Single nucleotide polymorphism arrays (SNP-A) have recently been widely applied as a powerful karyotyping tool in numerous translational cancer studies. SNP-A complements traditional metaphase cytogenetics with the unique ability to delineate a previously hidden chromosomal defect, copy neutral loss of heterozygosity (CN-LOH). Emerging data demonstrate that selected hematologic malignancies exhibit abundant CN-LOH, often in the setting of a normal metaphase karyotype and no previously identified clonal marker. In this review, we explore emerging biologic and clinical features of CN-LOH relevant to hematologic malignancies. In myeloid malignancies, CN-LOH has been associated with the duplication of oncogenic mutations with concomitant loss of the normal allele. Examples include JAK2, MPL, c-KIT, and FLT3. More recent investigations have focused on evaluation of candidate genes contained in common CN-LOH and deletion regions and have led to the discovery of tumor suppressor genes, including c-CBL and family members, as well as TET2. Investigations into the underlying mechanisms generating CN-LOH have great promise for elucidating general cancer mechanisms. We anticipate that further detailed characterization of CN-LOH lesions will probably facilitate our discovery of a more complete set of pathogenic molecular lesions, disease and prognosis markers, and better understanding of the initiation and progression of hematologic malignancies.

Single nucleotide polymorphism arrays (SNP-A) are currently in use as a powerful genotyping tool for a variety of whole-genome association studies. With resolution as good as one marker per every 100 bp of the genome and quantitative analysis of DNA copy number as well as genotypic information provided by polymorphic SNP markers, an outgrowth of this technology was the realization that SNP-A could potentially provide a powerful karyotyping tool.1  For example, application of SNP-A karyotyping to a large series of pediatric acute lymphocytic leukemia cases identified recurrent subcytogenetic deletions and target genes in the minimal deletion intervals, and new cancer-specific aberrant genetic networks that include PAX5, IKZF1, and CDKN2A.2-4  In addition to the identification of cryptic amplifications and deletions, SNP-A also complements traditional metaphase cytogenetics with the unique ability to delineate a previously underappreciated chromosomal defect, copy neutral loss of heterozygosity (CN-LOH). There are many technical challenges to accurately identifying CN-LOH regions from SNP-A raw data, and CN-LOH results were not reported in the initial acute lymphocytic leukemia (ALL) studies.5  For acute myeloid leukemia (AML) and myelodysplastic syndromes (MDSs), however, even early evaluations using low-density arrays demonstrated CN-LOH as a recurrent finding of potential interest.6  Here, we review the mechanisms that lead to CN-LOH, explore disease-specific CN-LOH patterns and potential biologic implications, and examine the evolving potential clinical significance of this unique genomic finding.

CN-LOH, also often referred as to uniparental disomy (UPD),7  leads to LOH by duplication of a maternal (unimaternal) or paternal (unipaternal) chromosome or chromosomal region and concurrent loss of the other allele. Whereas CN-LOH was initially identified by studying microsatellites, systematic application of SNP-A as a karyotyping tool led to the realization that the previously undetected areas of clonal CN-LOH are frequently encountered in various cancers, including hematologic malignancies.1,8  SNP-A–identified CN-LOH in various malignancies1,9  can be described according to its derivation or its location (Figure 1). CN-LOH, either chromosomal or segmental, can have constitutional or acquired origins. Constitutional UPD is associated with meiotic errors, resulting in developmental diseases, and was first described in humans by Engel10 ; however, it can also be observed in healthy controls, probably because of diverse mechanisms, including early mitotic errors and autozygosity.11-13 

Figure 1

Classification of CN-LOH. CN-LOH can be classified by either its origin or its location. CN-LOH can have an acquired, clonal derivation or a constitutional, nonclonal derivation. Nonclonal CN-LOH can be the result of an early embryonic mitotic event, leading to mosaicism, or may be truly constitutional. This constitutional CN-LOH can arise from autozygosity or meiotic events, including trisomic rescue, gamete complementation, duplication of a monosomic chromosome in an aneuploid zygote, or nonhomologous recombination. In addition, CN-LOH can be either segmental or numerical. Segmental CN-LOH arising from one crossing over will be telomeric (a, bottom left), whereas 2 crossing-over events will lead to interstitial CN-LOH (b). CN-LOH can also involve an entire chromosome (numeric; c, d, and e).

Figure 1

Classification of CN-LOH. CN-LOH can be classified by either its origin or its location. CN-LOH can have an acquired, clonal derivation or a constitutional, nonclonal derivation. Nonclonal CN-LOH can be the result of an early embryonic mitotic event, leading to mosaicism, or may be truly constitutional. This constitutional CN-LOH can arise from autozygosity or meiotic events, including trisomic rescue, gamete complementation, duplication of a monosomic chromosome in an aneuploid zygote, or nonhomologous recombination. In addition, CN-LOH can be either segmental or numerical. Segmental CN-LOH arising from one crossing over will be telomeric (a, bottom left), whereas 2 crossing-over events will lead to interstitial CN-LOH (b). CN-LOH can also involve an entire chromosome (numeric; c, d, and e).

Close modal

Events leading to CN-LOH in developmental disorders help to clarify the pathways that could lead to constitutional UPD observed in healthy persons and distinguish them from acquired UPD (aUPD) in malignancies that, similar to other acquired chromosomal defects, constitute markers of clonality (Figure 1). UPD can occur during meiosis or as a postfertilization mitotic event at a very early embryonic stage, leading to mosaicism.14  Trisomy rescue (loss of a chromosome from a trisomic zygote) of a meiotic nondisjunction error leads to CN-LOH and, depending on the meiotic stage at which the nondisjunction event occurred, results in uniparental isodisomy (meiosis II error) or heterodisomy (meiosis I error).15  In two-thirds of the cases, trisomy rescue would result in a normal diploid, biparental karyotype. A meiosis I nondisjunction error can be followed by a mitotic crossing over between 2 nonuniparental chromatids followed by a loss of chromosomal material originating from a disomic gamete. In a similar setting, UPD can also be associated with an isochromosome arising from meiotic misdivision of the centromeres, with fertilization by a normal gamete and subsequent loss of the homologous chromosome. In addition, fertilization between disomic and nullisomic gametes can produce a similar result. Finally, mitotic misdivision at the centromere in a monosomic zygote can result in CN-LOH (Figure 1).

