Compositions and methods for the detection of DNA topoisomerase II complexes with DNA

Compositions, methods, and kits for detecting DNA topoisomerase II-DNA complexes are disclosed.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description

This application claims priority under 35 U.S.C. § 119(e) to U.S. Provisional Patent Application No. 60/608,331, filed on Sep. 9, 2004. The foregoing application is incorporated by reference herein.

Pursuant to 35 U.S.C. §202(c) it is acknowledged that the U.S. Government has certain rights in the invention described herein, which was made in part with funds from the National Institutes of Health, Grant Numbers RO1 CA77683, CA85469, and CA80175.

FIELD OF THE INVENTION

The present invention relates to the fields of molecular biology and oncology. More specifically, the invention provides compositions and methods for detection of DNA topoisomerase II complexes with genomic DNA.

BACKGROUND OF THE INVENTION

Several publications and patent documents are cited throughout the specification in order to describe the state of the art to which this invention pertains. Each of these citations is incorporated herein by reference as though set forth in full.

Epipodophyllotoxins and anthracyclines, which are commonly used chemotherapeutic DNA topoisomerase II inhibitors, more accurately are called DNA topoisomerase II poisons because they increase the concentration of DNA topoisomerase II cleavage complexes and have the overall effect of enhancing cleavage, which is cytotoxic (1). These agents are associated with leukemia as a treatment complication (2). Most DNA topoisomerase II inhibitor-related leukemias have MLL (myeloid lymphoid leukemia) translocations (3). The translocations disrupt an 8.3 kb bcr between exons 5-11 of the ˜100 kb MLL gene at chromosome band 11q23. The association of DNA topoisomerase II inhibitors with leukemia has suggested a translocation mechanism that involves chromosomal breakage from drug-stabilized DNA topoisomerase II cleavage and formation of the breakpoint junctions when the breakage is repaired (2). The drug stabilized complexes have been called ternary (drug-DNA-topoisomerase II) complexes in the literature. In previous reports, MLL translocations have been characterized and tracked in leukemias in two patients receiving chemotherapy (Megonigal 2000; Blanco 2001), and the role of chemotherapy-stabilized DNA topoisomerase II cleavage in the translocation process has been investigated in in vitro assays of DNA substrates outside of the cellular context (Lovett, 2001; Lovett 2001; Whitmarsh 2003). However, cell death from chemotherapy also forces bone marrow progenitor cell proliferation (4) and native DNA topoisomerase IIα expression is highest in proliferating cells (5, 6).

Genomic breakpoint junctions on derivative chromosomes arising from MLL translocations were cloned in two cases of leukemia following intensive neuroblastoma regimens (7, 8). Such chemotherapy regimens have been associated with a high incidence of leukemia as a treatment complication (9).

SUMMARY OF THE INVENTION

It is an objective of the present invention to provide compositions and methods for detecting ternary topoisomerase II/DNA complexes with cytotoxic chemotherapy drugs (e.g., DNA topoisomerase II poisons) or with natural compounds that have similar activity, as well as topoisomerase II complexes with DNA formed by the native enzyme as a means to assess strategies to prevent the oncogenic events associated with chemotherapy and exposure to natural topoisomerase II inhibitors.

In addition, it is known that cytotoxic chemotherapy may cause cell death that is followed by bone marrow progenitor cell proliferation with accompanying increased native DNA topoisomerase II expression. A patient has recently bee identified with leukemia with an MLL translocation which emerged after chemotherapy that did not include a DNA topoisomerase II poison (Langer, 2003). Accordingly, it is another objective of the present invention to provide compositions and methods for detecting topoisomerase II/DNA complexes that are formed by the native enzyme at elevated levels, and as a means to prevent leukemogenic events associated with chemotherapy.

Thus, in accordance with the present invention, compositions, methods, and kits are provided for the detection of topoisomerase II-genomic DNA complexes. The detection of such complexes can be indicative of DNA damage, particularly in relation to treatment-related leukemia. An exemplary method entails providing cells which express topoisomerase II and exposing the cells to an agent suspected of inducing formation of agent-topoisomerase-DNA cleavage complexes for a time period sufficient for such complexes to form. The cells are then lysed and DNA-topoisomerase cleavage complexes are isolated. Alternatively, the cells can be exposed to agents that result in increased expression of topoisomerase II without formation of agent-topoisomerase-DNA cleavage complexes, in which topoisomerase II-DNA cleavage complexes can be measured. Following isolation, the DNA present in the isolated complexes can be amplified by degenerative oligonucleotide PCR and then assayed by real-time PCR with primers specific for the region of interest, such as, for example, the MLL breakpoint cluster region (bcr). Alternatively or additionally, the DNA present in the isolated complexes that has been amplified by degenerative oligonucleotide PCR can be labeled with a detectable label. The optionally labeled DNA is then characterized via hybridization to predetermined sequences present in the region of genomic DNA where the complexes form in order to characterize the sites of complex formation with DNA at the sequence level. In one aspect of the invention, the predetermined sequences are present on a microarray. According to another aspect, the genomic DNA sequences are from the MLL bcr. According to another aspect, the genomic DNA sequences, with or without DNA sequences from the MLL bcr, comprise partner genes which are fused with MLL in certain leukemias. In accordance with another aspect of the invention, the genomic DNA sequences are sequences associated with leukemia. In accordance with yet another aspect of the invention, the genomic DNA sequences comprise the entire human genome.

In accordance with another aspect of the instant invention, a method for identifying sequences present in DNA topoisomerase II-DNA complexes in cells is provided. The method comprises: a) providing cells suspected of containing DNA topoisomerase II-DNA complexes; b) isolating DNA topoisomerase II-DNA complexes from the cell; c) amplifying the DNA present in isolated DNA topoisomerase II-DNA complexes via polymerase chain reaction; and d) identifying the sequences present in said amplified DNA, thereby identifying the sequences present in said DNA topoisomerase II-DNA complexes. In a preferred embodiment, the DNA in the DNA topoisomerase II-DNA complexes is genomic DNA. In a particular embodiment, the identification of the sequences in step d) comprises 1) further amplifying the amplified DNA from step c) with gene specific primers; 2) further amplifying the amplified DNA from step c) by real-time PCR; and/or 3) hybridizing the amplified DNA from step c) with a microarray, such as those described hereinbelow. In another embodiment, the cells are CD34+ cells. Additionally, the amplified DNA of step c) may comprise sequences from the MLL gene. According to another aspect of the invention, the cells are exposed to an agent suspected of modulating formation of topoisomerase cleavage complexes.

In yet another aspect of the invention, microarrays are provided. In a particular embodiment, the microarray comprises at least one of the group consisting of MLL bcr oligonucleotide sequences and oligonucleotide sequences of MLL partner genes. In another embodiment, the MLL bcr oligonucleotide sequences hybridize to non-repetitive MLL bcr sequences. The microarrays may also further comprise oligonucleotide sequences from the Alu region between nucleotide positions 663-1779 in the MLL bcr and/or control sequences which are not involved in MLL translocations. Examples of control sequences include, without limitation, MLL exon 25, MLL exon 3, GAPDH, c-myc, and bacterial gene sequences. MLL partner genes include, without limitation, LAF-4, AF4 (MLLT2, FEL), AF5α, AF5q31, AF6q21 (FKHRL1), AF9 (MLLT3), AF10, MLL, AF17, ENL (MLLT1, LTG19), AFX, CBP, ELL (MEN), p300, AF3p21, LCX (TET1), AF15q14, AF1p (eps15), AF1q, GMPS, LPP, GRAF, AF6, CDK6, FBP17, ABI-1, CBL, MPFYVE, GAS7, LASP1, MSF, EEN, hCDCrel, SEPTIN6, CALM, LARG, GPHN, MYO1F, Alkaline Ceramidase, RPS3, and MIFL. In a particular embodiment of the invention, the microarray comprises oligonucleotide sequences of at least one of SEQ ID NOs 1-162. In another particular embodiment of the invention, the microarray comprises oligonucleotide sequences of at least one of SEQ ID NOs 163-246.

In accordance with yet another aspect of the instant invention, kits are provided for performing the methods of the instant invention, such as for the detection of DNA-DNA topoisomerase II complexes and the sequences of the DNA of these complexes. The kits may comprise any or all of the following: an agent and a buffer for lysing cells; a solid support; a buffer for isolating DNA-DNA topoisomerase II complexes; at least one primer for use in whole genome amplification method; a buffer for PCR amplification of isolated topoisomerase II bound DNA; primers and buffer for real-time PCR analysis of specific genes in the isolated, topoisomerase II-bound DNA that has been amplified by a whole genome amplification method; at least one detectable label for comparative labeling of topoisomerase II bound DNA from two cell populations that has been amplified by a whole genome amplification method or comparative labeling of topoisomerase II bound DNA from one cell population that has been amplified by a whole genome amplification method and a calibrated control DNA; calibrated control DNA for labeling and hybridization to the microarray at the same time as the test sample labeled with another detectable label; a microarray comprising sequences from at least one of the group consisting of the MLL bcr and MLL partner genes and, optionally, control sequences; and instruction material. In a particular aspect of the invention, the kit comprises: a) an agent and a buffer for lysing cells; b) a solid support and a buffer for isolating DNA-DNA topoisomerase II complexes; c) at least one primer and a buffer for PCR amplification of isolated DNA by whole genome amplification; d) at least one first detectable label for incorporation into products of whole genome amplification; e) calibrated reference standard DNA comprising a second detectable label; f) a microarray comprising oligonucleotide sequences from the MLL bcr; and g) instruction material.

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1A is a summary of the clinical course and treatment of patient t-120 and the Southern blot and PCR analysis of sequential bone marrow specimens. The chemotherapy cycles administered according to the Memorial Sloan-Kettering N7 regimen were CAV (cyclophosphamide 4200 mg/m2, doxorubicin 75 mg/M2, vincristine 1.5 mg/m2) or PVP (cisplatin 200 mg/m2, etoposide 600 mg/m2). 3F8* indicates radiolabeled anti-GD2 monoclonal antibody treatment. (+) or (−) indicates detection of MLL translocation by Southern blot analysis or by nested PCR with gene-specific primers. FIG. 1B is a gel of a Southern blot analysis of MLL breakpoint cluster region (bcr) rearrangements in sequential marrow specimens. FIG. 1C is a gel of Clonotypic PCR analysis (Round 1) and nested clonotypic PCR analysis of the der(4) genomic breakpoint junction in sequential marrow specimens.

FIGS. 2A and 2B are images of autoradiographs showing the cleavage products after 10 min incubation at 37° C. of 25 ng (30,000 cpm) singly 5′ end-labeled DNA with 147 nM human DNA topoisomerase IIα, 1 mM ATP and, where indicated, 20 μM etoposide (VP16), etoposide catechol (VP16-OH), etoposide quinone (VP16-Q), or doxorubicin (ADR). FIG. 2A is an analysis of normal homologues of MLL genomic breakpoint sequences in ALL (acute lymphoblastic leukemia) of patient t-120. DNA topoisomerase II cleavage of MLL intron 8 coordinates 6513 to 6778. FIG. 2B is an analysis of normal homologues of AF-4 genomic breakpoint sequences in ALL of patient t-120 in DNA topoisomerase II in vitro cleavage assays. DNA topoisomerase II cleavage of AF-4 intron 3 coordinates 6956 to 7239. The indicated reactions were incubated for an additional 10 min at 75° C. before trapping of covalent complexes. The 5′ side (−1 position) of the cleavage sites are shown. Corkscrew arrows at far right indicate translocation breakpoints. FIGS. 2C and 2D demonstrate the effects of doxorubicin over a range of concentrations on the cleavage of the MLL (FIG. 2C) and AF-4 (FIG. 2D) substrates.

FIGS. 3A and 3B are schematic drawings of models for processing of cleavage sites to form der(11) and der(4) genomic breakpoint junctions in ALL of patient t-120. In FIG. 3A, DNA topoisomerase II cleavage sites at MLL intron 8 position 6588 and AF-4 intron 3 position 7126 with 4-base 5′ overhangs are shown at the top. The cleavage at MLL (fragment shown is SEQ ID NO: 258) intron 8 position 6588 was detected in the presence of etoposide, etoposide metabolites and doxorubicin. The cleavage at AF-4 (fragment shown is SEQ ID NO: 259) intron 3 position 7126 was detected in the presence of etoposide and etoposide metabolites. The processing includes exonucleolytic nibbling (italic) to form single-base homologies and create both breakpoint junctions of the t(4; 11) by error-prone non-homologous end joining (NHEJ) (boxes). In formation of the der(11) (SEQ ID NO: 260 shown) positions 6590 to 6592 on the antisense strand of MLL and positions 7127 to 7129 on the sense strand of AF-4 are lost by exonucleolytic nibbling (italic, middle) before NHEJ (box, middle) joins the indicated bases. Positions 7110 to 7126 on the sense strand of AF-4, positions 7111 to 7130 on the antisense strand of AF-4, positions 6589 to 6594 on the sense strand of MLL and positions 6593 to 6595 on the antisense strand of MLL are lost by exonucleolytic nibbling (italic, bottom) and the der(4) (SEQ ID NO: 261 shown) also forms by NHEJ (box, bottom). Similarly, FIG. 3B demonstrates the formation of der(11) (SEQ ID NO: 260 shown) and der(4) (SEQ ID NO: 261 shown) genomic breakpoint junctions wherein the DNA topoisomerase II cleavage sites at MLL (fragment shown is SEQ ID NO: 258) intron 8 position 6588 and AF-4 (fragment shown is SEQ ID NO: 259) intron 3 position 7114 are employed, the latter of which was detected in the presence of doxorubicin.

FIGS. 4A and 4B are images of autoradiographs of normal homologues of MLL (FIG. 4A) and GAS7 (FIG. 4B) genomic breakpoint sequences in acute myeloid leukemia (AML) of patient t-39 in DNA topoisomerase II in vitro cleavage assays. DNA topoisomerase II cleavage of MLL intron 8 coordinates 4589 to 4768 and GAS7 coordinates 1129 to 1440 upstream of exon 1. FIGS. 4C and 4D demonstrate the effects of doxorubicin over a range of concentrations on the cleavage of the MLL at position 4673 and 4675 (FIG. 4C) and GAS7 at position 1238 (FIG. 4D).

FIG. 5 is a schematic drawing of a model for processing of DNA topoisomerase II cleavage sites that were detected without drug and enhanced greatly with low concentration doxorubicin, (the only DNA topoiosomerase II targeted drug to which the patient was exposed before molecular detection of the translocation) to form der(11) (SEQ ID NO: 264 shown) and der(17) (SEQ ID NO: 265 shown) genomic breakpoint junctions in AML of patient t-39. Both genomic breakpoint junctions are formed by resolution of doxorubicin-stimulated DNA topoisomerase II cleavage sites via error-prone nonhomologous end-joining (NHEJ). DNA topoisomerase II cleavage sites at MLL (fragment shown is SEQ ID NO: 262) intron 8 position 4675 and GAS7 (fragment shown is SEQ ID NO: 263) position 1238 with 4-base 5′ overhangs are shown at the top. In formation of the der(11) positions 4664 to 4675 on the sense strand of MLL, positions 4665 to 4679 on the antisense strand of MLL and positions 1239 to 1240 on the sense strand of GAS7 are lost by exonucleolytic nibbling (italic, middle) before NHEJ (box, middle) and gap fill-in (black, middle) ensue. Positions 1204 to 1238 on the sense strand of GAS7, positions 1207 to 1242 on the antisense strand of GAS7, positions 4676 to 4679 on the sense strand of MLL and positions 4680 to 4682 on the antisense strand of MLL are lost by exonucleolytic nibbling (italic, bottom) and the der(17) forms by NHEJ (box, bottom) and mismatch repair (asterisks, bottom).

FIG. 6 is an image of a Western blot analysis of DNA topoisomerase IIα protein expression in ex vivo-expanded CD34+ cells and cultured hematopoietic cell lines. 3 μg of protein per lane were loaded on the gel. The filter was simultaneously hybridized to mouse anti-human DNA topoisomerase IIα (DAKO, Glostrup, Denmark) and mouse anti-human β-actin (Abcam, Cambridge, Mass.) antibodies.

FIGS. 7A through 7D are images of Western blots demonstrating DNA topoisomerase II-DNA complex formations in the presence of various drugs at various concentrations in a modified ICE bioassay. The assays were performed on CEM cells (FIG. 7A), K562 cells (FIGS. 7B and 7C), and KG-1 cells (FIG. 7D). Etoposide (VP16), etoposide catechol (VP16-OH), etoposide quinone (VP16-Q), and doxorubicin were tested.

FIG. 8 is an image of a gel showing a modified in vivo complex of enzyme (ICE) assay of DNA topoisomerase II covalent complexes in ex-vivo expanded CD34+ cells. The cells were untreated or treated for 2 hours with etoposide at 100 μM final concentration. Total genomic DNA containing protein-bound DNA was isolated on a CsCl cushion and 5.6 μg per well was loaded on a 4-12% Bis-Tris gradient gel. An anti-human DNA topoisomerase IIα antibody (DAKO, Glostrup, Denmark) was used to detect DNA topoisomerase II covalent complexes. Recombinant human DNA topoisomerase IIα is in lane at the far left. The molecular weight of the topoisomerase II band is >170 kD, which is the molecular weight of topoisomerase II. This is consistent with DNA bound to the enzyme.

FIGS. 9A, 9B, 9C, and 9D are plots from the real-time PCR analysis of DNA topoisomerase II covalent complexes with MLL and other sequences. CD34+ cells were untreated or treated for 2 hours with etoposide at a 100 μM final concentration. Total genomic DNA containing protein-bound DNA was isolated on a CsCl cushion, and DNA topoisomerase IIα-bound DNA was purified on an immunoaffinity column and then amplified by degenerative oligonucleotide PCR (DOP). 50 ng of DOP products ranging from 500 bp to 1 kb in size served as a template for amplification in real-time PCR with primers specific for MLL intron 8-exon 9 (FIG. 9A), GAPDH (FIG. 9B), MLL exon 25 (FIG. 9C), or MLL intron7-exon 8 (FIG. 9D). Products were not detectable in duplicate reagent-control reactions for these amplicons. ΔRn (measure of reporter signal) v. cycles are shown in the plots.

FIG. 10 is a schematic drawing of the methods of an in vivo complex of enzyme bioassay allowing for the detection of DNA-DNA topoisomerase II complexes.

FIG. 11 is a schematic drawing of the methods for the isolation of DNA-DNA topoisomerase II complexes.

FIG. 12A is an image of XeoChips™ containing different amounts of products from DOP amplification of MLL bcr plasmid that were labeled with AlexaFluor 546 or AlexaFluor 647 in order to establish conditions for calibrated standard. FIGS. 12B and 12C are Quantile-Quantile plots standardized by intensity comparing etoposide-treated and untreated cells and genistein-treated and untreated cells, respectively, when the test sample (treated or untreated) was labeled with AlexaFluor 546 and the MLL bcr calibrated reference sample was labeled with AlexaFluor 647. The points that deviate from the linear relationship suggests coldspots or hotspots for formation DNA topoisomerase II cleavage complexes in the treated compared to untreated cells.

DETAILED DESCRIPTION OF THE INVENTION

The emergence of the MLL translocation relative to intensive multimodality neuroblastoma therapy administered to two patients with secondary acute leukemia was traced to assess the role of specific agents in the genesis of the translocations. An in vitro assay was employed and a CD34+ cell model developed in order to pinpoint whether the translocations arose from drug stimulated DNA topoisomerase IIα cleavage and formation of drug-DNA-topoisomerase II complexes at or in close proximity to the translocation breakpoints. Variability in the molecular emergence of traceable translocations was found. In one case, the t(4; 11) was first detectable 6 months after neuroblastoma diagnosis, following completion of all chemotherapy and exposure to etoposide and doxorubicin. Processing of functional DNA topoisomerase II cleavage sites enhanced by etoposide or its metabolites or doxorubicin resulted in both breakpoint junctions. In another case, doxorubicin was the only DNA topoisomerase II inhibitor exposure before detection of the MLL-GAS7 translocation at six weeks after starting treatment (7). There were strong DNA topoisomerase II cleavage sites detected without drug and further enhanced by doxorubicin in MLL and GAS7 that resulted in both breakpoint junctions. This is the first functional demonstration of relevance of doxorubicin-DNA-topoisomerase II covalent complexes with MLL and with its partner gene in the genesis of MLL translocations in treatment-related AML.

