have the potential to greatly improve single-cell sequencing. Although many obstacles remain before single-cell sequencing can be routinely implemented as a standard procedure for detecting genome instability, it has the ability to provide an unprecedented view of genomic instability.

3.2 Multicellular approaches

Flow cytometry, which measures cells in suspension as they pass through a laser, scatter light, and emit fluorescence, can be used to approximate cellular aneuploidy. This strategy estimates cell ploidy based on DNA content (which correlates to the intensity of fluorescence) and the stage of cells in the cell cycle. Comparison of the estimated ploidy in the G0/G1 fraction of malignant and normal cells allows a gross estimate of genome instability in cancer cells [60, 61]. While flow cytometry is extremely accurate in its ability to estimate ploidy, it provides no information regarding NIN, MSI, or the segmental or whole-chromosome aberration components of CIN.
Array comparative genomic hybridization (aCGH) offers the ability to quantitatively detect and visualize whole and segmental chromosomal alterations such as gains, losses, amplifications, and LOH [62, 63]. Briefly, reference genomic DNA and test DNA are differentially labeled, pooled, and hybridized onto arrays comprising BAC, cDNA, or oligonucleotides, and imbalances are visualized as differences in fluorescence intensity. The advent of SNP arrays offered improved resolution, enabling more precise mapping of copy number alterations and the detection of uniparental disomy (copy neutral loss of heterozygosity) as well as the ability to distinguish alleles at specific polymorphic sites [6466]. However, neither aCGH nor SNP arrays are able to detect translocations, inversions, or somatic mutations.
PCR is the gold standard for detecting MSI. PCR is used to amplify known microsatellite regions, and the lengths of the short tandem repeats (PCR products) are compared in tumor and normal DNA to determine the state of MSI [37, 67, 68]. This approach is therefore limited to assessing MSI. PCR is also used routinely for the analysis of mitochondrial instability. The ability to isolate mtDNA from total DNA using mitochondrial-specific primers rather than through centrifugation not only reduced tissue requirements but also enabled the use of archival paraffin-embedded tissues, greatly expanding the number samples available for analysis [19]. Commonly used markers of mitochondrial genome instability detected by PCR and followed by direct sequencing include point mutations, insertions, deletions, and length changes in homopolymeric or dimeric nucleotide tracts. Competitive PCR, in which a competitor DNA fragment is added to the DNA sample, can be used to determine mitochondrial DNA copy number by determining the ratio between the intensities of the control and the sample PCR product band [18].
While not routinely performed at the single-cell level, whole-genome sequencing is arguably the most comprehensive and informative method of profiling the cancer genome. In a single experiment, sequencing is capable of identifying nucleotide substitutions, insertions or deletions, and larger genomic rearrangements such as copy number changes, inversions, and translocations, simultaneously capturing all levels of genomic instability for a given population of tumor cells (Fig. 3) [69, 70]. The detection and extent of somatic mutations is determined informatically using computational programs for variant calling. These programs compare the sequences of both the tumor and patient-matched normal sample to a reference genome to reveal somatic and germline alterations, providing confidence calls for each mutation [69, 71]. Copy number analysis by sequencing (both by high or low coverage) offers substantial benefits over array-based methods, including higher resolution (down to a single base) and precise delineation of breakpoints [72, 73]. Copy number ratios at each genomic locus are estimated by counting and comparing the number of reads in both tumor and normal samples. Furthermore, whole-genome sequencing provides data on non-coding regions (promoters, enhances, introns, and non-coding RNA) as well as un-annotated regions, requiring no a priori knowledge of genome sequence, facilitating the discovery of novel DNA sequences.
Sequencing studies have provided massive amounts of data on cancer genomes, revealing great diversity in the mutation frequency across tumor types and identifying novel rearrangements in epithelial cancers. As data from sequencing studies continue to emerge in the public domain, a large-scale pan-cancer comparison of genomic instability in different cancers will be feasible. Such an analysis may shed more light on the mechanistic differences of cancer development in different tissues, which itself will improve our understanding of cancer biology and our ability to develop rationally designed therapies. The interpretation of whole-genome sequencing data in the context of heterogeneous tumors, however, remains a considerable challenge to the application of such data to patient care.
The fundamental limitation of these multicellular approaches is that they provide only a snapshot of the state of alterations in a tumor sample and are incapable of defining the rate of chromosomal changes within a tumor—two features that define genomic instability. While single-cell approaches such as karyotyping or single-cell array-CGH allow for unbiased comparisons of variability in chromosomal alterations between cells, they are not amenable to automation and are therefore time-consuming and labor-intensive. Collection of repeated tumor biopsy samples and advances in single-cell profiling technologies will help generate more accurate metrics of genomic instability.
It is well established that vast genome instability exists at different levels and to different extents in various tumor types. In the last decade, several large-scale sequencing studies have been undertaken in an attempt to characterize recurrent alterations in cancer genomes [7479]. While thousands of mutations have been identified, these studies have shown that very few genes are recurrently mutated, deleted, or amplified at high frequencies within a tumor type. Of the handful of recurrently altered genes, TP53 is the most frequently altered gene in all tumor types, while the others (CDKN2A, PTEN, EGFR, and RAS) have roles in regulating growth and encode classical tumor suppressors and oncogenes [74, 76, 80, 81].
In general, epithelial tumors are thought to be more genomically unstable than hematologic and mesenchymal malignancies, in which a high proportion of cases are characterized by specific genetic rearrangements such as translocations [82]. Interestingly, certain cancer types display characteristic instability phenotypes. For instance, BRCA-associated breast and ovarian cancers demonstrate high levels of CIN, whereas lung cancer in smokers and never smokers differs in the extent of segmental alterations and subsequently, genome instability [8386]. Moreover, specific subtypes of breast, ovarian, and lung cancers exhibit distinct patterns of alterations; the basal-like subtype of breast cancers (typically estrogen receptor-negative) have greater CIN than luminal subtypes, while type II high-grade serous ovarian carcinomas have greater CIN than type I serous ovarian cancers [87, 88]. In lung cancer, adenocarcinoma and squamous cell carcinoma demonstrate distinct patters of genomic alterations, and within lung adenocarcinoma, the magnoid subtype displays higher CIN than other adenocarcinoma subtypes [89, 90]. A review of genome sequencing studies revealed that epithelial-derived cancers such as breast, non-small cell lung, small-cell lung, melanoma, and prostate cancers have a greater number of somatic mutations than blood cancers including acute myeloid leukemia [91], which could suggest that epithelial cancers have greater nucleotide instability. However, specific environmental exposures, such as tobacco smoke, can have specific signatures in terms of epigenetic and genetic alterations in tumors, making it difficult to determine whether the mutations detected arose from nucleotide instability within a tumor or from carcinogen exposure [91]. As more cancer genome sequence data become publicly available, it will be interesting to determine whether specific cancer types exhibit a mutator phenotype and harbor greater nucleotide instability than others.
To compare CIN trends in a pan-cancer manner, we accessed and interrogated copy number data for a set of 2,201 tumor samples representing 24 cancer types made publically available by the Broad and Dana Farber Cancer Institutes (Table 2). Segmented tumor data were downloaded (http://​www.​broadinstitute.​org/​tumorscape/​pages/​portalHome.​jsf); any segment with a log2 ratio exceeding ±0.1 was defined as a segmental alteration. Next, we calculated the fraction of each cancer genome encompassed by segmental alterations to determine the proportion of the genome altered (PGA) and summarized the PGA across the various malignancies (Table 2) [86]. Cancer cell lines were not included in our analysis.
Table 2
Proportion of genome altered (log2 ratio ± 0.1) for various cancer types (n = 2,201)
Type
Count
Median
Average
SD
Mesothelioma
18
0.579305
0.539195
0.261045
Lung SC
17
0.574731
0.539328
0.265144
Melanoma
3
0.533935
0.63286
0.178692
Breast
193
0.361504
0.359875
0.228679
Ovarian
95
0.330162
0.348426
0.298813
Lung NSC
629
0.318028
0.324707
0.234076
Esophageal squamous
2
0.293933
0.293933
0.415683
Hepatocellular
110
0.242876
0.288188
0.222686
Glioma
28
0.198592
0.22249
0.143089
Neuroblastoma
25
0.196659
0.287776
0.24191
Colorectal
128
0.189425
0.261818
0.244976
Renal
99
0.130901
0.226044
0.224458
Medulloblastoma
119
0.110602
0.208003
0.250298
Meningioma
6
0.088144
0.228436
0.347694
Endometrial
1
0.07679
0.07679
N/A
Prostate
83
0.065822
0.180566
0.231366
Synovial sarcoma
2
0.030057
0.030057
0.042508
Acute lymphoblastic leukemia
378
0.02309
0.120554
0.256989
Schwannoma
5
0.011336
0.015021
0.012035
Sarcoma NOS
1
0.009686
0.009686
N/A
Myelodysplasia
19
0.000621
0.007157
0.009818
Thyroid
9
0.000445
0.083437
0.157782
GIST
16
0.000207
0.1654
0.341026
Myeloproliferative disorder
215
0.000135
0.011227
0.048053
Of the 18 cancer types with at least six representative samples, mesothelioma and small-cell lung cancer had the greatest PGA and average number of copy number alterations (CNAs), suggesting that they may be the most genomically unstable in the context of CIN (Fig. 4). These two cancers were followed by breast, ovarian, non-small cell lung cancer, and liver, all epithelial cancers. The number of CNAs was highly correlated with PGA as greater PGAs were associated with a greater number of CNAs (Pearson’s correlation: r = 0.77) across all tumor samples. Of note is that hematological malignancies including acute lymphoblastic leukemia, myelodysplasia, and myeloproliferative disorder harbored some of the lowest PGAs. Thus, these results suggest that CNAs are highly correlated with PGA and that CIN may be greater in epithelial tumors than in hematological cancers, consistent with previously reported trends (Table 2 and Fig. 4).