Theoretically, CN-LOH can lead to either gain of imprinting through duplication of a methylated allele or loss of imprinting through duplication of unmethylated allele.16  For this mechanism to be pathogenic, the CN-LOH region would have to involve genes that are subject to genomic imprinting, such as those in Prader-Willi/Angelman syndromes.17  Detailed descriptions of the discovery of genomic imprinting and pathogenesis of uniparental disomy-related disorders are presented elsewhere.18 

Autozygosity is the inheritance of the same ancestral genomic region from both parents. Small stretches (1-5 Mb) of CN-LOH are thought to be the result of autozygosity, in particular in inbred ethnic groups.13  Theoretically, reference samples used as “controls” in some LOH studies may indeed have unexpectedly high genetic similarity from undiscovered consanguinity.19  However, the cause of larger regions of CN-LOH seen in significant proportions of healthy unrelated controls studied in various projects has not been precisely identified, and the size of regions of CN-LOH is expected to get smaller with the increasingly outbred nature of urban human populations.20  Of great interest, forms of constitutional LOH have been implicated in predisposition to malignancies, a fact best illustrated in inbred ethnic populations.21-24  Comparisons of germline SNP-A data of 74 colorectal cancer patients identified that the percentage of those with autozygous segments of 4 Mb or more is at least twice as high as in control groups.25  However, specific locations and disease associations, including their contribution to disease risk, are currently not well defined.

Extrinsic processes that lead to genomic DNA damage upstream of sites of mitotic recombination have also been implicated in the generation of acquired CN-LOH. Whether treatment with DNA-damaging chemotherapy contributes to the increase in aUPD seen in secondary AML, transformed follicular lymphoma or relapsed versus de novo ALL is unknown. Specific mechanisms responsible for the propensity for chromosomal breaks and subsequent uniallelic strand loss that might occur in MDS or AML before therapy are not well understood, but it is probable that a variety may be involved. These may include acquired or inherited weakness of various components of DNA repair machinery or mitotic spindle machinery or telomere shortening.26  Finally, a supersaturating overwhelming of rate-limiting DNA repair components could theoretically provide another mechanism. The cumulative acquisition of lesions fits well with current models of the association of age with accumulated genetic and epigenetic lesions but remains to be experimentally investigated. For example, whether there is specifically more CN-LOH in disease-associated or even hematologically disease-free aged persons is currently unknown. Chronic lymphocytic leukemia (CLL), myeloproliferative neoplasia (MPN), MDS, and AML are all associated with significant increased incidence with age.

The interpretation of the origin, location, frequency, and clinical implications of CN-LOH ultimately requires an understanding of the potential extrinsic disease-related and intrinsic chromosomal determinants associated with CN-LOH/aUPD development. Unfortunately, there are many more questions than answers related to these issues. Various mechanisms can be responsible for this form of LOH, now found to be very prevalent in various myeloid malignancies. These mechanisms include a cellular attempt to correct a deletion because of mitotic errors, such as anaphase lag through reduplication of the remaining chromosome. In addition, an attempted repair of double-strand breaks resulting in losses whereby the lost region is replaced using the remaining allele as a template can lead to segmental LOH (Figure 2).

Figure 2

Mitotic mechanisms of formation of CN-LOH. (A) CN-LOH can occur as the result of mitotic recombination between homologous chromosomes. Depending on how the chromosomes are sorted during mitosis, daughter cells with CN-LOH can arise. (B) CN-LOH can also arise as the consequence of deletion followed by recombination using the homolog as a template for correction.

Figure 2

Mitotic mechanisms of formation of CN-LOH. (A) CN-LOH can occur as the result of mitotic recombination between homologous chromosomes. Depending on how the chromosomes are sorted during mitosis, daughter cells with CN-LOH can arise. (B) CN-LOH can also arise as the consequence of deletion followed by recombination using the homolog as a template for correction.

Close modal

Mitotic recombination is considered a major contributor to acquired CN-LOH.27  In the first whole-genome SNP-A study of aUPD in AML, large regions of CN-LOH were identified in 20% of the patients studied; 8 separate nonrandom regions were identified,6  differing in locations from those identified in similar studies of epithelial cancers.28,29  Most regions of homozygosity were partial but extended to the telomere, consistent with UPD occurring as a result of a single mitotic recombination. Subsequent studies of other myeloid malignancies also identified partial aUPD as the most frequent type of CN-LOH in these disorders.1  However, investigations of the molecular basis for somatic NF1 inactivation in childhood leukemias associated with neurofibromatosis type I elegantly illustrated that interstitial UPD can also represent a pathway of aUPD by double-homologous recombination events30 ; 8 of 10 cases had large LOH, half were partial UPD, but 4 had interstitial UPD. Preferred sites of mitotic recombination were also identified, with a clustering of the centromeric and telomeric breakpoints. Similarly, a careful analysis of the sites of aUPD origin in low-risk MDS showed that 43% of UPD regions were localized to within or as part of a previously identified fragile site.31  Fragile locations correspond to known sites of frequent genomic instability. They are associated with the breakpoints of chromosomal aberrations in hematologic malignancies32  and often track with regulatory microRNA amplifications and deletions.33 

A high-resolution SNP-A profiling of mantle cell lymphoma (MCL) cell lines and primary tumors34  has provided additional insight into mechanics of aUPD. In MCL, pathognomonic t(11;14)(q13;q32) are associated with secondary chromosomal alterations, including frequent areas of partial UPD, such as UPD17p coinciding with homozygous TP53 inactivation. The breakpoints flanking the genomic alterations, including regions of UPD, were significantly associated with genomic regions enriched in copy number variants and segmental duplications, suggesting that recombination at these regions may play a role in the genetic instability. Thus, copy number variants and segmental duplications may represent DNA breakage-prone regions that may contribute to the generation of chromosomal alterations by facilitating nonallelic homologous recombination, similar to fragile sites.35-37  Additional mechanistic clues have been derived from colon cancer, in which mitotic recombination appears to be important for inactivating tumor suppressor genes. The APC gene located at 5q21 is frequently mutated, often in a homozygous constellation resulting from CN-LOH, through a mitotic recombination mechanism.38  Studies demonstrate that copy number changes in aneuploid/polyploid colorectal tumors generally occur as additional genetic events, whereas LOH by mitotic recombination is an early event that initiates tumorigenesis.39  By mapping mitotic recombination breakpoints between the centromere and APC, they were again found to be nonrandom, with the highest frequency close to locus control regions at 68 to 71 Mb, far from APC. Low copy repeats predispose to chromosomal breakage, perhaps via replication fork stalling, leading to mitotic recombination and ultimately CN-LOH. In contrast, breakpoints involved in APC copy number loss clustered to a different, more centromeric region near a suggested area of the greatest chromosomal fragility. Consequently, it is possible that breakage through the centromere cannot be readily repaired by mitotic recombination because the exposure of pericentromeric repeats produces a chromosome that is prone to nonspecific pairings and recombination.

Specific oncogenes have also been implicated in mechanisms of genomic instability, which make these intriguing candidates to consider as potentially mechanistically involved in the generation of CN-LOH. For example, BRCA1/2 has been linked to UPD seen in ovarian cancer.40  Cyclin D1 is overexpressed in the majority of MCLs, which have abundant aUPD.34  UPD has also been associated with microsatellite instability in AML with normal karyotype.41  The JAK2 V617F mutation is frequently observed in classic MPN, and disease progression is associated with biallelic acquisition of the mutation through mitotic recombination and aUPD. However, in manipulated cell lines and CD34+ cells from patients with JAK2 V617F, an increase in homologous recombination activity in the presence of erythropoietin was observed, without modifications in nonhomologous end joining efficiency.42 

Traditional karyotyping techniques, including metaphase cytogenetics, cannot detect CN-LOH. In the past, microsatellite analysis was used for detection of CN-LOH. Informative microsatellites present in a germline control sample in a heterozygous constellation were compared with the tumor sample from the same patient to identify LOH. A diploid copy number was shown by karyotyping.43  This approach is cumbersome, and the boundaries of the defect cannot be precisely delineated because of natural limitations of the number of informative microsatellites present in the genome.