In the ALL of patient t-120 described above, the translocation breakpoint in the leukemia and the site of in vitro drug-DNA-topoisomerase II complex formation both were at the translocation breakpoint hotspot region 3′ in intron 8 in the MLL bcr (reviewed in Whitmarsh 2003). In the recent treatment-related AML described by Langer (2003) where the leukemia occurred after cytotoxic chemotherapy without DNA topoisomerase II poisons, the translocation breakpoint also was at the same translocation breakpoint hotspot region. The in vitro assays described above as well as in Whitmarsh (2003) showed that not only drug-DNA-topoisomerase II complexes but also DNA-native topoisomerase II complexes were formed in this translocation breakpoint hotspot region.

It was important to devise a cellular model to further study the role of DNA topoisomerase II cleavage in the genesis of MLL translocations because cleavage sites in the cellular context should be more restricted than in vitro (Capranico et al., 1990). In the development of this cellular model it was important to focus on the breakpoint hotspot region 3′ in intron 8 of the MLL bcr. DNA topoisomerase II was highly expressed in CD34+ cells and formed cleavage complexes with the MLL bcr at the 3′ breakpoint hotspot region in untreated CD34+ cells and CD34+ cells treated with etoposide. These findings establish that DNA topoisomerase II forms cleavage complexes with the MLL bcr in bone marrow stem cells, and implicate not only drug-stabilized DNA topoisomerase II cleavage but also native DNA topoisomerase II cleavage as mechanisms to damage MLL.

A modified in vivo complex of enzyme (ICE) bioassay, which entails trapping and immunodetection of DNA covalently bound to DNA topoisomerase II, has been developed to examine native and chemotherapy-stabilized DNA topoisomerase II cleavage complexes in ex vivo stimulated CD34+ progenitor cells from normal human marrow. The details of the ICE assay as applied to cell lines were described in (Whitmarsh 2003). However, the ICE bioassay detects overall formation of DNA topoisomerase II covalent complexes genome-wide but does not address the enzyme-DNA interaction at the sequence level.

In accordance with the present invention, a new approach was created to detect DNA topoisomerase II covalent complexes with MLL bcr in CD34+ cells at the sequence level. Total genomic DNA samples including protein-bound DNA from untreated or etoposide-treated ex vivo-expanded human CD34+ cells were prepared on a CsCl cushions according to the ICE assay protocol. However, the ICE assay then utilizes immunoblotting to detect all DNA in total genomic DNA bound to DNA topoisomerase IIα by the covalent phosphotyrosine linkage formed between this enzyme and DNA (Whitmarsh et al., 2003) without attention to any specific sequences. The instant assay diverges, for example, from the ICE assay in the following manner: DNA topoisomerase IIα covalent complexes in the total genomic DNA isolated using the CsCl cushion are purified on an immuno-affinity column consisting of Protein A-Sepharose beads covalently coupled to a mouse anti-human DNA topoisomerase IIα antibody. Eluted material from the column is then subjected to degenerative oligonucleotide PCR (DOP) in order to generate template DNAs in sufficient quantity for real-time PCR. MLL bcr primers for real-time PCR were designed to amplify positions 6784 to 6851 at the junction of intron 8-exon 9 near the previously described translocation breakpoint hotspot (Whitmarsh et al., 2003). Other primer pairs would amplify a genomic region of GAPDH and a genomic region of MLL exon 25, which are not involved in MLL translocations (Langer et al., 2003). Yet other primers would amplify a region of the MLL bcr at the junction of intron 7-exon 8 near a region where other MLL translocation breakpoints have been identified in cases of leukemia in infants.

The finding of native DNA topoisomerase II complexes and etoposide-DNA-topoisomerase II complexes in total genomic DNA (without regard to any specific sequences in the genome) of CD34+ cells by ICE had never been described. Moreover, the assay disclosed herein that incorporates immuno affinity purification, whole genome amplification and real-time PCR after isolation of total cellular DNA including protein bound DNA, to rapidly detect DNA topoisomerase II covalently bound specifically to the MLL bcr in human CD34+ cells. Using this new assay, native as well as etoposide-stabilized DNA topoisomerase II covalent complexes with the MLL bcr were detectable in the immuno-affinity purified DNA topoisomerase II covalent complexes, demonstrating for the first time that DNA topoisomerase II forms covalent complexes with MLL at sites of translocation breakpoints in human CD34+ cells. Since DNA templates containing double-strand breaks from DNA topoisomerase II cleavage would not be amplified by PCR, these data indicate the presence of DNA-DNA topoisomerase II covalent complexes proximal to the amplicon (MLL bcr positions 6784 to 6851) where products were detected with resolution at the size of the DOP template. The use of chemotherapy creates a risk for leukemia as a treatment complication. Tracing of the temporal molecular emergence of the leukemia-associated MLL translocations relative to chemotherapy administration and analysis of the genomic breakpoint junction sequences in the leukemias and functional DNA topoisomerase II cleavage assays suggest that not only drug-stabilized but also native DNA topoisomerase II cleavage can result in translocations. In particular, a rare case of treatment-related leukemia recently was described where the prior chemotherapy exposure did not include a DNA topoisomerase II poison (Langer 2003) Native DNA topoisomerase II cleavage can be the cause of the DNA damage in such cases because cytotoxic chemotherapy in general typically is followed by bone marrow progenitor cell proliferation (Knudson, 1992), which would be associated with high DNA topoisomerase IIα expression (Isaacs et al., 1998; Woessner et al., 1991).

In accordance with the present invention, a custom oligonucleotide array comprising sequences that span the breakpoint cluster region of the MLL gene that is disrupted in infant leukemias and treatment-related leukemias is provided. This custom oligonucleotide array can be used as an alternative to the real time PCR approach to detect the specific sequences that are involved in the formation of DNA-topoisomerase complexes or drug-DNA-topoisomerase complexes. This microarray facilitates analysis of the formation of DNA topoisomerase II complexes with MLL upon various different treatments of primary human hematopoietic cells and hematopoietic cell lines or non-hematopoietic cells. Briefly, such complexes can be detected by 1) treating the cells (in vitro, ex vivo, or in vivo), 2) isolating total genomic DNA including protein bound DNA, 3) isolating DNA topoisomerase II-bound DNA (e.g., on an immunoaffinity column), 4) amplifying the DNA by degenerative oligonucleotide PCR or by alternative whole-genome amplification methods, 5) labeling the test sample and a calibrated reference sample with different detectable labels (e.g., two different fluorescent dyes), and 6) hybridizing the labeled test sample and the calibrated standard or, alternatively, two different test samples labeled with different dyes with an MLL bcr microarray with two different channels, one channel for each one of the two dyes. The Examples describe experiments utilizing real-time PCR and microarrays to further characterize topoisomerase II-genomic DNA complexes and the sequences bound by topoisomerase II. Partner genes identified by various panhandle PCR techniques (see, e.g., U.S. Pat. No. 6,368,791; U.S. patent application Ser. No. 10/118,783; and U.S. Provisional Application 60/599,385), or by other techniques that lead to the identification of the sequences of translocation breakpoints may also be used with the real-time PCR and microarrays (see Table 6 for list of exemplary partner genes). The partner genes can be employed, alone or in combination with the MLL bcr in order to determine the relationship of translocation breakpoints to sites of DNA topoisomerase complex formation in different types of cells.

Notably, DNA damage mediated by aberrant topoisomerase activity can occur following exposure to naturally occurring topoisomerase poisons/inhibitors rather than chemotherapeutic agents, and exposure of pregnant women to such agents has been linked to infant leukemias. The methods and compositions of the invention can be utilized to characterize this DNA damage and provide the means to develop strategies to prevent topoisomerase II mediated alteration of the fetal chromosomal DNA during pregnancy.

Definitions

As used herein, the term “microarray” refers to an ordered arrangement of hybridizable array elements. The array elements are arranged so that there are preferably at least one or more different array elements, more preferably at least 100 array elements, and most preferably at least 1,000 array elements on a solid support. Preferably, the hybridization signal from each of the array elements is individually distinguishable, the solid support is a chip, and the array elements comprise oligonucleotide probes.

The term “MLL partner gene” refers to the gene or genomic DNA sequence fused with MLL after a translocation, such as those fusions present in certain leukemias.

“Nucleic acid” or a “nucleic acid molecule” as used herein refers to any DNA or RNA molecule, either single or double stranded and, if single stranded, the molecule of its complementary sequence in either linear or circular form. In discussing nucleic acid molecules, a sequence or structure of a particular nucleic acid molecule may be described herein according to the normal convention of providing the sequence in the 5′ to 3′ direction. With reference to nucleic acids of the invention, the term “isolated nucleic acid” is sometimes used. This term, when applied to DNA, refers to a DNA molecule that is separated from sequences with which it is immediately contiguous in the naturally occurring genome of the organism in which it originated. For example, an “isolated nucleic acid” may comprise a DNA molecule inserted into a vector, such as a plasmid or virus vector, or integrated into the genomic DNA of a prokaryotic or eukaryotic cell or host organism.

The term “oligonucleotide” as used herein refers to sequences, primers and probes of the present invention, and is defined as a nucleic acid molecule comprised of two or more ribo- or deoxyribonucleotides, preferably more than three. The exact size of the oligonucleotide will depend on various factors and on the particular application and use of the oligonucleotide.

The phrase “specifically hybridize” refers to the association between two single-stranded nucleic acid molecules of sufficiently complementary sequence to permit such hybridization under pre-determined conditions generally used in the art (sometimes termed “substantially complementary”). In particular, the term refers to hybridization of an oligonucleotide with a substantially complementary sequence contained within a single-stranded DNA or RNA molecule of the invention, to the substantial exclusion of hybridization of the oligonucleotide with single-stranded nucleic acids of non-complementary sequence.

For instance, one common formula for calculating the stringency conditions required to achieve hybridization between nucleic acid molecules of a specified sequence homology is set forth below (Sambrook et al., 1989):
Tm=81.5° C.+16.6 Log [Na+]+0.41(%G+C)−0.63(%formamide)−600/#bp in duplex

As an illustration of the above formula, using [Na+]=[0.368] and 50% formamide, with GC content of 42% and an average probe size of 200 bases, the Tm is 57° C. The Tm of a DNA duplex decreases by 1-1.5° C. with every 1% decrease in homology. Thus, targets with greater than about 75% sequence identity would be observed using a hybridization temperature of 42° C.

The stringency of the hybridization and wash depend primarily on the salt concentration and temperature of the solutions. In general, to maximize the rate of annealing of the probe with its target, the hybridization is usually carried out at salt and temperature conditions that are 20-25° C. below the calculated Tm of the hybrid. Wash conditions should be as stringent as possible for the degree of identity of the probe for the target. In general, wash conditions are selected to be approximately 12-20° C. below the Tm of the hybrid. In regards to the nucleic acids of the current invention, a moderate stringency hybridization is defined as hybridization in 6×SSC, 5× Denhardt's solution, 0.5% SDS and 100 μg/ml denatured salmon sperm DNA at 42° C., and washed in 2×SSC and 0.5% SDS at 55° C. for 15 minutes. A high stringency hybridization is defined as hybridization in 6×SSC, 5× Denhardt's solution, 0.5% SDS and 100 μg/ml denatured salmon sperm DNA at 42° C., and washed in 1×SSC and 0.5% SDS at 65° C. for 15 minutes. A very high stringency hybridization is defined as hybridization in 6×SSC, 5× Denhardt's solution, 0.5% SDS and 100 μg/ml denatured salmon sperm DNA at 42° C., and washed in 0.1×SSC and 0.5% SDS at 65° C. for 15 minutes.

The term “primer” as used herein refers to a DNA oligonucleotide, either single-stranded or double-stranded, either derived from a biological system, generated by restriction enzyme digestion, or produced synthetically which, when placed in the proper environment, is able to functionally act as an initiator of template-dependent nucleic acid synthesis. When presented with an appropriate nucleic acid template, suitable nucleoside triphosphate precursors of nucleic acids, a polymerase enzyme, suitable cofactors and conditions such as a suitable temperature and pH, the primer may be extended at its 3′ terminus by the addition of nucleotides by the action of a polymerase or similar activity to yield a primer extension product. The primer may vary in length depending on the particular conditions and requirement of the application. For example, in diagnostic applications, the oligonucleotide primer is typically 15-25 or more nucleotides in length. The primer must be of sufficient complementarity to the desired template to prime the synthesis of the desired extension product, that is, to be able anneal with the desired template strand in a manner sufficient to provide the 3′ hydroxyl moiety of the primer in appropriate juxtaposition for use in the initiation of synthesis by a polymerase or similar enzyme. It is not required that the primer sequence represent an exact complement of the desired template. For example, a non-complementary nucleotide sequence may be attached to the 5′ end of an otherwise complementary primer. Alternatively, non-complementary bases may be interspersed within the oligonucleotide primer sequence, provided that the primer sequence has sufficient complementarity with the sequence of the desired template strand to functionally provide a template-primer complex for the synthesis of the extension product.

Polymerase chain reaction (PCR) has been described in U.S. Pat. Nos. 4,683,195, 4,800,195, and 4,965,188, the entire disclosures of which are incorporated by reference herein.

The term “isolated” may refer to a compound or complex that has been sufficiently separated from other compounds with which it would naturally be associated. “Isolated” is not meant to exclude artificial or synthetic mixtures with other compounds or materials, or the presence of impurities that do not interfere with fundamental activity or ensuing assays, and that may be present, for example, due to incomplete purification, or the addition of stabilizers.

As used herein, an “instructional material” includes a publication, a recording, a diagram, or any other medium of expression which can be used to communicate the usefulness of the composition of the invention for performing a method of the invention.

The phrase “solid support” refers to any solid surface including, without limitation, any chip (for example, silica-based, glass, or gold chip), glass slide, membrane, bead, solid particle (for example, agarose, sepharose, polystyrene or magnetic bead), column (or column material), test tube, or microtiter dish.

The following materials and methods are provided to facilitate the practice of the present invention.

Cloning and Detection of MLL Genomic Breakpoint Junction Sequences in Sequential Bone Marrows

Characterization of genomic breakpoint junction sequences of the der(11) and other derivative chromosomes in the leukemias of the patients designated patients t-120 and t-39 by panhandle PCR approaches and PCR with gene-specific primers was previously described (7, 8). Backtracking to examine molecular emergence of the translocation in sequential bone marrow specimens from patient t-39 and to date the translocation in relation to chemotherapy administration has been reported (7). For patient t-120 diagnosed with treatment-related ALL harboring t(4; 11), BamHI-digested DNAs from sequential marrow samples were analyzed with the B859 fragment of ALL-1 cDNA corresponding to the MLL bcr. The der(4) genomic breakpoint junction sequence obtained by reverse panhandle PCR (8) provided sequences for forward AF-4 primer 5′-ATTGTTCTGCCCCCAACATA-3′ (SEQ ID NO: 247) and reverse MLL primer 5′-AGAGGCCCAGCTGTAGTTCT-3′ (SEQ ID NO: 248), and nested forward AF-4 primer 5′-TCCGTAAGCTCGACCCTAGT-3′ (SEQ ID NO: 249) and nested reverse MLL primer 5′-GCGCTCGTTCTCCTCTAAAC-3′ (SEQ ID NO: 250), which were used for clonotypic PCR to examine sequential bone marrow samples for molecular emergence of the t(4;11) translocation in relation to chemotherapy administered.

DNA Topoisomerase II In Vitro Cleavage Assays

DNA topoisomerase II in vitro cleavage assays were performed using previously described methods (10, 11). DNA fragments containing the normal homologues of each genomic breakpoint in MLL or GAS7 were subcloned into the EcoRI and BamHI sites, and the DNA fragment containing the normal homologue of the AF-4 genomic breakpoint into the BamHI and NotI sites of pBluescript II SK (Stratagene; La Jolla, Calif.). Twenty-five ng of singly 5′ end-labeled DNA substrates from these plasmids were incubated with human DNA topoisomerase IIα, ATP and MgCl2 in absence of drug or in the presence of etoposide, etoposide catechol, etoposide quinone or doxorubicin at 20 μM final concentration for 10 min at 37° C. DNA topoisomerase II cleavage complexes were irreversibly trapped by adding SDS, without heating or after subsequent incubation for 10 min at 75° C. to evaluate heat stability (12). Cleavage complexes were deproteinized and electrophoresed in a DNA sequencing gel alongside a dideoxy sequencing ladder to locate the cleavage sites (10, 11). In addition, doxorubicin was studied over a range of lower concentrations because of its known mixed effects of DNA topoisomerase II cleavage enhancement at low concentrations and DNA topoisomerase II catalytic inhibition due to intercalation at high concentrations (Capranico, 1998).

Ex Vivo Expansion of CD34+ Cells

Ten×106 primary human cadaveric bone marrow CD34+ cells obtained through NHLBI were established in culture at a density of 2×104 to 2×105 cells per ml and expanded in two T-75 flasks for 8 days in HPGM serum-free medium (Cambrex, Walkersville, Md.) supplemented with 100 ng/ml each of stem cell factor, Flk2/Flt3 ligand (FL) and IL-6, 10 ng/ml thrombopoietin and 1 μg/ml soluble IL-6 receptor (R&D Systems, Minneapolis, Minn.), conditions that promote minimal differentiation (13).

Western Blot Analysis

DNA topoisomerase II protein expression was examined with a mouse anti-human DNA topoisomerase IIα antibody (DAKO, Glostrup, Denmark) in 3 μg of total cellular protein from untreated ex vivo-expanded CD34+ cells and compared to that in the hematopoietic cell lines RS4:11, SEM-K2 and K562 (14-16). Protein concentrations were measured using the RCDC protein assay kit (Biorad, Hercules, Calif.) and Western blot analysis was performed with the Western Breeze kit (Invitrogen, Carlsbad, Calif.). The filter was simultaneously hybridized to a mouse anti-human β-actin antibody (Abcam, Cambridge, Mass.).

Real-Time PCR Analysis of DNA Topoisomerase IIα mRNA Expression

Quantitative real-time PCR analysis was performed on 10 ng of cDNA prepared from total RNA from untreated ex vivo-expanded CD34+ cells by ‘Assays on Demand’ for DNA topoisomerase IIα and GAPDH (Applied Biosystems, Foster City, Calif.) and compared to that in cultured hematopoietic cell lines using 2−ΔΔCt analysis of relative gene expression data (17).

ICE (In Vivo Complex of Enzyme) Bioassay

Modified ICE assays were performed exactly as described (11). Five×106 ex vivo expanded CD34+ cells were incubated without drug or with 100 μM final concentration of etoposide at 37° C. in 2 ml of HPGM medium for 2 hours. Percentages of viable cells were assessed by Trypan Blue exclusion. Treated and untreated cells were pelleted and lysed as described (11). After flash-freezing and thawing at 37° C., the DNA was sheared by passage through a 251/2G needle. Supernatants were collected and layered onto a CsCl cushion, and total genomic DNA including protein-bound DNA was isolated by ultracentrifugation at 80,000×g in an NVT90 rotor (Beckman; Palo Alto, Calif.) (11). The pellet was dissolved in 200 μl of 10 mM Tris-HCl (pH 8.0) 1 mM EDTA buffer, the DNA was quantified and 5.6 μg of DNA was analyzed on a Western blot with a mouse anti-human DNA topoisomerase IIα antibody (DAKO, Glostrup, Denmark) to detect DNA topoisomerase IIα-bound DNA (11).