5 Conclusion

Genomic instability occurs early in tumorigenesis, increasing the spontaneous mutation rate and enabling the acquisition of DNA alterations that promote the hallmarks of cancer, thereby driving tumor development. While the molecular basis of instability is well understood in hereditary cancers, where it is linked to mutations in DNA repair genes, the basis of instability in sporadic cancers remains poorly defined. This limited understanding is due both to the genomic heterogeneity in different tumor types as well as within individual tumors and a lack of methods capable of capturing both the state and rate of instability, which are required to determine the true measure of instability. Genome sequencing studies have provided a wealth of information regarding the state of instability in a variety of cancers, highlighting the diversity in both the types and amounts of instability observed in tumor genomes. As the amount of starting materials for whole-genome sequencing experiments continues to decrease, single-cell sequencing will become feasible for solid tumors; with this will come an expanded understanding of which mechanisms of genomic instability are selected for and precisely how specific patterns of instability support tumor growth in unique systems. In combination with repeat biopsies and sequencing of multiple areas in a single tumor, detailed maps of how genomic instability changes over time will emerge, which can then be interpreted in the context of unique selective pressures in the tumor microenvironment (e.g., the immune system, chemotherapy) or correlated to specific clinical features (e.g., tumor progression). Genomic instability remains an important, yet poorly defined, mechanism by which tumors accelerate their own evolution and survival. At the same time, once uncovered, these same mechanisms will undoubtedly present to the researcher a host of novel therapeutic opportunities.

Acknowledgments

This work was supported by funds from the Canadian Institutes for Health Research (CIHR; MOP 86731, MOP 94867, MOP-110949), Canadian Cancer Society Research Institute (no. 700809), U.S Department of Defence (CDMRP W81XWH-10-1-0634), NCI Early Detection Research Network, and the Canary Foundation. LAP and KLT are supported by Vanier Canada Graduate Scholarships. EAV is supported by Frederick Banting and Charles Best Canada Graduate Scholarship from CIHR.