SNP-A combines genotyping (classification of a homozygous or heterozygous constellation at a polymorphic locus) and copy number analysis (intensity of hybridization signal).7  A number of general and technical reviews have outlined the conceptual and technical aspects of performing SNP-A karyotyping.1,44  The precision of this technology is increased by the use of bioanalytic algorithms, facilitating a very accurate diagnosis of LOH (Figure 3). Current algorithms are based on the deviation between observed and expected frequencies of homozygous SNPs, which should be approximately 1 of 30 consecutive SNPs tested.45  Consequently, the longer a region of LOH, the higher the number of homozygous SNPs and herewith the statistical improbability of encountering such a situation per chance rises. When a homozygous constellation of genotyping calls is combined with diploid copy number, the diagnosis of CN-LOH can be established (Figure 3). By comparison, comparative genomic hybridization arrays rely solely on analysis of hybridization signals and CN-LOH cannot be detected. The presence of nonclonal diploid cells in the tested DNA sample complicates the SNP-A–based detection algorithm, and in general, the larger the aberrant clone, the better the detection of CN-LOH. In our hands, mixing experiments have shown that even dilution of the clonal, aberrant cells to 30% of the total sample can still allow reliable detection of an acquired abnormality.46 

Figure 3

Determination of acquired versus germline nature of CN-LOH. Acquired CN-LOH (red bar, top left) is identified when allelic imbalance (as shown by genotyping calls) with normal copy number (top track) in bone marrow and not CD3+ cells (representing the germline configuration). Top left: An example of acquired CN-LOH of chromosome 7. A region of homozygosity and diploid copy number (as indicated by the red bar) are seen in bone marrow only. Top right: An example of germline CN-LOH of chromosome 20. Runs of homozygosity (red bars) are present in both bone marrow and CD3+ cells. Among a cohort of 1003 healthy controls, CN-LOH was mainly interstitial (bottom left) and ranged in size from 0.3 to 65 Mb (median, 8.7 Mb; bottom center). Bottom right: The distribution of nonclonal, germline CN-LOH in controls on an exemplary chromosome (chromosome 6).

Figure 3

Determination of acquired versus germline nature of CN-LOH. Acquired CN-LOH (red bar, top left) is identified when allelic imbalance (as shown by genotyping calls) with normal copy number (top track) in bone marrow and not CD3+ cells (representing the germline configuration). Top left: An example of acquired CN-LOH of chromosome 7. A region of homozygosity and diploid copy number (as indicated by the red bar) are seen in bone marrow only. Top right: An example of germline CN-LOH of chromosome 20. Runs of homozygosity (red bars) are present in both bone marrow and CD3+ cells. Among a cohort of 1003 healthy controls, CN-LOH was mainly interstitial (bottom left) and ranged in size from 0.3 to 65 Mb (median, 8.7 Mb; bottom center). Bottom right: The distribution of nonclonal, germline CN-LOH in controls on an exemplary chromosome (chromosome 6).

Close modal

Of utmost importance for the detection of tumor-associated, clonal, acquired CN-LOH is its distinction from germline-encoded forms of CN-LOH, seen in up to 15% of control persons.31  Analysis of 1000 nonclonal control samples (Figure 3; C.O., unpublished results, April 2009) shows that germline regions of CN-LOH usually are smaller (median, 8.7 Mb) and primarily interstitial; whereas in our more than 600 cases of myeloid malignancies, acquired clonal CN-LOH seem virtually uniformly telomeric.46-50  Regions of germline CN-LOH identified in controls show a characteristic size and distribution across the chromosome as show for exemplary chromosome 6 (Figure 3). Interstitial CN-LOH is difficult to explain based on homologous recombination, in particular for LOH regions that are genetically small. Thus, smaller (< 24.6 Mb or 95th percentile of distribution) regions of CN-LOH in particular must be confirmed by analysis of germline DNA to be unambiguously defined as acquired CN-LOH (Figure 3).

Conceptually, CN-LOH may have several implications. In one way, duplications of chromosomal material constitute UPD as it occurs with the retention of the homologous chromosome, thus implying that UPD without LOH is also possible (Figure 1). In addition, we have observed uniparental trisomy of various chromosomes, including chromosome 21. Although theoretically all chromosomes can be affected, UPD of certain chromosomes is more common. For example, in AML and MDS, chromosomes 4, 7, 11, 13, and 21 show frequently recurrent aUPD (Figure 4).

Figure 4

Genomic distribution of acquired CN-LOH in MDS/secondary AML and primary AML. CN-LOH is nonrandomly distributed across the genome in both MDS/secondary AML (blue lines) and primary AML (red lines), with some chromosomes and chromosomal regions being more frequently affected.

Figure 4

Genomic distribution of acquired CN-LOH in MDS/secondary AML and primary AML. CN-LOH is nonrandomly distributed across the genome in both MDS/secondary AML (blue lines) and primary AML (red lines), with some chromosomes and chromosomal regions being more frequently affected.

Close modal

It is increasingly clear that a number of identified and theoretical situations provide a biologic milieu out of which cells may obtain a clonal advantage through the acquisition or expansion of aUPD in hematologic cells. Since the discovery of UPD9p and the corresponding duplication of JAK2 V617F mutations, a number of recurrent areas of UPD associated with homozygous gene mutations have been found,47,51  suggesting that this is a common mechanism leading to homozygosity (Table 1). UPD maps can be constructed for specific malignancies, similar to a map presented in Figure 4 for MDS and AML. This is a new paradigm that brought about the realization that, because many mutations have a pathologic effect only in a homozygous form, areas of CN-LOH point toward the presence of such mutations. It is probable that duplication of a mutated allele is beneficial in the selection process. Thus, the presence of UPD implies at least 2 mutational steps, including acquisition of a mutation followed by homologous recombination as a secondary event resulting in aUPD. Additional steps may be possible as described for patients with concomitant homozygous JAK2 and c-MPL mutations and corresponding areas of CN-LOH.64  However, one could also assume that the initial mutational event is followed by deletion of the normal allele as a secondary step with a subsequent duplication of the retained allele as a tertiary step. Consequently, deletion may reflect an inability to repair genomic losses.