Affinity Column Purification of DNA-DNA Topoisomerase IIα Covalent Complexes and Detection of MLL-Bound DNA Topoisomerase Ia

Each 1 ml affinity column consisted of Protein A-Sepharose Beads (Pharmacia Biotech, Upsala, Sweden) covalently coupled to mouse anti-human DNA topoisomerase IIα antibody (DAKO, Glostrup, Denmark) in PBS/0.02% sodium azide (18). The column was pre-washed with 10 volumes of 1×PBS. Total genomic DNA samples including protein-bound DNA from 32.5×106 untreated or etoposide-treated ex vivo-expanded human CD34+ cells was prepared according to the ICE assay protocol and diluted to a volume of 10 ml with cold 1×PBS. Samples were pre-cleared by incubation with Protein A Sepharose for 30 min while rotating at 4° C. in a 15 ml conical tube (18) and then centrifuged at 1200 rpm for 5 min in a Beckman G6 low-speed centrifuge. Supernatants were transferred to clean 15 ml conical tubes and incubated with the 1 ml of anti-human DNA topoisomerase IIα antibody-conjugated beads for 2 hrs while rotating at 4° C. and then run by gravity flow over affinity columns, followed by washing of the columns with 10 volumes of cold 1×PBS. Antibody-bound DNA topoisomerase IIα-DNA complexes from the untreated and etoposide-treated CD34+ cells were eluted with 1 ml of 100 mM glycine pH 2.7 and collected in 1.5 ml Eppendorf tubes containing a few drops of 1×PBS pH 11 for neutralization (18). The eluted material was PCI-extracted, ethanol-precipitated with NaOAc, washed with 70% EtOH and resuspended in 25 μl of dH2O and 1 μl was subjected to degenerative oligonucleotide PCR (DOP) using the primer 5′-CCGACTCGAGNNNNNNATGTGG-3′ (SEQ ID NO: 251) (19) to generate template DNAs in sufficient quantity for real-time PCR. The products were quantified by OD260 measurements and their sizes determined on an agarose gel.

MLL bcr primers for real-time PCR were selected using Primer Express v. 2.0 software, and their specificity confirmed using the BLAST algorithm. Forward and reverse primers 5′-ATAGTTTGTGTATTGCCAAGTCTGTTG-3′ (SEQ ID NO: 252) and 5′-GGCGCTCGTTCTCCTCTAAA-3′ (SEQ ID NO: 253), respectively, spanned MLL bcr positions 6784 to 6851 at the junction of intron 8-exon 9. The primer pair 5′-ACCACCGGGACCGCTACT-3′ (SEQ ID NO: 254) and 5′-GTGGCCCTAAGACATGATCAACT-3′ (SEQ ID NO: 255), was designed to amplify a genomic region of MLL exon 25, and a genomic region of GAPDH was examined by an intra-exon ‘Assay on Demand’ (Applied Biosystems); these genomic regions are not involved in MLL translocations. MGB oligonucleotide fluorogenic probes, which were synthesized by Applied Biosystems, were non-overlapping with the respective primer pairs and were designed according to Applied Biosystems guidelines using Primer Express v. 2.0 (20-23). Fluorogenic probe sequences for the amplicons at the MLL intron 8-exon 9 junction and MLL exon 25, respectively, were 5′-CCCTTCCACAAGTTTT-3′ (SEQ ID NO: 256) and 5′-ATCTTGAATCAAGTGCCAAA-3′ (SEQ ID NO: 257). Real-time PCR reactions were performed in duplicate in a MicroAmp 96-well plate using 50 ng of DOP-generated template, and the ABI Prism 7700 Sequence Detection System was used to examine product accumulation.

The following examples are provided to illustrate various embodiments of the present invention. They are not intended to limit the invention in any way.

EXAMPLE I Identification and Tracing of Translocation Breakpoint Sequences in Leukemias in Patients, and Evidence from In Vitro Assays to Suggest that Translocation Breakpoints are Topoisomerase II Cleavage Sites

Therapy, Clinical Course and Detection of MLL-AF-4 Translocation in Sequential Bone Marrow Specimens of Patient t-120

In patient t-120, rearrangements consistent with both derivative chromosomes from the t(4;11) translocation were detected by Southern blot analysis in the bone marrow from ALL diagnosis, and both breakpoint junctions were characterized by panhandle PCR approaches (Raffini et al., 2002). The clinical course, primary neuroblastoma therapy and molecular analyses are summarized in FIG. 1A. All available sequential bone marrows were examined for the presence of the translocation. The translocation also was detectable by Southern blot at 9 months from the start of treatment. The translocation was not detectable by Southern blot analysis in the cells used for autologous marrow rescue or in any other marrow samples (FIG. 1B, 1C). By first-round PCR analysis of the der(4) breakpoint junction in 200 ng genomic DNA prepared from cryopreserved sequential bone marrow samples (˜20,000 cell equivalent), only the marrows at 9 months after neuroblastoma diagnosis and at ALL diagnosis contained the translocation. By nested PCR (FIG. 1B, 1C) the translocation also was detectable in the marrow from 6 months after neuroblastoma diagnosis, after all 7 chemotherapy cycles and bone marrow harvest. This was before local radiation therapy, radiolabeled 3F8 monoclonal antibody and autologous marrow rescue, 5 months before leukemia was diagnosed. Spiking DNA from the ALL sample into peripheral blood lymphocyte DNA from a normal subject and serial dilutions indicated that the sensitivity of the nested PCR for detection of the translocation was between 1 cell in 105 and 1 cell in 106 cells.

Chemotherapy Enhances DNA Topoisomerase II Cleavage at MLL and AF-4 Genomic Translocation Breakpoints in all of Patient t-120

From prior sequencing the der(11) MLL breakpoint was position 6588 or 6589 in intron 8 and the der(11) AF-4 breakpoint was position 7130 or 7131 in intron 3. The der(4) AF-4 breakpoint was position 7108, 7109 or 7110 in intron 3 and the der(4) MLL breakpoint was position 6594, 6595 or 6596 in intron 8 (Raffini et al., 2002). Although ‘A’ nucleotides at the breakpoints in both genes at the der(11) breakpoint junction and 5′-CA-3′ sequences at the breakpoints in both genes at the der(4) breakpoint junction precluded more precise breakpoint assignments, 4-7 bases and 19-22 bases, respectively, were deleted from MLL and AF-4 (Raffini et al., 2002).

These near-precise recombinations with few bases lost relative to the normal sequences indicated that the translocation breakpoints were in close proximity to the sites of damage. The first molecular detection of the translocation was after all 7 chemotherapy cycles, which included etoposide and doxorubicin as DNA topoisomerase II poisons. Therefore, DNA topoisomerase II in vitro cleavage assays were performed without drug or with etoposide, its catechol or quinone metabolites or doxorubicin on double stranded DNA substrates containing the normal homologues of the respective MLL (FIG. 2A, 2C) and AF-4 (FIG. 2B, 2D) translocation breakpoints to locate where the drugs to which the patient was exposed stimulated cleavage complexes. DNA topoisomerase II creates staggered nicks in duplex DNA with 4-base 5′ overhangs (Fortune and Osheroff, 2000); cleavage site locations were defined by the base at the 5′ side of cleavage (−1 position) on the sense strand of DNA. DNA topoisomerase II cleavage sites in MLL (FIG. 2A, 2C) and AF-4 (FIG. 2B, 2D) were identified at or proximal to the translocation breakpoints.

Because the der(11) and der(4) MLL breakpoints were at a hotspot for translocation breakpoints in treatment-related leukemia, cleavage assays of the relevant MLL substrate with all drugs at 20 μM final concentration had already been performed (Whitmarsh et al., 2003). In the present study, however, doxorubicin was studied over a range of concentrations (FIG. 2C) because of its dual effects of cleavage stimulation at low concentrations and DNA topoisomerase II catalytic inhibition due to intercalation at high concentrations (Capranico and Binaschi, 1998). MLL bcr position 6588 ranked 5th of 8 cleavage sites detected without drug in the MLL substrate (Whitmarsh et al., 2003). Etoposide, etoposide catechol and etoposide quinone each at 20 μM, enhanced cleavage at this site 7.9-, 4.4- and 9.8-fold, respectively, over cleavage without drug (FIG. 2A). The especially strong cleavage detected at this position ranked 3rd of cleavage sites in the substrate in the presence of etoposide quinone (FIG. 2A) (Whitmarsh et al., 2003). The enzyme-only cleavage and cleavage with etoposide or its metabolites at position 6588 remained detectable after heating, indicating resistance to religation and stability of the cleavage complexes. Although position 6588 was one of only two cleavage sites detected with doxorubicin at 20 μM, the cleavage was weak compared to enzyme-only cleavage (0.4-fold), indicating catalytic inhibition at high concentration (FIG. 2A). At concentrations from 0.01 μM to 0.5 μM doxorubicin resulted in dose-dependent increases over enzyme-only cleavage at MLL bcr position 6588 consistent with poisoning effects, whereas the cleavage enhancement at this site began to decrease at 2.5 μM (FIG. 2C). Thus the intercalating agent doxorubicin has site-specific, concentration-dependent, mixed effects of a poison and a catalytic inhibitor of DNA topoisomerase II at this position in the MLL translocation breakpoint hotspot region. In contrast, at positions 6587 and 6589, only catalytic inhibition was observed at all concentrations tested (FIG. 2C).

In the AF-4 intron 3 substrate spanning positions 6956 to 7239, there was no detectable cleavage at position 7126 without drug or with doxorubicin at 20 μM (FIG. 2B) or lower concentrations (FIG. 2D), but cleavage at this position ranked first in relative intensity among all cleavage sites with etoposide and etoposide catechol and 5th among all cleavage sites with etoposide quinone (FIG. 2B). There was 3.2-, 5.9- and 3.4-fold cleavage at position 7126 in the presence of etoposide, etoposide catechol and etoposide quinone, respectively, relative to the strongest enzyme-only cleavage in the substrate, which occurred at position 7114. Cleavage at position 7126 in the presence of etoposide and both etoposide metabolites were not only especially strong, but also especially heat-stable. Examination of doxorubicin-associated cleavage over a range of concentrations with particular attention to the region of the breakpoints showed dose-dependent increases in cleavage stimulation over enzyme-only cleavage at AF-4 intron 3 positions 7111, 7114 and 7119, consistent with poisoning effects, while cleavage at these sites began to decrease at 0.5 μM, consistent with the mixed effect of catalytic inhibition (FIG. 2D).

Processing of DNA Topoisomerase II Cleavage Sites Enhanced by Etoposide or its Metabolites or Doxorubicin Forms Both Genomic Breakpoint Junctions in all of Patient t-120

The model shown in FIG. 3A for formation of the observed der(11) and der(4) genomic breakpoint junctions was derived from the cleavage sites at MLL bcr position 6588, which was enhanced by etoposide, both etoposide metabolites and doxorubicin, and AF-4 intron 3 position 7126, which was enhanced by etoposide and both etoposide metabolites but not doxorubicin. Processing of the 4-base 5′ overhangs from DNA topoisomerase II cleavage at these sites would generate the der(11) and der(4) sequences observed in the leukemia. In the model shown, exonucleolytic nibbling creates single-base homologies and base-pairing promotes the formation of both breakpoint junctions by error-prone nonhomologous end-joining (NHEJ). Consistent with the genomic sequencing and relative to the sense strands, 6 bases from MLL and 20 bases from AF-4 are lost during the processing, MLL bcr position 6588 and AF-4 intron 3 position 7130 are joined to form the der(11), and AF-4 intron 3 position 7109 and MLL bcr position 6595 are joined to form the der(4) (FIG. 3A).

The especially strong doxorubicin-stabilized cleavage sites at MLL bcr position 6588 and AF-4 intron 3 position 7114 were used to develop the alternative model in FIG. 3B for formation of the observed der(11) and der(4) genomic breakpoint junctions by error-prone NHEJ. In this model, 6 bases from MLL and 20 bases from AF-4 relative to the sense strands again are lost during the processing (FIG. 3B), and the same bases in MLL bcr and AF-4 intron 3 as described above are joined to form both breakpoint junctions. These models demonstrate that interchromosomal recombination by repair of chemotherapy-stabilized DNA topoisomerase II cleavage could be the translocation mechanism in this ALL, although models using other DNA topoisomerase II cleavage sites proximal to the translocation breakpoints are possible as well (not shown). However, it cannot be determined in this case whether etoposide or its metabolites or doxorubicin led to the relevant damage resulting in the translocation in the leukemia in this patient since each compound stimulated strong DNA topoisomerase II cleavage complexes proximal to the breakpoints.

Therapy, Clinical Course and Molecular Detection of MLL-GAS7 Translocation in Sequential Bone Marrow Specimens of Patient t-39

Patient t-39 was a 13-year old boy with stage 4 neuroblastoma treated with 4 cycles of cyclophosphamide, doxorubicin and vincristine (CAV), 1 cycle of cyclophosphamide and doxorubicin in which vincristine was omitted for toxicity, 3 cycles of cisplatin and etoposide (PVP), surgical resection, local radiation, and 3F8 monoclonal antibody with GM-CSF (Megonigal et al., 2000). The clinical diagnosis of secondary AML and molecular analyses of sequential bone marrow samples relative to the neuroblastoma treatment have been described (Megonigal et al., 2000). The MLL translocation was not PCR-detectable at neuroblastoma diagnosis, but was detectable by clonotypic PCR analysis of the der(11) genomic breakpoint junction in all marrow specimens obtained at and after 6 weeks from the start of treatment, which was after two cycles of CAV (Megonigal et al., 2000). AML was diagnosed 15.5 months after the translocation was PCR-detectable (Megonigal et al., 2000).

MLL and GAS7 Genomic Translocation Breakpoints in AML of Patient t-39 are Proximal to Doxorubicin-Stabilized DNA Topoisomerase II Cleavage Sites

From previously described genomic sequencing, the der(11) MLL breakpoint in the treatment-related AML was position 4662, 4663, or 4664 in intron 8 and the der(11) GAS7 breakpoint was position 1240, 1241 or 1242 upstream of exon 1 (Megonigal et al., 2000). MLL positions 4663-4664 and GAS7 positions 1240-1241 were 5′-AT-3′, precluding more precise breakpoint assignments (Megonigal et al., 2000). The der(17) GAS7 breakpoint was position 1203 and the der(17) MLL breakpoint was position 4680 (Megonigal et al., 2000). 15-17 bp from MLL and 36-38 bp from GAS7 were deleted during this translocation (Megonigal et al., 2000). The involved region of MLL was more 5′ in the bcr and was not the translocation breakpoint hotspot.

Although doxorubicin was the only DNA topoisomerase II poison to which the patient was exposed before molecular emergence of the translocation (Megonigal et al., 2000), DNA topoisomerase II in vitro cleavage assays were performed without drug, with doxorubicin or, as references for cleavage site intensities, with etoposide or its metabolites. The translocation breakpoints were near strong, enzyme-only cleavage sites at MLL bcr position 4675 (FIG. 4A) and GAS7 position 1238 (FIG. 4B) that were enhanced substantially by doxorubicin at low concentrations (FIG. 4C, 4D). In the MLL bcr substrate, the enzyme-only cleavage at position 4675 was 0.27-fold relative to cleavage with etoposide, and this position ranked 3rd among all enzyme-only cleavage sites (FIG. 4A). GAS7 position 1238, where the enzyme-only cleavage was 0.44-fold relative to cleavage with etoposide, was the strongest enzyme-only cleavage site in the GAS7 substrate (FIG. 4B). The enzyme-only cleavage complexes at MLL bcr position 4675 and GAS7 position 1238 were highly heat-resistant (FIG. 4A, 4B), indicating these cleavage complexes were particularly stable. Consistent with behavior as a poison, doxorubicin at 0.01 μM was associated with quantifiably enhanced cleavage over the already very strong enzyme-only cleavage, not only at MLL bcr position 4675, but also at MLL bcr position 4673 (FIG. 4C) and GAS7 position 1238 (FIG. 4D). The site-specific enhancement of cleavage over enzyme-only cleavage at these sites in the presence of doxorubicin began to decrease at 0.1 μM (FIG. 4C, D) and there was complete diminution at 20 μM (FIG. 4A, 4B), indicating mixed effects of a poison and catalytic inhibitor of DNA topoisomerase II.

Processing of Doxorubicin-Stabilized DNA Topoisomerase II Cleavage Sites in MLL and GAS7 Generates Genomic Breakpoint Junctions in AML of Patient t-39

A model for processing of the doxorubicin-stimulated cleavage sites at MLL intron 8 position 4675 and GAS7 position 1238 to form both genomic breakpoint junctions in the AML of patient t-39 is shown in FIG. 5. Exonucleolytic nibbling of the indicated bases (italic, middle) creates a single-base homology (box, middle), and NHEJ and gap fill-in ensue, resulting in the der(11) breakpoint junction. The processing to create the observed der(17) genomic breakpoint junction also includes exonucleolytic nibbling (italic, bottom) to form a single-base homology (box), followed by NHEJ and mismatch repair (bottom). Relative to the sense sequences, the exonucleolytic nibbling in the model results in loss of 16 bases from MLL and 37 bases from GAS7 during processing of the overhangs from the cleavage (FIG. 5). MLL bcr position 4663 and GAS7 position 1241 are joined to form the der(11), and GAS7 position 1203 and MLL bcr position 4680 are joined to form the der(17), which is consistent with prior genomic sequencing of the breakpoint junctions in the AML (Megonigal et al., 2000).

EXAMPLE II Hematopoietic Progenitor Cells—a Model Cellular System for Analysis of Chromosomal Translocations Mediated by Elevated Expression of Topoisomerase II

Characterization of Native DNA Topoisomerase IIα mRNA and Protein Expression in Human CD34+ Cells

By quantitative real-time PCR analysis, DNA topoisomerase IIα mRNA expression in the cell lines RS4:11, SEM-K2 and K562 cells was 1.54(1.16+2.03)-fold, 1.55(1.14-2.10)-fold and 2.75(2.53-2.99)-fold relative to ex vivo-expanded human CD34+ cells (Table 1), indicating that DNA topoisomerase IIα mRNA in proliferating bone marrow stem cells is in the range of that in hematopoietic cell lines. Western blot analysis suggested that DNA topoisomerase IIα protein expression in the ex vivo-expanded human CD34+ cells was also high and comparable to that in hematopoietic cell lines (FIG. 6).