Conflict of interest

The authors declare that they have no conflict of interest.
Open Access This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Sieber, O. M., Heinimann, K., & Tomlinson, I. P. (2003). Genomic instability—the engine of tumorigenesis? Nature Reviews. Cancer, 3(9), 701–708.PubMedCrossRef Sieber, O. M., Heinimann, K., & Tomlinson, I. P. (2003). Genomic instability—the engine of tumorigenesis? Nature Reviews. Cancer, 3(9), 701–708.PubMedCrossRef
2.
Zurück zum Zitat Nowell, P. C. (1976). The clonal evolution of tumor cell populations. Science, 194(4260), 23–28.PubMedCrossRef Nowell, P. C. (1976). The clonal evolution of tumor cell populations. Science, 194(4260), 23–28.PubMedCrossRef
3.
Zurück zum Zitat Lengauer, C., Kinzler, K. W., & Vogelstein, B. (1997). Genetic instability in colorectal cancers. Nature, 386(6625), 623–627.PubMedCrossRef Lengauer, C., Kinzler, K. W., & Vogelstein, B. (1997). Genetic instability in colorectal cancers. Nature, 386(6625), 623–627.PubMedCrossRef
4.
Zurück zum Zitat Gorgoulis, V. G., Vassiliou, L. V., Karakaidos, P., Zacharatos, P., Kotsinas, A., Liloglou, T., et al. (2005). Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. Nature, 434(7035), 907–913.PubMedCrossRef Gorgoulis, V. G., Vassiliou, L. V., Karakaidos, P., Zacharatos, P., Kotsinas, A., Liloglou, T., et al. (2005). Activation of the DNA damage checkpoint and genomic instability in human precancerous lesions. Nature, 434(7035), 907–913.PubMedCrossRef
5.
Zurück zum Zitat Loeb, L. A. (2001). A mutator phenotype in cancer. Cancer Research, 61(8), 3230–3239.PubMed Loeb, L. A. (2001). A mutator phenotype in cancer. Cancer Research, 61(8), 3230–3239.PubMed
6.
Zurück zum Zitat Bartkova, J., Horejsi, Z., Koed, K., Kramer, A., Tort, F., Zieger, K., et al. (2005). DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature, 434(7035), 864–870.PubMedCrossRef Bartkova, J., Horejsi, Z., Koed, K., Kramer, A., Tort, F., Zieger, K., et al. (2005). DNA damage response as a candidate anti-cancer barrier in early human tumorigenesis. Nature, 434(7035), 864–870.PubMedCrossRef
7.
Zurück zum Zitat Bartkova, J., Rezaei, N., Liontos, M., Karakaidos, P., Kletsas, D., Issaeva, N., et al. (2006). Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature, 444(7119), 633–637.PubMedCrossRef Bartkova, J., Rezaei, N., Liontos, M., Karakaidos, P., Kletsas, D., Issaeva, N., et al. (2006). Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature, 444(7119), 633–637.PubMedCrossRef
8.
Zurück zum Zitat Halazonetis, T. D., Gorgoulis, V. G., & Bartek, J. (2008). An oncogene-induced DNA damage model for cancer development. Science, 319(5868), 1352–1355.PubMedCrossRef Halazonetis, T. D., Gorgoulis, V. G., & Bartek, J. (2008). An oncogene-induced DNA damage model for cancer development. Science, 319(5868), 1352–1355.PubMedCrossRef
9.
Zurück zum Zitat Janssen, A., van der Burg, M., Szuhai, K., Kops, G. J., & Medema, R. H. (2011). Chromosome segregation errors as a cause of DNA damage and structural chromosome aberrations. Science, 333(6051), 1895–1898.PubMedCrossRef Janssen, A., van der Burg, M., Szuhai, K., Kops, G. J., & Medema, R. H. (2011). Chromosome segregation errors as a cause of DNA damage and structural chromosome aberrations. Science, 333(6051), 1895–1898.PubMedCrossRef
10.
Zurück zum Zitat Hackett, J. A., & Greider, C. W. (2002). Balancing instability: dual roles for telomerase and telomere dysfunction in tumorigenesis. Oncogene, 21(4), 619–626.PubMedCrossRef Hackett, J. A., & Greider, C. W. (2002). Balancing instability: dual roles for telomerase and telomere dysfunction in tumorigenesis. Oncogene, 21(4), 619–626.PubMedCrossRef
11.
Zurück zum Zitat Stephens, P. J., Greenman, C. D., Fu, B., Yang, F., Bignell, G. R., Mudie, L. J., et al. (2011). Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell, 144(1), 27–40.PubMedCrossRef Stephens, P. J., Greenman, C. D., Fu, B., Yang, F., Bignell, G. R., Mudie, L. J., et al. (2011). Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell, 144(1), 27–40.PubMedCrossRef
12.
Zurück zum Zitat Lengauer, C., Kinzler, K. W., & Vogelstein, B. (1998). Genetic instabilities in human cancers. Nature, 396(6712), 643–649.PubMedCrossRef Lengauer, C., Kinzler, K. W., & Vogelstein, B. (1998). Genetic instabilities in human cancers. Nature, 396(6712), 643–649.PubMedCrossRef
13.
Zurück zum Zitat Boland, C. R., & Goel, A. (2010). Microsatellite instability in colorectal cancer. Gastroenterology, 138(6), 2073–2087. e2073.PubMedCrossRef Boland, C. R., & Goel, A. (2010). Microsatellite instability in colorectal cancer. Gastroenterology, 138(6), 2073–2087. e2073.PubMedCrossRef
14.
Zurück zum Zitat Di Nicolantonio, F., Martini, M., Molinari, F., Sartore-Bianchi, A., Arena, S., Saletti, P., et al. (2008). Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology, 26(35), 5705–5712.CrossRef Di Nicolantonio, F., Martini, M., Molinari, F., Sartore-Bianchi, A., Arena, S., Saletti, P., et al. (2008). Wild-type BRAF is required for response to panitumumab or cetuximab in metastatic colorectal cancer. Journal of Clinical Oncology: Official Journal of the American Society of Clinical Oncology, 26(35), 5705–5712.CrossRef
15.
Zurück zum Zitat Carter, S. L., Eklund, A. C., Kohane, I. S., Harris, L. N., & Szallasi, Z. (2006). A signature of chromosomal instability inferred from gene expression profiles predicts clinical outcome in multiple human cancers. Nature Genetics, 38(9), 1043–1048.PubMedCrossRef Carter, S. L., Eklund, A. C., Kohane, I. S., Harris, L. N., & Szallasi, Z. (2006). A signature of chromosomal instability inferred from gene expression profiles predicts clinical outcome in multiple human cancers. Nature Genetics, 38(9), 1043–1048.PubMedCrossRef
16.
Zurück zum Zitat Dworaczek, H., & Xiao, W. (2007). Xeroderma pigmentosum: a glimpse into nucleotide excision repair, genetic instability, and cancer. Critical Reviews in Oncogenesis, 13(2), 159–177.PubMedCrossRef Dworaczek, H., & Xiao, W. (2007). Xeroderma pigmentosum: a glimpse into nucleotide excision repair, genetic instability, and cancer. Critical Reviews in Oncogenesis, 13(2), 159–177.PubMedCrossRef
17.
Zurück zum Zitat Al-Tassan, N., Chmiel, N. H., Maynard, J., Fleming, N., Livingston, A. L., Williams, G. T., et al. (2002). Inherited variants of MYH associated with somatic G:C→T:A mutations in colorectal tumors. Nature Genetics, 30(2), 227–232.PubMedCrossRef Al-Tassan, N., Chmiel, N. H., Maynard, J., Fleming, N., Livingston, A. L., Williams, G. T., et al. (2002). Inherited variants of MYH associated with somatic G:C→T:A mutations in colorectal tumors. Nature Genetics, 30(2), 227–232.PubMedCrossRef
18.