Table 1

Mutated genes in regions of UPD

Mutated geneRegion of UPDDescribedReferences
MPL UPD1p MPN, RARSt 52  
NRAS UPD1p JMML, CMML 49  
TET2 UPD4q sAML, MPN, MDS/MPD, 53, 54  
JAK2 UPD9p PV, ET, IMF, RARSt 55,57  
CDKN2B UPD9p ALL 4  
WT1 UPD11p AML 6  
c-CBL UPD11q CMML, MDS/MPN, JMML 49, 50, 58,60  
FLT3ITD UPD13 AML 6, 49  
TP53 UPD17p MDS, sAML 61  
NF1 UPD17q JMML 62  
CEBPa UPD19q AML 6, 63  
RUNX1 UPD21q AML 6  
Mutated geneRegion of UPDDescribedReferences
MPL UPD1p MPN, RARSt 52  
NRAS UPD1p JMML, CMML 49  
TET2 UPD4q sAML, MPN, MDS/MPD, 53, 54  
JAK2 UPD9p PV, ET, IMF, RARSt 55,57  
CDKN2B UPD9p ALL 4  
WT1 UPD11p AML 6  
c-CBL UPD11q CMML, MDS/MPN, JMML 49, 50, 58,60  
FLT3ITD UPD13 AML 6, 49  
TP53 UPD17p MDS, sAML 61  
NF1 UPD17q JMML 62  
CEBPa UPD19q AML 6, 63  
RUNX1 UPD21q AML 6  

RARSt indicates refractory anemia with ring sideroblasts in transformation; JMML, juvenile myelomonocytic leukemia; sAML, secondary acute myeloid leukemia; PV, polycythemia vera; ET, essential thrombocytopenia; and IMF, idiopathic myelofibrosis.

Principally, we have observed 2 types of recurrent LOH resulting from deletions: those associated with frequent CN-LOH of the corresponding region, for example, UPD7q and del7q, UPD17p and del17, and those that are never or rarely associated with corresponding UPD (Figure 4). In a cohort of more than 600 patients, we have never encountered acquired UPD5q or UPD20q, whereas deletions of the long arm of these chromosomes are frequent. One could stipulate that, should a region deleted from one homolog contain a hemizygous mutation on the other homolog, selection pressure may favor duplication of the mutated copy; consequently, in addition to deletions, corresponding regions of UPD would also be found. However, UPD and deletions affecting the same chromosomal region may also reflect distinct pathogenetic mechanisms. For example, whereas homozygous c-CBL mutations are frequent, hemizygous mutations have not been observed,49  thus implying that deletion and UPD in this region correspond to the presence of different mutations. In contrast, in TET2 mutant cells, homozygous, hemizygous, and heterozygous as well as biallelic mutations were found,53  suggesting that various pathways can be exploited during malignant evolution to inactivate the mutated allele.

As indicated by the identification of homozygous mutations, acquired CN-LOH can convey a selection advantage through total knockout of a tumor suppressor gene by duplication of inactivating mutation (eg, TP53 mutations and UPD17p) or an activating mutation (eg, JAK2 V617F) (Figure 5; Table 1). In addition to mutations, deletions can lead to either a loss or retention of the methylated (silenced) allele, whereas CN-LOH can result in duplication of either the methylated or the unmethylated allele. As a consequence, LOH can lead to either effective knockout or enhanced expression. Clearly, such a mechanism could be operative for genes that can be regulated by methylation-based promoter silencing. The third mechanism by which CN-LOH can become permissive in the context of the selection process is duplication of a minor disease-prone allele present in the germline constellation. Informative loci would have to have a very low homozygous frequency to allow for analysis (Figure 5). A recent remarkable case of aUPD leading to homozygous selection of HLA surface markers that allowed an AML leukemia to escape immune surveillance65  illustrates yet another powerful example of clonal selection and the biologic consequences of CN-LOH as a disease resistance/disease progression mechanism.

Figure 5

Pathogenic actions of LOH, both CN-LOH and deletion. CN-LOH can lead to the duplication of a disease-linked minor germline variant (top line, left) or an acquired mutation (top line, right). It can also lead to increased gene expression by the loss of a negative epigenetic mark (second line, left) or decreased gene expression by the duplication of a repressive epigenetic mark (second line, right). Deletion of chromosomal material can lead to the unveiling of a minor germline variant (third line, left) of acquired mutation (third line, right), resulting in hemizygosity. Furthermore, deletion can affect gene expression: it can lead to increased expression through loss of an imprinted allele, repression by loss of the expressed allele, or haploinsufficiency.

Figure 5

Pathogenic actions of LOH, both CN-LOH and deletion. CN-LOH can lead to the duplication of a disease-linked minor germline variant (top line, left) or an acquired mutation (top line, right). It can also lead to increased gene expression by the loss of a negative epigenetic mark (second line, left) or decreased gene expression by the duplication of a repressive epigenetic mark (second line, right). Deletion of chromosomal material can lead to the unveiling of a minor germline variant (third line, left) of acquired mutation (third line, right), resulting in hemizygosity. Furthermore, deletion can affect gene expression: it can lead to increased expression through loss of an imprinted allele, repression by loss of the expressed allele, or haploinsufficiency.

Close modal

Traditional cytogenetic technologies, including metaphase cytogenetics, fluorescence in situ hybridization, and comparative genomic hybridization, do not detect CN-LOH, and it was not until the description of UPD9p in the setting of MPN55  that the clinical significance of this type of the lesion was begun to be appreciated. With the broader application of SNP-A came the realization as to how widely spread this balanced acquired lesion is in various malignancies, including AML, MDS, MPN, and the overlap syndromes but also ALL, CLL, and multiple myeloma5,31,46-48,55,66-68  (Table 2). Representative examples of SNP-A karyograms of acquired areas of UPD found particularly frequently in myeloid malignancies from our studies are illustrated in Figure 4.