TABLE 1 Relative DNA topoisomerase IIα mRNA expression in leukemia cell lines compared to CD34+ cells ΔCT Normalized DNA DNA (Avg. DNA topoisomerase IIα topoisomerase topoisomerase ΔΔCT amount relative to IIα CT GAPDH CT IIα CT − Avg. (Avg. ΔCT − Avg. CD34+ cells Cells (Avg. ± S.D.) (Avg. ± S.D.) GAPDH CT) ΔCTCD34+ cells) 2−ΔΔCT CD34+ 25.69 ± 0.26 20.58 ± 0.06 5.11 ± 0.23 0 1 cells RS4: 11 25.28 ± 0.38 20.79 ± 0.15 4.49 ± 0.40 −0.62 ± 0.40 1.54(1.16-2.03) SEM- 24.50 ± 0.28 20.02 ± 0.18 4.48 ± 0.44 −0.63 ± 0.44 1.55(1.14-2.10) K2 K562 25.61 ± 0.23 21.96 ± 0.12 3.65 ± 0.12 −1.46 ± 0.12 2.75(2.53-2.99)

EXAMPLE III Ice Bioassay Detects Topoisomerase—DNA and Topoisomerase-DNA-Drug Covalent Complexes in Total Genomic DNA of Hematopoietic Cell Lines and CD34 Cells

DNA Topoisomerase II Covalent Complexes are Detected in Hematopoietic Cell Lines Treated with Chemotherapy

Although etoposide has been previously studied in ICE assays of, eg., the hematopoietic cell line CEM, induction of DNA topoisomerase II cleavage complexes by etoposide metabolites had not been previously studied in a cellular context. A modified in vivo complex of enzyme (ICE) bioassy was employed to determine whether etoposide metabolites induce DNA topoisomerase II covalent complexes in the chromatin context of hematopoietic cell lines (11). The assay entails trapping the DNA covalently bound to DNA topoisomerase II by phosphotyrosine linkage using protein denaturants and detection on a Western blot. Isolation of the protein-bound DNA using a CsCl cushion, as elaborated by Topogene, streamlines the methodology compared to previous methods. In addition, the approach was changed further from use of the slot blot analysis to detect the cleavage complexes to detection of the complexes by Western blot analysis, which enables size separation of the DNA-protein complexes. ICE bioassys demonstrated significant induction of DNA topoisomerase II cleavage complexes in CEM cells after treatment for 2 hours with etoposide or its catechol or quinone metabolites at 20 μM (FIG. 7A). The cleavage complexes induced by etoposide catechol were comparable in amount to the parent drug. Induction of DNA cleavage complexes was also observed in K562 cells treated with 20 μM etoposide or etoposide quinone (FIG. 7B). Consistent with the presence of DNA bound to the enzyme, the DNA topoisomerase IIα protein ran higher than its known molecular weight of 170 kDa. Notably, no induction of DNA topoisomerase II cleavage complexes was detected in either CEM or K562 cells after treatment with doxorubicin at the same concentration within the sensitivity of the assay. However, doxorubicin is an intercalative DNA topoisomerase II poison that induces DNA topoisomerase II cleavage with different sequence site-selectivity (Capranico et al., 1990) and that also is associated with leukemia as a treatment complication, albeit less often (Sandoval et al., 1993). Its behavior as a site-specific poison of DNA topoisomerase II in in vitro assays of translocation breakpoints was established herein. Additional ICE assays in K562 cells showed a concentration dependent increase in the induction of DNA topoisomerase II cleavage complexes treated with etoposide quinone for 2 hours, consistent with its behavior as a DNA topoisomerase II poison (FIG. 7C). Unlike in CEM cells and K562 cells, in KG1 cells not only etoposide and its metabolites, but also doxorubicin at 100 μM induced DNA II covalent complexes (FIG. 7D).

Native and Etoposide-Stabilized DNA Topoisomerase II Covalent Complexes are Detected In Ex Vivo Expanded Human CD34+ Cells

ICE assays were performed on ex vivo expanded CD34+ cells from cadaveric human bone marrow that were either treated for 2 hours with etoposide or untreated. Cell viability assessed by Trypan blue exclusion was ˜70% for both conditions. Results for the untreated cells and cells treated with etoposide at 100 μM final concentration are shown in FIG. 8. As predicted in proliferating cells (6), native DNA topoisomerase II covalent complexes were detected in the untreated cells. Etoposide treatment resulted in increased formation of DNA topoisomerase II covalent complexes. The DNA topoisomerase IIα protein ran higher than its known molecular weight of 170 kDa, which is consistent with the presence of DNA bound to the enzyme. Etoposide catechol treatment also showed increased formation of topoisomerase-DNA cleavage complexes in CD34+ cells.

EXAMPLE IV Real-Time PCR with MLL Specific Primers Detects Topoisomerase-DNA and Topoisomerase-DNA-Drug Covalent Complexes with Specific Sequences in MLL Gene in Total Genomic DNA of Hematopoietic Cell Lines and CD34 Cells After Immunoaffinity Purification and DOP Amplification

DNA Topoisomerase II Covalent Complexes with MLL bcr are Detected in Untreated and Etoposide-Treated Ex Vivo-Expanded CD34+ Cells

A real-time PCR approach was developed to detect DNA topoisomerase II covalent complexes at the sequence level with the MLL breakpoint cluster region (bcr) in CD34+ cells. Total genomic DNA samples, including protein bound DNA from untreated or etoposide-treated, ex-vivo expanded human CD34+ cells, were prepared on CsCl cushions according to the ICE assay protocol. DNA-DNA topoisomerase IIα covalent complexes in the total genomic DNA were purified on an immuno-affinity column consisting of Protein A-Sepharose beads covalently coupled to a mouse anti-human DNA topoisomerase IIα antibody. The eluted material from the column was subjected to degenerative oligonucleotide PCR in order to generate template DNAs in sufficient quantity for real-time PCR. MLL bcr primers for real-time PCR were designed to amplify MLL bcr positions 6784 to 6851 at the junction of intron 8-exon 9 near the translocation breakpoint, which is in proximity to the hotspot region for translocation breakpoints occurring in cases of leukemia in patients exposed to DNA topoisomerase II poisons (Whitmarsh 2003) and, in at least one patient after chemotherapy without such agents (Langer 2003).

Real-time PCR products were detected with the primer pair for the junction of MLL intron 8-exon 9 in the DOP-amplified immunoaffinity-purified DNA topoisomerase IIα-bound DNA from untreated and etoposide-treated CD34+ cells (FIG. 9A). The DOP template DNA used for real-time PCR was 500 bp to 1 kb. Since DNA templates containing double-strand breaks from DNA topoisomerase II cleavage would not be amplified by PCR, these data indicate the presence of DNA-DNA topoisomerase II covalent complexes proximal to the amplicon (MLL bcr positions 6784 to 6851) where products were detected with resolution at the size of the DOP template. No products were detected in reactions assaying the GAPDH gene (FIG. 9B), which is not involved in MLL translocations. Although no product was detected in untreated CD34+ cells, etoposide induced DNA-DNA topoisomerase II covalent complexes proximal to the MLL exon 25 amplicon (FIG. 9C), indicating that the formation of DNA topoisomerase II cleavage complexes in MLL is not limited to the bcr.

DNA Topoisomerase II Covalent Complexes with MLL bcr are Detected in Etoposide-Treated and Genistein-Treated Ex Vivo-Expanded CD34+ Cells

In cases of leukemia in infants, the MLL translocation breakpoints are distributed heterogeneously in the bcr (Felix, 1998.) Genistein is one example of a naturally-occurring DNA topoisomerase II poison that can stimulate formation of cleavage complexes with DNA and to which the fetus can be exposed in utero via the maternal diet. FIG. 9D shows real-time PCR products that were detected with primers at the junction of MLL intron 7-exon 8 in the DOP-amplified immunoaffinity-purified DNA topoisomerase IIα-bound DNA after treatment of CD34+ cells with either etoposide or genistein.

2. Discussion

The temporal emergence of MLL translocations has been characterized herein with respect to the timing of administration of specific anti-cancer drugs and mechanistic studies of DNA topoisomerase II cleavage were performed in order to link specific DNA damage to the genesis of MLL translocations in two cases of treatment-related leukemia. There was variability in molecular emergence of the translocations; after all 4 cycles of CAV and 3 cycles of PVP in one case, but after only 2 cycles of CAV in the other (Megonigal et al., 2000). In both cases the translocation was absent at neuroblastoma diagnosis, suggesting that the treatment caused and did not select for a pre-existing translocation. These results are consistent with other observations on a patient diagnosed with primary ALL and MLL-rearranged treatment-related AML where the MLL translocation was found to emerge during the course of treatment (Blanco et al., 2001). Absence of the MLL translocation at neuroblastoma diagnosis and the short latency in the secondary cases under study here also relate to the time of acquisition of the translocation in MLL-rearranged de novo cases. In MLL-rearranged infant leukemias the translocation is a somatic, in utero event and the latency is short: specifically from some time in pregnancy to the time of leukemia diagnosis in the infant host (Ford et al., 1993; Gale et al., 1997; Megonigal et al., 1998). However, MLL-rearranged leukemias in older children generally are not traceable to birth (Maia et al., 2004). Factors that determine latency from acquisition of the translocation to emergence of leukemia are unknown but the variable latencies in the present study, 5 months and 15.5 months in the respective cases where the partner genes were AF-4 and GAS7, may indicate differences in sufficiency of the varied MLL gene fusions. Latency also may be a function of the subsequent primary cancer treatment, which would not only contribute to secondary alterations but also affect selection and survival of the preleukemia clone.

Patient t-120 was managed with autologous marrow rescue after monoclonal antibody 3F8-targeted 131I-radioimmunotherapy. The ALL was diagnosed 11 months after neuroblastoma diagnosis, only 2 weeks after transplant. It has been suggested that autologous stem-cell collection following etoposide is associated with an increased risk of leukemia with characteristic balanced translocations (Krishnan et al., 2000). However, by a sensitive PCR-based assay the t(4;11) was not detectable in the unpurged or the purged marrow autograft harvested after the second cycle of etoposide-containing PVP, and did not emerge until after all chemotherapy cycles were complete. The t(4;11) occurred before the local radiation and radioimmunotherapy during the intensive N7 neuroblastoma regimen. The MLL-GAS7 translocation also was present before any radiation (Megonigal et al., 2000), indicating that the chemotherapy but not radiation contributed to the damage that caused these translocations.

The DNA topoisomerase II inhibitor exposures before molecular emergence of the t(4;11) in the case of patient t-120 were doxorubicin and etoposide, and there were etoposide-, etoposide metabolite- and doxorubicin-stimulated DNA topoisomerase II in vitro cleavage sites proximal to the translocation breakpoints that could be repaired to form both breakpoint junctions. The alternative models for formation of the der(11) and der(4) genomic breakpoint junctions were based on cleavage sites at MLL bcr position 6588, which was enhanced by all of the drug exposures, and either the cleavage site at AF-4 intron 3 position 7126 enhanced only by etoposide and its metabolites (FIG. 3A) or AF-4 intron 3 position 7114 enhanced only by doxorubicin (FIG. 3B). Since this was a later-occurring translocation during the treatment and emerged only after exposure to both DNA topoisomerase II poisons in the regimen, it is possible that etoposide or its metabolites or doxorubicin caused the relevant damage. Nonetheless, in the ALL of patient t-120, the processing of functional drug-stabilized DNA topoisomerase II cleavage sites in MLL and AF-4 generated the observed genomic breakpoint junctions.

These results differ from the findings in the second case where the translocation first became detectable when doxorubicin was the only DNA topoisomerase II poison to which the patient was exposed. The translocation breakpoints in MLL and GAS7 were proximal to strong doxorubicin-stimulated DNA topoisomerase II in vitro cleavage sites at MLL bcr position 4675 and GAS7 position 1238 that could be resolved to form both breakpoint junctions. This is the first functional demonstration of relevance of doxorubicin-stimulated DNA topoisomerase II covalent complexes with MLL and with its partner gene in the genesis of MLL translocations in treatment-related AML.

Another consideration was that anthracyclines are known to exhibit both cleavage stimulation characteristic of DNA topoisomerase II poisons at low concentrations but, at high concentrations, catalytic inhibition of DNA topoisomerase II function due to intercalation (Capranico and Binaschi, 1998). Analysis of DNA topoisomerase II cleavage with doxorubicin over a range of concentrations unmasked doxorubicin-stimulated cleavage sites proximal to the translocation breakpoints in MLL, AF-4 and GAS7 that were not detected at higher concentrations. The balance of dose-dependent dual effects of doxorubicin as a poison and a catalytic inhibitor of DNA topoisomerase II function has substantial implications for its role in the genesis of translocations in a cellular context because the model for formation of the translocations is based on poisoning effects. The site selectivity of the doxorubicin-stimulated in vitro cleavage sites proximal to the breakpoints also is of interest. Different patterns of cleavage stimulation by various DNA topoisomerase II poisons at preferred sites in any given substrate has suggested that local base sequences enable formation of ternary drug-DNA-enzyme complexes (Capranico and Binaschi, 1998). The previously reported site selectivity of doxorubicin was A at position −1 and T at position −2 relative to the cleavage (Capranico and Binaschi, 1998). Here, however, only 2 of the 7 cleavage sites in MLL, AF-4 and GAS7 (shown in FIGS. 2C, 2D, 4C, and 4D) where doxorubicin behaved as a poison (specifically, MLL bcr position 4673 and AF-4 intron 3 position 7119), fulfilled these sequence preferences on either the sense strand or the antisense strand of the DNA. Thus the sequence preferences for poisoning effects of doxorubicin may be less stringent than previously thought.

Non-homologous end joining (NEJ) repair events, which have been implicated in the resolution of DNA damage in the MLL translocation process (Lovett et al., 2001), are often imprecise and ensue after small deletions or insertions at the site of damage (Liang et al., 1998). The models in FIGS. 3A, 3B, and 5 invoked small deletions at the DNA topoisomerase II cleavage sites in MLL and its partner genes to create homologous overhangs that formed characteristic breakpoint junctions. Single-base homologies, as would be present after exonucleolytic nibbling at these staggered nicks, are sufficient for error-prone NHEJ (Liang et al., 1998).

The formation of drug-stabilized and native DNA topoisomerase II cleavage complexes in ex vivo-expanded human CD34+ cells was also investigated as a cellular model for analysis of MLL translocations. It was important to devise a cellular model to further study the role of DNA topoisomerase II cleavage in the genesis of MLL translocations because cleavage sites in the cellular context should be more restricted than in vitro (Capranico et al., 1990). In a murine retroviral transplant model it recently was shown that leukemias with MLL translocations can arise either in self-renewing stem cells or in committed myeloid progenitor cell populations (Cozzio et al., 2003), corroborating relevance of the human CD34+cell model system. The analyses of DNA topoisomerase IIα mRNA (Table 1) and protein (FIG. 6) expression indicate that in human CD34+ cells grown in short-term culture DNA topoisomerase IIα is highly expressed at levels comparable to human hematopoietic cell lines. Human CD34+ cells had not been previously studied in the ICE bioassay, which detects any DNA in total genomic DNA bound to DNA topoisomerase IIα by the covalent phosphotyrosine linkage formed between this enzyme and DNA (Whitmarsh et al., 2003). Both native and etoposide-stabilized DNA topoisomerase IIα cleavage complexes were demonstrable in the CD34+ cells.

In addition, a new approach was devised for detecting DNA topoisomerase IIα covalently bound specifically to the MLL bcr in untreated or etoposide-treated CD34+ cells. This assay, which is similar to a ChIP assay (Boyd et al., 1998) without cross-linking, was accomplished by immuno-affinity purification of DNA topoisomerase II covalent complexes from the total genomic DNA of the CD34+ cells followed by DOP and then real-time PCR using primers proximal to the hotspot for MLL translocation breakpoints (Whitmarsh et al., 2003) in intron 8 3′ in the bcr. Native as well as etoposide-stabilized DNA topoisomerase II covalent complexes with the MLL bcr were detectable in the immuno-affinity purified DNA topoisomerase II covalent complexes, demonstrating that DNA topoisomerase II forms covalent complexes with MLL at sites of translocation breakpoints in human CD34+ cells. That MLL exon 25 is distal to the bcr and not involved in leukemia-associated translocations, yet etoposide induced DNA topoisomerase II cleavage complexes proximal to the MLL exon 25 amplicon, may suggest that translocations resulting from repair of DNA topoisomerase II-mediated damage in MLL exon 25 would not provide a selective or proliferative advantage for leukemogenesis. Detection of DNA topoisomerase II covalent complexes at the hotspot for MLL translocation breakpoints in intron 8 3′ in the bcr in the presence of etoposide establishes that drug-stabilized DNA topoisomerase II cleavage is a mechanism to damage MLL at a relevant site and in a relevant cellular model system.

In addition, formation of DNA topoisomerase IIα covalent complexes at the hotspot for MLL translocation breakpoints by the native enzyme is especially important. Translocations of the MLL gene are the hallmark aberrations in leukemias that follow chemotherapy with DNA topoisomerase II poisons (Rowley and Olney, 2002) and several treatment-related leukemias occurring after DNA topoisomerase II poisons have translocation breakpoints at this hotspot region (reviewed in (Whitmarsh et al., 2003)). However, not all MLL-rearranged treatment-related leukemias occur after exposure to these agents. For example, a case of secondary AML with t(9;11) and der(11) and der(9) MLL breakpoints in this hotspot region has been reported after chemotherapy for primary Hodgkin's disease without DNA topoisomerase II poisons (Langer et al., 2003). Formation of DNA topoisomerase IIα covalent complexes in this genomic region by the native enzyme may be the cause of the breakage in such cases. Furthermore, native DNA topoisomerase II cleavage also should be active during the bone marrow repopulation and recovery after cytotoxic chemotherapy in general because DNA topoisomerase IIα expression is cell cycle-dependent and highest in proliferating cells (Isaacs et al., 1998).

The results of the present study imply two mechanisms for DNA topoisomerase II involvement in the DNA damage that results in MLL translocations. The first mechanism implicates direct poisoning effects of drug-stabilized DNA topoisomerase II cleavage complexes at the translocation breakpoint sites as the damage mechanism. In the second mechanism, native DNA topoisomerase II mediates cleavage at the translocation breakpoints. This second mechanism is relevant to the cases where prior cytotoxic chemotherapy without DNA topoisomerase II poisons was administered. Ternary DNA topoisomerase II cleavage complexes with MLL sequences involved in translocations can be stimulated by poisons of the enzyme, but the native enzyme alone forms cleavage complexes with the MLL translocation breakpoint hotspot; either can be important in the DNA damage that leads to translocations.

Another example was shown of real-time PCR detection of DNA topoisomerase II complexes in proximity to the MLL intron 7-exon 8 junction in CD34+ cells that were treated either with etoposide or genistein, indicating that the naturally-occurring compound that is found in diet also is associated with formation of functional DNA topoisomerase cleavage complexes proximal to this amplicon in the MLL bcr.

EXAMPLE V Microarrays

One goal of these assays is the global identification of DNA topoisomerase II mediated damage from etoposide and its metabolites in the MLL bcr and in the genome in general. Such damage can be studied in ex vivo-expanded human bone marrow progenitor cells as representative targets for translocations. A direct interaction of DNA topoisomerase II with specific MLL bcr sequences has not been investigated in a cellular context except in the real-time PCR assays described herein. The generation of a custom MLL bcr-specific oligonucleotide array would further streamline and improve upon the present detection method.

Employing XeoChip™ technology (Xeotron, Houston, Tex.), a first generation MLL bcr oligonucleotide array containing replicate slots of each of 162 probes was created. The 162 probes included 73 probes for the non-repetitive MLL bcr sequences, 8 probes for the Alu region between nucleotide positions 663-1779 in the MLL bcr, 57 probes for MLL exon 25 which is unaffected by the translocations, and 24 probes for 9 bacterial control genes (see Tables 2 and 3; Tables 4 and 5 provide another example of a series of oligonucleotides that can be employed in a microarray). The 8.3 kb MLL bcr is represented by replicates of each of the 73 50mers at average intervals of 115 bases to span the bcr. Because it has been well documented in the literature that the printing, general hybridization conditions, and scanning of microarrays introduce systemic effects that are related to the position on the chip, replicates of each of the probes in the array have been spaced throughout the chip.

MLL-specific probes without homology to each other or to Alu sequences were designed to be 50mer sense-strand sequences in accordance with Xeotron parameters. A BLAST search of the probes against the human genome sequence confirmed that the probes were MLL-specific. Special probe design was utilized for the Alu-rich region from MLL bcr positions 663-1779 where no consideration for cross-homology was possible. Bacterial control gene sequences were BLAST searched against the human genome sequence database. Probes were not designed to regions where BLAST hits were found and then designed to be sense-strand 50mer sequences using the Xeotron parameters. Except in the instance of the Alu-rich region probes, this probe design, under stringent hybridization and washing conditions, should enable distinguishing between distinct sequences where the overall homology is <80% (i.e. 10 mismatches out of 50 bases) and where there are no stretches of >25 bases of homology.