Zurück zum Zitat Lee, H. C., Li, S. H., Lin, J. C., Wu, C. C., Yeh, D. C., & Wei, Y. H. (2004). Somatic mutations in the D-loop and decrease in the copy number of mitochondrial DNA in human hepatocellular carcinoma. Mutation Research, 547(1–2), 71–78.PubMedCrossRef Lee, H. C., Li, S. H., Lin, J. C., Wu, C. C., Yeh, D. C., & Wei, Y. H. (2004). Somatic mutations in the D-loop and decrease in the copy number of mitochondrial DNA in human hepatocellular carcinoma. Mutation Research, 547(1–2), 71–78.PubMedCrossRef
19.
Zurück zum Zitat Lee, H. C., Yin, P. H., Lin, J. C., Wu, C. C., Chen, C. Y., Wu, C. W., et al. (2005). Mitochondrial genome instability and mtDNA depletion in human cancers. Annals of the New York Academy of Sciences, 1042, 109–122.PubMedCrossRef Lee, H. C., Yin, P. H., Lin, J. C., Wu, C. C., Chen, C. Y., Wu, C. W., et al. (2005). Mitochondrial genome instability and mtDNA depletion in human cancers. Annals of the New York Academy of Sciences, 1042, 109–122.PubMedCrossRef
20.
Zurück zum Zitat Weissenbach, J., Gyapay, G., Dib, C., Vignal, A., Morissette, J., Millasseau, P., et al. (1992). A second-generation linkage map of the human genome. Nature, 359(6398), 794–801.PubMedCrossRef Weissenbach, J., Gyapay, G., Dib, C., Vignal, A., Morissette, J., Millasseau, P., et al. (1992). A second-generation linkage map of the human genome. Nature, 359(6398), 794–801.PubMedCrossRef
21.
Zurück zum Zitat Thibodeau, S. N., Bren, G., & Schaid, D. (1993). Microsatellite instability in cancer of the proximal colon. Science, 260(5109), 816–819.PubMedCrossRef Thibodeau, S. N., Bren, G., & Schaid, D. (1993). Microsatellite instability in cancer of the proximal colon. Science, 260(5109), 816–819.PubMedCrossRef
22.
Zurück zum Zitat Ionov, Y., Peinado, M. A., Malkhosyan, S., Shibata, D., & Perucho, M. (1993). Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis. Nature, 363(6429), 558–561.PubMedCrossRef Ionov, Y., Peinado, M. A., Malkhosyan, S., Shibata, D., & Perucho, M. (1993). Ubiquitous somatic mutations in simple repeated sequences reveal a new mechanism for colonic carcinogenesis. Nature, 363(6429), 558–561.PubMedCrossRef
23.
Zurück zum Zitat Risinger, J. I., Berchuck, A., Kohler, M. F., Watson, P., Lynch, H. T., & Boyd, J. (1993). Genetic instability of microsatellites in endometrial carcinoma. Cancer Research, 53(21), 5100–5103.PubMed Risinger, J. I., Berchuck, A., Kohler, M. F., Watson, P., Lynch, H. T., & Boyd, J. (1993). Genetic instability of microsatellites in endometrial carcinoma. Cancer Research, 53(21), 5100–5103.PubMed
24.
Zurück zum Zitat Rhyu, M. G., Park, W. S., & Meltzer, S. J. (1994). Microsatellite instability occurs frequently in human gastric carcinoma. Oncogene, 9(1), 29–32.PubMed Rhyu, M. G., Park, W. S., & Meltzer, S. J. (1994). Microsatellite instability occurs frequently in human gastric carcinoma. Oncogene, 9(1), 29–32.PubMed
25.
Zurück zum Zitat Halling, K. C., Harper, J., Moskaluk, C. A., Thibodeau, S. N., Petroni, G. R., Yustein, A. S., et al. (1999). Origin of microsatellite instability in gastric cancer. The American Journal of Pathology, 155(1), 205–211.PubMedCrossRef Halling, K. C., Harper, J., Moskaluk, C. A., Thibodeau, S. N., Petroni, G. R., Yustein, A. S., et al. (1999). Origin of microsatellite instability in gastric cancer. The American Journal of Pathology, 155(1), 205–211.PubMedCrossRef
26.
Zurück zum Zitat Kim, W. S., Park, C., Hong, S. K., Park, B. K., Kim, H. S., & Park, K. (2000). Microsatellite instability (MSI) in non-small cell lung cancer (NSCLC) is highly associated with transforming growth factor-beta type II receptor (TGF-beta RII) frameshift mutation. Anticancer Research, 20(3A), 1499–1502.PubMed Kim, W. S., Park, C., Hong, S. K., Park, B. K., Kim, H. S., & Park, K. (2000). Microsatellite instability (MSI) in non-small cell lung cancer (NSCLC) is highly associated with transforming growth factor-beta type II receptor (TGF-beta RII) frameshift mutation. Anticancer Research, 20(3A), 1499–1502.PubMed
27.
Zurück zum Zitat Fishel, R., Lescoe, M. K., Rao, M. R., Copeland, N. G., Jenkins, N. A., Garber, J., et al. (1993). The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer. Cell, 75(5), 1027–1038.PubMedCrossRef Fishel, R., Lescoe, M. K., Rao, M. R., Copeland, N. G., Jenkins, N. A., Garber, J., et al. (1993). The human mutator gene homolog MSH2 and its association with hereditary nonpolyposis colon cancer. Cell, 75(5), 1027–1038.PubMedCrossRef
28.
Zurück zum Zitat Leach, F. S., Nicolaides, N. C., Papadopoulos, N., Liu, B., Jen, J., Parsons, R., et al. (1993). Mutations of a mutS homolog in hereditary nonpolyposis colorectal cancer. Cell, 75(6), 1215–1225.PubMedCrossRef Leach, F. S., Nicolaides, N. C., Papadopoulos, N., Liu, B., Jen, J., Parsons, R., et al. (1993). Mutations of a mutS homolog in hereditary nonpolyposis colorectal cancer. Cell, 75(6), 1215–1225.PubMedCrossRef
29.
Zurück zum Zitat Bronner, C. E., Baker, S. M., Morrison, P. T., Warren, G., Smith, L. G., Lescoe, M. K., et al. (1994). Mutation in the DNA mismatch repair gene homologue hMLH1 is associated with hereditary non-polyposis colon cancer. Nature, 368(6468), 258–261.PubMedCrossRef Bronner, C. E., Baker, S. M., Morrison, P. T., Warren, G., Smith, L. G., Lescoe, M. K., et al. (1994). Mutation in the DNA mismatch repair gene homologue hMLH1 is associated with hereditary non-polyposis colon cancer. Nature, 368(6468), 258–261.PubMedCrossRef
30.
Zurück zum Zitat Nicolaides, N. C., Papadopoulos, N., Liu, B., Wei, Y. F., Carter, K. C., Ruben, S. M., et al. (1994). Mutations of two PMS homologues in hereditary nonpolyposis colon cancer. Nature, 371(6492), 75–80.PubMedCrossRef Nicolaides, N. C., Papadopoulos, N., Liu, B., Wei, Y. F., Carter, K. C., Ruben, S. M., et al. (1994). Mutations of two PMS homologues in hereditary nonpolyposis colon cancer. Nature, 371(6492), 75–80.PubMedCrossRef
31.
Zurück zum Zitat Palombo, F., Gallinari, P., Iaccarino, I., Lettieri, T., Hughes, M., D'Arrigo, A., et al. (1995). GTBP, a 160-kilodalton protein essential for mismatch-binding activity in human cells. Science, 268(5219), 1912–1914.PubMedCrossRef Palombo, F., Gallinari, P., Iaccarino, I., Lettieri, T., Hughes, M., D'Arrigo, A., et al. (1995). GTBP, a 160-kilodalton protein essential for mismatch-binding activity in human cells. Science, 268(5219), 1912–1914.PubMedCrossRef
32.
Zurück zum Zitat Papadopoulos, N., Nicolaides, N. C., Wei, Y. F., Ruben, S. M., Carter, K. C., Rosen, C. A., et al. (1994). Mutation of a mutL homolog in hereditary colon cancer. Science, 263(5153), 1625–1629.PubMedCrossRef Papadopoulos, N., Nicolaides, N. C., Wei, Y. F., Ruben, S. M., Carter, K. C., Rosen, C. A., et al. (1994). Mutation of a mutL homolog in hereditary colon cancer. Science, 263(5153), 1625–1629.PubMedCrossRef
33.