Table 2

UPD in hematologic disorders

DiseaseReferencesArrayFrequency, %Notes
AML 47  250K 81 Secondary, includes MPN 
 46  250K 23 Secondary 
 69  10K 17 Primary, UPD13q limited to intermediate-risk group, all UPD lost on remission 
 6  10K 12 Primary, lost on remission 
 41  10K 22.7 All normal karyotype 
 70  10K 11; 48 At diagnosis; at relapse 
 49  250K 34 Secondary 
ALL 5  10K 40; 80 At diagnosis; at relapse 
 3  500K 32.5  
 2  100K; 500K 21  
 71  50K; 250K 24  
 72  250K 21; 36 At diagnosis; at relapse 
MM 68  50K 10  
CLL 73  50K Untreated 
 67  10K; 50K 20  
MDS 54  250K All telomeric, > 21 Mb 
 66  50K 33 Low-risk MDS 
 46  250K 20  
 31  10K; 50K; 250K 46 Low-risk MDS 
 74  250K 12 Normal karyotype by MC 
 75  500K 62 CD34+, all small (< 5 Mb) 
 49  250K 18  
 76  50K 33  
MDS/MPN 46  250K 35 Most prevalent in CMML 
 47  250K 36; 11 MDS/MPNu; RARSt 
 49  250K 44 MDS/MPNu, CMML, sAML 
 54  6.0 19 JMML, UPD11q 
 57  250K JMML, UPD11q 
 61  100K 80; 0 JMML with NF1 mutation; without NF1 mutation 
 52  250K 28 RARSt 
DiseaseReferencesArrayFrequency, %Notes
AML 47  250K 81 Secondary, includes MPN 
 46  250K 23 Secondary 
 69  10K 17 Primary, UPD13q limited to intermediate-risk group, all UPD lost on remission 
 6  10K 12 Primary, lost on remission 
 41  10K 22.7 All normal karyotype 
 70  10K 11; 48 At diagnosis; at relapse 
 49  250K 34 Secondary 
ALL 5  10K 40; 80 At diagnosis; at relapse 
 3  500K 32.5  
 2  100K; 500K 21  
 71  50K; 250K 24  
 72  250K 21; 36 At diagnosis; at relapse 
MM 68  50K 10  
CLL 73  50K Untreated 
 67  10K; 50K 20  
MDS 54  250K All telomeric, > 21 Mb 
 66  50K 33 Low-risk MDS 
 46  250K 20  
 31  10K; 50K; 250K 46 Low-risk MDS 
 74  250K 12 Normal karyotype by MC 
 75  500K 62 CD34+, all small (< 5 Mb) 
 49  250K 18  
 76  50K 33  
MDS/MPN 46  250K 35 Most prevalent in CMML 
 47  250K 36; 11 MDS/MPNu; RARSt 
 49  250K 44 MDS/MPNu, CMML, sAML 
 54  6.0 19 JMML, UPD11q 
 57  250K JMML, UPD11q 
 61  100K 80; 0 JMML with NF1 mutation; without NF1 mutation 
 52  250K 28 RARSt 

MM indicates multiple myeloma; MDS/MPNu, MDS/MPN unclassifiable; RARSt, refractory anemia with ring sideroblasts in transformation; sAML, secondary AML; and JMML, juvenile myelomonocytic leukemia.

Studies in MDS and AML show that the addition of SNP-A karyotyping to standard metaphase cytogenetics increases the ability to identify a clonal marker from 50% to approximately 80%.46,48,66  This is particularly helpful in cases with normal cytogenetics, which is still associated with a wide range of clinical outcomes in individual patients, such as patients with intermediate-risk AML and a normal karyotype, some mixed MDS/MPN such as chronic myelomonocytic leukemia (CMML),49  and JAK2-negative MPN.58,72  The technical aspects of the SNP-A technology, including not requiring cell culture, provides a significant increase in the number of evaluable samples, particularly in hematologic malignancies associated with bone marrow fibrosis, through the use of peripheral blood cells in selected situations.72 

Identification of recurrent CN-LOH raises the question of their clinical impact. Similar to other cytogenetic lesions, areas of aUPD can be mapped as, for example, in myeloid malignancies (Figure 4). Recurrent deletions and gains have an established prognostic and diagnostic value and have been incorporated in many prognostic schemes, including those for multiple myeloma and CLL or the International Prognostic Scoring System in MDS. However, the clinical impact of only a few recurrent areas of CN-LOH has been established. For example, homozygous JAK2 mutations indicate the presence of UPD9p and per inference this “doubled” dose of mutant JAK2 has been shown to be associated with more serious prognosis.56  Similarly, UPD13q associated with homozygous FLT-3 ITD or UPD17p and homozygous TP53 mutation, as well as UPD11q and C-CBL mutations, have been attributed with unfavorable outcomes probably because of the presence of the corresponding mutation.49  However, CN-LOH can by itself convey poor prognosis; we have shown that UPD7q in MDS is associated with equally poor prognosis as is corresponding del7q.48  Assignment of prognostic significance will require systematic application of SNP-A as a routine diagnostic tool complementing metaphase cytogenetics. In particular, in patients with normal cytogenetics or lesions associated with favorable prognosis, detection of UPD may effectively upstage the prognosis and allow for better prognostic resolution explaining heterogeneity of outcomes in patients with otherwise comparable clinical features.

  1. CN-LOH acquired during malignant progression must be distinguished from germline areas of CN-LOH, which may be a result of an early embryonic mitotic homologous recombination event or inherited autozygosity. The latter findings can represent important cancer predisposition events. Acquired CN-LOH, resulting from mitotic homologous recombination, has certain genetic characteristics. Factors that might promote clonal selection with CN-LOH include duplication of acquired epigenetic or mutational events, disease predisposing polymorphisms, or improper imprinting patterns.

  2. First identified in JAK2, c-CBL and TET2 are the most recent examples of mutations found in a homozygous constellation in myeloid malignancies resulting from acquired CN-LOH.

  3. CN-LOH is common in myeloid disorders, even in those patients who are often cytogenetically normal. CN-LOH identified by SNP-A is especially common in mixed MDS/MPN, acute CML, CMML, JAK2-negative MPN, MDS, and AML, all of which have increased incidence with age. For these disorders, CN-LOH provides a novel and often unique molecular marker. As such, this marker has the potential to be diagnostic and prognostic, supported by emerging data from MDS, AML, and CMML, and specific CN-LOH regions, such as 7q, may complement existing methodologies and approaches.

  4. Whether CN-LOH explains the worst prognosis of persons with normal metaphase cytogenetics but abnormal SNP-A karyotype, including aging persons, either as a molecular marker of genomic damage or directly related based on clonal selection of acquired molecular lesions, remains to be formally investigated.

  5. Investigations into the underlying mechanisms generating CN-LOH have great promise for elucidating general cancer mechanisms. They can lead to the discovery of new tumor suppressor genes.

  6. We anticipate that further detailed characterization of CN-LOH lesions will probably facilitate our discovery of a more complete set of pathogenic molecular lesions, discovery of disease and prognosis markers, and better understanding of the initiation and progression of hematologic malignancies.

This work was supported by the National Institutes of Health (R01 HL082983 and K24 HL077522, to J.P.M.; DOD MPO48018, to M.A.M.) and the Robert Duggan Cancer Research Foundation.

National Institutes of Health

Contribution: C.O., M.A.M., and J.P.M. wrote and edited the manuscript.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Jaroslaw P. Maciejewski, Taussig Cancer Institute/R40, 9500 Euclid Ave, Cleveland, OH 44195; e-mail: maciejj@ccf.org.