Primary CD34+ selected human bone marrow cells were obtained through the NHLBI or purchased commercially. The cells were expanded ex vivo using conditions optimized to promote minimal differentiation in order to increase cell numbers as required to generate a useful model system. Briefly, the general strategy is: 1) ex vivo-expand CD34+ cells or other relevant cells to be treated with etoposide or different test compound, 2) treat the cells with etoposide or reserve as untreated, 3) lyse the cells, 4) shear the DNA, 5) isolate total genomic DNA including any protein-bound DNA as for ICE bioassays (see FIG. 10), 6) isolate DNA-DNA topoisomerase II covalent complexes with an immunoaffinity column according to the scheme summarized in FIG. 11 and elute DNA-DNA topoisomerase II covalent complexes from the column, 7) perform whole genome amplification by degenerative oligonucleotide PCR or another suitable technique, 8) quantify DOP products by measuring the OD260, 9) measure fragment sizes of DOP products in an agarose gel, and 10) analyze the DOP products from the experimental sample (which can either be untreated cells or cells treated with specific agent) and the calibrated standard sample (i.e. plasmid containing genomic sequence of the MLL bcr) labeled with a detectable label such as Alexa Fluor dyes (Molecular Probes, Eugene, Oreg.), heat-denatured, and hybridized in different channels to a pre-hybridized MLL bcr XeoChip™.

Components of the experimental design, including statistical and computational tools to use MLL bcr DNA oligonucleotide arrays to study human CD34+ cells treated with etoposide and compare treated with untreated cells, also were developed. A degenerative oligonucleotide PCR-(DOP-) amplified plasmid DNA template containing an 8.3 kb genomic DNA insert of the entire MLL bcr in PBSK II+ is employed for use as a labeled reference sample calibrated against every probe in the array in order to enable better accuracy in spot intensity information and control for any unequal labeling of specific regions in the bcr in the DOP-amplified experimental samples. By 1) serial dilutions of AlexaFluor dye-labeled DOP products of the reference sample, 2) acquisition of MLL bcr XeoChip™ images with the Cy-3 and Cy-5 channels of a GenePix® (Axon Instruments, Foster City, Calif.) laser scanner, and 3) analysis using the GenePix® program, it has been established that hybridization of 0.02 μg of the reference sample gives a non-saturating hybridization signal (FIG. 12).

In additional experiments, CD34+ cells were expanded in culture, treated or untreated for 2 hours with etoposide at 100 μM final concentration. The total cellular DNA was harvested including any protein-bound DNA on CsCl cushions, and then immunoaffinity column purification of DNA covalently bound to DNA topoisomerase IIα was performed in etoposide-treated or untreated samples according to the schematics summarized in FIG. 10 and FIG. 11. DOP of the immunoaffinity-purified DNA topoisomerase II cleavage complexes from the treated and untreated cells was performed exactly as was done with the reference sample. In one such experiment the samples from untreated cells or cells treated with 100 μM etoposide were labeled with AlexaFluor 546 (equivalent of Cy-3; green) and the MLL bcr reference sample was labeled with AlexaFluor 647 (equivalent of Cy-5; red). Dye-swap experiments were also performed. MLL bcr XeoChip™ hybridizations were then performed with 0.02 μg of AlexaFluor dye-labeled DOP products of etoposide-treated or untreated samples in one channel of the chip and the reference sample labeled with the reverse AlexaFluor dye in the other channel.

GeneSpring (Silicon Genetics, Redwood City, Calif.) was employed as a computational tool to visualize and analyze the data from MLL bcr XeoChips™ with either treated or untreated samples in one channel and the reference sample in the other. Non-parametric statistical regression tools can also be applied in order to identify hybridization hotspots that suggest effects of treatment. The preliminary data analysis suggests that the MLL Xeochips™ microarray is useful for detecting hotspots and coldspots for DNA topoisomerase II complex formation with the MLL bcr.

A Quantile-Quantile (Q-Q) plot (44, 45) standardized by intensity was employed to compare etoposide-treated and untreated cells (FIG. 12B). DOP-amplified DNA from the DNA topoisomerase II cleavage complexes in treated or untreated cells on separate chips was labeled with AlexaFluor 546. The MLL bcr reference sample was labeled with AlexaFluor 647. The null hypothesis represented by the straight line is that there is no effect at any probe, whereas upward departure points are potential hotspots and downward departure points are potential coldspots. As seen in FIG. 12B, probes 1-41 and 1-51, for example, appear as hotspots and probes 2-28, 1-30, 1-18, and 1-15, for example, appears as a cold spot.

A similar Q-Q plot was performed on untreated cells and cells treated with 100 μM genistein (FIG. 12C). Probes 1-68 (SEQ ID NO: 157), 1-41 (SEQ ID NO: 49), 1-51 (SEQ ID NO: 141), and 1-33 (SEQ ID NO: 132) were identified as potential hotspots and probes 1-31 (SEQ ID NO: 27), 2-50 (SEQ ID NO: 162), 1-74 (SEQ ID NO: 62), 2-28 (SEQ ID NO: 34), 1-10 (SEQ ID NO: 83), and 1-22 (SEQ ID NO: 71) were identified as potential coldspots. Notably, probes 1-41 (SEQ ID NO: 49) and 1-51 (SEQ ID NO: 141) were identified as hotspots in both etopside and genistein treated cells while probe 2-28 (SEQ ID NO: 34) was identified as a coldspot. It is also noteworthy that probes 1-68 (SEQ ID NO: 157) and 1-33 (SEQ ID NO: 132) correspond to nearby oligonucleotides. Additionally, these results mirror in vitro cleavage studies with etoposide and genistein as there is some overlap in cleavage sites induced by the different agents as well as differences in site selectivity.

TABLE 2 Probe Ranked Position Random Original order of Sequence within Sorter Sorter Probe ID preference Sequence Description Length sequence 1 93 0002-12 12 MLL Exon 25 4249 2662 2 66 0001-66 24 MLL Exon-intron 5 to 11 8342 3682 3 113 0002-32 32 MLL Exon 25 4249 2450 4 24 0001-24 24 MLL Exon-intron 5 to 11 8342 5309 5 32 0001-32 32 MLL Exon-intron 5 to 11 8342 5938 6 106 0002-25 25 MLL Exon 25 4249 436 7 54 0001-54 12 MLL Exon-intron 5 to 11 8342 4395 8 161 LysX-M 1 LysX-M 270 132 9 118 0002-37 37 MLL Exon 25 4249 2748 10 112 0002-31 31 MLL Exon 25 4249 334 11 110 0002-29 29 MLL Exon 25 4249 2829 12 157 TrpnX-3 1 TrpnX-3 579 128 13 86 0002-5 5 MLL Exon 25 4249 3476 14 160 BioB-3 1 BioB-3 298 61 15 30 0001-30 30 MLL Exon-intron 5 to 11 8342 2460 16 61 0001-61 19 MLL Exon-intron 5 to 11 8342 3149 17 39 0001-39 39 MLL Exon-intron 5 to 11 8342 3214 18 81 0001-81 8 >MLL Exon-intron 5 to 11 8342 1578 19 12 0001-12 12 MLL Exon-intron 5 to 11 8342 4666 20 2 0001-2 2 MLL Exon-intron 5 to 11 8342 7974 21 73 0001-73 31 MLL Exon-intron 5 to 11 8342 1875 22 130 0002-49 12 MLL Exon 25 4249 160 23 85 0002-4 4 MLL Exon 25 4249 4048 24 103 0002-22 22 MLL Exon 25 4249 1285 25 46 0001-46 4 MLL Exon-intron 5 to 11 8342 314 26 57 0001-57 15 MLL Exon-intron 5 to 11 8342 3349 27 31 0001-31 31 MLL Exon-intron 5 to 11 8342 88 28 139 BioB-5 1 BioB-5 274 83 29 52 0001-52 10 MLL Exon-intron 5 to 11 8342 5089 30 72 0001-72 30 MLL Exon-intron 5 to 11 8342 4585 31 20 0001-20 20 MLL Exon-intron 5 to 11 8342 7877 32 13 0001-13 13 MLL Exon-intron 5 to 11 8342 5549 33 155 ThrX-5 1 ThrX-5 645 391 34 109 0002-28 28 MLL Exon 25 4249 2314 35 21 0001-21 21 MLL Exon-intron 5 to 11 8342 7776 36 45 0001-45 3 MLL Exon-intron 5 to 11 8342 4501 37 65 0001-65 23 MLL Exon-intron 5 to 11 8342 5347 38 34 0001-34 34 MLL Exon-intron 5 to 11 8342 4361 39 15 0001-15 15 MLL Exon-intron 5 to 11 8342 1779 40 119 0002-38 1 MLL Exon 25 4249 3435 41 35 0001-35 35 MLL Exon-intron 5 to 11 8342 6403 42 18 0001-18 18 MLL Exon-intron 5 to 11 8342 6578 43 99 0002-18 18 MLL Exon 25 4249 3937 44 126 0002-45 8 MLL Exon 25 4249 857 45 94 0002-13 13 MLL Exon 25 4249 181 46 7 0001-7 7 MLL Exon-intron 5 to 11 8342 7 47 105 0002-24 24 MLL Exon 25 4249 3395 48 100 0002-19 19 MLL Exon 25 4249 1182 49 41 0001-41 41 MLL Exon-intron 5 to 11 8342 343 50 116 0002-35 35 MLL Exon 25 4249 1984 51 117 0002-36 36 MLL Exon 25 4249 523 52 149 DapX-M 1 DapX-M 561 367 53 150 LysX-3 1 LysX-3 283 171 54 5 0001-5 5 MLL Exon-intron 5 to 11 8342 2361 55 162 LysX-5 1 LysX-5 273 12 56 67 0001-67 25 MLL Exon-intron 5 to 11 8342 3471 57 53 0001-53 11 MLL Exon-intron 5 to 11 8342 7636 58 88 0002-7 7 MLL Exon 25 4249 3557 59 27 0001-27 27 MLL Exon-intron 5 to 11 8342 3584 60 153 PheX-M 1 PheX-M 392 238 61 152 PheX-5 1 PheX-5 348 86 62 74 0001-74 1 >MLL Exon-intron 5 to 11 8342 779 63 83 0002-2 2 MLL Exon 25 4249 3839 64 75 0001-75 2 >MLL Exon-intron 5 to 11 8342 870 65 158 TrpnX-5 1 TrpnX-5 531 218 66 101 0002-20 20 MLL Exon 25 4249 2956 67 44 0001-44 2 MLL Exon-intron 5 to 11 8342 516 68 76 0001-76 3 >MLL Exon-intron 5 to 11 8342 991 69 114 0002-33 33 MLL Exon 25 4249 3303 70 19 0001-19 19 MLL Exon-intron 5 to 11 8342 7541 71 22 0001-22 22 MLL Exon-intron 5 to 11 8342 3022 72 135 0002-54 17 MLL Exon 25 4249 2457 73 90 0002-9 9 MLL Exon 25 4249 70 74 47 0001-47 5 MLL Exon-intron 5 to 11 8342 6398 75 144 BioDn-5 1 BioDn-5 277 221 76 123 0002-42 5 MLL Exon 25 4249 1264 77 79 0001-79 6 >MLL Exon-intron 5 to 11 8342 1343 78 122 0002-41 4 MLL Exon 25 4249 1556 79 111 0002-30 30 MLL Exon 25 4249 909 80 120 0002-39 2 MLL Exon 25 4249 2280 81 50 0001-50 8 MLL Exon-intron 5 to 11 8342 680 82 56 0001-56 14 MLL Exon-intron 5 to 11 8342 5466 83 10 0001-10 10 MLL Exon-intron 5 to 11 8342 2611 84 91 0002-10 10 MLL Exon 25 4249 604 85 124 0002-43 6 MLL Exon 25 4249 2016 86 37 0001-37 37 MLL Exon-intron 5 to 11 8342 6683 87 8 0001-8 8 MLL Exon-intron 5 to 11 8342 169 88 107 0002-26 26 MLL Exon 25 4249 1656 89 25 0001-25 25 MLL Exon-intron 5 to 11 8342 663 90 140 BioB-M 1 BioB-M 258 80 91 38 0001-38 38 MLL Exon-intron 5 to 11 8342 4480 92 26 0001-26 26 MLL Exon-intron 5 to 11 8342 3921 93 98 0002-17 17 MLL Exon 25 4249 2540 94 142 BioC-5 1 BioC-5 349 193 95 97 0002-16 16 MLL Exon 25 4249 1575 96 102 0002-21 21 MLL Exon 25 4249 3753 97 14 0001-14 14 MLL Exon-intron 5 to 11 8342 2699 98 16 0001-16 16 MLL Exon-intron 5 to 11 8342 6769 99 151 PheX-3 1 PheX-3 442 353 100 42 0001-42 42 MLL Exon-intron 5 to 11 8342 4574 101 55 0001-55 13 MLL Exon-intron 5 to 11 8342 2829 102 64 0001-64 22 MLL Exon-intron 5 to 11 8342 2515 103 127 0002-46 9 MLL Exon 25 4249 2636 104 134 0002-53 16 MLL Exon 25 4249 1392 105 84 0002-3 3 MLL Exon 25 4249 3101 106 108 0002-27 27 MLL Exon 25 4249 705 107 78 0001-78 5 >MLL Exon-intron 5 to 11 8342 1256 108 63 0001-63 21 MLL Exon-intron 5 to 11 8342 2940 109 138 0002-57 20 MLL Exon 25 4249 3351 110 141 BioC-3 1 BioC-3 253 5 111 4 0001-4 4 MLL Exon-intron 5 to 11 8342 6963 112 49 0001-49 7 MLL Exon-intron 5 to 11 8342 5675 113 77 0001-77 4 >MLL Exon-intron 5 to 11 8342 1153 114 28 0001-28 28 MLL Exon-intron 5 to 11 8342 5669 115 136 0002-55 18 MLL Exon 25 4249 278 116 92 0002-11 11 MLL Exon 25 4249 1437 117 89 0002-8 8 MLL Exon 25 4249 4165 118 128 0002-47 10 MLL Exon 25 4249 3269 119 3 0001-3 3 MLL Exon-intron 5 to 11 8342 250 120 11 0001-11 11 MLL Exon-intron 5 to 11 8342 517 121 62 0001-62 20 MLL Exon-intron 5 to 11 8342 3913 122 58 0001-58 16 MLL Exon-intron 5 to 11 8342 2299 123 125 0002-44 7 MLL Exon 25 4249 2118 124 148 DapX-5 1 DapX-5 499 2 125 146 CreX-5 1 CreX-5 535 229 126 95 0002-14 14 MLL Exon 25 4249 2206 127 147 DapX-3 1 DapX-3 447 85 128 132 0002-51 14 MLL Exon 25 4249 3757 129 137 0002-56 19 MLL Exon 25 4249 3037 130 71 0001-71 29 MLL Exon-intron 5 to 11 8342 5171 131 48 0001-48 6 MLL Exon-intron 5 to 11 8342 5258 132 33 0001-33 33 MLL Exon-intron 5 to 11 8342 2827 133 145 CreX-3 1 CreX-3 407 220 134 82 0002-1 1 MLL Exon 25 4249 1890 135 43 0001-43 1 MLL Exon-intron 5 to 11 8342 6966 136 23 0001-23 23 MLL Exon-intron 5 to 11 8342 7450 137 17 0001-17 17 MLL Exon-intron 5 to 11 8342 6493 138 115 0002-34 34 MLL Exon 25 4249 1775 139 129 0002-48 11 MLL Exon 25 4249 3568 140 6 0001-6 6 MLL Exon-intron 5 to 11 8342 3104 141 51 0001-51 9 MLL Exon-intron 5 to 11 8342 8087 142 121 0002-40 3 MLL Exon 25 4249 641 143 159 TrpnX-M 1 TrpnX-M 489 165 144 143 BioDn-3 1 BioDn-3 286 73 145 60 0001-60 18 MLL Exon-intron 5 to 11 8342 8175 146 133 0002-52 15 MLL Exon 25 4249 1748 147 80 0001-80 7 >MLL Exon-intron 5 to 11 8342 1493 148 70 0001-70 28 MLL Exon-intron 5 to 11 8342 5791 149 1 0001-1 1 MLL Exon-intron 5 to 11 8342 6881 150 87 0002-6 6 MLL Exon 25 4249 1033 151 9 0001-9 9 MLL Exon-intron 5 to 11 8342 5126 152 40 0001-40 40 MLL Exon-intron 5 to 11 8342 2278 153 156 ThrX-M 1 ThrX-M 571 485 154 104 0002-23 23 MLL Exon 25 4249 3201 155 96 0002-15 15 MLL Exon 25 4249 2073 156 154 ThrX-3 1 ThrX-3 463 135 157 68 0001-68 26 MLL Exon-intron 5 to 11 8342 3257 158 69 0001-69 27 MLL Exon-intron 5 to 11 8342 435 159 59 0001-59 17 MLL Exon-intron 5 to 11 8342 3818 160 36 0001-36 36 MLL Exon-intron 5 to 11 8342 8055 161 29 0001-29 29 MLL Exon-intron 5 to 11 8342 8293 162 131 0002-50 13 MLL Exon 25 4249 3121