Zurück zum Zitat Toyota, M., Ahuja, N., Ohe-Toyota, M., Herman, J. G., Baylin, S. B., & Issa, J. P. (1999). CpG island methylator phenotype in colorectal cancer. Proceedings of the National Academy of Sciences of the United States of America, 96(15), 8681–8686.PubMedCrossRef Toyota, M., Ahuja, N., Ohe-Toyota, M., Herman, J. G., Baylin, S. B., & Issa, J. P. (1999). CpG island methylator phenotype in colorectal cancer. Proceedings of the National Academy of Sciences of the United States of America, 96(15), 8681–8686.PubMedCrossRef
34.
Zurück zum Zitat Issa, J. P. (2004). CpG island methylator phenotype in cancer. Nature Reviews. Cancer, 4(12), 988–993.PubMedCrossRef Issa, J. P. (2004). CpG island methylator phenotype in cancer. Nature Reviews. Cancer, 4(12), 988–993.PubMedCrossRef
35.
Zurück zum Zitat Goel, A., Nagasaka, T., Arnold, C. N., Inoue, T., Hamilton, C., Niedzwiecki, D., et al. (2007). The CpG island methylator phenotype and chromosomal instability are inversely correlated in sporadic colorectal cancer. Gastroenterology, 132(1), 127–138.PubMedCrossRef Goel, A., Nagasaka, T., Arnold, C. N., Inoue, T., Hamilton, C., Niedzwiecki, D., et al. (2007). The CpG island methylator phenotype and chromosomal instability are inversely correlated in sporadic colorectal cancer. Gastroenterology, 132(1), 127–138.PubMedCrossRef
36.
Zurück zum Zitat Ogino, S., Nosho, K., Kirkner, G. J., Kawasaki, T., Meyerhardt, J. A., Loda, M., et al. (2009). CpG island methylator phenotype, microsatellite instability, BRAF mutation and clinical outcome in colon cancer. Gut, 58(1), 90–96.PubMedCrossRef Ogino, S., Nosho, K., Kirkner, G. J., Kawasaki, T., Meyerhardt, J. A., Loda, M., et al. (2009). CpG island methylator phenotype, microsatellite instability, BRAF mutation and clinical outcome in colon cancer. Gut, 58(1), 90–96.PubMedCrossRef
37.
Zurück zum Zitat Vilar, E., & Gruber, S. B. (2010). Microsatellite instability in colorectal cancer—the stable evidence. Nature Reviews. Clinical Oncology, 7(3), 153–162.PubMedCrossRef Vilar, E., & Gruber, S. B. (2010). Microsatellite instability in colorectal cancer—the stable evidence. Nature Reviews. Clinical Oncology, 7(3), 153–162.PubMedCrossRef
38.
Zurück zum Zitat Ward, R., Meagher, A., Tomlinson, I., O'Connor, T., Norrie, M., Wu, R., et al. (2001). Microsatellite instability and the clinicopathological features of sporadic colorectal cancer. Gut, 48(6), 821–829.PubMedCrossRef Ward, R., Meagher, A., Tomlinson, I., O'Connor, T., Norrie, M., Wu, R., et al. (2001). Microsatellite instability and the clinicopathological features of sporadic colorectal cancer. Gut, 48(6), 821–829.PubMedCrossRef
39.
Zurück zum Zitat Gagos, S., & Irminger-Finger, I. (2005). Chromosome instability in neoplasia: chaotic roots to continuous growth. The International Journal of Biochemistry & Cell Biology, 37(5), 1014–1033.CrossRef Gagos, S., & Irminger-Finger, I. (2005). Chromosome instability in neoplasia: chaotic roots to continuous growth. The International Journal of Biochemistry & Cell Biology, 37(5), 1014–1033.CrossRef
40.
Zurück zum Zitat Lee, A. J., Endesfelder, D., Rowan, A. J., Walther, A., Birkbak, N. J., Futreal, P. A., et al. (2011). Chromosomal instability confers intrinsic multidrug resistance. Cancer Research, 71(5), 1858–1870.PubMedCrossRef Lee, A. J., Endesfelder, D., Rowan, A. J., Walther, A., Birkbak, N. J., Futreal, P. A., et al. (2011). Chromosomal instability confers intrinsic multidrug resistance. Cancer Research, 71(5), 1858–1870.PubMedCrossRef
41.
Zurück zum Zitat Walther, A., Houlston, R., & Tomlinson, I. (2008). Association between chromosomal instability and prognosis in colorectal cancer: a meta-analysis. Gut, 57(7), 941–950.PubMedCrossRef Walther, A., Houlston, R., & Tomlinson, I. (2008). Association between chromosomal instability and prognosis in colorectal cancer: a meta-analysis. Gut, 57(7), 941–950.PubMedCrossRef
42.
Zurück zum Zitat Solomon, D. A., Kim, T., Diaz-Martinez, L. A., Fair, J., Elkahloun, A. G., Harris, B. T., et al. (2011). Mutational inactivation of STAG2 causes aneuploidy in human cancer. Science, 333(6045), 1039–1043.PubMedCrossRef Solomon, D. A., Kim, T., Diaz-Martinez, L. A., Fair, J., Elkahloun, A. G., Harris, B. T., et al. (2011). Mutational inactivation of STAG2 causes aneuploidy in human cancer. Science, 333(6045), 1039–1043.PubMedCrossRef
43.
Zurück zum Zitat O'Sullivan, R. J., & Karlseder, J. (2010). Telomeres: protecting chromosomes against genome instability. Nature Reviews Molecular Cell Biology, 11(3), 171–181.PubMedCrossRef O'Sullivan, R. J., & Karlseder, J. (2010). Telomeres: protecting chromosomes against genome instability. Nature Reviews Molecular Cell Biology, 11(3), 171–181.PubMedCrossRef
44.
Zurück zum Zitat Levitt, N. C., & Hickson, I. D. (2002). Caretaker tumour suppressor genes that defend genome integrity. Trends in Molecular Medicine, 8(4), 179–186.PubMedCrossRef Levitt, N. C., & Hickson, I. D. (2002). Caretaker tumour suppressor genes that defend genome integrity. Trends in Molecular Medicine, 8(4), 179–186.PubMedCrossRef
45.
Zurück zum Zitat Rotman, G., & Shiloh, Y. (1998). ATM: from gene to function. Human Molecular Genetics, 7(10), 1555–1563.PubMedCrossRef Rotman, G., & Shiloh, Y. (1998). ATM: from gene to function. Human Molecular Genetics, 7(10), 1555–1563.PubMedCrossRef
46.
Zurück zum Zitat Cahill, D. P., Lengauer, C., Yu, J., Riggins, G. J., Willson, J. K., Markowitz, S. D., et al. (1998). Mutations of mitotic checkpoint genes in human cancers. Nature, 392(6673), 300–303.PubMedCrossRef Cahill, D. P., Lengauer, C., Yu, J., Riggins, G. J., Willson, J. K., Markowitz, S. D., et al. (1998). Mutations of mitotic checkpoint genes in human cancers. Nature, 392(6673), 300–303.PubMedCrossRef
47.
Zurück zum Zitat Hanks, S., Coleman, K., Reid, S., Plaja, A., Firth, H., Fitzpatrick, D., et al. (2004). Constitutional aneuploidy and cancer predisposition caused by biallelic mutations in BUB1B. Nature Genetics, 36(11), 1159–1161.PubMedCrossRef Hanks, S., Coleman, K., Reid, S., Plaja, A., Firth, H., Fitzpatrick, D., et al. (2004). Constitutional aneuploidy and cancer predisposition caused by biallelic mutations in BUB1B. Nature Genetics, 36(11), 1159–1161.PubMedCrossRef
48.
Zurück zum Zitat Li, Y., & Benezra, R. (1996). Identification of a human mitotic checkpoint gene: hsMAD2. Science, 274(5285), 246–248.PubMedCrossRef Li, Y., & Benezra, R. (1996). Identification of a human mitotic checkpoint gene: hsMAD2. Science, 274(5285), 246–248.PubMedCrossRef
49.
Zurück zum Zitat Negrini, S., Gorgoulis, V. G., & Halazonetis, T. D. (2010). Genomic instability—an evolving hallmark of cancer. Nature Reviews Molecular Cell Biology, 11(3), 220–228.PubMedCrossRef Negrini, S., Gorgoulis, V. G., & Halazonetis, T. D. (2010). Genomic instability—an evolving hallmark of cancer. Nature Reviews Molecular Cell Biology, 11(3), 220–228.PubMedCrossRef