1
Maciejewski
 
JP
Mufti
 
GJ
Whole genome scanning as a cytogenetic tool in hematologic malignancies.
Blood
2008
, vol. 
112
 
4
(pg. 
965
-
974
)
2
Mullighan
 
CG
Goorha
 
S
Radtke
 
I
, et al. 
Genome-wide analysis of genetic alterations in acute lymphoblastic leukaemia.
Nature
2007
, vol. 
446
 
7137
(pg. 
758
-
764
)
3
Kuiper
 
RP
Schoenmakers
 
EF
van Reijmersdal
 
SV
, et al. 
High-resolution genomic profiling of childhood ALL reveals novel recurrent genetic lesions affecting pathways involved in lymphocyte differentiation and cell cycle progression.
Leukemia
2007
, vol. 
21
 
6
(pg. 
1258
-
1266
)
4
Sulong
 
S
Moorman
 
AV
Irving
 
JA
, et al. 
A comprehensive analysis of the CDKN2A gene in childhood acute lymphoblastic leukemia reveals genomic deletion, copy number neutral loss of heterozygosity, and association with specific cytogenetic subgroups.
Blood
2009
, vol. 
113
 
1
(pg. 
100
-
107
)
5
Irving
 
JA
Bloodworth
 
L
Bown
 
NP
Case
 
MC
Hogarth
 
LA
Hall
 
AG
Loss of heterozygosity in childhood acute lymphoblastic leukemia detected by genome-wide microarray single nucleotide polymorphism analysis.
Cancer Res
2005
, vol. 
65
 
8
(pg. 
3053
-
3058
)
6
Raghavan
 
M
Lillington
 
DM
Skoulakis
 
S
, et al. 
Genome-wide single nucleotide polymorphism analysis reveals frequent partial uniparental disomy due to somatic recombination in acute myeloid leukemias.
Cancer Res
2005
, vol. 
65
 
2
(pg. 
375
-
378
)
7
LaFramboise
 
T
Single nucleotide polymorphism arrays: a decade of biological, computational and technological advances.
Nucleic Acids Res
2009
, vol. 
37
 
13
(pg. 
4181
-
4193
)
8
Maciejewski
 
JP
Tiu
 
RV
O'Keefe
 
C
Application of array-based whole genome scanning technologies as a cytogenetic tool in haematological malignancies.
Br J Haematol
2009
, vol. 
146
 
5
(pg. 
479
-
488
)
9
Bacolod
 
MD
Schemmann
 
GS
Giardina
 
SF
Paty
 
P
Notterman
 
DA
Barany
 
F
Emerging paradigms in cancer genetics: some important findings from high-density single nucleotide polymorphism array studies.
Cancer Res
2009
, vol. 
69
 
3
(pg. 
723
-
727
)
10
Engel
 
E
A new genetic concept: uniparental disomy and its potential effect, isodisomy.
Am J Med Genet
1980
, vol. 
6
 
2
(pg. 
137
-
143
)
11
Broman
 
KW
Weber
 
JL
Long homozygous chromosomal segments in reference families from the centre d'Etude du polymorphisme humain.
Am J Hum Genet
1999
, vol. 
65
 
6
(pg. 
1493
-
1500
)
12
Simon-Sanchez
 
J
Scholz
 
S
Fung
 
HC
, et al. 
Genome-wide SNP assay reveals structural genomic variation, extended homozygosity and cell-line induced alterations in normal individuals.
Hum Mol Genet
2007
, vol. 
16
 
1
(pg. 
1
-
14
)
13
McQuillan
 
R
Leutenegger
 
AL
Abdel-Rahman
 
R
, et al. 
Runs of homozygosity in European populations.
Am J Hum Genet
2008
, vol. 
83
 
3
(pg. 
359
-
372
)
14
Kotzot
 
D
Complex and segmental uniparental disomy (UPD): review and lessons from rare chromosomal complements.
J Med Genet
2001
, vol. 
38
 
8
(pg. 
497
-
507
)
15
Robinson
 
WP
Mechanisms leading to uniparental disomy and their clinical consequences.
Bioessays
2000
, vol. 
22
 
5
(pg. 
452
-
459
)
16
Kotzot
 
D
Abnormal phenotypes in uniparental disomy (UPD): fundamental aspects and a critical review with bibliography of UPD other than 15.
Am J Med Genet
1999
, vol. 
82
 
3
(pg. 
265
-
274
)
17
Horsthemke
 
B
Wagstaff
 
J
Mechanisms of imprinting of the Prader-Willi/Angelman region.
Am J Med Genet A
2008
, vol. 
146A
 
16
(pg. 
2041
-
2052
)
18
Engel
 
E
Antonarakis
 
SE
Genomic Imprinting and Uniparental Disomy in Medicine, Clinical and Molecular Aspects
2002
New York, NY
Wiley-Liss
19
Bacolod
 
MD
Schemmann
 
GS
Wang
 
S
, et al. 
The signatures of autozygosity among patients with colorectal cancer.
Cancer Res
2008
, vol. 
68
 
8
(pg. 
2610
-
2621
)
20
Nalls
 
MA
Simon-Sanchez
 
J
Gibbs
 
JR
, et al. 
Measures of autozygosity in decline: globalization, urbanization, and its implications for medical genetics.
PLoS Genet
2009
, vol. 
5
 
3
pg. 
e1000415
 
21
Shami
 
SA
Qaisar
 
R
Bittles
 
AH
Consanguinity and adult morbidity in Pakistan.
Lancet
1991
, vol. 
338
 
8772
pg. 
954
 
22
Simpson
 
JL
Martin
 
AO
Elias
 
S
Sarto
 
GE
Dunn
 
JK
Cancers of the breast and female genital system: search for recessive genetic factors through analysis of human isolate.
Am J Obstet Gynecol
1981
, vol. 
141
 
6
(pg. 
629
-
636
)
23
Lebel
 
RR
Gallagher
 
WB
Wisconsin consanguinity studies: II. familial adenocarcinomatosis.
Am J Med Genet
1989
, vol. 
33
 
1
(pg. 
1
-
6
)
24
Rudan
 
I
Inbreeding and cancer incidence in human isolates.
Hum Biol
1999
, vol. 
71
 
2
(pg. 
173
-
187
)
25
Olshen
 
AB
Gold
 
B
Lohmueller
 
KE
, et al. 
Analysis of genetic variation in Ashkenazi Jews by high density SNP genotyping.
BMC Genet
2008
, vol. 
9
 pg. 
14
 
26
Jankowska
 
AM
Gondek
 
LP
Szpurka
 
H
Nearman
 
ZP
Tiu
 
RV
Maciejewski
 
JP
Base excision repair dysfunction in a subgroup of patients with myelodysplastic syndrome.
Leukemia
2008
, vol. 
22
 
3
(pg. 
551
-
558
)
27
Young
 
BD
Debernardi
 
S
Lillington
 
DM
, et al. 
A role for mitotic recombination in leukemogenesis.
Adv Enzyme Regul
2006
, vol. 
46
 (pg. 
90
-
97
)
28
Murthy
 
SK
DiFrancesco
 
LM
Ogilvie
 
RT
Demetrick
 
DJ
Loss of heterozygosity associated with uniparental disomy in breast carcinoma.
Mod Pathol
2002
, vol. 
15
 
12
(pg. 
1241
-
1250
)
29
Teh
 
M
Blaydon
 
D
Chaplin
 
T
, et al. 
Genomewide single nucleotide polymorphism microarray mapping in basal cell carcinomas unveils uniparental disomy as a key somatic event.
Cancer Res
2005
, vol. 
65
 
19
(pg. 
8597
-
8603
)
30
Stephens
 
K
Weaver
 
M
Leppig
 
KA
, et al. 
Interstitial uniparental isodisomy at clustered breakpoint intervals is a frequent mechanism of NF1 inactivation in myeloid malignancies.
Blood
2006
, vol. 
108
 