TABLE 3 SEQ ID Probe ID NO Probe Sequence 0002-12 1 GTCCTGGCCCGTCTCAGATTTCCAATGCAGCTGTCCAGACCACTCCACCC 0001-66 2 AAAGAATCCTGAATAAATGGGGACTTTCTGTTGGTGGAAAGAAATATAGA 0002-32 3 TCCAACTCCTGAAGGCCACATGACTCCTGATCATTTTATCCAAGGACACA 0001-24 4 ATTCAGTCTACAAGTGCCAGGGGTCTACTGTATCCTCTTTTCCGTCTTAA 0001-32 5 AGGCCTTATTTAGGTTTGACCAATTGTCCCAATAATTCCTTTATGGCAAA 0002-25 6 TGAGTTCCAAGAGCTCAGAGGGATCTGCACATAATGTGCCTTACCCTGGA 0001-54 7 AGAGCAGGTTACAAGATAATATATAAAGCACAATCCCATCTTAGTTTGCA LysX-M 8 CGCATACGCATGACTACATTACAACGGGCCAGGAAGATTCAAAGTTTGGT 0002-37 9 AACCAGAACATGCAGCCACTTTATGTTCTCCAAACTCTTCCAAATGGAGT 0002-31 10 ACAGTCACTTGGATGGATCTTCATCTTCAGAAATGAAGCAGTCCAGTCCT 0002-29 11 AGTTCTACACCCAGTGTGATGGAGACAAATACTTCAGTATTGGGACCCAT TrpnX-3 12 AAATATTGCGGTATTCGGTCACTAAAGGATTTGCAGCTTGCGGCGGAATC 0002-5 13 CCACCTCACATCAGGGTCTGTGTCTGGCTTGGCATCCAGTTCCTCTGTCT BioB-3 14 GAACGAACAGACTCAGGCGATGTGCTTTATGGCAGGCGCAAACTCGATTT 0001-30 15 CCACAGGATCAGAGTGGACTTTAAGGTAAAGGTGTTCAGTGATCATAAAG 0001-61 16 AGGCATCCTGCTTCTTTGTACCCCAGGAAGTACATAAATGATTGATCTGG 0001-39 17 AGTCTGTTTTGTTGGTATTTAGCAGGTACTATTCCCTGTTTAAACCAGCT 0001-81 18 CCTGTACTCCCAGCTACTCAGGAGAGTGAGCCAGGAGAATCGCGTGAACC 0001-12 19 TCCTACATCCTTTACAGTTCTTAAATTCCTGGCAGATACCTCTTTGCCTT 0001-2 20 TGGAGTGTAATAAGTGCCGAAACAGCTATCACCCTGAGTGCCTGGGACCA 0001-73 21 AAATCACCCTTCCCTGTATTCACTATTTTTATTTATTATGGATAAAGAGA 0002-49 22 ACTCTAGGAATAATGTTTCCTCAGTCTCCACCACCGGGACCGCTACTGAT 0002-4 23 CTCCATCCTCTCCATCTTCTGGACAGCGGTCAGCAAGCCCTTCAGTGCCG 0002-22 24 AGGACAGAAACCTAATGCTTCCAGATGGCCCCAAACCTCAGGAGCATGGC 0001-46 25 AAGCACTGATGTCTCAAACAGCATTTGAAAGCAGGAAATGTATGATTTGA 0001-57 26 AATCCCATCTCTCTTAAATTCAGTCTTTATTAGAGTTCTGATCTTTCTGT 0001-31 27 TCGGCCTGAATCCAAACAGGCCACCACTCCAGCTTCCAGGAAGTCAAGCA BioB-5 28 TTCGATCCTCGTCAGGTGCAGGTCAGCACGTTGCTGTCGATTAAGACCGG 0001-52 29 AGAAAATCCAAGCTAGGTTGAAATCTGAATGTTGAGCAGTCAGTGAGACA 0001-72 30 TGTACCACCTTTACAATGAGGAAGGAAAAAGTAGCACAATTTTAAATAGG 0001-20 31 TGCCAGTAAATGTGAAATGGGGTACTAAGTAATAGGTGTTGGCTGAAGGT 0001-13 32 ACTGCACTCCTAAAGCATGACCAGTGCTTGATAAACTCTCCTCCATGCGA ThrX-5 33 AGAAATCACCGATTGCCCTTGTCAACTCAGTCAACCCTTACCGCATTGAA 0002-28 34 GGAGTCCCACTGTCCCCAGCCAGAATCCCAGTAGACTAGCTGTTATCTCA 0001-21 35 ACTGAGTGCCTTTGGCAGGAAATAAATCTATCTCAATGCGTTAATTGGGA 0001-45 36 ATTAAGAGTGTGGTTGGATTATGGGTGACCTTTATTTGTTTCTCTGGTTT 0001-65 37 TTTCCGTCTTAATACAGTGCTTTGCACCCATATATATGCCACCCACAGGA 0001-34 38 TGGAAAGATGTCCATGACATATCACTGAGTGAAAAGAGCAGGTTACAAGA 0001-15 39 CCCACATGTTCTAGCCTAGGAATCTGCTTATTCTAAAGGCCATTTGGCGT 0002-38 40 GCCACAGCGGCAGGCACATCAACAATAAGCCAGGATACTAGCCACCTCAC 0001-35 41 TGGAAAGGACAAACCAGACCTTACAACTGTTTCGTATATTACAGAAAACG 0001-18 42 TTTCCACTGGTATTACCACTTTAGTACTCTGAATCTCCCGCAATGTCCAA 0002-18 43 CCCAGGTATCCAACTTTACCCAGACCGTAGACGCTCCTAATAGCATGGGA 0002-45 44 CGTGACAACTGGTGAGGAAGGAAACTTCAAGCCAGAGTTTATGGATGAGG 0002-13 45 CAGTCTCCACCACCGCGACCGCTACTGATCTTGAATCAAGTGCCAAAGTA 0001-7 46 TGCCCCAAAGAAAAGCAGTAGTGAGCCTCCTCCACGAAAGCCCGTCGAGG 0002-24 47 ACCGGCACCCCTGTTACCACAGAGTGTGGGAGGAACTGCTGCCACAGCGG 0002-19 48 TCAGCCTCTGAAAATCCAGGAGATGGTCCAGTGGCCCAACCAAGCCCCAA 0001-41 49 AGCAGGAAATGTATGATTTGAAGTCTTCAGTTCAAGAAAATCAGCTCTCT 0002-35 50 AGCTCCTGAAATCAGATTCAGACAATAACAACAGTGATCACTGTGGGAAT 0002-36 51 CTAGAGAACTGAATGTTAGTAAAATCGGCTCCTTTGCTGAACCCTCTTCA DapX-M 52 TGTCTCGGCATTAATCCGTTTATGTGATCTGTATTCCATTCCGCTCGCCA LysX-3 53 GATCGAACCGGGCCGTTCTCTCGTGGGAGACGCAGGCACAACTCTTTATA 0001-5 54 ACCTCCGGTCAATAAGCAGGAGAATGCAGGCACTTTCAACATCCTCAGCA LysX-5 55 CAACATGGTCATTTAGAAATCGGAGGTGTGGATGCTCTCTATTTAGCGGA 0001-67 56 TGTATATCAAAGCCTCTTCATCTATAAGGAGCTCTTACCAATTAATAAGA 0001-53 57 TTTACTTAGTCTGTCTTTAGCATTTAATTGGGTGTAATCAGTTGCCTATT 0002-7 58 CCCTACAAGTAGTGCGTCAGTTCCAGGACACGTCACCTTAACCAACCCAA 0001-27 59 GAAATAAATACATGTTGGGTGGCAGGGGGAGGTGAAGGGAGGGTGTCTGT PheX-M 60 TTTAATACATGAACAGCCTTTCCCAATCGTCGGTGAAATGACGTTGCCGA PheX-5 61 GCAGATTCAGTAGCAGATGCCGTTCAAAAGGTCGATTTAAGTAGAAGTGC 0001-74 62 AACTTCAAGTTTAGGCTTTTAGCTGGGCACGGTCGCTCACGCTGGTAATC 0002-2 63 AGCAGACGAACACTATCAGCTTCAGCATGTGAACCAGCTCCTTGCCAGCA 0001-75 64 CAGCAGTTCAAGACCAGCCTGGGCAACATAGCAAGACCCTGTCTTTATTT TrpnX-5 65 AAACCCTTACTGCCGGTGAGGCTGAAACGCTGATGAATATGATGATGGCA 0002-20 66 AAGGATTGCTACCCATGTCTCATCACCAGCACTTACATTCCTTCCCTGCA 0001-44 67 GCCCTTTCTTCACAGGTCAGTCAGTACTAAAGTAGTCGTTGCCAGCATCT 0001-76 68 TCAGGAGGCTGAGATAGAAGGATTGTCTTGAGCCCACGAATTCAAGGCTG 0002-33 69 AGTCTGTTAGATTTGGGGTCACTTAATACTTCATCTCACCGAACTGTCCC 0001-19 70 GGAAACCAAGGATGACTGTGCTTAGAGTATTGCTTTCTTTCTTGATTTGT 0001-22 71 CTCTCCACAGGAGGATTGTGAAGCACAAAATGTGTGGGAGATGGGAGGCT 0002-54 72 CCTGAAGGCCACATGACTCCTGATCATTTTATCCAAGGACACATGGATGC 0002-9 73 GCATTGGCTCCAGGCGTCACAGTACCTCTTCCTTATCACCCCACCGCTCC 0001-47 74 ACAAATGGAAAGGACAAACCAGACCTTACAACTGTTTCGTATATTACAGA BioDn-5 75 CGCAAGAGGGCAGACCGATAGAATCATTGGTAATGAGCGCCGGATTACGC 0002-42 76 ACTATCAGAATCTTCCAGTACAGGACAGAAACCTAATGCTTCCAGATGGC 0001-79 77 GCAGTGAGCCGAGATTGCATCATTGCACTCTAGCCTGGACAACAGAGCTA 0002-41 78 CACTAGAACAGTGATTTCTTCAGGTGGAGAGCAACGACTGGCATCCCATA 0002-30 79 TTGACTCCTGAGTATATGGGCCAACGACCATGTAACAATGTTTCTTCTGA 0002-39 80 GAGCTACCATCTGATCTGTCTGTCTTGACCACCCGCAGTCCCACTGTCCC 0001-50 81 TGTTTCTCTGCCATTTCTCAGGGATGTATTCTATTTTGTAGGGAAAAGCC 0001-56 82 TTCTGATCTAAATTCTTTATAGTTGTACATAGCAATCTCACAGGGTTCCT 0001-10 83 AGCAGGTGGGTTTAGCGCTGGGACAGCTTTGGACAGTGTTGTTAGGTCAC 0002-10 84 TCCCACACCTCCATTTGAGAGGGCAAAGGAATGATCGAGACCAACACACA 0002-43 85 AGTGATGACTGTGGGAATATCCTGCCTTCAGACATTATGGACTTTGTACT 0001-37 86 TGCCAGTGGACTACTAAAACCCAAAGTATATAAGAAGGGTATGGTTGATT 0001-8 87 GCCTCAGCCACCTACTACAGGACCGCCAAGAAAAGAAGTTCCCAAAACCA 0002-26 88 GATGGTGTTGATGATGGGACAGAGAGTGATACTAGTGTCACAGCCACAAC 0001-25 89 AAAGGTGAGGAGAGATTTGTTTCTCTGCCATTTCTCAGGGATGTATTCTA BioB-M 90 GGGATCAAAGTCTGTTCTGGCGGCATTGTCGGCTTAGGCGAAACCGTAAA 0001-38 91 TGGAAGGATTCACACCAAAATATTAAGAGTGTGGTTGGATTATGGGTGAC 0001-26 92 ACCCGAAAGTCCATCTATAGGGAGCATGGGTTAAAATAAGCATAGGGCAT 0002-17 93 AGAGCAAGGTCATGGCAACAATCAGGATTTAACTAGGAACAGTAGCACCC BioC-5 94 CGCCAATGCTTGTTCAGGCACGCCAGAAGGATGCCGCAGACCATTATCTG 0002-16 95 TCAGGTGGAGAGGAACGACTGGCATCCCATAATTTATTTCGGGAGGAGGA 0002-21 96 ACTGCTGCAATAACAGCGGCATCTAGCATCTGTGTGCTCCCCTCCACTCA 0001-14 97 TTCCTATCCATCCTGAGGAGTATCAGAGGAAGTAATTCCTTCACATGGAA 0001-16 98 TCCCATGTTCTTACTATAGTTTGTCTATTGCCAAGTCTGTTGTGAGCCCT PheX-3 99 CGTTTGATGATGTATTGATTCCAGGGGCCATGCAGGAGCTTGAAGCACTC 0001-42 100 TTTTTGGAGTATGTACCACCTTTACAATGAGGAAGGAAAAAGTAGCACAA 0001-55 101 CTATGAATTGAACAACTAGGTGAGCCTTTTAATAGTCCGTGTCTGAGATT 0001-64 102 TGAGTGTCAAAGACTTTAAATAAAGAAAATGCTACTACCAAAGGTGTTGA 0002-46 103 GAAGTATGTGCCCAATTCTACTGATAGTCCTGGCCCGTCTCAGATTTCCA 0002-53 104 GGGCTTACCCCACTCTATGGAGTAAGATCCTATGGTGAAGAAGACATTCC 0002-3 105 AGAATCCAGCCAGAGGACAGACCTCAGTACCACAGTAGCCACTCCATCCT 0002-27 106 ACCTTGAAGCTATCTGGAATGAGCAACAGATCATCCATTATCAACGAACA 0001-78 107 CCGGTTGTGGTAGTGGGTGCTTGGTAATCCTAGCTACTTGGGAGCCTGAG 0001-63 108 ATGTCACACTAATTTTATGCTTTTCATCCTTATTTTCCATCCAAAGTTGT 0002-57 109 CCCAACATCATAAAAAGATCTAAATCTAGCATCATGTATTTTGAACCGGC BioC-3 110 CGAACGTCATCAGGCGTGGCAGGCGGTGCACGAGCGTCCGCATGCTAATC 0001-4 111 TGGGCCTCTGTATCAGTGGGTTCTGTATCCCTGGACTCAACCAACCTTGG 0001-49 112 CCCTCACCCAAATTCCCTAAGTGTTAATATGTTTCTCTGTGTGTATATAT 0001-77 113 CACTTTGGGAAGCCGAAGCAGGCAGATCACTTGAGGTCAGGAGTTGGAGA 0001-28 114 AAGTACCCCTCACCCAAATTCCCTAAGTGTTAATATGTTTCTCTGTGTGT 0002-55 115 AAACACTTCCACCTCTTCAAATTTGCAAAGGACAGTGGTTACTGTAGGCA 0002-11 116 ATTCCATTCTACAGCAGCTCAACTGGGAAGAAGCGAGGCAAGAGATCAGC 0002-8 117 AGCACAAAGTTTCCCATTTGCGGACCAGTTCTTCTGAAGCACACATTCCA 0002-47 118 TAACTTCACACCCTCCCAGCTTCCTAATCATCCAAGTCTGTTAGATTTGG 0001-3 119 ACCACCAGAATCAGGTGAGTGAGGAGGGCAAGAAGGAATTGCTGACCCAC 0001-11 120 CCCTTTCTTCACACGTCAGTCAGTACTAAAGTAGTCGTTGCCAGCATCTG 0001-62 121 TGGAAACAACCCGAAAGTCCATCTATAGGGAGCATGGGTTAAAATAAGCA 0001-58 122 TGTGAAGGCAAATAGGGTGTGATTTTGTTCTATATTCATCTTTTGTCTCC 0002-44 123 TCAGAACTCCTGAATCTTGGTGAAGGATTGGGTCTTCACAGTAATCGTGA DapX-5 124 GGCAGAACGAACACCACATTTTGACCTTGTAGGGGCCATAGACCATACAT CreX-5 125 AAACTATCCAGCAACATTTGGGCCAGCTAAACATGCTTCATCGTCGGTCC 0002-14 126 TGCCTACAACAGAACCTGTGGATAGTAGTGTCTCTTCCTCTATCTCAGCA DapX-3 127 TGATCAGGCAATTCGGTCAATTGTCACTGTCATCAGAATCTGTCGGCCAA 0002-51 128 CTGCAATAACAGCGGCATCTAGCATCTGTGTGCTCCCCTCCACTCAGACT 0002-56 129 GCAATCCTCCTTCAGGCCTGCTTATTGGGGTTCAGCCTCCTCCGGATCCC 0001-71 130 TGCCATTTGAAGTTATTACTAGCAAAATTACAAATTATTGCCTACTATTC 0001-48 131 ACAACTTATTGTTCTAAGTGCAGAAGTTCAGATATCATTGAGACTGAGAA 0001-33 132 CACTATGAATTGAACAACTAGGTGAGCCTTTTAATAGTCCGTGTCTGAGA CreX-3 133 CTAAGGATGACTCTGGTCAGAGATACCTGGCCTGGTCTGGACACAGTGCC 0002-1 134 TCCTTGGAAGCTCAGCTCAGCTCATTGGAGTCAAGCCGCAGAGTCCACAC 0001-43 135 GCCTCTGTATCAGTGGGTTCTGTATCCCTGGACTCAACCAACCTTGGATT 0001-23 136 TGTTATATGCAAATGCTGCACCATTTTGTCTAGGGACTTGGGCATCCATG 0001-17 137 TCCCATAGCTCTTTGTTTATACCACTCTTAGGTCACTTAGCATGTTCTGT 0002-34 138 ACCTGAAGATGCTGGGGAGAAAGAACATGTCACTAAGAGTTCTGTTGGCC 0002-48 139 GTGCGTCAGTTCCAGGACACGTCACCTTAACCAACCCAAGGTTGCTTGGT 0001-6 140 AGGGTGGTTTGCTTTCTCTGTGCCAGTAGTGGGCATGTAGAGGTAAGGCA 0001-51 141 TTATAGAGAACCACCATGTGACTATTGGACTTATGTAACTTGTATTACAA 0002-40 142 AGACCAACACACAGATTCTACCCAATCAGCAAACTCCTCTCCAGATGAAG TrpnX-M 143 CTCACTGAAGGAAGGAACGGAGCTGGCGTTAGAGACGATTACAACCGGAG BioDn-3 144 CCGGTGATACTGGTAGTTGGTGTGAAACTCGGCTGTATTAATCACGCGAT 0001-60 145 CCCTCATTACTAGGAAATCATCTCAGGAGAGAAATTAAATCTATAAATGG 0002-52 146 TGGAACAGAGAACTTAAAGATTGATAGACCTGAAGATGCTGGGGAGAAAG 0001-80 147 AGGAGATCGAGACCATCCTGGCTAACACGGTGAAACCCTGTCTCTACTAA 0001-70 148 TGAGAACAAGTTGGAGACATAAACCATTTTACCTCTAAATATTTTAGTGT 0001-1 149 TGTCGTCGCTGCAAATTCTGTCACGTTTGTGGAGGGCAACATCACGCTAC 0002-6 150 AGGAATTACAGGCACCACGGAAACGCACAGTCAAAGTGACACTGACACCT 0001-9 151 AGTCAGTGAGACACAAACTAGCTAAGAAAGTCAACCCTGCCCACTTGCCA 0001-40 152 TGCATTATTATCTGTTGCAAATGTGAAGGCAAATAGGGTGTGATTTTGTT ThrX-M 153 TACAGATAACCTGATCTACCAAGTGGCTAAACGGACCGCAGATTTGTACG 0002-23 154 AAACTTGCTCCCTCTAGTACCCCTTCAAACATTGCCCCTTCTGATGTGGT 0002-15 155 ACTCCATCCATGCAGGCTTTGGGTGAGAGCCCAGAGTCATCTTCATCAGA ThrX-3 156 AAGTGCTGACAAGAGACGCGAGAGACGTGCTTCCGAAGGAGTTTCCATAT 0001-68 157 ACCAGCTAAAGAAATGTTTTGAAGTATTTTAGAGATTTTAGGAAGGAATC 0001-69 158 AAACAGTTAAATTGGAGGTATTGTTTTAATTTCCTGTTCGAAGCCTAGAG 0001-59 159 GCACTTCAAACACTTATGGATATAATTAGATAAATTGGCAAATCTGTAGA 0001-36 160 AAGTCTGGGTGAGTTATACACATGATGCTCTTTTATAGAGAACCACCATG 0001-29 161 TTCTTTTCTAGATCTGTACCAAGTGTGTTCGCTGTAAGAGCTGTGGATCC 0002-50 162 ACCTCAGTACCACAGTAGCCACTCCATCCTCTGGACTCAAGAAAAGACCC