50.
Zurück zum Zitat Maher, C. A., & Wilson, R. K. (2012). Chromothripsis and human disease: piecing together the shattering process. Cell, 148(1–2), 29–32.PubMedCrossRef Maher, C. A., & Wilson, R. K. (2012). Chromothripsis and human disease: piecing together the shattering process. Cell, 148(1–2), 29–32.PubMedCrossRef
51.
Zurück zum Zitat Forment, J. V., Kaidi, A., & Jackson, S. P. (2012). Chromothripsis and cancer: causes and consequences of chromosome shattering. Nature Reviews. Cancer, 12(10), 663–670.PubMedCrossRef Forment, J. V., Kaidi, A., & Jackson, S. P. (2012). Chromothripsis and cancer: causes and consequences of chromosome shattering. Nature Reviews. Cancer, 12(10), 663–670.PubMedCrossRef
52.
Zurück zum Zitat Abdel-Rahman, W. M., Katsura, K., Rens, W., Gorman, P. A., Sheer, D., Bicknell, D., et al. (2001). Spectral karyotyping suggests additional subsets of colorectal cancers characterized by pattern of chromosome rearrangement. Proceedings of the National Academy of Sciences of the United States of America, 98(5), 2538–2543.PubMedCrossRef Abdel-Rahman, W. M., Katsura, K., Rens, W., Gorman, P. A., Sheer, D., Bicknell, D., et al. (2001). Spectral karyotyping suggests additional subsets of colorectal cancers characterized by pattern of chromosome rearrangement. Proceedings of the National Academy of Sciences of the United States of America, 98(5), 2538–2543.PubMedCrossRef
53.
Zurück zum Zitat Muresu, R., Sini, M. C., Cossu, A., Tore, S., Baldinu, P., Manca, A., et al. (2002). Chromosomal abnormalities and microsatellite instability in sporadic endometrial cancer. European Journal of Cancer, 38(13), 1802–1809.PubMedCrossRef Muresu, R., Sini, M. C., Cossu, A., Tore, S., Baldinu, P., Manca, A., et al. (2002). Chromosomal abnormalities and microsatellite instability in sporadic endometrial cancer. European Journal of Cancer, 38(13), 1802–1809.PubMedCrossRef
54.
Zurück zum Zitat Geigl, J. B., Obenauf, A. C., Schwarzbraun, T., & Speicher, M. R. (2008). Defining ‘chromosomal instability’. Trends in Genetics: TIG, 24(2), 64–69.PubMedCrossRef Geigl, J. B., Obenauf, A. C., Schwarzbraun, T., & Speicher, M. R. (2008). Defining ‘chromosomal instability’. Trends in Genetics: TIG, 24(2), 64–69.PubMedCrossRef
55.
Zurück zum Zitat Wan, T. S., & Ma, E. S. (2012). Molecular cytogenetics: an indispensable tool for cancer diagnosis. Chang Gung Medical Journal, 35(2), 96–110.PubMed Wan, T. S., & Ma, E. S. (2012). Molecular cytogenetics: an indispensable tool for cancer diagnosis. Chang Gung Medical Journal, 35(2), 96–110.PubMed
56.
Zurück zum Zitat Beheshti, B., Park, P. C., Sweet, J. M., Trachtenberg, J., Jewett, M. A., & Squire, J. A. (2001). Evidence of chromosomal instability in prostate cancer determined by spectral karyotyping (SKY) and interphase FISH analysis. Neoplasia, 3(1), 62–69.PubMedCrossRef Beheshti, B., Park, P. C., Sweet, J. M., Trachtenberg, J., Jewett, M. A., & Squire, J. A. (2001). Evidence of chromosomal instability in prostate cancer determined by spectral karyotyping (SKY) and interphase FISH analysis. Neoplasia, 3(1), 62–69.PubMedCrossRef
57.
Zurück zum Zitat Bayani, J., & Squire, J. A. (2001). Advances in the detection of chromosomal aberrations using spectral karyotyping. Clinical Genetics, 59(2), 65–73.PubMedCrossRef Bayani, J., & Squire, J. A. (2001). Advances in the detection of chromosomal aberrations using spectral karyotyping. Clinical Genetics, 59(2), 65–73.PubMedCrossRef
58.
Zurück zum Zitat Ye, C. J., Stevens, J. B., Liu, G., Bremer, S. W., Jaiswal, A. S., Ye, K. J., et al. (2009). Genome based cell population heterogeneity promotes tumorigenicity: the evolutionary mechanism of cancer. Journal of Cellular Physiology, 219(2), 288–300.PubMedCrossRef Ye, C. J., Stevens, J. B., Liu, G., Bremer, S. W., Jaiswal, A. S., Ye, K. J., et al. (2009). Genome based cell population heterogeneity promotes tumorigenicity: the evolutionary mechanism of cancer. Journal of Cellular Physiology, 219(2), 288–300.PubMedCrossRef
59.
Zurück zum Zitat Zong, C., Lu, S., Chapman, A. R., & Xie, X. S. (2012). Genome-wide detection of single-nucleotide and copy-number variations of a single human cell. Science, 338(6114), 1622–1626.PubMedCrossRef Zong, C., Lu, S., Chapman, A. R., & Xie, X. S. (2012). Genome-wide detection of single-nucleotide and copy-number variations of a single human cell. Science, 338(6114), 1622–1626.PubMedCrossRef
60.
Zurück zum Zitat Darzynkiewicz, Z., Halicka, H. D., & Zhao, H. (2010). Analysis of cellular DNA content by flow and laser scanning cytometry. Advances in Experimental Medicine and Biology, 676, 137–147.PubMedCrossRef Darzynkiewicz, Z., Halicka, H. D., & Zhao, H. (2010). Analysis of cellular DNA content by flow and laser scanning cytometry. Advances in Experimental Medicine and Biology, 676, 137–147.PubMedCrossRef
61.
Zurück zum Zitat D'Urso, V., Collodoro, A., Mattioli, E., Giordano, A., & Bagella, L. (2010). Cytometry and DNA ploidy: clinical uses and molecular perspective in gastric and lung cancer. Journal of Cellular Physiology, 222(3), 532–539.PubMed D'Urso, V., Collodoro, A., Mattioli, E., Giordano, A., & Bagella, L. (2010). Cytometry and DNA ploidy: clinical uses and molecular perspective in gastric and lung cancer. Journal of Cellular Physiology, 222(3), 532–539.PubMed
62.
Zurück zum Zitat Ishkanian, A. S., Malloff, C. A., Watson, S. K., DeLeeuw, R. J., Chi, B., Coe, B. P., et al. (2004). A tiling resolution DNA microarray with complete coverage of the human genome. Nature Genetics, 36(3), 299–303.PubMedCrossRef Ishkanian, A. S., Malloff, C. A., Watson, S. K., DeLeeuw, R. J., Chi, B., Coe, B. P., et al. (2004). A tiling resolution DNA microarray with complete coverage of the human genome. Nature Genetics, 36(3), 299–303.PubMedCrossRef
63.
Zurück zum Zitat Pinkel, D., Segraves, R., Sudar, D., Clark, S., Poole, I., Kowbel, D., et al. (1998). High resolution analysis of DNA copy number variation using comparative genomic hybridization to microarrays. Nature Genetics, 20(2), 207–211.PubMedCrossRef Pinkel, D., Segraves, R., Sudar, D., Clark, S., Poole, I., Kowbel, D., et al. (1998). High resolution analysis of DNA copy number variation using comparative genomic hybridization to microarrays. Nature Genetics, 20(2), 207–211.PubMedCrossRef
64.
Zurück zum Zitat Heinrichs, S., & Look, A. T. (2007). Identification of structural aberrations in cancer by SNP array analysis. Genome Biology, 8(7), 219.PubMedCrossRef Heinrichs, S., & Look, A. T. (2007). Identification of structural aberrations in cancer by SNP array analysis. Genome Biology, 8(7), 219.PubMedCrossRef
65.