5
(pg. 
1684
-
1689
)
31
Mohamedali
 
A
Gaken
 
J
Twine
 
NA
, et al. 
Prevalence and prognostic significance of allelic imbalance by single nucleotide polymorphism analysis in low risk myelodysplastic syndromes.
Blood
2007
, vol. 
110
 
9
(pg. 
3365
-
3373
)
32
Gumus
 
G
Sunguroglu
 
A
Tukun
 
A
Sayin
 
DB
Bokesoy
 
I
Common fragile sites associated with the breakpoints of chromosomal aberrations in hematologic neoplasms.
Cancer Genet Cytogenet
2002
, vol. 
133
 
2
(pg. 
168
-
171
)
33
Calin
 
GA
Sevignani
 
C
Dumitru
 
CD
, et al. 
Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers.
Proc Natl Acad Sci U S A
2004
, vol. 
101
 
9
(pg. 
2999
-
3004
)
34
Bea
 
S
Salaverria
 
I
Armengol
 
L
, et al. 
Uniparental disomies, homozygous deletions, amplifications, and target genes in mantle cell lymphoma revealed by integrative high-resolution whole-genome profiling.
Blood
2009
, vol. 
113
 
13
(pg. 
3059
-
3069
)
35
Stankiewicz
 
P
Shaw
 
CJ
Dapper
 
JD
, et al. 
Genome architecture catalyzes nonrecurrent chromosomal rearrangements.
Am J Hum Genet
2003
, vol. 
72
 
5
(pg. 
1101
-
1116
)
36
Goidts
 
V
Cooper
 
DN
Armengol
 
L
, et al. 
Complex patterns of copy number variation at sites of segmental duplications: an important category of structural variation in the human genome.
Hum Genet
2006
, vol. 
120
 
2
(pg. 
270
-
284
)
37
Bailey
 
JA
Eichler
 
EE
Primate segmental duplications: crucibles of evolution, diversity and disease.
Nat Rev Genet
2006
, vol. 
7
 
7
(pg. 
552
-
564
)
38
Howarth
 
K
Ranta
 
S
Winter
 
E
, et al. 
A mitotic recombination map proximal to the APC locus on chromosome 5q and assessment of influences on colorectal cancer risk.
BMC Med Genet
2009
, vol. 
10
 pg. 
54
 
39
Middeldorp
 
A
van Puijenbroek
 
M
Nielsen
 
M
, et al. 
High frequency of copy-neutral LOH in MUTYH-associated polyposis carcinomas.
J Pathol
2008
, vol. 
216
 
1
(pg. 
25
-
31
)
40
Walsh
 
CS
Ogawa
 
S
Scoles
 
DR
, et al. 
Genome-wide loss of heterozygosity and uniparental disomy in BRCA1/2-associated ovarian carcinomas.
Clin Cancer Res
2008
, vol. 
14
 
23
(pg. 
7645
-
7651
)
41
Serrano
 
E
Carnicer
 
MJ
Orantes
 
V
, et al. 
Uniparental disomy may be associated with microsatellite instability in acute myeloid leukemia (AML) with a normal karyotype.
Leuk Lymphoma
2008
, vol. 
49
 
6
(pg. 
1178
-
1183
)
42
Plo
 
I
Nakatake
 
M
Malivert
 
L
, et al. 
JAK2 stimulates homologous recombination and genetic instability: potential implication in the heterogeneity of myeloproliferative disorders.
Blood
2008
, vol. 
112
 
4
(pg. 
1402
-
1412
)
43
Mutirangura
 
A
Greenberg
 
F
Butler
 
MG
, et al. 
Multiplex PCR of three dinucleotide repeats in the Prader-Willi/Angelman critical region (15q11-q13): molecular diagnosis and mechanism of uniparental disomy.
Hum Mol Genet
1993
, vol. 
2
 
2
(pg. 
143
-
151
)
44
Gupta
 
M
Young
 
BD
Application of SNP genotype arrays to determine somatic changes in cancer.
Methods Mol Biol
2009
, vol. 
538
 (pg. 
179
-
206
)
45
Li
 
X
Self
 
SG
Galipeau
 
PC
Paulson
 
TG
Reid
 
BJ
Direct inference of SNP heterozygosity rates and resolution of LOH detection.
PLoS Comput Biol
2007
, vol. 
3
 
11
pg. 
e244
 
46
Gondek
 
LP
Tiu
 
R
O'Keefe
 
CL
Sekeres
 
MA
Theil
 
KS
Maciejewski
 
JP
Chromosomal lesions and uniparental disomy detected by SNP arrays in MDS, MDS/MPD, and MDS-derived AML.
Blood
2008
, vol. 
111
 
3
(pg. 
1534
-
1542
)
47
Gondek
 
LP
Dunbar
 
AJ
Szpurka
 
H
McDevitt
 
MA
Maciejewski
 
JP
SNP array karyotyping allows for the detection of uniparental disomy and cryptic chromosomal abnormalities in MDS/MPD-U and MPD.
PLoS ONE
2007
, vol. 
2
 
11
pg. 
e1225
 
48
Tiu
 
RV
Gondek
 
LP
O'Keefe
 
CL
, et al. 
New lesions detected by SNP array-based chromosomal analysis have important clinical impact in AML.
J Clin Oncol
2009
, vol. 
27
 
31
(pg. 
5219
-
5226
)
49
Dunbar
 
AJ
Gondek
 
LP
O'Keefe
 
CL
, et al. 
250K SNP array karyotyping identifies acquired uniparental disomy and homozygous mutation, including novel missense substitutions of c-Cbl, in myeloid malignancies.
Cancer Res
2008
, vol. 
15
 (pg. 
10349
-
10357
)
50
Makishima
 
H
Cazzolli
 
H
Dunbar
 
A
, et al. 
Mutations of e3 ubiquitin ligase cbl family members constitute a novel common pathogenic lesion in myeloid malignancies.
J Clin Oncol
2009
, vol. 
27
 
36
(pg. 
6109
-
6116
)
51
Yamamoto
 
G
Nannya
 
Y
Kato
 
M
, et al. 
Highly sensitive method for genomewide detection of allelic composition in nonpaired, primary tumor specimens by use of affymetrix single-nucleotide-polymorphism genotyping microarrays.
Am J Hum Genet
2007
, vol. 
81
 
1
(pg. 
114
-
126
)
52
Szpurka
 
H
Gondek
 
LP
Mohan
 
SR
, et al. 
UPD1p indicates the presence of MPL W515L mutation in RARS-T, a mechanism analogous to UPD9p and JAK2 V627F mutation.
Leukemia
2008
, vol. 
23
 
3
(pg. 
610
-
6144
)
53
Jankowska
 
AM
Szpurka
 
H
Tiu
 
RV
, et al. 
Loss of heterozygosity 4q42 and TET2 mutations associated with myelodysplatic/myeloproliferative neoplasms.
Blood
2009
, vol. 
113
 (pg. 
6403
-
6410
)
54
Langemeijer
 