TABLE 4 SEQ ID NO Sequence 163 CACTTTGCACTGGAACTTACAACACCCGAGCAAGGACCCGACTCTCCCGA 164 GACACTTCCCCGCCGCTGCCAGGACCCGCTTCTCTGAAAGGCTCTCCTTG 165 CCAGCCAGCGGTCCGCAACCCTTGCCGCATCCACGAAACTTTGCCCATAG 166 CTTTGCACTGGAACTTACAACACCCGAGCAAGGAC 167 CAACCCTTGCCGCATCCACGAAACTTTGCCCATAG 168 CTCAACGTTAGCTTCACCAACAGGAACTATGACCTCGACTACGACTCGGT 169 TTAGCTTCACCAACAGGAACTATGACCTCGACTACGACTC 170 CGAGACCTTCATCAAAAACATCATCATCCAGGACTGTATG 171 GTATTTCTACTGCGACGAGGAGGAGAACTTCTACCAGCAG 172 CGTTTATAGCAGTTACACAGAATTTCAATCCTAGTATATAGTACCTAGTA 173 GAGACTGAAAGATTTAGCCATAATGTAAACTGCCTCAAATTGGACTTTGG 174 CCTTCTAACAGAAATGTCCTGAGCAATCACCTATGAACTTGTTTCAAATG 175 TTACACAATGTTTCTCTGTAAATATTGCCATTAAATGTAAATAACTTTAA 176 CATCTCCGTATTGAGTGCGAAGGGAGGTCCCCCTATTATTATTTGACACC 177 GCCACTCCAGCCGGCGAGAGAAAGAAGAAAAGCTGGCAAAAGGAGTGTTG 178 GTATTGAGTGCGAAGGGAGGTGCCCCTATTATTATTTG 179 CTTGTATTTATGGAGGGGTGTTAAAGCCCGCGGCTGAG 180 AAAACTTTGTGCCTTGGATTTTGGCAAATTGTTTTCCTCACCGCCACCTC 181 GAGATAGCAGGGGACTGTCCAAAGGGGGTGAAAGGGTGCTCCCTTTATTC 182 AAAACTTTGTGCCTTGGATTTTGGCAAATTGTTTTCCTC 183 GGAATGGTTTTTAAGACTACCCTTTCGAGATTTCTGCCTTATGAATATAT 184 TTTTATCACTTTAATGCTGAGATGAGTCGAATGCCTAAATAGGGTGTCTT 185 CTCCCATTCCTGCGCTATTGACACTTTTCTCAGACTAGTTATGGTAACTG 186 TTATCTTACAACTCAATCCACTTCTTCTTACCTCCCGTTAACATTTTAAT 187 GATCTTCTCAGCCTATTTTGAACACTGAAAAGCAAATCCTTCCCAAAGTT 188 TTTCATTGGCAGCTTATTTAACGGGCCACTCTTATTAGGAAGGAGAGATA 189 CATTAAGTCTTAGGTAAGAATTGGCATCAATGTCCTATCCTGGGAAGTTG 190 CATTTCCAGTAAAATAGGGAGTTGCTAAAGTCATACCAAGCAATTTGCAG 191 ATCATTTGCAACACCTGAAGTGTTCTTGGTAAAGTCCCTCAAAAATAGGA 192 AATCTGGTAATTGATTATTTTAATGTAACCTTGCTAAAGGAGTGATTTCT 193 GATAATTTTGTCCAGAGACCTTTCTAACGTATTCATGCCTTGTATTTGTA 194 GTCTAGAAAAACCTGCCAAATATGATGACATCAAGAAGGTGGTGAAGCAG 195 TACACTCAGCACCAGGTGCTCTCCTCTGACTTCAACAGCGACACCCACTC 196 CTGGGGCTGGCATTGCCCTCAACGACCACTTTGTCAAGCTCATTTCCTGG 197 CACTGCCAACGTGTCAGTGGTGGACCTGACCTGCCGTCTAGAAAAACCTG 198 AAATATGATGACATCAAGAAGGTGGTGAAGCAGGCGTCGGAGGGCCCCCT 199 CATTGCCCTCAACGACCACTTTGTCAAGCTCATTTCCTG 200 CTACACTGAGCACCAGGTGGTCTCCTCTGACTTCAACAG 201 TGATGCTTTTCCTAGATTATTCTCTGGTAAATCAAAGAAGTGGGTTTATG 202 CTGTCCAGTTAATTTCTGACCTTTACTCCTGCCCTTTGAGTTTGATGATG 203 TATTCTCTGGTAAATCAAAGAAGTGGGTTTATGGAGGTCCTCTTCTGTCC 204 GATTTCTGGAAAAGAGCTAGGAAGGACAGGCAACTTGGCAAATCAAAGCC 205 CACCGCACCCTGGTCTGAGGTTAAATATAGCTGCTGACCTTTCTGTAGCT 206 CAGAAGCTGAGTCATGGGTAGTTGGAAAAGGACATTTCCACCGCAAAATG 207 ATTATTCTCTGGTAAATCAAAGAAGTGGGTTTATGGAGGTCC 208 CTGTCCAGTTAATTTCTGACCTTTACTCCTGCCCTTTGAG 209 CCTGGTCTGAGGTTAAATATAGCTGCTGACCTTTCTGTAG 210 GAAAAGAGCTAGGAAGGACAGGCAACTTGGCAAATCAAAG 211 GGGAGAAGCTGAGTCATGGGTAGTTGGAAAAGGACATTTC 212 GAAAGACAAGGAAGGAACACCTCCACTTACAAAAGAAGATAAGACAGTTG 213 CAAAGTATGCCAAAGAAGGTCTTATTCGCAAACCAATATTTGATAATTTC 214 AAATAACACATGGAAAGGACATTTCAGAGTTACCAAAGGGAAACAAAGAA 215 GCTGTCAAAACCAAAATACTTATAAAGAAAGGGAGAGGAAATCTGGAAAA 216 CTCACTCTAGAATATTTGAGTCTGTAACCTTGCCTAGTAATCGAACTTCT 217 GTTTATAGAGGATGAGGATTATGACCCTCCAATTAAAATTGCCCGATTAG 218 AAATGAGAGTAATGATAGGAGAAGCAGAAGGTATTCAGTGTCGGAGAGAA 219 GACACATTACATAAACTAGGAGAACTTAAGACTACGAACAGAATATTTGG 220 TGTGTATGAATTCCTTCTTTCAAGTGAACTGATACTAGATTTATTTAAGA 221 CTTAATTATAAAGTTGGATGTCATTTGAGAAACTCTGGGAATTGGAAGTA 222 TAAATAAATTCTTATTCAGCTCCTCGAACCAATAATTACTTTCCAGTAGG 223 TTAAACCGAAATCAGGAGTAGTTGTGTAAAGAACTTATTGGTAATGATGG 224 TGCTTGTGGATACATTGTAACAAATGCTTATAAATCATTTCCAAACTAAT 225 CCAAGGAAAACAGTAGGTGTGGTATCAATATAGGAAACAAATAAGTATTT 226 TAATCATTAAGAAATACTAATTTAAGTATGGCAAAGGAAAGCACAGGTGC 227 TTTCTAGCCTATTCAAATCAATCTGGTCATTTATGGTACTTTCCTATTAG 228 CAATAACAGCATACATTTCTTGACTGGTTGAATTTCATTAACTATTTGGC 229 GCATTTGTGTTTCTTAGGTGACAGTTGCTAGGTAGAATTGAATTAAATAT 230 TTAACATTTCTCAATATCAGGCAGAGATCATATTTAAACAGTTTCAATCT 231 CTGTGTATTTGACTGTGCTTGGGTATATTATACTTTTCTTACTGATTGAG 232 AAACTGACTTTATGGAGACATAACCCTGTTTACCTTTAGAAAGAAGGAAG 233 AAAATAAACCCTGTGATTGTGATGCTTAACTTAATTTTCTACAGTGAATC 234 GAATAATTTGGGACATTGCCAGGAATCACAATAGTTTACTATCTGAAGTA 235 ATGAAATTGAGGGATAGATACAGTAAGTGAGTTGTCTAAGATTACATAGT 236 GCTTTCCAAGTGATTTCACATAAATTATTTATTCCTTACAGTGCTCAAAT 237 ATTGTGATAAGATTTTATATTAATTGTGCTGTTAGGAGTTTTGGCTGTTT 238 TTTCATGAGAAGTCATTCAGTATCATTAAGTATGCTGATTTGTCTCCTTT 239 AGAAATTTATTTGGGGTTCAGATTCACATGTTGTAGGTTAGTTATATACT 240 GGATCTCAAATGTACAGAAATCACATCTAAATGTCAATTCCTGAGTTAAG 241 CTGTATGTTTCTGCCATTATACTTATTTGCTTACCTGATTTAAAGTTGTC 242 TATTAATCAGTTTCTTTAAATAGACCATCTTTCTTGACAACTTGTGCAAA 243 GTATCTATAGTTTGAAATTAGGACTATCCTCTGTGTACTATGCACCAAAG 244 TCTGTAATAAAGCTGTATGGCTGGGTCCATTTATTTCAATATTAGTTATT 245 CCAACTATAACTGAAAATAGGATGCTTCCCTAAGTTTTAGTAAAGGATTT 246 GTCTTTGAAGAGGAGAATTTCAGCCTTTTCTTAAATAGTCCAATACTTTA

TABLE 5 Probe Probe position SEQ Position within ID Sequence within gene NO Name Length fragment sequence 163 CMYC_Exon1_NT008046.14[1_to_366]_1 366 195 195 164 CMYC_Exon1_NT008046.14[1_to_366]_2 366 273 273 165 CMYC_Exon1_NT008046.14[1_to_366]_3 366 134 134 166 CMYC_Exon1_NT008046.14[1_to_366]_short_1 366 197 197 167 CMYC_Exon1_NT008046.14[1_to_366]_short_2 366 149 149 168 CMYC_Exon2_NT008046.14[1991_to_2762]_1 772 22 2012 169 CMYC_Exon2_NT008046.14[1991_to_2762]_short_1 772 29 2019 170 CMYC_Exon2_NT008046.14[1991_to_2762]_short_2 772 378 2368 171 CMYC_Exon2_NT008046.14[1991_to_2762]_short_3 772 78 2068 172 CMYC_Exon3_NT008046.14[4141_to_5168]_1 1028 863 5003 173 CMYC_Exon3_NT008046.14[4141_to_5168]_2 1028 671 4811 174 CMYC_Exon3_NT008046.14[4141_to_5168]_3 1028 583 4723 175 CMYC_Exon3_NT008046.14[4141_to_5168]_4 1028 808 4948 176 CMYC_Intron1_NT008046.14[1110_to_1410]_1 301 158 1267 177 CMYC_Intron1_NT008046.14[1110_to_1410]_2 301 252 1361 178 CMYC_Intron1_NT008046.14[1110_to_1410]_short_1 301 165 1274 179 CMYC_Intron1_NT008046.14[1110_to_1410]_short_3 301 211 1320 180 CMYC_Intron1_NT008046.14[1551_to_1880]_1 330 54 1604 181 CMYC_Intron1_NT008046.14[1551_to_1880]_2 330 243 1793 182 CMYC_Intron1_NT008046.14[1551_to_1880]_short_1 330 54 1604 183 CMYC_Intron1_NT008046.14[367_to_1110]_1 744 222 588 184 CMYC_Intron1_NT008046.14[367_to_1110]_2 744 37 403 185 CMYC_Intron1_NT008046.14[367_to_1110]_4 744 89 455 186 CMYC_Intron2_NT008046.14[2763_to_3400]_1 637 262 3024 187 CMYC_Intron2_NT008046.14[2763_to_3400]_4 637 527 3289 188 CMYC_Intron2_NT008046.14[2763_to_3400]_5 637 55 2817 189 CMYC_Intron2_NT008046.14[3419_to_3709]_1 291 145 3563 190 CMYC_Intron2_NT008046.14[3419_to_3709]_2 291 1 3419 191 CMYC_Intron2_NT008046.14[3419_to_3709]_3 291 53 3471 192 CMYC_Intron2_NT008046.14[3996_to_4140]_1 145 78 4073 193 CMYC_Intron2_NT008046.14[3996_to_4140]_2 145 21 4016 194 GAPDH_Exon8_NT009759.15[3050_to_3462]_1 413 217 3266 195 GAPDH_Exon8_NT009759.15[3050_to_3462]_2 413 300 3349 196 GAPDH_Exon8_NT009759.15[3050_to_3462]_3 413 364 3413 197 GAPDH_Exon8_NT009759.15[3050_to_3462]_4 413 182 3231 198 GAPDH_Exon8_NT009759.15[3050_to_3462]_5 413 234 3283 199 GAPDH_Exon8_NT009759.15[3050_to_3462]_short_2 413 374 3423 200 GAPDH_Exon8_NT009759.15[3050_to_3462]_short_3 413 299 3348 201 GAPDH_Intron2_NT009759.15[328_to_1959]_1 1632 582 909 202 GAPDH_Intron2_NT009759.15[328_to_1959]_2 1632 1196 1523 203 GAPDH_Intron2_NT009759.15[328_to_1959]_3 1632 599 926 204 GAPDH_Intron2_NT009759.15[328_to_1959]_5 1632 1049 1376 205 GAPDH_Intron2_NT009759.15[328_to_1959]_6 1632 928 1255 206 GAPDH_Intron2_NT009759.15[328_to_1959]_7 1632 693 1020 207 GAPDH_Intron2_NT009759.15[328_to_1959]_short_1 1632 597 924 208 GAPDH_Intron2_NT009759.15[328_to_1959]_short_2 1632 1196 1523 209 GAPDH_Intron2_NT009759.15[328_to_1959]_short_3 1632 936 1263 210 GAPDH_Intron2_NT009759.15[328_to_1959]_short_4 1632 1057 1384 211 GAPDH_Intron2_NT009759.15[328_to_1959]_short_5 1632 691 1018 212 MLL_Exon3_AP001267.4[1_to_2654]_10 2654 491 491 213 MLL_Exon3_AP001267.4[1_to_2654]_2 2654 1402 1402 214 MLL_Exon3_AP001267.4[1_to_2654]_3 2654 199 199 215 MLL_Exon3_AP001267.4[1_to_2654]_4 2654 2361 2361 216 MLL_Exon3_AP001267.4[1_to_2654]_5 2654 1618 1618 217 MLL_Exon3_AP001267.4[1_to_2654]_6 2654 779 779 218 MLL_Exon3_AP001267.4[1_to_2654]_7 2654 1058 1058 219 MLL_Intron1_AP001267.4[10100_to_12332]_1 2233 339 10438 220 MLL_Intron1_AP001267.4[10100_to_12332]_10 2233 1609 11708 221 MLL_Intron1_AP001267.4[10100_to_12332]_2 2233 1809 11908 222 MLL_Intron1_AP001267.4[10100_to_12332]_3 2233 988 11087 223 MLL_Intron1_AP001267.4[10100_to_12332]_4 2233 803 10902 224 MLL_Intron1_AP001267.4[10100_to_12332]_6 2233 1270 11369 225 MLL_Intron1_AP001267.4[10100_to_12332]_7 2233 1147 11246 226 MLL_Intron1_AP001267.4[10100_to_12332]_8 2233 391 10490 227 MLL_Intron1_AP001267.4[12670_to_14434]_1 1765 401 13070 228 MLL_Intron1_AP001267.4[12670_to_14434]_2 1765 1204 13873 229 MLL_Intron1_AP001267.4[12670_to_14434]_3 1765 876 13545 230 MLL_Intron1_AP001267.4[12670_to_14434]_4 1765 1036 13705 231 MLL_Intron1_AP001267.4[12670_to_14434]_6 1765 1277 13946 232 MLL_Intron1_AP001267.4[12670_to_14434]_7 1765 89 12758 233 MLL_Intron1_AP001267.4[12670_to_14434]_8 1765 454 13123 234 MLL_Intron1_AP001267.4[12670_to_14434]_9 1765 625 13294 235 MLL_Intron1_AP001267.4[27613_to_29591]_1 1979 994 28606 236 MLL_Intron1_AP001267.4[27613_to_29591]_2 1979 939 28551 237 MLL_Intron1_AP001267.4[27613_to_29591]_3 1979 1304 28916 238 MLL_Intron1_AP001267.4[27613_to_29591]_7 1979 478 28090 239 MLL_Intron1_AP001267.4[27613_to_29591]_9 1979 1417 29029 240 MLL_Intron1_AP001267.4[30450_to_31832]_10 1383 844 31293 241 MLL_Intron1_AP001267.4[30450_to_31832]_2 1383 1027 31476 242 MLL_Intron1_AP001267.4[30450_to_31832]_3 1383 550 30999 243 MLL_Intron1_AP001267.4[30450_to_31832]_6 1383 1095 31544 244 MLL_Intron1_AP001267.4[30450_to_31832]_7 1383 1190 31639 245 MLL_Intron1_AP001267.4[30450_to_31832]_8 1383 680 31129 246 MLL_Intron1_AP001267.4[30450_to_31832]_9 1383 21 30470

TABLE 6 Chromosomal Location Structure/Function Occurrence Reference Nuclear transcription factors LAF-4 2q11 transcription factor ALL GenBank Accession No. AF422798 (Huret, 2001) AF4 4q21 transcription factor ALL (Nakamura, 1993) (MLLT2, t-ALL (Raffini, 2002) FEL) AML AF5α 5q12 (Taki, 1996) AF5q31 5q31 AF6q21 6q21 forkhead transcription t-AML (Hillion, 1997) (FKHRL1) factor AF9 9p22 transcriptional activator AML (Nakamura, 1993) (MLLT3) ALL (Langer, 2003) t-AML (Whitmarsh, 2003) AF10 10p12 leucine zipper protein t-AML (Megonigal, 2000) 2 α-helical domains MLL 11q23 de novo AML t-AML AF17 17q21 ENL 19p13.3 transcriptional activator ALL (Tkachuk, 1992) (MLLT1, AML (Yamamoto, 1993) LTG19) T-cell ALL (Iida, 1993) t-AML (Chervinsky, 1995) (Rubnitz, 1996) (Moorman, 1998) (LoNigro, 2002) AFX Xq13 forkhead transcription (Corral, 1993) factor Proteins involved in transcripttional regulation CBP 16p13 transcriptional adaptor/ MDS (Taki, 1997) co-activator; histone (RAEB-T) (Satake, 1997) acetyl transferase t-MDS (Sobulo, 1997) (RAEB-T) (Rowley, 1997) t-CMML (Hayashi, 2000) t-AML (Sugita, 2000) t-ALL (B- lineage) T-cell ALL ELL (MEN) 19p13.1 RNA polymerase II AML (Thirman, 1994) elongation t-AML (Mitani, 1995) factor (Rubnitz, 1996) (Shilatifard, 1996) (Johnstone, 2001) (Maki, 1999) (Moorman, 1998) (LoNigro, 2002) (Megonigal, 2000) p300 22q13 transcriptional co- activator Nuclear proteins of unknown function AF3p21 3p21 SH3 domain, bipartite t-AML (Sano, 2001) nuclear localization (Hayakawa, 2001) signal, proline rich domain, homo- oligomerization domain LCX (TET1) 10q22 CXXC domain, nuclear de novo (Ono, 2002) localization signals, AML (Lorsbach, 2003) coiled-coil motif AF15q14 (Hayette, 2000) Cytoplasmic proteins AF1p 1p32 EGFR pathway tyrosine AUL (M0) (Bernard, 1994) (eps15) kinase substrate CMML (Wong, 1994) ALL (Rogaia, 1997) AF1q 1q21 mRNA destabilizing AML (Tse, 1995) consensus (So, 2000) sequences (Busson-Le Coniat, cytokine-like features 1999) GMPS 3q24 amidotransferease t-AML (Pegram, 2000) LPP 3q28 GRAF 5q31 AF6 6q27 Ras binding protein AML (Prasad, 1993) t-AML (Taki, 1996) T-cell ALL (Martineau, 1998) B-lineage (Joh, 1997) ALL (Mitterbauer, 2000) (Akao, 2000) CDK6 7q21 kinase FBP17 9q34 ABI-1 10p11.2 CBL 11q23.3 proline-rich domain, de novo (Fu, 2003) ubiquitin- AML associated domain, leucine zipper domain, zinc finger domain, tyrosine kinase binding domain, linker region, ring finger domain MPFYVE 15q14 FYVE domain de novo (Chinwalla, 2003) phosphotidyl-inositol-3 AML phosphate (PtdIns(3)P binding protein GAS7 17p13 (Megonigal, 2000) LASP1 17q21 LIM and SH3 domains AML (Strehl, 2003) MSF 17q25 septin (McIlhatton, 2001) GTP-binding domain lacks coiled-coil domain in C- terminus EEN 19p13 Src homology 3 (SH3) AML (So, 1997) protein hCDCrel 22q11 (Megonigal, 1998) SEPTIN6 Xq23 (Slater, 2002) Cell membrane proteins CALM 11q14-q21 clathrin assembly AML (Wechsler, 2003) protein LARG 11q23 GPHN 14q23.3 MYO1F 19p13.2-19p13.3 head domain with AML (LoNigro, 2002) conserved ATP- and actin-binding sites, neck domain with IQ motif, tail domain Golgi/Endo plasmic reticulum ALKALINE 19p13 (LoNigro, 2002) CERAMIDASE Ribosomal protein RPS3 11q13.3-11q13.5 AML (LoNigro, 2003) MIFL U.S. Provisional Application No. 60/599,385 MAM L2 11q21 Mastermind-Like MLL exon 7 to position transcriptional 1799 of MAML2 coactivator for mammalian Notch receptors (GenBank No. AY040322)

While certain of the preferred embodiments of the present invention have been described and specifically exemplified above, it is not intended that the invention be limited to such embodiments. Various modifications may be made thereto without departing from the scope and spirit of the present invention, as set forth in the following claims.