Zurück zum Zitat Zhao, X., Li, C., Paez, J. G., Chin, K., Janne, P. A., Chen, T. H., et al. (2004). An integrated view of copy number and allelic alterations in the cancer genome using single nucleotide polymorphism arrays. Cancer Research, 64(9), 3060–3071.PubMedCrossRef Zhao, X., Li, C., Paez, J. G., Chin, K., Janne, P. A., Chen, T. H., et al. (2004). An integrated view of copy number and allelic alterations in the cancer genome using single nucleotide polymorphism arrays. Cancer Research, 64(9), 3060–3071.PubMedCrossRef
66.
Zurück zum Zitat Gondek, L. P., Tiu, R., Haddad, A. S., O'Keefe, C. L., Sekeres, M. A., Theil, K. S., et al. (2007). Single nucleotide polymorphism arrays complement metaphase cytogenetics in detection of new chromosomal lesions in MDS. Leukemia: Official Journal of the Leukemia Society of America, Leukemia Research Fund, UK, 21(9), 2058–2061.CrossRef Gondek, L. P., Tiu, R., Haddad, A. S., O'Keefe, C. L., Sekeres, M. A., Theil, K. S., et al. (2007). Single nucleotide polymorphism arrays complement metaphase cytogenetics in detection of new chromosomal lesions in MDS. Leukemia: Official Journal of the Leukemia Society of America, Leukemia Research Fund, UK, 21(9), 2058–2061.CrossRef
67.
Zurück zum Zitat Goel, A., Nagasaka, T., Hamelin, R., & Boland, C. R. (2010). An optimized pentaplex PCR for detecting DNA mismatch repair-deficient colorectal cancers. PloS One, 5(2), e9393.PubMedCrossRef Goel, A., Nagasaka, T., Hamelin, R., & Boland, C. R. (2010). An optimized pentaplex PCR for detecting DNA mismatch repair-deficient colorectal cancers. PloS One, 5(2), e9393.PubMedCrossRef
68.
Zurück zum Zitat Janavicius, R., Matiukaite, D., Jakubauskas, A., & Griskevicius, L. (2010). Microsatellite instability detection by high-resolution melting analysis. Clinical Chemistry, 56(11), 1750–1757.PubMedCrossRef Janavicius, R., Matiukaite, D., Jakubauskas, A., & Griskevicius, L. (2010). Microsatellite instability detection by high-resolution melting analysis. Clinical Chemistry, 56(11), 1750–1757.PubMedCrossRef
69.
Zurück zum Zitat Meyerson, M., Gabriel, S., & Getz, G. (2010). Advances in understanding cancer genomes through second-generation sequencing. Nature Reviews Genetics, 11(10), 685–696.PubMedCrossRef Meyerson, M., Gabriel, S., & Getz, G. (2010). Advances in understanding cancer genomes through second-generation sequencing. Nature Reviews Genetics, 11(10), 685–696.PubMedCrossRef
70.
Zurück zum Zitat Wheeler, D. A., Srinivasan, M., Egholm, M., Shen, Y., Chen, L., McGuire, A., et al. (2008). The complete genome of an individual by massively parallel DNA sequencing. Nature, 452(7189), 872–876.PubMedCrossRef Wheeler, D. A., Srinivasan, M., Egholm, M., Shen, Y., Chen, L., McGuire, A., et al. (2008). The complete genome of an individual by massively parallel DNA sequencing. Nature, 452(7189), 872–876.PubMedCrossRef
71.
Zurück zum Zitat Ng, P. C., & Kirkness, E. F. (2010). Whole genome sequencing. Methods in Molecular Biology, 628, 215–226.PubMedCrossRef Ng, P. C., & Kirkness, E. F. (2010). Whole genome sequencing. Methods in Molecular Biology, 628, 215–226.PubMedCrossRef
72.
Zurück zum Zitat Campbell, P. J., Stephens, P. J., Pleasance, E. D., O'Meara, S., Li, H., Santarius, T., et al. (2008). Identification of somatically acquired rearrangements in cancer using genome-wide massively parallel paired-end sequencing. Nature Genetics, 40(6), 722–729.PubMedCrossRef Campbell, P. J., Stephens, P. J., Pleasance, E. D., O'Meara, S., Li, H., Santarius, T., et al. (2008). Identification of somatically acquired rearrangements in cancer using genome-wide massively parallel paired-end sequencing. Nature Genetics, 40(6), 722–729.PubMedCrossRef
73.
Zurück zum Zitat Chiang, D. Y., Getz, G., Jaffe, D. B., O'Kelly, M. J., Zhao, X., Carter, S. L., et al. (2009). High-resolution mapping of copy-number alterations with massively parallel sequencing. Nature Methods, 6(1), 99–103.PubMedCrossRef Chiang, D. Y., Getz, G., Jaffe, D. B., O'Kelly, M. J., Zhao, X., Carter, S. L., et al. (2009). High-resolution mapping of copy-number alterations with massively parallel sequencing. Nature Methods, 6(1), 99–103.PubMedCrossRef
74.
Zurück zum Zitat Ding, L., Getz, G., Wheeler, D. A., Mardis, E. R., McLellan, M. D., Cibulskis, K., et al. (2008). Somatic mutations affect key pathways in lung adenocarcinoma. Nature, 455(7216), 1069–1075.PubMedCrossRef Ding, L., Getz, G., Wheeler, D. A., Mardis, E. R., McLellan, M. D., Cibulskis, K., et al. (2008). Somatic mutations affect key pathways in lung adenocarcinoma. Nature, 455(7216), 1069–1075.PubMedCrossRef
75.
Zurück zum Zitat Bell, D. (2011). Integrated genomic analyses of ovarian carcinoma. Nature, 474(7353), 609–615.CrossRef Bell, D. (2011). Integrated genomic analyses of ovarian carcinoma. Nature, 474(7353), 609–615.CrossRef
76.
Zurück zum Zitat McLendon, R. (2008). Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature, 455(7216), 1061–1068.CrossRef McLendon, R. (2008). Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature, 455(7216), 1061–1068.CrossRef
77.
Zurück zum Zitat Shah, S. P., Roth, A., Goya, R., Oloumi, A., Ha, G., Zhao, Y., et al. (2012). The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature, 486(7403), 395–399.PubMed Shah, S. P., Roth, A., Goya, R., Oloumi, A., Ha, G., Zhao, Y., et al. (2012). The clonal and mutational evolution spectrum of primary triple-negative breast cancers. Nature, 486(7403), 395–399.PubMed
78.
Zurück zum Zitat Seo JS, Ju YS, Lee WC, Shin JY, Lee JK, Bleazard T, Lee J, Jung YJ, Kim JO, Yu SB et al. (2012). The transcriptional landscape and mutational profile of lung adenocarcinoma. Genome Research, 22, 2109–2119. Seo JS, Ju YS, Lee WC, Shin JY, Lee JK, Bleazard T, Lee J, Jung YJ, Kim JO, Yu SB et al. (2012). The transcriptional landscape and mutational profile of lung adenocarcinoma. Genome Research, 22, 2109–2119.
79.
Zurück zum Zitat Berger, M. F., Lawrence, M. S., Demichelis, F., Drier, Y., Cibulskis, K., Sivachenko, A. Y., et al. (2011). The genomic complexity of primary human prostate cancer. Nature, 470(7333), 214–220.PubMedCrossRef Berger, M. F., Lawrence, M. S., Demichelis, F., Drier, Y., Cibulskis, K., Sivachenko, A. Y., et al. (2011). The genomic complexity of primary human prostate cancer. Nature, 470(7333), 214–220.PubMedCrossRef
80.
Zurück zum Zitat Jones, S., Zhang, X., Parsons, D. W., Lin, J. C., Leary, R. J., Angenendt, P., et al. (2008). Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science, 321(5897), 1801–1806.PubMedCrossRef Jones, S., Zhang, X., Parsons, D. W., Lin, J. C., Leary, R. J., Angenendt, P., et al. (2008). Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science, 321(5897), 1801–1806.PubMedCrossRef
81.