SM
Kuiper
 
RP
Berends
 
M
, et al. 
Acquired mutations in TET2 are common in myelodysplastic syndromes.
Nat Genet
2009
, vol. 
41
 
7
(pg. 
838
-
842
)
55
Kralovics
 
R
Guan
 
Y
Prchal
 
JT
Acquired uniparental disomy of chromosome 9p is a frequent stem cell defect in polycythemia vera.
Exp Hematol
2002
, vol. 
30
 
3
(pg. 
229
-
236
)
56
Kralovics
 
R
Passamonti
 
F
Buser
 
AS
, et al. 
A gain-of-function mutation of JAK2 in myeloproliferative disorders.
N Engl J Med
2005
, vol. 
352
 
17
(pg. 
1779
-
1790
)
57
Szpurka
 
H
Tiu
 
R
Murugesan
 
G
, et al. 
Refractory anemia with ringed sideroblasts associated with marked thrombocytosis (RARS-T), another myeloproliferative condition characterized by JAK2 V617F mutation.
Blood
2006
, vol. 
108
 
7
(pg. 
2173
-
2181
)
58
Grand
 
FH
Hidalgo-Curtis
 
CE
Ernst
 
T
, et al. 
Frequent CBL mutations associated with 11q acquired uniparental disomy in myeloproliferative neoplasms.
Blood
2009
, vol. 
113
 
24
(pg. 
6182
-
6192
)
59
Loh
 
ML
Sakai
 
DS
Flotho
 
C
, et al. 
Mutations in CBL occur frequently in juvenile myelomonocytic leukemia.
Blood
2009
, vol. 
114
 
9
(pg. 
1859
-
1863
)
60
Muramatsu
 
H
Makishima
 
H
Jankowska
 
A
, et al. 
Mutations of E3 ubiquitin ligase Cbl family members but not TET2 mutations are pathogenic in juvenile myelomonocytic leukemia.
Blood
2010
, vol. 
115
 
10
(pg. 
1969
-
1975
)
61
Jasek
 
M
Gondek
 
L
Bejanyan
 
N
, et al. 
TP53 mutations in myeloid malignancies are either homozygous or hemizygous due to copy number-neutral loss of heterozygosity or deletion of 17p.
Leukemia
2010
, vol. 
24
 
1
(pg. 
216
-
219
)
62
Flotho
 
C
Steinemann
 
D
Mullighan
 
CG
, et al. 
Genome-wide single-nucleotide polymorphism analysis in juvenile myelomonocytic leukemia identifies uniparental disomy surrounding the NF1 locus in cases associated with neurofibromatosis but not in cases with mutant RAS or PTPN11.
Oncogene
2007
, vol. 
26
 
39
(pg. 
5816
-
5821
)
63
Fitzgibbon
 
J
Smith
 
LL
Raghavan
 
M
, et al. 
Association between acquired uniparental disomy and homozygous gene mutation in acute myeloid leukemias.
Cancer Res
2005
, vol. 
65
 
20
(pg. 
9152
-
9154
)
64
Kralovics
 
R
Stockton
 
DW
Prchal
 
JT
Clonal hematopoiesis in familial polycythemia vera suggests the involvement of multiple mutational events in the early pathogenesis of the disease.
Blood
2003
, vol. 
102
 
10
(pg. 
3793
-
3796
)
65
Vago
 
L
Perna
 
SK
Zanussi
 
M
, et al. 
Loss of mismatched HLA in leukemia after stem-cell transplantation.
N Engl J Med
2009
, vol. 
361
 
5
(pg. 
478
-
488
)
66
Gondek
 
LP
Haddad
 
AS
O'Keefe
 
CL
, et al. 
Detection of cryptic chromosomal lesions including acquired segmental uniparental disomy in advanced and low-risk myelodysplastic syndromes.
Exp Hematol
2007
, vol. 
35
 
11
(pg. 
1728
-
1738
)
67
Pfeifer
 
D
Pantic
 
M
Skatulla
 
I
, et al. 
Genome-wide analysis of DNA copy number changes and LOH in CLL using high-density SNP arrays.
Blood
2007
, vol. 
109
 
3
(pg. 
1202
-
1210
)
68
Walker
 
BA
Leone
 
PE
Jenner
 
MW
, et al. 
Integration of global SNP-based mapping and expression arrays reveals key regions, mechanisms, and genes important in the pathogenesis of multiple myeloma.
Blood
2006
, vol. 
108
 
5
(pg. 
1733
-
1743
)
69
Gupta
 
M
Raghavan
 
M
Gale
 
RE
, et al. 
Novel regions of acquired uniparental disomy discovered in acute myeloid leukemia.
Genes Chromosomes Cancer
2008
, vol. 
47
 
9
(pg. 
729
-
739
)
70
Raghavan
 
M
Smith
 
LL
Lillington
 
DM
, et al. 
Segmental uniparental disomy is a commonly acquired genetic even in relapsed acute myeloid leukemia.
Blood
2008
, vol. 
112
 
3
(pg. 
814
-
821
)
71
Kawamata
 
N
Ogawa
 
S
Zimmerman
 
M
, et al. 
Molecular allelokaryotyping of pediatric acute lymphoblastic leukemias by high-resolution single nucleotide polymorphism oligonucleotide genomic microarray.
Blood
2008
, vol. 
111
 
6
(pg. 
776
-
784
)
72
Kawamata
 
N
Ogawa
 
S
Yamamoto
 
G
, et al. 
Genetic profiling of myeloproliferative disorders by single-nucleotide polymorphism oligonucleotide microarray.
Exp Hematol
2008
, vol. 
36
 
11
(pg. 
1471
-
1479
)
73
Lehmann
 
S
Ogawa
 
S
Raynaud
 
SD
, et al. 
Molecular allelokaryotyping of early-stage, untreated chronic lymphocytic leukemia.
Cancer
2008
, vol. 
112
 
6
(pg. 
1296
-
1305
)
74
Heinrichs
 
S
Kulkarni
 
RV
Bueso-Ramos
 
CE
, et al. 
Accurate detection of uniparental disomy and microdeletions by SNP array analysis in myelodysplastic syndromes with normal cytogenetics.
Leukemia
2009
, vol. 
23
 
9
(pg. 
1605
-
1613
)
75
Nowak
 
D
Nolte
 
F
Mossner
 
M
, et al. 
Genome-wide DNA mapping of CD34+ cells from patients with myelodysplastic syndrome using 500K SNP arrays identifies significant regions of deletion and uniparental disomy.
Exp Hematol
2009
, vol. 
37
 
2
(pg. 
215
-
224
)
76
Gondek
 
LP
Tiu
 
R
Haddad
 
AS
, et al. 
Single nucleotide polymorphism arrays complement metaphase cytogenetics in detection of new chromosomal lesions in MDS.
Leukemia
2007
, vol. 
21
 
9
(pg. 
2058
-
2061
)
Sign in via your Institution