REFERENCES

  • 1. Fortune, J. M. & Osheroff, N. (2000) Prog Nucleic Acid Res and Molecular Biology 64, 221-53.
  • 2. Felix, C. A. (2001) Med Pediatr Oncol 36, 525-535.
  • 3. Rowley, J. D. & Olney, H. J. (2002) Genes Chromosomes Cancer 33, 331-45.
  • 4. Knudson, A. G. (1992) Sem Cancer Biol 3, 99-106.
  • 5. Woessner, R. D., Mattern, M. R., Mirabelli, C. K., Johnson, R. K. & F. H., D. (1991) Cell Growth Differ 2, 209-14.
  • 6. Isaacs, R. J., Davies, S. L., Sandri, M. I., Redwood, C., Wells, N. J. & Hickson, I. D. (1998) Biochim Biophys Acta 1400, 121-37.
  • 7. Megonigal, M. D., Cheung, N. K., Rappaport, E. F., Nowell, P. C., Wilson, R. B., Jones, D. H., Addya, K., Leonard, D. G., Kushner, B. H., Williams, T. M., Lange, B. J. & Felix, C. A. (2000) Proc Natl Acad Sci USA 97, 2814-9.
  • 8. Raffini, L. J., Slater, D. J., Rappaport, E. F., Lo Nigro, L., Cheung, N.-K. V., Biegel, J. A., Nowell, P. C., Lange, B. J. & Felix, C. A. (2002) Proc Natl Acad Sci USA 99, 4568-4573.
  • 9. Kushner, B. H., Cheung, N. K., Kramer, K., Heller, G. & Jhanwar, S. C. (1998) J Clin Oncol 16, 3880-3889.
  • 10. Lovett, B. D., Lo Nigro, L., Rappaport, E. F., Blair, I. A., Osheroff, N., Zheng, N., Megonigal, M. D., Williams, W. R., Nowell, P. C. & Felix, C. A. (2001) Proc Natl Acad Sci USA 98, 9802-9807.
  • 11. Whitmarsh, R., Saginario, C., Zhuo, Y., Hilgenfeld, E., Rappaport, E. F., Megonigal, M. D., Carroll, M., Liu, M., Osheroff, N., Cheung, N.-K. V., Slater, D. J., Ried, T., Knutsen, T., Blair, I. A. & Felix, C. A. (2003) Oncogene 22, 8448-8459.
  • 12. Bromberg, K. D. & Osheroff, N. (2001) Biochem 40, 8410-8418.
  • 13. Ueda, T., Tsuji, K., Yoshino, H., Ebihara, Y., Yagasaki, H., Hisakawa, H., Mitsui, T., Manabe, A., Tanaka, R., Kimio, K., Ito, M., Yasukawa, K. & Nakahata, T. (2000) J Clin Invest 105, 1013-1021.
  • 14. Stong, R., Korsmeyer, S., Parkin, J., Arthur, D. & Kersey, J. (1985) Blood 65, 21-31.
  • 15. Pocock, C. F., Malone, M., Booth, M., Evans, M., Morgan, G., Greil, J. & Cotter, F. E. (1995) Br J Haematol 90, 855-67.
  • 16. Lozzio, B. B., Lozzio, C. B., Bamberger, E. G. & Feliu, A. S. (1981) Proc Soc Exp Biol Med 166, 546-50.
  • 17. Livak, K. J. & Schmittgen, T. D. (2001) Methods 25, 402-408.
  • 18. Harlow, E. & Lane, D. (1999) Using Antibodies: A Laboratory Manual (Cold Spring Harbor Laboratory.
  • 19. Cheung, V. G. & Nelson, S. F. (1996) Proc Natl Acad Sci USA 93, 14676-14679.
  • 20. Gelmini, S., Orlando, C., Sestini, R., Vona, G., Pinzani, P., Ruocco, L. & Pazzagli, M. (1997) Clin Chem 43, 752-8.
  • 21. Heid, C. A., Stevens, J., Livak, K. J. & Williams, P. M. (1996) Genome Res 6, 986-94.
  • 22. Holland, P. M., Abramson, R. D., Watson, R. & Gelfand, D. H. (1991) Proc Natl Acad Sci USA 88, 7276-80.
  • 23. Pongers-Willemse, M. J., Verhagen, O. J., Tibbe, G. J., Wijkhuijs, A. J., de Haas, V., Roovers, E., van der Schoot, C. E. & van Dongen, J. J. (1998) Leukemia 12, 2006-14.
  • 24. Lovett, B. D., Strumberg, D., Blair, I. A., Pang, S., Burden, D. A., Megonigal, M. D., Rappaport, E. F., Rebbeck, T. R., Osheroff, N., Pommier, Y. G. & Felix, C. A. (2001) Biochemistry 40, 1159-70.
  • 25. Blanco, J. G., Dervieux, T., Edick, M. J., Mehta, P. K., Rubnitz, J. E., Shurtleff, S. A., Raimondi, S. C., Behm, F. G., Pui, C.-H. & Relling, M. V. (2001) Proc Natl Acad Sci USA 98, 10338-43.
  • 26. Ford, A. M., Ridge, S. A., Cabrera, M. E., Mahmoud, H., Steel, C. M., Chan, L. C. & Greaves, M. (1993) Nature 363, 358-360.
  • 27. Gale, K., Ford, A., Repp, R., Borkhardt, A., Keller, C., Eden, O. & Greaves, M. (1997) Proc Natl Acad Sci USA 94, 13950-13954.
  • 28. Megonigal, M. D., Rappaport, E. F., Jones, D. H., Williams, T. M., Lovett, B. D., Kelly, K. M., Lerou, P. H., Moulton, T., Budarf, M. L. & Felix, C. A. (1998) Proc Natl Acad Sci USA 95, 6413-6418.
  • 29. Maia, A. T., Koechling, J., Corbett, R., Metzler, M., Wiemels, J. L. & Greaves, M. (2004) Genes, Chromosomes & Cancer 39, 335-340.
  • 30. Krishnan, A., Bhatia, S., Slovak, M. L., Arber, D. A., Niland, J. C., Nademanee, A., Fung, H., Bhatia, R., Kashyap, A., Molina, A., O'Donnell, M. R., Parker, P. A., Sniecinski, I. & Snyder, D. S. (2000) Blood 95, 1588-1593.
  • 31. Liang, F., Han, M., Romanienko, P. J. & Jasin, M. (1998) Proc Natl Acad Sci USA 95, 5172-5177.
  • 32. Capranico, G., Jaxel, C., Roberge, M., Kohn, K. W. & Pommier, Y. (1990) Nucleic Acids Res 18, 4553-4559.
  • 33. Cozzio, A., Passegue, E., Ayton, P. M., Karsunky, H., Cleary, M. L. & Weissman, I. L. (2003) Genes Dev 17, 3029-35.
  • 34. Boyd, K. E., Wells, J., Gutman, J., Bartley, S. M. & Farnham, P. J. (1998) Proc Natl Acad Sci USA 95, 13887-92.
  • 35. Langer, T., Metzler, M., Reinhardt, D., Viehmann, S., Borkhardt, A., Reichel, M., Stanulla, M., Schrappe, M., Creutzig, U., Ritter, J., Leis, T., Jacobs, U., Harbott, J., Beck, J. D., Rascher, W. & Repp, R. (2003) Genes Chromosomes Cancer 36, 393-401.
  • 36. Felix, C. A., Hosler, M. R., Slater, D. J., et al. (1998) J. Pediatr. Hematol/Oncol 20:299-308.
  • 37. Felix, C. A., Jones, D. H. (1998) Leukemia 12:976-981.
  • 38. Slater, D. J., Hilgenfeld, E., Rappaport, E. F. et al. (2002) Oncogene 21:4706-4714.
  • 39. Megonigal, M. D. Rappaport E. F., Wilson, R. B., et al. (2000) PNAS 97:9597-9602.
  • 40. Pegram, L. D., Megonigal, M. D., Lange, B. J., et al. (2000) Blood 96:4360-4362.
  • 41. Felix, C. A., Kim, C. S., Megonigal, M. D., et al. (1997) Blood 90:4679-4686.
  • 42. Lo Nigro, L., Rappaport E., Slater, D., et al. (1999) Proc ASCO 18:565a.
  • 43. Lo Nigro, L., Slater, D. J., Rappaport, E. F., et al. (2002) Blood 100(Suppl 1):531a.
  • 44. Dudoit, S. and Yang, Y. H. (2002) Bioconductor R packages for exploratory analysis and normalization of cDNA microarray data. In: The analysis of Gene Expression Data: Methods and Software; Parmigiani, et al. (Ed) pp. 73-101; New York:Springer.
  • 45. Dudoit, S., Speed, T. P., Callow, M. J. (2002) Statistica Sinica 12:111-139.
  • 46. Huret J L, et al. (2001) Leukemia, 15(6):987-9.
  • 47. Nakamura T, et al. (1993) Proc Natl Acad Sci USA. 15; 90(10):4631-5.
  • 48. Taki T, et al. (1996) Oncogene. 13(10):2121-30.
  • 49. Hillion J, et al. (1997) Blood. 90(9):3714-9.
  • 50. Moorman A V, et al. (1998) Leukemia. 12(5):805-10.
  • 51. Rubnitz J E, et al. (1996) Blood. 87(11):4804-8.
  • 52. Chervinsky D S, et al. (1995) Genes Chromosomes Cancer. 14(1):76-84.
  • 53. Yamamoto K, et al. (1993) Oncogene. 8(10):2617-25.
  • 54. Iida S, et al. (1993) Oncogene. 8(11):3085-92.
  • 55. Tkachuk D C, et al. (1992) Cell. 71(4):691-700.
  • 56. Corral J, et al. (1993) Proc Natl Acad Sci USA. 90(18):8538-42.
  • 57. Taki T, et al. (1997) Blood. 89(11):3945-50.
  • 58. Satake N, et al. (1997) Genes Chromosomes Cancer. 20(1):60-3.
  • 59. Sobulo O M, et al. (1997) Proc Natl Acad Sci USA. 94(16):8732-7.
  • 60. Rowley J D, et al. (1997) Blood. 90(2):535-41.
  • 61. Hayashi Y, et al. (2000) Cancer Res. 60(4):1139-45.
  • 62. Sugita K, et al. (2000) Genes Chromosomes Cancer. 27(3):264-9.
  • 63. Thirman M J, et al. (1994) Proc Natl Acad Sci USA. 91(25):12110-4.
  • 64. Shilatifard A, et al. (1996) Science. 271(5257):1873-6.
  • 65. Maki K, et al. (1999) Blood. 93(10):3216-24.
  • 66. Mitani K, et al. (1995) Blood. 85(8):2017-24.
  • 67. Sano K. (2001) Leuk Lymphoma. 42(4):595-602.
  • 68. Hayakawa A, et al. (2001) Genes Chromosomes Cancer. 30(4):364-74.
  • 69. Ono R, et al. (2002) Cancer Res. 62(14):4075-80.
  • 70. Lorsbach R B, et al. (2003) Leukemia. 17(3):637-41.
  • 71. Hayette S, et al. (2000) Oncogene. 19(38):4446-50.
  • 72. Bernard O A, et al. (1994) Oncogene. 9(4):1039-45.
  • 73. Wong W T, et al. (1994) Oncogene. 9(6):1591-7.
  • 74. Rogaia D, et al. (1997) Cancer Res. 57(5):799-802.
  • 75. Tse W, et al. (1995) Blood. 85(3):650-6.
  • 76. So C W, et al. (2000) Leukemia. 14(4):594-601.
  • 77. Busson-Le Coniat M, et al. (1999) Leukemia. 13(2):302-6.
  • 78. Pegram L D, et al. (2000) Blood. 96(13):4360-2.
  • 79. Prasad R, et al. (1993) Cancer Res. 53(23):5624-8.
  • 80. Taki T, et al. (1996) Oncogene. 13(10):2121-30.
  • 81. Martineau M, et al. (1998) Leukemia. 12(5):788-91.
  • 82. Joh T, et al. (1997) Oncogene. 15(14):1681-7.
  • 83. Mitterbauer G, et al. (2000) Br J Haematol. 109(3):622-8.
  • 84. Akao Y, et al. (2000) Genes Chromosomes Cancer. 27(4):412-7.
  • 85. Fu J F, et al. (2003) Genes Chromosomes Cancer. 37(2):214-9.
  • 86. Chinwalla V, et al. (2003) Oncogene. 22(9):1400-10.
  • 87. Strehl S, et al. (2003) Oncogene. 22(1):157-60.
  • 88. McIlhatton M A, et al. (2001) Oncogene. 20(41):5930-9.
  • 89. So C W, et al. (1997) Proc Natl Acad Sci USA. 94(6):2563-8.
  • 90. Wechsler D S, et al. (2003) Genes Chromosomes Cancer. 36(1):26-36.
  • 91. Lo Nigro L, et al. (2003) Blood. 102:184-185b.

Claims

1. A method for identifying sequences present in DNA topoisomerase H-DNA complexes in cells, comprising:

a) providing cells suspected of containing DNA topoisomerase H-DNA complexes;
b) isolating DNA topoisomerase H-DNA complexes from the cell;
c) amplifying the DNA present in said isolated DNA topoisomerase H-DNA complexes via polymerase chain reaction; and
d) identifying the sequences present in said amplified DNA, thereby identifying the sequences present in said DNA topoisomerase II-DNA complexes.

2. The method of claim 1, wherein the DNA in said DNA topoisomerase 11-DNA complexes is genomic DNA.

3. The method of claim 1, wherein the identification of the sequences in step d) comprises further amplifying the amplified DNA from step c) with gene specific primers.

4. The method of claim 1, wherein the identification of the sequences in step d) comprises further amplifying the amplified DNA from step c) by real-time PCR.

5. The method of claim 1, wherein the identification of the sequences in step d) comprises hybridizing the amplified DNA from step c) with a microarray.

6. The method of claim 1, wherein said cells are CD34+ cells.

7. The method of claim 1, wherein said amplified DNA of step c) comprises sequences from the MLL gene.

8. The method of claim 1, wherein said cells are exposed to an agent suspected of modulating formation of topoisomerase cleavage complexes.

9. The method of claim 5, wherein said microarray comprises MLL bcr oligonucleotide sequences.

10. The method of claim 9, wherein said MLL bcr oligonucleotide sequences hybridize to non-repetitive MLL bcr sequences.

11. The method of claim 9, wherein said microarray further comprises oligonucleotide sequences from the Alu region between nucleotide positions 663-1779 in the MLL bcr.

12. The method of claim 9, wherein said microarray further comprises control sequences which are not involved in MLL translocations.

13. The method of claim 13, wherein said control sequences are selected from the group consisting of MLL exon 25, MLL exon 3, GAPDH, c-myc, and bacterial gene sequences.

14. The method of claim 9, wherein said microarray further comprises oligonucleotide sequences from MLL partner genes.

15. The method of claim 14, wherein said MLL partner genes are selected from the group consisting of LAF-4, AF4 (MLLT2, FEL), AF5α, AF5q31, AF6q21 (FKHRL1), AF9 (MLLT3), AF10, MLL, AF17, ENL (MLLT1, LTG19), AFX, CBP, ELL (MEN), p300, AF3p21, LCX (TET1), AF15q14, AF1p (eps1S), AF1q, GMPS, LPP, GRAF, AF6, CDK6, FBP17, ABI-1, CBL, MPFYVE, GAS7, LASP1, MSF, EEN, hCDCrel, SEPTIN6, CALM, LARG, GPHN, MYO1F, Alkaline Ceramidase, RPS3, and MIFL.

16. The method of claim 5, wherein said microarray comprises oligonucleotide sequences from MLL partner genes.

17. The method of claim 16, wherein said MLL partner genes are selected from the group consisting of LAF-4, AF4 (MLLT2, FEL), AF5α, AF5q31, AF6q21 (FKHRL1), AF9 (MLLT3), AF10, MLL, AF17, ENL (MLLT1, LTG19), AFX, CBP, ELL (MEN), p300, AF3p21, LCX (TET1), AF15q14, AF1p (eps15), AF1q, GMPS, LPP, GRAF, AF6, CDK6, FBP17, ABI-1, CBL, MPFYVE, GAS7, LASP1, MSF, EEN, hCDCrel, SEPTIN6, CALM, LARG, GPHN, MYO1F, Alkaline Ceramidase, RPS3, and MIFL.

18. The method of claim 9, wherein the microarray comprises oligonucleotide sequences comprising SEQ ID NOs 1-162.

19. The method of claim 9, wherein the microarray comprises oligonucleotide sequences comprising SEQ ID NOs 163-246.

20. The method of claim 1, wherein said isolating of DNA topoisomerase II-DNA complexes of step b) comprises lysing said cells and immunoprecipitating said DNA topoisomerase 11-DNA complexes.

21. A microarray comprising MLL bcr oligonucleotide sequences.

22. The microarray of claim 21, wherein said MLL bcr oligonucleotide sequences hybridize to non-repetitive MLL bcr sequences.

23. The microarray of claim 21, wherein said microarray further comprises oligonucleotide sequences from the Alu region between nucleotide positions 663-1779 in the MLL bcr.

24. The microarray of claim 21, wherein said microarray further comprises control sequences which are not involved in MLL translocations.

25. The microarray of claim 24, wherein said control sequences are selected from the group consisting of MLL exon 25, MLL exon 3, GAPDH, c-myc, and bacterial gene sequences.

26. The microarray of claim 21, wherein said microarray further comprises oligonucleotide sequences from MLL partner genes.

27. The microarray of claim 26, wherein said MLL partner genes are selected from the group consisting of LAF-4, AF4 (MLLT2, FEL), AF5α, AF5q31, AF6q21 (FKHRL1), AF9 (MLLT3), AF10, MLL, AF17, ENL (MLLT1, LTG19), AFX, CBP, ELL (MEN), p300, AF3p21, LCX (TET1), AF15q14, AF1p (eps1S), AF1q, GMPS, LPP, GRAF, AF6, CDK6, FBP17, ABI-1, CBL, MPFYVE, GAS7, LASP1, MSF, EEN, hCDCrel, SEPTIN6, CALM, LARG, GPHN, MYO1F, Alkaline Ceramidase, RPS3, and MIFL.

28. A microarray comprising oligonucleotide sequences from MLL partner genes.

29. The microarray of claim 28, wherein said MLL partner genes are selected from the group consisting of LAF-4, AF4 (MLLT2, FEL), AF5α, AF5q31, AF6q21 (FKHRL1), AF9 (MLLT3), AF10, MLL, AF17, ENL (MLLT1, LTG19), AFX, CBP, ELL (MEN), p300, AF3p21, LCX (TET1), AF15q14, AF1p (eps15), AF1q, GMPS, LPP, GRAF, AF6, CDK6, FBP17, ABI-1, CBL, MPFYVE, GAS7, LASP1, MSF, EEN, hCDCrel, SEPTIN6, CALM, LARG, GPHN, MYO1F, Alkaline Ceramidase, RPS3, and MIFL.

30. The microarray of claim 21, wherein the microarray comprises oligonucleotide sequences comprising SEQ ID NOs 1-162.

31. The microarray of claim 21, wherein the microarray comprises oligonucleotide sequences comprising SEQ ID NOs 163-246.

32. A kit for performing the method of claim 1.

33. The kit of claim 32, comprising:

a) an agent and a buffer for lysing cells;
b) a solid support and a buffer for isolating DNA-DNA topoisomerase II complexes;
c) at least one primer and a buffer for PCR amplification of isolated DNA by whole genome amplification;
d) at least one first detectable label for incorporation into products of whole genome amplification;
e) calibrated reference standard DNA comprising a second detectable label;
f) a microarray comprising oligonucleotide sequences from the MLL bcr; and
g) instruction material.
Patent History
Publication number: 20060063184
Type: Application
Filed: Sep 9, 2005
Publication Date: Mar 23, 2006
Inventors: Carolyn Felix (Ardmore, PA), Donald Baldwin (Newtown Square, PA)
Application Number: 11/222,626
Classifications
Current U.S. Class: 435/6.000; 435/91.200
International Classification: C12Q 1/68 (20060101); C12P 19/34 (20060101);