Zurück zum Zitat Wood, L. D., Parsons, D. W., Jones, S., Lin, J., Sjoblom, T., Leary, R. J., et al. (2007). The genomic landscapes of human breast and colorectal cancers. Science, 318(5853), 1108–1113.PubMedCrossRef Wood, L. D., Parsons, D. W., Jones, S., Lin, J., Sjoblom, T., Leary, R. J., et al. (2007). The genomic landscapes of human breast and colorectal cancers. Science, 318(5853), 1108–1113.PubMedCrossRef
82.
Zurück zum Zitat Mitelman, F., Johansson, B., & Mertens, F. (2004). Fusion genes and rearranged genes as a linear function of chromosome aberrations in cancer. Nature Genetics, 36(4), 331–334.PubMedCrossRef Mitelman, F., Johansson, B., & Mertens, F. (2004). Fusion genes and rearranged genes as a linear function of chromosome aberrations in cancer. Nature Genetics, 36(4), 331–334.PubMedCrossRef
83.
Zurück zum Zitat Vollebergh, M. A., Jonkers, J., & Linn, S. C. (2012). Genomic instability in breast and ovarian cancers: translation into clinical predictive biomarkers. Cellular and Molecular Life Sciences: CMLS, 69(2), 223–245.PubMedCrossRef Vollebergh, M. A., Jonkers, J., & Linn, S. C. (2012). Genomic instability in breast and ovarian cancers: translation into clinical predictive biomarkers. Cellular and Molecular Life Sciences: CMLS, 69(2), 223–245.PubMedCrossRef
84.
Zurück zum Zitat Huang, Y. T., Lin, X., Liu, Y., Chirieac, L. R., McGovern, R., Wain, J., et al. (2011). Cigarette smoking increases copy number alterations in nonsmall-cell lung cancer. Proceedings of the National Academy of Sciences of the United States of America, 108(39), 16345–16350.PubMedCrossRef Huang, Y. T., Lin, X., Liu, Y., Chirieac, L. R., McGovern, R., Wain, J., et al. (2011). Cigarette smoking increases copy number alterations in nonsmall-cell lung cancer. Proceedings of the National Academy of Sciences of the United States of America, 108(39), 16345–16350.PubMedCrossRef
85.
Zurück zum Zitat Massion, P. P., Zou, Y., Chen, H., Jiang, A., Coulson, P., Amos, C. I., et al. (2008). Smoking-related genomic signatures in non-small cell lung cancer. American Journal of Respiratory and Critical Care Medicine, 178(11), 1164–1172.PubMedCrossRef Massion, P. P., Zou, Y., Chen, H., Jiang, A., Coulson, P., Amos, C. I., et al. (2008). Smoking-related genomic signatures in non-small cell lung cancer. American Journal of Respiratory and Critical Care Medicine, 178(11), 1164–1172.PubMedCrossRef
86.
Zurück zum Zitat Thu, K. L., Vucic, E. A., Chari, R., Zhang, W., Lockwood, W. W., English, J. C., et al. (2012). Lung adenocarcinoma of never smokers and smokers harbor differential regions of genetic alteration and exhibit different levels of genomic instability. PloS One, 7(3), e33003.PubMedCrossRef Thu, K. L., Vucic, E. A., Chari, R., Zhang, W., Lockwood, W. W., English, J. C., et al. (2012). Lung adenocarcinoma of never smokers and smokers harbor differential regions of genetic alteration and exhibit different levels of genomic instability. PloS One, 7(3), e33003.PubMedCrossRef
87.
Zurück zum Zitat Bowtell, D. D. (2010). The genesis and evolution of high-grade serous ovarian cancer. Nature Reviews. Cancer, 10(11), 803–808.PubMedCrossRef Bowtell, D. D. (2010). The genesis and evolution of high-grade serous ovarian cancer. Nature Reviews. Cancer, 10(11), 803–808.PubMedCrossRef
88.
Zurück zum Zitat Fang, M., Toher, J., Morgan, M., Davison, J., Tannenbaum, S., & Claffey, K. (2011). Genomic differences between estrogen receptor (ER)-positive and ER-negative human breast carcinoma identified by single nucleotide polymorphism array comparative genome hybridization analysis. Cancer, 117(10), 2024–2034.PubMedCrossRef Fang, M., Toher, J., Morgan, M., Davison, J., Tannenbaum, S., & Claffey, K. (2011). Genomic differences between estrogen receptor (ER)-positive and ER-negative human breast carcinoma identified by single nucleotide polymorphism array comparative genome hybridization analysis. Cancer, 117(10), 2024–2034.PubMedCrossRef
89.
Zurück zum Zitat Lockwood, W. W., Wilson, I. M., Coe, B. P., Chari, R., Pikor, L. A., Thu, K. L., et al. (2012). Divergent genomic and epigenomic landscapes of lung cancer subtypes underscore the selection of different oncogenic pathways during tumor development. PloS One, 7(5), e37775.PubMedCrossRef Lockwood, W. W., Wilson, I. M., Coe, B. P., Chari, R., Pikor, L. A., Thu, K. L., et al. (2012). Divergent genomic and epigenomic landscapes of lung cancer subtypes underscore the selection of different oncogenic pathways during tumor development. PloS One, 7(5), e37775.PubMedCrossRef
90.
Zurück zum Zitat Wilkerson, M. D., Yin, X., Walter, V., Zhao, N., Cabanski, C. R., Hayward, M. C., et al. (2012). Differential pathogenesis of lung adenocarcinoma subtypes involving sequence mutations, copy number, chromosomal instability, and methylation. PloS One, 7(5), e36530.PubMedCrossRef Wilkerson, M. D., Yin, X., Walter, V., Zhao, N., Cabanski, C. R., Hayward, M. C., et al. (2012). Differential pathogenesis of lung adenocarcinoma subtypes involving sequence mutations, copy number, chromosomal instability, and methylation. PloS One, 7(5), e36530.PubMedCrossRef
91.
Zurück zum Zitat Loeb, L. A. (2011). Human cancers express mutator phenotypes: origin, consequences and targeting. Nature Reviews. Cancer, 11(6), 450–457.PubMedCrossRef Loeb, L. A. (2011). Human cancers express mutator phenotypes: origin, consequences and targeting. Nature Reviews. Cancer, 11(6), 450–457.PubMedCrossRef
Metadaten
Titel
The detection and implication of genome instability in cancer
verfasst von
Larissa Pikor
Kelsie Thu
Emily Vucic
Wan Lam
Publikationsdatum
01.12.2013
Verlag
Springer US
Erschienen in
Cancer and Metastasis Reviews / Ausgabe 3-4/2013
Print ISSN: 0167-7659
Elektronische ISSN: 1573-7233
DOI
https://doi.org/10.1007/s10555-013-9429-5

Weitere Artikel der Ausgabe 3-4/2013

Cancer and Metastasis Reviews 3-4/2013 Zur Ausgabe

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Alectinib verbessert krankheitsfreies Überleben bei ALK-positivem NSCLC

25.04.2024 NSCLC Nachrichten

Das Risiko für Rezidiv oder Tod von Patienten und Patientinnen mit reseziertem ALK-positivem NSCLC ist unter einer adjuvanten Therapie mit dem Tyrosinkinase-Inhibitor Alectinib signifikant geringer als unter platinbasierter Chemotherapie.

Bei Senioren mit Prostatakarzinom auf Anämie achten!

24.04.2024 DGIM 2024 Nachrichten

Patienten, die zur Behandlung ihres Prostatakarzinoms eine Androgendeprivationstherapie erhalten, entwickeln nicht selten eine Anämie. Wer ältere Patienten internistisch mitbetreut, sollte auf diese Nebenwirkung achten.

ICI-Therapie in der Schwangerschaft wird gut toleriert

Müssen sich Schwangere einer Krebstherapie unterziehen, rufen Immuncheckpointinhibitoren offenbar nicht mehr unerwünschte Wirkungen hervor als andere Mittel gegen Krebs.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.